Clinical Hematology and Fundamentals of Hemostasis - Harmening
Clinical Hematology and Fundamentals of Hemostasis - Harmening
Harmening
ma @itercesi
eloeatalKe)(e
meberetslaalseleclicwey
-. Hemostasis
FIFTH EDITION
HEMATOLOGIC VALUES*
Reference Range
Determination Conventional SI
Hematology
“Complete” blood count
(CBC): Automated
Methodology Coulter
Model STKS
WBC 4.8-10.8 X 103/uL 4.8-10.8 X 10°/L
RBC Male: 4.7-6.1 * 10°/uL 4.7-6.1 * 10?/L
Female: 4.2-5.4 < 10°/uL 4.2-5.4 * 10?/L
Hemoglobin (Hgb) Male: 14-18 g/dL 140-180 g/L Direct measurement:
Female: 12-16 g/dL 120-160 g/L Cyanide pigment
Infant: 14-22 g/dL 140-220 g/L
Hematocrit (Hct) Male: 42%-52% 0.42-0.52 L/L Calculation:
Female: 37%-47% 0.37-0.47 L/L RBC X MCV/10
Mean corpuscular 80-100 fL 80-100 fL Derived from RBC
volume (MCV) histogram: Measured
volume of 1 RBC
Calculation: Hct/RBC x 10
Mean corpuscular 27-31 pg 27-31 pg Calculation:
hemoglobin (MCH) Hgb/RBC X 10
Mean corpuscular 32%-36% 32.0-36.0 g/dL Calculation:
hemoglobin concentration Hegb/Hct 100
(MCHC)
Red cell distribution 11.5%-14.5% 11.5-14.5 g/L Derived from RBC
width (RDW) histogram:
CV of RBC distribution =
(SD X 100)
Mean
Platelets (PLT) 150,000-400,000/uL 150-400 * 10°/L Direct count: Cells 2-20 fL
Mean platelet volume (MPV) 7.4-10.4 fL 7.4-10.4 fL Derived from PLT histogram
WBC Differential
Lymphocytes 20%-44%
Monocytes 2%-9%
Neutrophils 50%-10%
Bands 2%-6%
Eosinophils O%4%
Basophils 0%—2%
Absolute Counts
Lymphocytes 1.2-3.4 X 10%/uL 1.2-3.4 X 10°/L Lymphs %/100 X WBC
Monocytes 0.11-0.59 X 103/uL 0.11-0.59 X 10°/L Mono %/100 * WBC
Neutrophils 1.4-6.5 X 10% 1.4-6.5 X 10°/L Neutrophils %/100 * WBC
Bands 0-0.7 X 103/uL 0-0.7 X 10°/L Bands %/100 X WBC
Eosinophils 0-0.5 X 103/uL 0-0.5 X 10°/L Eos %/100 * WBC
Basophils 0-0.2 X 10°/uL 0-0.2 10°/L Baso %/100 X WBC
Manual Methodology: CBC
Hematocrit Newborn: 53%-65% 0.53-0.65 L/L Centrifugal microhemato-
Infant/child: 30%43% 0.30-0.43 L/L crit method
Adult male: 42%-—52% 0.42-0.52 L/L
Adult female: 37%47% 0.37-0.47 L/L
Hemoglobin Newborn: 17-23 g/dL 170-230 g/L Cyanmethemeglobin
3 mo: 9-14 g/dL 90-140 g/L method
10 yr: 12-14.5 g/dL 120-145 g/L
Adult male: 14-17 g/dL 140-170 g/L
Adult female: 12.5-15 g/dL 125-150 g/L
*Please note: Normal values vary by institution, patient population, and testing methodology.
Reference Range
RBC Enzymes
G6PD 3.4-8.0 U/g Hgb 0.22-0.52 MU/mol Hgb Bishop, modified method
Pyruvate kinase 13-17 U/g Hgb 0.84-1.1 MU/mol Hgb ICSH method
Iron Studies
Serum iron Adult male: 65-170 ug/dL 11.63-30.43 umol/L Colorimetric
Adult female: 50-170 pg/dL 8.95-30.43 pmol/L
TIBC 250-450 g/dL 44.75-80.55 umol/L Colorimetric
Transferrin saturation Male: 20%-50%
Female: 15%—50%
Serum TfR (transferrin 1.5-2.75 mg/L
receptor)
Ferritin (serum) 12-30 ng/mL 12 ug/L-30 ug/L
Iron deficiency 0-12 ng/mL 12 ug/L
Borderline 13-20 ng/mL 13-20 ug/L
Iron excess >400 ng/L >400 pg/L
Zinc protoporphyrin 16-65 pg/dL 0.28 pmol/L-1.17 mol/L
(free erythrocyte
protoporphyrin)
Folic Acid
Normal = s.onp/18 >7.3 nmol/L RIA
Borderline 2.5—3.2 ng/mL 5.75-7.39 nmol/L
Red cell folate 150-450 mg/mL 340-1020 nmol/L Collect in EDTA
httos://archive.org/details/clinicalhematolo0000unse_d5f7
Clinical
Hematology and
Fundamentals of
Hemostasis
FIFTH EDITION
~
FIFTH EDITION
Copyright © 2009 by F. A. Davis Company. All rights reserved. This product is protected by copyright. No part
of it may be reproduced, stored in a retrieval system, or transmitted in any form or by any means, electronic,
mechanical, photocopying, recording, or otherwise, without written permission from the publisher.
As new scientific information becomes available through basic and clinical research, recommended treatments
and drug therapies undergo changes. The author(s) and publisher have done everything possible to make
this book accurate, up to date, and in accord with accepted standards at the time of publication. The author(s),
editors, and publisher are not responsible for errors or omissions or for consequences from application of the
book, and make no warranty, expressed or implied, in regard to the contents of the book. Any practice described
in this book should be applied by the reader in accordance with professional standards of care used in regard
to the unique circumstances that may apply in each situation. The reader is advised always to check product
information (package inserts) for changes and new information regarding dose and contraindications before
administering any drug. Caution is especially urged when using new or infrequently ordered drugs.
Clinical hematology’ and fundamentals of hemostasis / [edited by] Denise M. Harmening. —Sth ed.
p.;cm.
Includes bibliographical references and index.
ISBN-13: 978-0-8036-1732-2
ISBN-10: 0-8036-1732-1
1. Hematology. 2. Blood—Diseases. 3. Homeostasis. I. Harmening, Denise.
[DNLM: |. Hematologic Diseases. 2. Blood Cells—physiology. 3. Hemostasis. WH 100 C6413 2009]
RB145.C536 2009
616.1'5—de22 2008048004
Authorization to photocopy items for internal or personal use, or the internal or personal use of specific clients,
is granted by F. A. Davis Company for users registered with the Copyright Clearance Center (CCC) Transactional
Reporting Service, provided that the fee of $.25 per copy is paid directly to CCC, 222 Rosewood Drive, Danvers,
MA 01923. For those organizations that have been granted a photocopy license by CCC, a separate system of
payment has been arranged. The fee code for users of the Transactional Reporting Service is: 0-8036-1732/09
[ete O:20)
To all students—full-time, part-time, past, present,
and future—who have touched and will continue to
touch the lives of so many educators... .
The fifth edition of this text continues to set the standard for guide with student exercises; and all the color illustrations
teaching and learning in clinical hematology and fundamen- and figures by chapter to use in PowerPoint lectures. A stu-
tals of hemostasis. Thorough, yet concise, a total of 36 chap- dent website is also available for this edition which includes
ters serve as a combination of text, laboratory procedure Picture Match, Flash Cards, and Case Histories.
manual, and atlas of cell morphology. This textbook is unique This new edition, like the first, second, third, and fourth,
in its five-part format, featuring an introduction to clinical is a culmination of the dedicated efforts of many prominent
hematology and sections on anemias, white blood cell disor- laboratory professionals. More than fifty-five contributors
ders, hemostasis and thrombosis, and laboratory methods. committed themselves to this project by donating their time
Outlines, educational objectives, case histories, summary and expertise out of concern for a common goal: improving
charts, and study guide questions support each chapter. patient care by providing a high-quality, practical, and usable
The laboratory methods section includes two new chap- textbook. To all of these contributors, thank you and congrat-
ters, “Quality Management, Quality Assurance, and Quality ulations on meeting this goal. A very special thank you and
Control” and “Body Fluid Examination,” in addition to chap- acknowledgment must also be given to my husband, Jesse
ters on hematology methods, automated differential analysis, Javens, who always supports my writing and provides en-
coagulation methods, flow cytometry, molecular diagnostic couragement which always motivates me. I would also like to
techniques in hematopathology, and cytochemistry. Numer- thank the following students who helped with this edition:
ous illustrations, including 375 color photographs, 600 line Ernest Frank, Laura Little, Nathan Marchiano, Abiola
drawings, and 355 tables throughout the book, aid in the Oderinlo, and especially Laura Simpson, who spent many
process of learning. The text has retained the popular listing hours assisting me.
of normal hematologic values on the inside covers for a quick Finally, my sincere appreciation is also extended to
reference. In addition, an invaluable glossary provides easy the educators, clinicians, and industry representatives
access to definitions of medical terms unique to hematology who gave thorough and thoughtful reviews of selected
and hemostasis. Summary charts have been added at the end manuscripts.
of each chapter to identify for students the most important In summary, this book has been designed to inspire an
information to know for clinical rotation. ACD-Rom is avail- unquenchable thirst for knowledge in every medical technol-
able to educators who adopt the text. This CD includes an ogist, hematologist, clinician, and practitioner whose knowl-
interactive test-generating question bank containing over edge, skills, and ongoing education provide the public with
2000 questions with assigned taxonomy levels; an instructor’s excellent health care.
D.M. Harmening, PhD, MT(ASCP), CLS(NCA)
vil
— = 7 “@
~~
:
es LVS BI ivy
ts. 7 Pi =
: 7 f Nii (Ee F
= = = ms tet mre _
-
——- , on ive oa7
i ! am a 7 ¥
= = ? ; iin
a = a a a)
SS aay ‘
' | Agius ell
= —
SS
=
ny
SS
" ' i
1 Ja2
=
Cte. o¥
oe Shien,
- - a
a 7
: ~ Be
+3) -
— ~
: ASS aM tape is
ener ew aes
«
—
Contributors
Marvin D. Bearden, MA, CLS(NCA), MT(ASCP) Jan Fox, BSC, ART MLT
Assistant Professor, Education Coordinator Coordinator
Clinical Laboratory Sciences Medical Laboratory Sciences
The University of Texas Health Science Center St. Lawrence College
at San Antonio Kingston, Ontario, Canada
San Antonio, Texas
Mildred K. Fuller, PHD, MT(ASCP)
Bernadette Bekken, CLS(NCA), MT(ASCP)BB Chair and Professor
Program Director * Allied Health
Clinical Laboratory Sciences Norfolk State University
Augusta Medical Center Norfolk, Virginia
Fishersville, Virginia
Andrea G. Gordon, MED, MT(ASCP)SH
Rose V. Brown, MED, CLS, MT(ASCP) Professor
Program Director Health and Human Services
Clinical Laboratory Sciences New Hampshire Community Technical College
Penrose-St. Francis Hospital Claremont, New Hampshire
Colorado Springs, Colorado
Karen G. Gordon, BS, CLS, MLT(ASCP)SLS
Donna D. Castellone, MS, MT(ASCP)SH Program Manager, Assistant Professor
Clinical Project Manager Medical Laboratory Technology
Hemostasis/Hematology Northern Virginia Community College
Siemens Healthcare Diagnostics Springfield, Virginia
Tarrytown, New York
Michele G. Harms, MS, MT(ASCP)
Faye E. Coleman, MS, CLS, MT(ASCP) Program Director
Program Director, Associate Professor Medical Technology
Medical Laboratory and Radiation Sciences Women’s Christian Association Hospital
Old Dominion University Jamestown, New York
Norfolk, Virginia
Sheryl B. Herring, BS, MSA, MT(ASCP)
Linda Collins, MS, MT(ASCP) Program Coordinator
Instructor Clinical Laboratory Technology
Medical Laboratory Technology Louisiana State University
Delaware Technical & Community College Alexandria, Virginia
Dover, Delaware
Virginia C. Hughes, Ms, MT(ASCP)SBB, CLS(NCA)I
Jane B. Finley, BS, MT(ASCP) Assistant Professor
Clinical Assistant Professor Division of Clinical Laboratory Sciences
Clinical Laboratory Sciences Auburn University at Montgomery
The University of Texas Medical Branch Montgomery, Alabama
Galveston, Texas Xiil
X1V Reviewers
Captor 13
Chantal Ricaud Harrison, MD
The Red Blood Cell: Structure and Function 64
Denise M. Harmening, PHD, MT(ASCP), CLS(NCA)
Hemolytic Anemias: Extracorpuscular
Chapter 4
Defects 252
Anemia: Diagnosis and Clinical Denise M. Harmening, PHD, MT(ASCP), CLS(NCA)
Considerations 82 Louann W. Lawrence, DRPH, MT(ASCP)SH, CLSPH(NCA)
Armand B. Glassman, MD Ralph Green, BAPPSCI(MLS), FAIMLS
Chapter 5 Carl R. Schaub, MD
XV
XVI Contents
Chapter 18
Chronic Myeloproliferative Disorders II: PART 5
Polycythemia Vera, Essential Thrombocythemia, LABORATORY METHODS 697
and Idiopathic Myelofibrosis 3585 Chapter 29
Kathrina Chua, MD Quality Management, Quality Assurance,
Meyer R. Heyman, MD and Quality Control 697
Chapter 19 Kim A. Przekop, BS, MT(ASCP)
Myelodysplastic Syndromes 412 Chapter 30
Giovanni D’Angelo, FCMLS Body Fluid Examination: The Qualitative,
Luigina Mollica, MD, FRCP(C), PHD
Quantitative, and Morphologic Analysis
Josée Hébert, MD, FRCP(C)
Lambert Busque, MD, FRCP(C)
of Serous, Cerebrospinal, and Synovial
Fluids 720
Chapter 20 Sharon L. Schwartz, MS, MT(ASCP)SH, CLS(NCA)
Chronic Lymphocytic Leukemia and Related
Chapter 31
Lymphoproliferative Disorders 440
Laurel D. Holmer, MEb, MT(ASCP)SH Hematology Methods 759
Virginia C. Hughes, MS, MT(ASCP)SBB, CLS(NCA)I
Carlos E. Bueso-Ramos, MD, PHD
Chapter 21 Chapter 32
The Lymphomas 466 Principles of Automated Differential
Dan M. Hyder, MD Analysis 793
Denise M. Harmening, PHD, MT(ASCP), CLS(NCA)
Chapter 22
Angelica Black, USAF, BSc, MS, MT(ASCP)
Multiple Myeloma and Related Plasma Cell Naomi B. Culp, DA, MT(ASCP)SH
Disorders 500 Jolanta Kunicka, PHD
AshrafZ. Badros, MD Krista M. Curcio, MT(ASCP)
Chapter 25 Tan Giles, MD
Richard Kendall, FIBMS, PHD
Lipid (Lysosomal) Storage Diseases
Rick Gooch, MBA, MT(ASCP)
and Histiocytosis 525
Denise M. Harmening, PHD, MT(ASCP), CLS(NCA) Chapter 35
Catherine M. Spier, MD Coagulation Methods 849
Melissa Bethel, MT(ASCP)
Saas
PART 4 Chapter 34
HEMOSTASIS AND INTRODUCTION Applications of Flow Cytometry
TO THROMBOSIS 543 to Hematopathology 882
Donna M. Gandour, PHD
Chapter 24
Chapter 35
Introduction to Hemostasis 543
Denise M. Harmening, PHD, MT(ASCP), CES(NCA) Molecular Diagnostic Techniques
Claudia E. Escobar, MT(ASCP)SH in Hematopathology 911
David L. McGlasson, MS, CLS(NCA) Margaret L. Gulley, MD
Chapter 25 Chapter 36
Disorders of Primary Hemostasis: Quantitative Special Stains/Cytochemistry 929
and Qualitative Platelet Disorders and Vascular Mary Loring Perkins, MS, MT(ASCP)SH
Denise M. Harmening, PHD, MT(ASCP), CLS(NCA)
Disorders 577
Frank Xianfeng Zhao, MD, PHD
Darla K. Liles, MD
Charles L. Knupp, MD
Chapter
Morphology of Human
Blood and Marrow Cells
Hematopoiesis
Ann Bell, MS, SH(ASCP), CLSpH(NCA)
Denise M. Harmening, PhD, MT(ASCP), CLS(NCA)
Virginia C. Hughes, Ms, MT(ASCP)SBB, CLS(NCA)I
Proerythroblast) 9. List the proper cell maturation sequence of the erythroid series.
Basophilic Normoblast '
10. Recognize each cell in the erythrocytic series.
(Prorubricyte, Basophilic
Erythroblast) 11. Give two or more characteristics of each nucleated red cell in the erythrocytic series.
Polychromatophilic
Normoblast (Rubricyte, 12. List the proper cell sequence for myelopoiesis (granulocytopoiesis).
Erythrocyte (Red Blood Cell, 19. Distinguish between osteoblasts and osteoclasts.
Discocyte)
20. Name two differences between a megakaryocyte and an osteoclast.
Myelopoiesis
(Granulocytopoiesis)
Morphological Changes
Stages of Differentiation and
Maturation
Monopoiesis
Monoblasts and
Promonocytes
Monocytes and
Macrophages
Lymphopoiesis
Lymphoblasts and
Prolymphocytes
Lymphocytes
Plasmablasts and
Proplasmacytes
Plasmacytes (Plasma Cells)
Megakaryocytopoiesis
Bone-derived Cells
Osteoblasts
Osteoclasts
Molecular Hematology and
Advanced Concepts
Introduction to the Cell
Cycle
Specific Cell Line Ontogeny
Trends in Therapeutic
Manipulation of
Hematopoiesis
Recombinant Cytokines
Clinical Trials of
Recombinant Cytokines
CD Nomenclature
Clinical Applications of Cell
Surface Markers
Water (91.5%)
Plasma Proteins (7%)
Other Solutes (1.5%)
Absolute Values
(per mm*)
N. segmented 2400-7500
Eosinophil 0-450
Basophil 0-200
Lymphocyte 1200-3400
Monocyte 100-900
filament. Lobes of nuclei often touch each other or overlap, granules vary in size from 0.2 to 1.0 um.? They are coarse and
and it may be impossible to see the connecting filaments. In a unevenly distributed; vary in number, shape, and color; and
band neutrophil, there are two distinct margins with nuclear are less numerous than eosinophil granules (see Figs. 1—-2/
chromatin material visible between the margins. If the margin and 1—7). These granules have an affinity for blue or basic thi-
of a lobe can be traced as a definite and continuing line from azine dyes.' Basophil granules are algo water soluble. In cells
one side of the nucleus across the isthmus to the other side, then that are poorly fixed during staining, the center of the granule
it may be assumed that a filament is present even though it is may disappear or the entire granule may be washed away,
not visible. In attempting to differentiate between a segmented leaving a small colorless cytoplasmic area.
and a band neutrophil, identification should not be made on a Basophils show a diurnal variation similar to that of
single morphological characteristic but on combined features. eosinophils, increasing at night and decreasing in the morning.*
In case of doubt regarding a borderline cell, the questionable
cell should be placed into the mature category.’ LYMPHOCYTES
Lymphocytes are the second most numerous cells in the blood,
EOSINOPHILS comprising from 20% to 44% of the adult blood cells (see Table
Eosinophils are usually easily recognizable because of the 1-2). Most lymphocytes are small, varying from 7 to 10 um.
large, round, secondary, refractile granules that have an affin- There are also intermediate sizes and some large lymphocytes
ity for the acid eosin stain (see Figs. |-2D and 1-6). With (Fig. 1-8). Size is not a reliable basis for determining the age
Wright’s stain, normal eosinophilic granules become orange of metabolic activity of lymphocytes because their size varies
to reddish-orange. The granules are spherical, uniform in with the thickness of the smear. Lymphocytes tend to become
size, and evenly distributed. Because of the size and round- spherical and small in thick areas of the smear; in the thinnest
ness of the granules, eosinophils may be recognized in end of the smear, lymphocytes may spread out and appear
unstained moist preparations of blood on light microscopy large. Small lymphocytes are usually round with smooth
and via phase microscopy. The crystalloid core of the gran-
ule is composed mainly of major basic protein (MBP), which
binds to acid aniline dyes and which may help to explain the
staining qualities of the granule.'
Normal adult peripheral blood contains 0 to 4%
eosinophils (see Table 1-2). Normal blood eosinophils are
about the size of or slightly larger than neutrophils and have
a band or two-lobed nucleus with condensed chromatin;
rarely does an eosinophil have three lobes. There is a diurnal
variation in the percentage of circulating eosinophils, which
increases at night and decreases in the morning.
BASOPHILS
Although basophils constitute only 0 to 2% of normal blood
cells (see Table 1—2), the large, abundant, violet-blue (or
purple-black) granules aid in the immediate recognition of
this cell.! These granules are visible above the nucleus as well
Figure 1-7 @ Basophil.
as lateral to it, and they obscure most of the nucleus. The
6 Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis
MONOCYTES
In the thin areas of the peripheral blood smear, a monocyte mea-
sures about 12 to 18 um* and is larger than the mature neu-
trophil.* Monocytes have abundant cytoplasm in relation to the
nucleus (N:C ratio is 1:1 or 2:1). With Wright’s stain the cyto-
plasm turns a dull gray-blue, in contrast to the pink cytoplasm
of the neutrophils. Numerous fine, small, reddish- or purplish-
stained, evenly distributed granules in the cytoplasm give the
cell a ground-glass, cloudy appearance (see Figs. 1-2F and
1-10). There may be varying numbers of prominent granules in
addition to the small granules. Some monocytes may appear
J
nongranular, suggesting rapid turnover. Digestive vacuoles may
Figure 1-8 ™ Lymphocytes. A. Small mature lymphocyte. be observed in the cytoplasm. In disease states, phagocytized
B. Lymphocyte of intermediate size. C. Lymphocyte with indented erythrocytes, nuclei, cell fragments, bacteria, fungi, and pigment
nucleus. D. Lymphocyte of intermediate size. E. Lymphocyte with may be present.*
pointed cytoplasmic projections (frayed cytoplasm); typical nucleus. The nuclei of monocytes frequently may be kidney
F. Spindle-shaped and pointed cytoplasmic projections. G. Large
lymphocyte with indented nucleus and pointed cytoplasmic projec-
shaped, deeply folded or indented, or occasionally lobular. One
tions. H. Large lymphocyte. /. Large lymphocyte with purplish- of the distinctive features of the monocyte is the appearance of
red (azurophilic) granules. J. Large lymphocyte with irregular
cytoplasmic contours. K, Large lymphocyte with purplish-red
(azurophilic) granules and with indentations caused by pressure of
erythrocytes. L. Large lymphocyte with purplish-red (azurophilic)
granules. (From Diggs, LW, et al: The Morphology of Human Blood
Cells, ed 5. Abbott Laboratories, Abbott Park, IL, 1985, pp 1-18,
25-27, with permission.)
Figure 1-10 ® Monocytes. A. Monocyte with “ground-glass” appearance, evenly distributed fine granules, occasional azurophilic granules,
and vacuoles in cytoplasm. B. Monocyte with opaque cytoplasm and granules and with lobulation of nucleus and linear chromatin.
C. Monocyte with prominent granules and deeply indented nucleus. D. Monocyte without nuclear indentations. E. Monocyte with gray-blue
color, band type of nucleus linear chromatin, blunt pseudopods, and granules. F Monocyte with gray-blue color, irregular shape, and multilob-
ulated nucleus. G. Monocyte with segmented nucleus. H. Monocyte with multiple blunt nongranular pseudopods, nuclear indentations, and
folds. |. Monocyte with vacuoles and with nongranular ectoplasm and granular endoplasm. (From Diggs, LW, et al: The Morphology of Human
Blood Cells, ed 5. Abbott Laboratories, Abbott Park, IL, 1985, pp 1-18, 25-27, with permission.)
oa
convolutions (like those in the brain) in the nucleus (see Figs. It is helpful to memorize four helpful characteristic fea-
1-10 and I-11). Another characteristic is the lacy, often deli- tures of the monocytes: nuclear convolutions; lacy, often deli-
cate chromatin network of intermingled fine strands with small cate chromatin; dull gray-blue cytoplasm; and blunt pseudopods
chromatin clumps. (see Fig. |-10H). Kinetic studies have revealed that the half-life
The shape of the monocyte is variable. Many cells are of monocytes in the circulation ranges from 8 hours to 3 days
round; other cells reveal blunt pseudopods that are manifesta- before these cells enter tissues and are transformed into
tions of their slow mobility. These ameboid cells continue to macrophages.*
move while the blood film is drying and become fixed before Monocytes account for 2% to 9% of normal blood leuko-
the cytoplasmic extensions are retracted. Pseudopods vary in cytes (see Table 1-2).
size and number; the outer portion of the outstretched cyto-
plasm may have a hyaline appearance without granules, in LARGE LYMPHOCYTES VERSUS MONOCYTES
contrast to the inner granular cytoplasm. A monocyte (see Fig. 1-10D) is often mistaken for a large
lymphocyte (see Figs. 1-8G through L) because the mono-
cytic cytoplasm may be blue, the granules may be indis-
tinct, the nucleus is round, and the blunt pseudopods and
digestive vacuoles are missing. To distinguish monocytes
from large lymphocytes, it is useful to observe the nuclear
chromatin structure, character of the cytoplasm, and shape
of the cells. The nucleus of a lymphocyte tends to be
clumped, rather than linear or lacy as it is in a monocyte
(Fig. 1-12). There is a greater tendency for the nuclear
chromatin to be condensed at the periphery of the nucleus
in the lymphocyte. The brain-like convolutions present in a
monocyte (Fig. 1-13) are not observed in a lymphocyte
(Fig. 1-14).
Large lymphocytes and monocytes may have distinct
bluish-red granules. In a monocyte the large bluish-red granules
are interspersed with numerous fine granules in the cytoplasm
and cannot be enumerated (see Figs. 1-10B and C). In a lym-
phocyte these large granules are prominent (sometimes at the
Figure 1-11 ®& Monocytes. periphery of the cytoplasm) and can be counted easily because
8 Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis
there are no other granules (see Figs. 1-8/ and K). Because of
the finely granular cytoplasm the monocyte has a ground-glass
appearance; the cytoplasm of the lymphocyte has a relatively
clear, nongranular background. Large lymphocytes are often
deeply indented by neighboring RBCs (see Fig. 1-8K). Mono-
cytes tend to project blunt pseudopods between cells or to
compress cells, rather than being indented by them. The mor-
phological characteristics of large lymphocytes versus mono-
cytes are summarized and compared in Table 1-3.
Hematopoiesis
Definition
Hematopoiesis is the name given to the dynamic processes of
Figure 1-12 @ A. Lymphocytes with azure granules. B. Monocyte.
blood cell production and development of the various cells of
the blood. Strong evidence exists that blood cells are the prog-
eny of a hematopoietic stem cell.* Hematopoiesis is character-
ized by a constant turnover of cells. The normal hematopoietic
system continuously maintains a cell population of erythro-
cytes, leukocytes, and platelets through a complex network
of tissues, organs, stem cells, and regulatory factors.’ This
network is responsible for the maturation and division of
hematopoietic stem cells into the lineage-committed stages that
transport oxygen and excrete carbon dioxide (RBCs), fight
infection (granulocytes), perform immune functions (lympho-
cytes), and maintain hemostasis, a process in which blood clots
and bleeding is halted (platelets) (Table 1-4).
Hematopoietic stem cells duplicate themselves during
division, as shown by the curved arrow in Figure 1—15.° The
hematopoietic stem cell has the capacity for continuous self-
replication and proliferation, together with the ability to differ-
entiate into committed progenitor cells of lymphoid and
myeloid lineages.° Under the influence of growth factors
(cytokines) such as colony-stimulating factors and interleukins,
progenitor cells divide and differentiate to form the mature cel-
Figure 1-135 & Two monocytes. lular elements of the peripheral blood (see Fig. 1—15).’
The hematopoietic system consists of the bone marrow,
liver, spleen, lymph nodes, and thymus. These tissues and
organs are involved in the production, maturation, and destruc-
tion of blood cells. The entire process of hematopoiesis evolves
from the stem cells that support hematopoiesis, the progenitor
cells that are committed to particular cell lines, and the regula-
tory factors (growth factors) to which the hematopoietic system
responds. These features enable the hematopoietic system to
respond to stimuli such as infection, bleeding, or hypoxia by
increasing hematopoiesis with emphasis on the cell type needed.
becomes filled with RBCs during hematopoiesis. Bones of the Granulocytes Fight infection
toes, fingers, vertebrae, ribs, pelvis, long bones, and cranium Lymphocytes Cellular and humoral immunity
are filled with erythroid cells; early lymphocytic cells also Erythrocytes Transport oxygen and excrete
may be formed during fetal life. A few megakaryocytes (pre- (red cells) carbon dioxide
Platelets Maintain hemostasis
cursors to platelets) first appear at approximately 3 months of
fetal life, and granulocytes are observed at about 5 months.”
At birth, the liver and spleen have ceased hematopoietic
cell development, and the active sites of hematopoiesis are in
e
Hematopoietic Lineage _
Commitment |
Maturation Stages
of Committed
Stem Cells
Stages - Progenitor Cells
7@---- eo
B-cell progenitors |
B-Aympnocyte
e
: —
@~
Monocyte-macrophage progenitors
@- - -
Erythroid progenitors
+ >
|
|
|
-
asrecne
Ke)
| Erythrocytes
Figure 1-15 @ Diagram of hematopoietic cell differentiation. Hematopoietic stem cells can duplicate themselves during cell division (self-
replicate), as indicated by the curved arrow. Most descendants of the stem cells are committed to differentiate. This commitment process occurs
through a series of steps or stages, each of which leads to further restriction of lineage choice, until finally the descendant cells are limited to a
single lineage. After lineage commitment, the progenitor cells continue to differentiate and mature into the terminally differentiated cells found
in the blood. The diagram shows only the steps of commitment and does not depict the proliferation of cells that occurs throughout the process.
The amplification of cell numbers accompanying differentiation is very large. (From Koury, M, and Bondurant, M. News Physiol Sci 8:170-174,
1993, with permission.)
10. Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis
HEMATOPOIESIS
50% 50%
For platelets (also known as thrombocytes), the periph- NORMAL ERYTHROCYTIC SEQUENCE
eral blood contains 70% of platelets that circulate, with 30%
being stored in the spleen.' Figure 1-17 demonstrates that the
bone marrow pool consists of only proliferating and maturat-
ing platelet precursor cells.
One hundred percent of RBCs, known as erythrocytes,
circulate in the peripheral blood in a functional state and in
the bone marrow pool. Erythrocytes in various stages of
development are a large component of proliferating and
maturing red cell precursors found in the bone marrow. At
birth the normal cellularity is 100%.° Afterwards, the cellularity
gradually decreases with age. Marrow cellularity in adults is
approximately 50% (+ 10%). The general rule to estimate age-
related normal ranges is 100 minus the age + 10.'' For exam-
ple, the estimated normal marrow cellularity for a 50-year-old Basophilic normoblast
person would be 100 — 50 + 10; representing a range from
40% to 60%. In the bone marrow, an average myeloid to ery-
throid ratio is 4:1 in terms of cellularity.'* Therefore in an adult
with 50% marrow cellularity, approximately 40% represents
granulopoiesis and 10% represents erythropoiesis.
é =
> ee
A
; &
« os
|
D
Cc
»,
+
£
Figure 1-19 ® A. Two pronormoblasts (note the perinuclear halo). Figure 1-21 ™ Center: pronormoblasts; upper center: plasmacyte.
B. Two polychromatophilic normoblasts. C. Neutrophilic band.
D. Segmented neutrophil. £. Smudge cell.
from the pronormoblast by the coarsening of the chromatin pat- and a smaller nucleus than basophilic normoblasts.'* The
tern and the nucleoli, which are ill defined or not visible under cytoplasm contains a varying mixture of pink due to hemo-
light microscopy. As the basophilic normoblast matures, it globin and blue due to RNA; in the late polychromatophilic
accumulates more RNA and hemoglobin (Figs. 1-24, 1-25A, normoblast, the pinkish color is usually predominant.
1-26, and 1-27). The predominant color of the cytoplasm is Nuclear chromatin is thickened and irregularly con-
blue due to the staining of RNA, but there may be a pinkish densed in the polychromatophilic normoblast. Light-staining
tinge reflecting the presence of varying amounts of hemoglo- parachromatin areas are visible among the dark blue-staining,
bin. The N:C ratio in the basophilic normoblast is 6:1 to 4:1.'* irregular pyknotic masses. Nucleoli are no longer visible.
A basophilic normoblast is somewhat smaller than a pronor- The N:C ratio in a polychromatophilic normoblast is 4:1 to
moblast with a size of 12 to 17 um. Normal bone marrow 2:1'* (see Figs. 1-18, 1-19B, 1-20C, 1-25B, 1-28, and
contains 1% to 5% basophilic normoblasts (see Table 1-5). The 1-29A).
division of the basophilic normoblasts forms polychro- The maturation time for polychromatophilic normoblasts
matophilic normoblasts, which are smaller than basophilic in bone marrow is about 30 hours, and there are approximately
normoblasts but have twice the amount of hemoglobin. three times as many polychromatophilic normoblasts as
basophilic normoblasts in the bone marrow. Bone marrow in a
Polychromatophilic Normoblast (Rubricyte, normal adult contains 5% to 30% polychromatophilic nor-
Polychromatophilic Erythroblast) moblasts (see Table 1-5). Polychromatophilic normoblasts are
not present in the normal peripheral blood of adults, but they
Polychromatophilic normoblasts are smaller than basophilic may appear in small numbers in the peripheral blood of nor-
normoblasts (10 to 15 um), having relatively more cytoplasm mal newborn infants.’
Cell Percent
Figure 1-26 m Left: Basophilic normoblast; center: plasmacyte. Figure 1-28 m Polychromatophilic normoblasts: early and late
stages.
itself. Some of the bluish-staining color remains because Erythrocyte (Red Blood Cell, Discocyte)
of the presence of RNA. The erythrocyte contains approxi-
mately two-thirds of its total hemoglobin content by the The morphological characteristics of a normal erythrocyte are
time the nucleus is lost. The RNA content soon begins to presented at the beginning of this chapter.
decrease. A mature erythrocyte is not able to synthesize hemoglobin,
A diffusely basophilic erythrocyte is larger than a mature because it is without a nucleus, mitochondria, or ribosomes, but
red cell (8 to 10 um).'* It is released in 2 to 3 days from the it has a unique, yet limited, metabolism to sustain itself while
marrow and circulates for 1 or 2 days before maturing into an traversing the microvasculature. The erythrocyte carries oxygen
erythrocyte. Only rarely are diffusely basophilic erythrocytes from the lungs to the tissues where it is exchanged for carbon
found in the blood of normal adults; however, polychro- dioxide. Erythrocytes are pliable or flexible and deformable,
matophilic cells are frequently seen in the blood of normal making them capable of unusual changes in shape that are nec-
newborn infants.’ essary for the passage through the microcirculation to transport
When stained with new methylene blue, these polychro- oxygen. Refer to Table 1-6 for a summary of the morphological
matophilic erythrocytes reveal ribosomes in a granulofila- characteristics of each stage of maturation of the red cell.
mentous arrangement (or network of strands and granules)
and are classified as reticulocytes (Fig. 1-30). As ribosomes
Myelopoiesis (Granulocytopoiesis)
disappear, the diffusely basophilic cell changes into a mature
erythrocyte." Myelopoiesis or granulocytopoiesis refers to the production
With anemia or hypoxia, erythropoietin stimulates mar- of neutrophils, eosinophils, and basophils (Fig. 1-31).
row erythroid precursors to proliferate and to increase the Mature neutrophils, eosinophils, and basophils have similar
number of early erythroid cells. An increased number of poly-
chromatophilic cells are delivered early from the marrow and,
therefore, the reticulocyte count is increased.
Fi er
= a .s
Figure 1-27 ™ Center: Basophilic normoblast; right: Orthochro- Figure 1-29 mA. Polychromatophilic normoblasts. B. Lymphocyte.
matic normoblast. C. Segmented neutrophil.
IvapynN eIpNN
* Akapony/AOjOR
sade, uonsog uneUo1g?)
N
=
/10jOD
Jo”
JUNI
JUNOUWY
punoy [enusg anyq-ystppoy
youy
ioapan
10
| c0
pojddns Jejnueis
ausejdoisp
yeq
oUON
[eAol
usefdoyh3
uneuojryo .
(Qsey[quqni)
IysTys/anytq
}Sv[GOULIOUOIg
g arydose punoy fenuad ‘paseaiouy
Jase] —-YSTTS/otTTydoseg
sUON
yse]qouwou Ayiepnuers [BUOISBIDO
“‘quou
Aotignio0id)
(a Jo Ieajonu jounsIpul
uneworyo
Ajjfensp
snjooyonu
sgnuery
(ayAorIqn.t) ‘unewoiyoeied
pasvarour pedwin]s
Chapter 1
uneworys
vu
Alo A00.1y}
ay -o1yoApod
QUON
NINA) (ajA00]
sey
oyrydoyeur
udaq
snayonu
0} yuid
:e/u popn.yxo
vu vu dUON uid UON
vu
JIN}eIA
ajAd0.1YAlO
WY= sug ‘MoLeUI
v/u= jou a]qQeordde
% Ul WA= asvJUDIIOg
JO [k}O}] payvajonu
S{[99
UI Quo MOLIEU JUAasaidar[PULIOU 9oUdIOJOISasues
10J sI[Npe UdYL} WO OY} ATISIAATUF)
JO SCXAL_ P]POH DOUDIOG
1ajuaD
pure AjissoalupTeudsoyY
ueg o1Moluy
Morphology of Human Blood and Marrow Cells: Hematopoiesis
15
16 Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis
Morphological Changes
Many morphological changes occur during maturation of
granulocytes. These include a reduction in nuclear volume,
condensation of chromatin, change in nuclear shape,
appearance and disappearance of primary granules, appear-
ance of secondary granules, color changes in cytoplasm
from blue to pinkish-red, and change in the size of cells?
(Table 1-7).
Maturation of the granulocytic series of cells is char-
acterized by the development of primary blue-staining
Figure 1-351 ® Granulocytopoiesis: myelocytic (granulocytic) system. granules, which are replaced by secondary granules that
A. Myeloblast. B. Promyelocyte (progranulocyte). C. Basophilic myelo- differ in their affinity for various dyes. Cells with an affin-
cyte. D. Basophilic metamyelocyte. E. Basophilic band. F Segmented ity for basic dyes are basophils; the cells that stain reddish-
basophil. G. Neutrophilic myelocyte. H. Neutrophilic metamyelocyte.
orange with the acid dye eosin are eosinophils; the cells
!. Neutrophilic band. J. Segmented neutrophil. K. Eosinophilic myelocyte.
L. Eosinophilic metamyelocyte. M. Eosinophilic band. N. Segmented that do not stain intensely with either acid or basic dyes
eosinophil. (From Diggs, LW, et al: The Morphology of Human Blood are called neutrophils. As these motile cells mature, the
Cells, ed 5. Abbott Laboratories, Abbott Park, IL, 1985, pp 1-18, nucleus undergoes progressive changes from round to
25-27, with permission.)
multilobular forms.
Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis 17
yuid-}a]
‘sopnueis
ouy
ouy ‘oruoquy
‘orprydoinze
oytoadsuou
‘sopnuess
oruiseydoyAg
auy ‘Uasatg
saynueis
‘JUISoIg “‘WUISd1g
So|NUBLS)
uvs
1A
oytoads
sopnueis (oytoeds)
‘ourydoinze
oytoads orydo.nnouyurd-}a[O1a
‘saynueis ‘fedsoyY
JuasSgy oytoads
Ists/orrydoseg
fjurd-ysinpg AjSIOATUA)
JUNOULY /10[0D
posvoroul
aye1Opoul
/ortydosegq o}e1opoul PUR
juepunqge/yulg yuepunqe/yxuld IAIUaD
req)
quid
Avwi
BOUIIOS
10
Avi
dary
Jousnjoajonu
syqeystnsunsip
rll
YI[PAH{
YoOIpPny QuON QUON QUON
Ajisea
Sexe
uleworys
ueworys
Salas (d11ydouynaN) IADOjNUeIH ay} JO SdNSa}IeueYD jed1Hojoydsaopy 2-1 e1qeL
ouy
‘an[q-YSstppot ISI]
JO
‘yJOMYsoW oUY
sn[q-Ystppoy ATISIOATUL)
por-ystjding
/LOJORD AeapnN
oueIquoul
‘por-ystpding
podunys
Jejnueis
YIM
OU]uonesoaisse
YIOMYSOU
Ou¥ [eLIoyeul
JO WIMpajesaisse
uNeuUorys
AYslys
Ieynueis
usayjed
10 yyw
orydoseg uneworysdy)
peduinyo
Iejnuess
UONVSIISSL
JYSIS
APU
aq
Ieajonu
ye
uses
UNBUOIYD 148rT]
aydind-anyq WOIF
ye-QuUad
SIQUIIN7A
Ys]
‘an|q-Ystppos
USye}
s
JO Jo IO
[enuag [enuag [eyuag
sINpe
uOnIsOg
IeIpNnN
10
yenuad
10 dLQUaN99 d1QUINIE oLQuaa09 dLMUII99
ouacog Aqyens JO
sasuLs
:punol QoUdIOJOI
Koupry)
[Bao 0}
odeys
[BAo
0} Apystys
pojuopul poyuopul
(podeys pueq
MoieUjo
odeys
wuojrun
(20Ysesioy) Sogo]
SSOUYoIY} Jegponu
JTeIpPNN punoy [RAO
IO Aypensq ‘polesuoly C—Z
JOUNSTP [LULIOU
punoy
CEL
JUASaIdar
ae
ON
eI
[-T
ouey
eee
MOLILUT
“SZIS [JID
9I-Ol
9I-OL
8I-Ol
sI-0l
Oc-SI
ui
BUOY
UT
S][99
%S1-S
%O0C-S
payeafonu
%S1-S
ee
[e}O}
JO
VsvjUSIIOg
JSP]QOTAAJY
Io
sjtydonnou)
otydo.nnan poayuswises
aj AoojaAw
ottydonnan
=
WE
Ul
AdojaAurej}our
a pueg (sAjod%
WEN Hp-]
aAd0]aAWO.1g %SE-Ol
orrydo.nnoN SPX],
omrydonnan
18 Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis
mn
aes
= .
Figure 1-55 mA. Neutrophilic myelocyte. B. Neutrophilic Figure 1-37 ™ A. Two neutrophilic metamyelocytes. B. Three
metamyelocyte. C. Plasmacyte. D. Orthochromatic normoblasts. neutrophilic bands. C. Two segmented neutrophils.
E. Segmented neutrophils. F Nucleus of a degenerated cell.
primary granules. As myelocytes divide and age, the primary neutrophilic cell. Metamyelocytes do not divide, nor do they
granules become fewer and the secondary (specific) neu- have nucleoli.* The N:C ratio is 1:1.'*
trophilic granules predominate.* Secondary granules are consid- Many small, pinkish secondary granules fill the cytoplasm,
ered to be specific granules for neutrophils, and they contain and there may be a few primary darker granules remaining (see
collagenase, lysozyme, lactoferrin, plasminogen activators, and Figs. 1-31H, 1-35B, and 1—37A). These maturing cells remain
aminopeptidase.* in the bone marrow and represent a portion of the granulocytic
The nuclei of myelocytes may be round, oval, or flat- reserve.”
tened on one side and usually are eccentrally located? (see Metamyelocytes are somewhat smaller than myelocytes
Figs. 1-31G and 1-35A). Chromatin strands become con- (10 to 18 um) and are larger than the band neutrophil or seg-
densed, partly clumped, and thickened, and are unevenly mented cell.'* These cells are usually absent in normal periph-
stained. Nucleoli are absent or indistinct in myelocytes. The eral blood. There are approximately 5% to 20% metamyelocytes
neutrophilic myelocyte is the last myeloid precursor capable in normal bone marrow.
of division.*
Neutrophilic myelocytes are often smaller than promye- BAND NEUTROPHILS When the stage is reached in which
locytes (10 to 18 um) and have relatively large amounts of the nuclear indentation in the early granulocyte is greater than
cytoplasm (N:C ratio is 2:1 to 1:1),’* which gradually half the width of the nucleus (see Fig. 1—36), the cell is iden-
becomes less basophilic and more pinkish. The normal tified as a band neutrophil.’
peripheral blood does not contain neutrophilic myelocytes. The opposite edges of the nucleus become almost paral-
There are 5% to 20% myelocytes in normal bone marrow. lel, giving the appearance of a horseshoe, hot dog, or a curved
link of sausage. The shape of the nucleus of a band neutrophil
NEUTROPHILIC METAMYELOCYTES As maturation pro- is often folded or twisted, giving rise to difficulty in distin-
ceeds, the nucleus becomes slightly indented (bean- or kidney- guishing a band from a segmented neutrophil. The nuclear
shaped), and this shape serves to identify the cell as a chromatin is pyknotic, and there is usually a dark condensed
metamyelocyte. The indentation is less than half the width of mass at each end where the lobe is destined to be.* The small
an arbitrary round nucleus (Fig. 1—36).* There is noticeable secondary neutrophilic granules are evenly distributed and
condensation with clumping of the chromatin, but the chro- stain various shades of pink (see Figs. 1-31/ and 1—37B). An
matin structure is not as dense as that of the segmented occasional dark primary granule may be observed. The N:C
ratio of a neutrophilic band is 1:1 to 1:2."
Neutrophilic band cells are often slightly smaller than
TERMINOLOGY BASED ON INDENTATION OF NUCLEI metamyelocytes. Band forms constitute from 10% to 35% of
the nucleated cells in the bone marrow.
Figure 1-38 m Tissue neutrophil (large center cell). Figure 1-40 @ A. Eosinophilic metamyelocyte. B. Neutrophilic
band. C. Polychromatophilic normoblast.
Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis 21
O% i el
Figure 1-41 @ A. Eosinophilic band. B. Neutrophilic band. BASOPHILS Basophils may be identified as basophilic
C. Lymphocyte. D. NRBC (orthochromatic normoblast). £. Seg- myelocytes, metamyelocytes, bands, and segmented cells
mented neutrophil. based upon the shape of their nuclei. The shape of the
nucleus is, however, often masked by large basophilic gran-
The granules of eosinophils contain various hydrolytic ules (see Figs. 1-31C through F). The specific violet-blue
enzymes, including peroxidase, acid phosphatase, aryl sulfa- granules of basophils are formed in the myelocytic stage and
tase, beta-glucuronidase, phospholipase, cathepsin, and ribonu- continue to be produced throughout all later maturation
clease, but they lack lysosome, cationic proteins, and alkaline stages (see Figs. 1-31C through F).
phosphatase.* Maturation of basophils in the bone marrow takes place
Normal adult bone marrow contains 0 to 3% eosinophils. over 7 days.* Mature basophils rarely have more than two
The morphological characteristics of the eosinophilic series segments. Basophils circulate for a few hours in blood, then
are summarized in Table 1-8. migrate into skin, mucosa, and other serous membranes.*
Basophils in all stages of maturation are smaller than
Tissue Eosinophils promyelocytes and neutrophil myelocytes; their size approx1-
In smears of bone marrow, occasionally there may be a large mates that of neutrophils. The morphological characteristics
cell with elongated and tapering cytoplasmic extensions, con- of the basophilic series are summarized in Table 1-9. Normal
taining typical reddish-orange granules of the type seen in the bone marrow has 0 to 1% basophils.
eosinophils of the circulating blood? (Fig. 1-43). The nucleus of
such cells, instead of being indented or lobulated, is round or Tissue Basophil (Mast Cell)
oval and has a well-defined reticular chromatin and, often, Tissue basophils (mast cells) (Fig. 1-44) and blood basophils
nucleoli. Such cells are identified as tissue eosinophils and are are closely related in their functions and biochemical character-
thought to be fixed tissue variants of the more motile eosinophils istics, but the relationship between them is still being studied.
of the circulating blood.* Tissue eosinophils arise from the same Both cells participate in a similar manner in acute and delayed
progenitor cells as eosinophils in the marrow and blood.’ allergic reactions. The granules of both cells have similar
morphological characteristics, and each cell contains hista-
mine and heparin and is water soluble.*
%
ut
Jo
UT
Wa
Jo
Joy
[e}0)
om)
JUG
S[[99
pue
uo
ues
ee
s][Npe
[eWOU
MOLL
sosuvl
Sexo]
ee
“SPXO],
nleeH
poyeajonu
pees
JUaseidar
se
oe
oBvJUIdI0g
passe
Sega
Beas
Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis 23
sojnueis sopnue.is
yUNOWY/1OJOD
wisejdoyAZ
pasevoioul
soutydose ‘ged 91e.1Opoulr o1e.1opoul
‘anyq ‘an{q
ysintg
aed aed
Jo g
snyoojonu
TJOopPTNY
Avy
[KEIN QUON QUON
TI
UIM
WSIS
‘an[q-YsIppal Jepnueis Iejnuess
SyJOMYsoU ye ‘ajdind-anjq
aqeysinsunsip ‘gidind-anjq
JO
/AOOD UONeSAIBSe Apyst[s
WLI ‘an[q-YysIppoy
YIoMYsou payesoisse uneulo1ys uneUIOIYS
UneUIOLQ Ajasreoo Ajasreoo
1eoyonu
Teapanyy uA
sUy IYsiT] daaq daaq
Ou [euayeur
Jo
‘
uonesalsse sopnueis
reynueis
usoned
=,
yeanou [enuad A[UOUIWUOD O1IUIIIO oLQuacce
oLUIINY oq [e-nuss JO JO
UOISOg
IvIpPON [euag [enue
AvUI
10 10
=
JOUTSIP
poyuopul ‘(podeys _.podeys Jeoponu
Apystys OSUSNRS,,
IwapnN JO soqo]
adeys punoy
[VAQ OME,
wojunuoU
Aaupry)
uneuro3yo
Ajiseo [eAo
QZIS ©
pl-Zl 1-01 91-01 91-01
wu
IQ
IPO
pueg
ay poyusuises
({1ydoseq)
Ad0[aAWIOIg
ortydoseg orydoseg orptydose
sve
aye19poUul
(oytoeds)
uy
uneulolys
A00]aAuUr
‘punol
‘OEIUIIIO
jou
a)
Ae aye1opoul
poquoput
219909
IM
orprydoseg
A007
“N[Q-}O[OLA
ay
we}
0A
OW
g
ysry]
ajdind-anyq
QUON
aed
‘anyq
‘JUaSdIg
arpiydose
%1-O
SI-Ol
ei
Ayyensp)
[euag
gs1e0d
JO g
=
mal!
quasqy
ystys/orftydoseg
Ise[qo[ah
WYysr]
‘anyg-YsIppos
%C-O
OS!
[Slee
punoy
USSSA
OP
24 Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis
Tissue basophils are derived from the pluripotential stem and sometimes contain a few peroxidase-positive granules.
cell and are fixed tissue cells. The cytoplasm of the tissue The N:C ratio in promonocytes is 4:1 to 2:1 (see Fig. |-45£)."*
basophils is filled with large, prominent, intensely stained Promonocytes are slightly motile and may infrequently take
violet-blue granules. The granules are usually round and about part in phagocytosis.
the same size (0.1 to 0.3 um). They may overlay the margins of Promonocytes and monoblasts are not easily identifiable
the palely stained nucleus or obscure the nucleus completely. in bone marrow or peripheral blood smears except in disor-
The nucleus is small, round or oval, and not segmented.* ders in which there is marked proliferation of monocytic cells.
Tissue basophils are widely scattered in the connective The identification of early monocytic cells is based on slightly
tissue of various organs, bone marrow, and the mucosal area indented, or folded, large nuclei and on association with more
of serous membranes. In bone marrow tissue basophils are mature cells that have pseudopods and brain-like convolu-
usually observed in the hypercellular area and can be located tions in the nucleus.”
in the “squashed” smear made from a marrow particle. Some
tissue basophils have spindle shapes and jagged margins
resulting from trauma in the process of aspiration.’
Monocytes and Macrophages
Promonocytes develop into monocytes. Monocytes enter the
circulation for a short time and then migrate into tissue to
Monopoiesis
transform into tissue macrophages.’
The mononuclear phagocyte system (MPS) is composed of The characteristic features of monocytes in normal periph-
monocytes, macrophages, and their precursors—monoblasts eral blood are given in the earlier part of this chapter (see
and promonocytes.* The cells composing this system arise in Fig. 1-10). The scanning electron microscope shows the mono-
the bone marrow from progenitor cells that are committed to cyte to have a ruffled plasma membrane with long, thin
monocyté-macrophage production.* microvilli.’
As monocytes mature, they become too large to pass read-
Monoblasts and Promonocytes ily through capillaries, and so they move into tissue and convert
into macrophages in many organs (e.g., pulmonary alveolar
Monoblasts are large and have an eccentrically placed nucleus macrophages, peritoneal macrophages, splenic macrophages,
that may be minimally indented, one or two large prominent Kupffer cells in the liver, and connective tissue macrophages). '”
nucleoli, a fine, lacy nuclear chromatin, and a nongranular cyto- This transformation involves rapid growth, enlargement, and
plasm that stains a deep blue (Fig. 1458). The N:C ratio in intensified phagocytic activity. Macrophages do not normally
these cells is 7:1 to 4:1 (Table 1—10).'* Monoblasts are non- re-enter the bloodstream but may re-enter the circulation during
motile and nonphagocytic cells. Monoblasts divide and give rise inflammation.*
to promonocytes and then to monocytes. Macrophages are large, irregularly shaped tissue cells (25
Promonocytes also are large and have indented or folded to 80 um) with a round or reniform nucleus; and contain one
nuclei and fine chromatin. They often have a visible nucleolus or two nucleoli, clumped chromatin, abundant cytoplasm with
Figure 1-45 = Lymphocytic, monocytic, and plasmacytic systems. A. Lymphoblast. B. Monoblast. C. Plasmablast. D. Prolymphocyte.
E. Promonocyte. Ff. Proplasmacyte. G. Lymphocyte with clumped chromatin. H. Monocyte. /. Plasmacyte. (From Diggs, LW, et al: The
Morphology of Human Blood Cells, ed 5. Abbott Laboratories. Abbott Park, IL, 1985, pp 1-18, 25-27, with permission.)
Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis 2
vacuoles, and numerous azurophilic granules.* Macrophages nucleolus'® (see Fig. 1-45C). The cytoplasm is blue.
are also called histiocytes (histio = tissue; cyte = cell). The Plasmablasts are identified primarily in the presence of
morphological characteristics of the monocytic macrophage proplasmacytes and plasmacytes but cannot be easily differen-
series are summarized in Table 1-10. tiated from other blasts. The plasmablast appears slightly
larger than the more mature plasmacyte. The plasmablast is 16
to 25 um and the mature plasma cell is 10 to 20 um.'°
Lymphopoiesis Proplasmacytes and plasmacytes differ from plas-
The lymphoid progenitor cell is derived from the hematopoietic mablasts in that the color of the cytoplasm is deep blue, the
stem cell. The common lymphoid progenitor cell can differen- juxtanuclear light areas are prominent, and the nuclei are
tiate into either T or B cells, depending on the microenviron- eccentric.* The chromatin structure of the nuclei in proplas-
ment. T cells differentiate in the thymus, B cells in adult bone macytes is intermediate between that of plasmablasts and
marrow. Null cells, or third-population cells, originate in the plasmacytes. In proplasmacytes the nucleolus may be ill
bone marrow, although the maturation sequence is unknown.'” defined or absent.* The N:C ratio in proplasmacytes is 4:1 to
T, B, and null cells cannot be separately identified morpholog- 3:1_(seé Fig. 1-45 F).*°
ically but can be distinguished functionally and by immuno- Plasmablasts and proplasmacytes, although not observed
logic marker studies. (See the section on CD Nomenclature in normal bone marrow, are seen in diseases associated with
Jater in this chapter.) abnormal immunoglobulin production, especially multiple
In primary lymphoid organs stich as the thymus and bone myeloma.*
marrow, lymphocytes differentiate, proliferate, and mature into
fully functional immune cells. In secondary lymphoid organs Plasmacytes (Plasma Cells)
such as lymph nodes, spleen, and mucosal tissues (tonsils,
Peyer’s patches), lymphocytes communicate and interact with Plasmacytes represent the end stage of B-lymphocyte lineage.
antigen-presenting cells (APCs), phagocytes, and macrophages They are not observed in the peripheral blood smears of normal
in an active immune response.* individuals but constitute about 1% of the nucleated cells in
normal marrow.’ Mature plasmacytes range in size from 10 to
20 um.'® They may be round or oval, with slightly irregular
Lymphoblasts and Prolymphocytes margins.
The cytoplasm is nongranular and usually stains a deep or
The earliest lymphocytes are identified as lymphoblasts vibrant blue. The cytoplasm adjacent to the nucleus is pale, with
and prolymphocytes. Lymphoblasts contain a large, round a perinuclear clear zone containing the Golgi apparatus, and at
nucleus with a small or moderate amount of basophilic the cell periphery there are secretory vesicles. Fibrillar structures
cytoplasm. The nuclear chromatin strands in lymphoblasts that stain blue may be demonstrable in the cytoplasm. One or
are thin, loose, evenly stained, and not clumped. One or sev-
several small vacuoles may be observed. There is no evidence of
eral nucleoli are usually demonstrable.* These cells measure phagocytosis of visible particles.*
10 to 20 um in diameter and have a N:C ratio of 7:1 to 4:1" The nucleus of a plasmacyte is relatively small, round, or
(see Fig. 1-45A). oval, and eccentrically placed in the cell. The nuclear chro-
Prolymphocytes have an intermediate chromatin pattern matin is clumped or coarse and lumpy, similar to that of a
that has clumps in some areas of the nucleus but does not appear lymphocyte (see Figs. 1-45/, 1-46, and 1-47). The N:C ratio
as clumped as in mature lymphocytes.’ Parachromatin, which of a plasma cell is 1:1 to 1:2.'°
appears reddish-purple, may be present in the nucleus. Nucleoli
are less distinct than in lymphoblasts. Prolymphocytes are
slightly smaller than lymphoblasts, approximately 9 to 18 um,
and have a N:C ratio of 5:1 to 3:1'* (see Fig. 145D). Differ-
ences are subtle, and in case of doubt the cell should be called a
lymphocyte.” The morphological characteristics of the lympho-
cytic series are summarized in Table 1-11.
Lymphocytes
The morphological description of lymphocytes may be found
in the first part of this chapter.
0} orprydoinze
soynuRs
aye1apoul
Ayueosanyq
uses
puno1 10
‘uasqe
poluopul ayAooydwiA]
Ajueos/orprydoseq May}
“snoussowloy
atdind-por
uneuoryo
puno.
yim
afdind
‘posuaspuoZ
peduinyo
pajuopul
10
aps duo 0}
-anjq
juosqy
tuseydoyAo Aqueos
Ie
Ajueos/otrydoseq qjoours/atdind-poar
atuuseydoyAy
Ut EL
sajnueis
adeyg
respon
uiseydoyk3Jo “yoann UNeULOAYD uonISOg oney
fLOJOD TeITINN] IvapNny O:N
Morphology of Human Blood and Marrow Cells: Hematopoiesis
oyrydoinze
wuse{doyAd soynuris
WLIOJUAL
SnoJOUNN
soyonoea
(PIM Juepungy TI uneworyo podunio d1NUIDIq JO punoy = tOr ail O8-SC ssvydoioryy
oredopsnpjnoysno1y)
JO poayesuoyja
soponsed vipodopnosd SULIOJ OINVBUULUT
SNOIOUINN
SUIP[O}
posevoroUuy
«OTTG914,, uey) ulayyed
‘poi
ajyA00uoWlo.ldg
O1TG9I9,, WIOJLIQOII9
ayly-Jonp ‘por qm JuepuNge JO sasraio
‘quy ule}UO0S /ourydoseq ujoyed repnono1 uNeWOIYyo
you Aeul/Aeyy Avis
1o[eg ‘gtdind-pai o]ed jenusg YIM punoy, Lc-Lv Oc TI
JO Apystys
P9plOF
JUIB }Se]qouo|y
oLuacoq
uneuloly9s
‘[BAO
Chapter 1
‘punoy
o)1e1OpOul Apeoiy) ‘ouy
QUON
sorrydoseg ZI ‘g1dind-pai ajeg beote Gas)
wonsod
adeys
IvapnN
sopnuessy uisejdozAy jo aeMoyQ oney it
A
6 diuisejdozy<D junoury/10joD /AOJOR Avapny Ivapny IN 8215 11D
)
Salas WAIOUOP| AU} $O SIIVSLaIDeIeYD Jer21Hojoydsop O1-1 PIGeL
Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis PAT
[JID
adeys
“OZIS
UOTpISOg uneUo.yD HooponNJo wusvjdoy45 sopnuessy
ual
ysv[qeuiselg Seas)! yes, punoy [enuag apeg ‘o[dind-pai
auy €-1 aed Ajueos/anyq
01 oUON
pojddnys uneutorys ayeropour juonboly
Fea;OnNuLIE
Iva[dd 2UOZ
ayAovurse[dorg OGEST LE-Ly punoy omnus904q ‘aydind-poyposvaiour Te) yAeq a}eOPOU/antq
I0
QUON
[BAO Ayue[nuess
Morphology of Human Blood and Marrow Cells: Hematopoiesis
Jo unewo1yo
oINeIy Oc-OL CIEL punoy Ayjensq ‘aydind-ont
asuep
gq QUuON yeq aeJopow/onyq
vuseyd
[[99 0 OUON
[BAO oLuea9a UNeUTOTYS
YIM ase] wseydoys
yimo Jeajonutiod
sduinyo
Jeou Ieajonu Ieayo ‘QUOZ ABUI
uIsIeUr ureyUo sojonoea
Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis 29
Figure 1-48 @ Megakaryocytic system. A. Megakaryoblast with single oval nucleus, nucleoli, and bluish foamy marginal cytoplasmic struc-
tures. B. Promegakaryocyte with two nuclei, granular blue cytoplasm, and marginal bubbly cytoplasmic structures. C. Megakaryocyte with
granular cytoplasm and without discrete thrombocytes (platelets). D. Megakaryocyte with multiple nuclei and with thrombocytes (platelets).
E. Megakaryocyte nucleus with attached thrombocytes. F Thrombocytes (platelets). (From Diggs, LW, et al: The Morphology of Human Blood
Cells, ed 5. Abbott Laboratories, Abbott Park, IL, 1985, pp 33-34, 48-50, 85, with permission.)
Figure 1-49 ®@ Center: early megakaryocyte; top left: segmented BSUS lw Center: early megakaryouyte.
neutrophil; bottom center: neutrophilic metamyelocyte.
30 Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis
Ss
pgs
aa
Re,
Pigure 1-51 ™ Megakaryocytes without platelets. Figure 1-53 ™ Megakaryocyte with platelets.
Ultrastructural features and marker studies on megakary- Studies have shown that bone formation and hematopoiesis are
ocytes aid in identification of all stages. Ultrastructural fea- closely linked, in that the degree of hematopoiesis correlates
tures are the demarcation membrane, alpha (u) granules, and with the rate of bone turnover. Two normal, nonhematopoietic
platelet peroxidase activity. Monoclonal antibodies for cells that exhibit different functions, yet play essential roles in
platelet glycoproteins Ib, Ib, and IIa; factor VIII antigen; the formation of the bone cavity, are the osteoblast and the
beta-thromboglobulin, and factor V are markers for all stages osteoclast.
of megakaryocytes.'’ In marrow smears of normal individu-
als, there are approximately | to 4 megakaryocytes per 100
Osteoblasts
nucleated cells, and these cells are in the late stage of matura-
tion.2 The morphological characteristics of the megakary- An osteoblast is a large cell that can measure up to 30 um, with
ocytic series are summarized in Table 1-13. ample cytoplasm and a small, round, eccentrically placed
nucleus. These cells may be traumatized in the process of mar-
row aspiration and smearing and often have irregular shapes and
Bone-Derived Cells
cytoplasmic streamers. The cells may have comet or tadpole
The formation of the bone marrow cavity results from a com- shapes. The nucleus may be partially extruded, similar to a
plex process in which hematopoietic cells migrate and colonize small, round head on a round body. The nuclear chromatin
spaces originally occupied by cartilage and bone. This process strands and nuclear margins are well defined and stain purple-
occurs both in the long bones of the limbs and in the membra- red. Usually there is a distinct blue nucleolus? (Fig. 1-55).
nous bones of the skull, which develop directly into bone. Throughout the blue cytoplasm there are small spherical
bodies that are colorless and give a bubbly appearance to the
cytoplasm. Within the cytoplasm there is a prominent round
or oval chromophobic zone that stains lighter than the rest of
the cytoplasm. This area is usually away from the nucleus but
may be adjacent to it.”
Osteoblasts, more often seen in marrow from young
children, are responsible for the formation, calcification, and
maintenance of trabeculae and cancellous bone. Osteoblasts
secrete large amounts of collagen and proteoglycans, con-
tributing to structure of the bone and stromal matrix.4
Osteoblasts morphologically resemble plasmacytes,
both having irregular shapes, eccentric nuclei, cytoplasmic
protrusions, blue cytoplasmic fibrils, and vacuoles. The rel-
atively unstained zone of the plasmacyte is adjacent to the
Bape
oruuseydoyAg
‘on[q-Ysippod
Veer ear
Ie]NueISUON
Sure
Seem
orrydoinze
soynueis sopnueis “ouy
ouyorrydoinze ATudA9
sanuesy SnoJOWNN pasiadsip
:|
eipodopnasd ‘orrydoseg
juepunqesjseo yurd
ANIROSCHANEL SIR |
uisejdozAZD jo
uuseydoyAo
fae
JO
ayAo0Areyesoul
eee
==
——=
isvjq
se.
Ae
eS
UdY}
SSO]-oAreyesoul
Ayyeusn
‘{—0 QUON eu
UNRUIOIYS JOUNSIP YIM
Iegjonu Jo Ayaepnuess
unewuorys afdind-poy
/LOJOR espINNY
‘ajdind-on|g
oe ait sree Seer
unewiolyo
uneuoly9
Iepnurss
See
ere Se
ajdind-pay
pasvosour
Jo
919099
JO [eus)
[euo+)
uonIsOg
Ivan
[enuag
exer
eit eetin
o1ua909
sears
eee
=
-
CYN
ao
Sees
Matar
|
Dee
eee
poqynqoy
C
g
ikEee
canes
passa
fs
neni:
5
eer es
‘punol
[Baopojuapul
JO podeys-Aoupry
IO (s9qo|
Z)
o1OW
eee Seat
¢
payuopul
JO
podeys-Aoupry
‘punol
‘[eAo
Ayjens~
gjsurs AypensA
g[surs
eee
le
Gaede:
(liaMelt
este
onry
ON
Sy-0C
‘IZIS [IO
0O0I-0€
08-0C
wd
PRT
TN
ajA00quiosy
eeu)
-OAIBYRSIIN
Figure 1-54 ™ Naked nuclei, megakaryocyte. Figure 1-56 @ Group of osteoblasts (center) aspirated from the
marrow of a child. (magnification x 400)
Osteoclasts
Figure 1-57 @ The osteoclast is usually seen as a single giant cell
Osteoclasts are giant (greater than 100 um), multinucleated, with multiple and separated nuclei and basophilic granular
irregularly shaped marrow phagocytes that are capable of cytoplasm (center). (magnification 640)
Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis eS)ioe)
MEGAKARYOCYTE OSTEOCLAST (Go) after division. From the Gp state the resting cell
enters the G, phase, which is the postmitotic rest phase and
which directly precedes the DNA synthesis phase. The cell
proceeds into the synthesis (S) phase of active DNA synthe-
ag
sis, where the DNA content is doubled. The next phase is the
premitotic rest period (G;) as the cell prepares to enter the
mitotic period (M). During the final, or M, phase, there is cel-
lular division of the chromosomes in the nucleus and the
cytoplasm, resulting in two daughter cells.' T, is the cycle of
Figure 1-58 m Osteoclast versus a megakaryocyte. one complete mitotic division. After final differentiation, the
cell leaves the cycle as a nondividing cell (Gy) (see Fig. 1-59).
Differentiation is associated with cell-cycle arrest.*? In
(OAF), secreted by plasmacytes in myeloma, is a lymphokine erythropoiesis, terminal differentiation is coupled to prolifer-
that stimulates osteoclastic activity in the endosteum near ation.*? Erythroid precursors can undergo “renewal divisions”
groups of myeloma cells.‘ The presence of OAF helps to which is defined as divisions that double the number of
explain the development of osteolytic bone lesions observed erythroid precursors (progenitors), while maintaining the
in myeloma.* differentiation potential.*? This occurs when the expansion of
These cells are involved in the degradation (or reabsorp- erythroid compartment is required during fetal erythropoiesis
tion) of bone, which is essential for the formation of the bone or following hypoxic stress. Renewal divisons comform to
marrow cavity and bone remodeling. Osteoclasts are derived standard cell cycle characteristics.”° Several specific regula-
from the monocyte/macrophage hematopoietic cell lineage.'* tory proteins have been recently shown to regulate genes
Osteoclasts adhere to the bone matrix and secrete lytic involved in development and differentiation.*'! This provides
enzymes that degrade it. Osteoclast proliferation and survival a critical link between the cell cycle and differentiation
of their precursors depend on the cytokine, macrophage during development?!
colony-stimulating factor (M-CSF).'? The morphological
characteristics of the osteoblast and osteoclast are compared
in Table 1-15.
Specific Cell Line Ontogeny
MULTIPOTENTIAL STEM CELLS—COLONY-FORMING
UNITS
Molecular Hematology and Advanced The morphology of the recognizable stages of peripheral
Concepts blood and marrow cells has been described. As stated ear-
Introduction to the Cell Cycle lier, these cells come from an unrecognizable pluripotential
stem cell. The pluripotential stem cell has the capacity for
THE GENERATIVE (G) CELL CYCLE KINETICS continuous self-replication and also for differentiation into
When stimulated by hematopoietic growth factors (see the the multipotential stem cell and the lymphoid stem cell
discussion of colony-stimulating factors and interleukins later without capacity for self-renewal.*? The multipotential stem
in this section), hematopoietic cells undergo a continuous cell becomes committed to support progenitor cells for
generative (G) cycle in which the cells divide, differentiate, or myelopoiesis, erythropoiesis, monopoiesis, and megakary-
remain dormant”? (Fig. 1-59). The bone marrow contains cell opoiesis. The lymphoid stem cell line supports lym-
populations in all phases of cell development. The generative phopoiesis. The multipotential stem cell was shown to exist
cell cycle is divided into five phases: Gp, G,, S, Gs, and M.' in a classic experiment in 1961 by Till and McCullock,?*
The cells able to proliferate enter a resting or dormant phase who irradiated mice to empty the hematopoietic organs and
Cell
Size, pm Shape Cytoplasm Nuclei
then injected a suspension of marrow cells intravenously. other immunocompetent cells. B cells differentiate into plas-
About a week later, nodules of injected marrow could be macytes, which secrete specific immunoglobulins important in
observed on the cut surface of the spleen colonies. All cell the host’s defense against infection. Another population of
lines found in normal marrow were generated from the lymphocytes, called null cells, has none of the characteristics
multipotential stem cells in the marrow suspension. The mul- of either the T or the B cells. The null cell category includes
tipotential stem cell giving rise to several cell lines was killer (K) cells, which interact with antibody to cause destruc-
called the colony-forming unit—granulocyte-erythrocyte- tion of antibody coated targets, and natural killer (NK) cells,
monocyte-macrophage-megakaryocyte (CFU-GEMM).** which can lyse target cells through direct cytotoxic activity.
The CFU-GEMM in a colony assay forms a series of The different types of lymphocytes and their functions are
progenitor cells (CFU-GM, CFU-Eo, CFU-Bas, CFU-Meg, listed in Table 1-17.
BFU-E, CFU-E) under appropriate growth conditions (see
Table 1-16 for acronyms). CFU-GM makes colonies of gran- COLONY-STIMULATING FACTORS AND INTERLEUKINS
ulocytes and monocytes and/or macrophages (Fig. 1-60). Each cell line is dependent on cytokines, which are soluble
CFU-Eo forms colonies of eosinophils. CFU-Bas makes early mediators secreted by cells for the purpose of cell-to-cell com-
basophils and mast cells. CFU-Meg forms megakaryocyte munication. The different cell types and the cytokines they pro-
colonies. There are two colonies of erythroid progenitor cells: duce are listed in Table 1-18. The characteristics of cytokines
the early burst-forming unit-erythroid (BFU-E) (Fig. 1-61) are listed in Table 1-19. Cytokines act on multipotential stem
and the more mature colony-forming unit-erythroid (CFU-E). cells to stimulate their proliferation and differentiation to
The lymphoid stem cell is also derived from the pluripo- committed cell lines (Fig. 1-62).™ For a cell to develop from
tential stem cell. The lymphoid stem cell has the potential to a multipotential cell to a myeloblast, monoblast, erythroblast,
differentiate into a T or B cell. T cells participate in immune or megakaryoblast, cytokines are necessary. Colony-stimulat-
functions of a cellular nature, either directly cytotoxic, or help- ing factors (CSFs) or growth factors and interleukins (ILs) are
ing or suppressing immune activities through interaction with two types of cytokines. The cytokines involved in hematopoi-
etic blood cell development are summarized in Table 1—20,
DNA
synthesis
fe)
CFU-GEMM Colony-forming unit—granulocyte,
erythrocyte, macrophage—
Interphase” Nondividing monocyte, megakaryocyte
cell CFU-GM Colony-forming unit—granulocyte,
macrophage—monocyte
Postmitotic CFU-Eo Colony-forming unit-eosinophil
rest period CFU-Bas Colony-forming unit—basophil
CFU-Meg Colony-forming unit—
Figure 1-59 m Cell cycle kinetics. Tg = one complete mitotic megakaryocyte
cycle; Gy = resting or dormant phase; G, = postmitotic rest period; BFU-E Burst-forming unit-erythrocyte
S = active DNA synthesis phases; G, = premitotic rest period; M = CFU-E Colony-forming unit-erythrocyte
mitotic period; Gy, = nondividing cell.
Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis ios)Nn
/Functions
of Lymphocytes —
Lymphocyte Function
IL-1
IL-6 Phitinotent IL-1
IL-3 Stem Cell IL-6
fs)
4 he
() , BS
IL-6
foo
Lymphoid IL-1, 1L-2
seas resteee IL-7/ Stem Cell \ IL-6, IL-7
IL-3 [Thymus]
a —
CFU-Bas
IL-1
IL-2 rg:
BFU-E CFU-Meg
IL-4 IL-4
GM-CSF GM-CSF GM-CSF iL-5
GM-CSF
IL-3 IL-3 IL-3 act IL-6
IL-3/IL-4
CFU-G
GM-CSF
G-CSF
GS antigen antigen
q , driven driven
7 3% & @ @
a a>
=
a i y 4
Erythrocyte Thrombocytes Polymorpho- Monocyte Eosinophil Basophil B Cell T Cell
nucleated GM-CSF
Neutrophil |M-CSF IL-3/IL-4
Figure 1-62 ® Regulation of hematopoiesis by cytokines. BFU-E = burst-forming unit-erythroid; CFU-Bas = colony-forming unit-basophil; CFU-E
= colony-forming unit-erythroid; CFU-Eo = colony-forming unit-eosinophil; CFU-G = colony-forming unit-granulocyte; CFU-GEMM = colony-
forming unit-granulocyte, erythroid, monocyte macrophage, megakaryocyte; CFU-M = colony-forming unit-monocyte; CFU-Meg = colony-
forming unit-megakaryocyte; EPO = erythropoietin; G-CSF = granulocyte colony-stimulating factor; M-CSF = monocyte-colony-stimulating
factor. (Reprinted from Sandoz Pharmaceuticals Corporation and Schering-Plough, with permission.)
Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis = 37
Growth Factors
Epo Erythropoietin Kidney, liver Erythroid progenitors
G-CSF Granulocyte colony- Macrophages, Stem cells, neutrophil
stimulating factor endothelial cells, precursors
fibroblasts
GM-CSF Granulocyte-macrophage- T lymphocytes, Progenitors for neutrophils,
colony-stimulating factor macrophages, eosinophils, monocytes
endothelial cells,
fibroblasts
CSF-1, Monocyte- Endothelial cells, Mononuclear phagocytes
macrophage CSF fibroblasts, B cells,
monocytes-macrophages,
stromal cells
SCF Stem cell factor, Fibroblasts Stem cells
c-kit ligand
Interleukins
mei Hematopoietic-1; Macrophages, fibroblasts, Mononuclear phagocytes
response modulator endothelial cells progenitor cells
IL-2 T-cell growth factor T lymphocytes, T cells, B cells
macrophages
IL-3 Multi-CSF T lymphocytes Precursors of neutrophils,
platelets, monocytes,
eosinophils, basophils,
stem cells
IL-4 B-cell stimulatory factor I T lymphocytes B cells, mast cells, T cells
IL-5 B-cell growth factor II, T lymphocytes B cells, eosinophils
eosinophil differentiation
factor
IL-6 Interferon 8 hybridoma T lymphocytes, Stem cells, B cells
growth factor macrophages
IL-7 Lymphopoietin- 1 Stromal cells Pre-B cells, T cells,
early granulocytes
IL-8 Granulocyte Monocytes, T cells, Neutrophils, T cells,
chemotactic factor fibroblasts basophils
IL-9 T-cell growth factor II T cells BFU-E, T cells, mast cells
IL-10 Cytokine synthesis T cells, macrophages, B cells, macrophage,
inhibitory factor B cells T cells, mast cells
IL-11 Adipogenesis Stromal, fibroblasts Megakaryocyte, B cells,
inhibitory factor mast cells
IL-12 NK cell stimulatory factor B cells, macrophages T cells, NK cells Not reported
IL-13 IL-13 T cells B cells 5q
IL-14 High-molecular-weight T cells Activated B cells Not reported
B- cell growth factor
w Normal eosinophil granules are large, round, and stain # A neutrophilic metamyelocyte has a bean-shaped nucleus
orange to reddish-orange; eosinophils are found in 0 to with the indentation less than half the width of the arbi-
4% of the blood cells in an adult. trary round nucleus, noticeable chromatin clumping, and
cytoplasm filled with pinkish (neutrophilic) secondary
a The majority of lymphocytes on an adult blood smear are
granules.
small, have a relatively round nucleus with clumped chro-
matin and a small amount of pale blue cytoplasm; lympho- = Lymphoblasts contain a large, round nucleus with thin,
cytes comprise 20% to 44% of the blood cells in an adult. evenly stained, nonclumped chromatin strands; one or
more nucleoli; and a small amount of blue cytoplasm.
mA monocyte is larger than the mature neutrophil; has
abundant gray blue cytoplasm with fine, reddish or pur- m= Monoblasts are large and demonstrate a large nucleus
plish, evenly distributed granules; and has a nucleus with (sometimes minimally indented), one or two prominent
folds or brain-like convolutions, and lacy, often delicate, nucleoli, fine lacy nuclear chromatin, and nongranular,
chromatin. Monocytes comprise 2% to 9% of blood cells often deep blue cytoplasm.
in an adult. mPlasmacytes are characterized by an _ eccentrically
m Large lymphocytes may reveal a few well defined placed, round, small nucleus with lumpy chromatin;
purplish-red granules that can be easily counted, whereas nongranular deep or vibrant blue cytoplasm; perinuclear
numerous fine indistinct granules that cannot be enumer- clear area; occasional vacuoles; and slightly irregular
ated are present in a monocyte. margins.
m Hematopoiesis is defined as the dynamic processes of pro- a The mature megakaryocyte, the largest of the hematopoi-
duction and development of the various blood and mar- etic cells (range is 30 to 100 um) in the bone marrow, has
row cells. a multilobulated nucleus with coarse linear chromatin
and bluish cytoplasm, containing numerous small, dense,
m Hematopoiesis begins in the mesoderm of the yolk sac
reddish-blue granules, which fragment to form platelets.
and, after 2 months, migrates to the liver and spleen,
Chapter 1 Morphology of Human Blood and Marrow Cells: Hematopoiesis 4]
REFERENCES . Anderson, SC, and Poulsen, KG: Ander- 30. Ringden, OT, et al: Granulocyte and
son’s Atlas of Hematology. Lippincott granulocyte-macrophage colony-
1 . Greer, JP, et al (Eds): Wintrobe’s Clinical
Williams & Wilkins, Baltimore, 2003. stimulating factors in allografts: uses,
Hematology, ed 11. Lippincott Williams
. Colman, RW, et al (Eds): Hemostasis misuses, misconceptions, and future appli-
& Wilkins, Philadelphia, 2004.
and Thrombosis: Basic Principles and cations. Exp Hematol 33(5):505, 2005.
. Diggs, LW, et al: The Morphology of
Clinical Practice. Lippincott, Williams & . Lee, C, et al: Generation of a conditionally
Human Blood Cells, ed 7. Abbott Labo-
Wilkins, Philadelphia, 2001. immortalized myeloid progenitor cell line
ratories, Abbott Park, IL, 2005.
. Boyle, WJ, et al: Osteoclast differentiation requiring the presence of both
. Lichtman, MAT: Williams Hematology,
and activation. Nature 423:337, 2003. interleukin-3 and stem cell factor to sur-
digital edition. McGraw-Hill, New York,
. Aeschlimann, D, and Evans, BAJ: The vive and proliferate. Br JHematol
2006.
vital osteoclast: how is it regulated? Cell 122(6):985, 2003.
. Hoffbrand, AV, et al: Essential Hematol-
Death Differ 11:S5, 2004. . Bunn, HF: New agents that stimulate
ogy. Oxford, England, 2006.
. Von Lindern, M: Cell-cycle control in erythropoiesis. Blood 109:868, 2007.
. Barria, E, et al: Maintenance and self-
erythropoiesis. Blood 108(3):71, 2006. . Zola, H, et al: CD molecules 2005:
renewal of long-term reconstituting
. Kinross, MK, et al: E2f4 regulates fetal human cell differentiation molecules.
hematopoietic stem cells supported by
erythropoiesis through the promotion of Blood 106(9):3123, 2005S.
amniotic fluid. Stem Cells Dev 13(5):548,
cellular proliferation. Blood 108(3):886, . Zola, H: The 8th human leucocyte differ-
2004.
2006. entiation antigens workshop: The evolving
. Bruno, L, et al: Molecular signatures of
. Okubo, T, et al: Self-renewal and differ- role of antibodies in research, diagnosis
self-renewal, differentiation, and lineage
entiation of a novel bipotent myeloid and therapy. J Biol Regul Homeost
choice in multipotential hemopoietic
progenitor clone in the stroma-dependent Agents 19(1—2):72, 2005.
progenitor cells in vitro. Mol Cell Biol
culture. Exp Hematol 28(6):61, 2000. . Salganik, MP, et al: Detecting antibodies
24(2):71, 2004.
. Till, JE, and McCulloch, EA: A direct with similar reactivity patterns in the
—. Bryder, D, and Jacobsen, SE: Interleukin-3
measurement of the radiation sensitivity HLDAS8 blind panel of flow cytometry
supports expansion of long-term multilin-
of normal mouse bone marrow cells. data. J Immunol Methods 305(1):67,
eage repopulating activity after multiple
Radiat Res 14:213, 1961. 2005.
stem cell divisions in vitro. Blood 24. Schuringa, JJ, et al: Enforced activation of . Swart, B, et al: The HLDA8 blind panel:
96(5):1748, 2000.
STATSA facilitates the generation of findings and conclusions. J Immunol
Majumdar, MK, et al: Human marrow-
embryonic stem-derived hematopoietic Methods 305(1):75, 2005S.
derived mesenchymal stem cells (MSCs)
stem cells that contribute to hematopoiesis . Zola, H, and Swart, B: The human leuco-
express hematopoietic cytokines and
in vivo. Stem Cells 22(7):119, 2004. cyte differentiation antigens (HLDA)
support long-term hematopoiesis when
Yo Hess, DA, et al: Functional analysis of workshops: The evolving role of anti-
differentiated toward stromal and human hematopoietic repopulating cells bodies in research, diagnosis and ther-
osteogenic lineages. J Hematother Stem
mobilized with granulocyte colony- apy. Cell Res 15(9):691, 2005.
Cell Res 9(6):84, 2000. stimulating factor alone versus granulo- 3h Bensussan, A, and Olive, D: T-cell:
. de Alarcon, PA, and Werner, EJ (Eds):
cyte colony-stimulating factor in combina- section report. Cell Immunol 236(1—2):35,
Neonatal Hematology. Cambridge tion with stem cell factor. Blood 2005.
University Press, Cambridge, UK, 2005. 100(3):869, 2002. 3) Dasari, P, et al: Expression of T-like
. Erslev, AJ, and Gabuzda, TG: Patho-
26. Ribas, A, et al: Current developments receptors on B lymphocytes. Cell Immunol
physiology of Blood, ed 3. WB Saun- in cancer vaccines and cellular 2005.
ders, Philadelphia, 1995. immunotherapy. J Clin Oncol 40. Vidal-Laliena, M, et al: Characterization
. Stabin, MG, et al: Sensitivity of model- 21(12):2415, 2003. of antibodies submitted to the B cell sec-
based calculations of red marrow dosimetry ale Dudley, ME, et al: Adoptive cell transfer tion of the 8th Human Leukocyte Differ-
to changes in patient-specific parameters. therapy following non-myeloablative but entiation Antigens Workshop by flow
Cancer Biother Radio 17(5):535, 2002. lymphodepleting chemotherapy for the cytometry and immunohistochemistry.
. Beutler, E: Williams Hematology, ed 6. treatment of patients with refractory Cell Immunol 236(1—2):6, 2005.
McGraw-Hill, New York, 2001. metastatic melanoma. J Clin Oncol 4 — . Hillman, RS, et al: Hematology in Clini-
. Wickramasinghe, SN, and McCullough, 23(10):2346, 2005. cal Practice. McGraw-Hill, New York,
J: Blood and Bone Marrow Pathology. 28. Rosenberg, SA: The emergence of mod- 2005.
Churchill Livingstone, New York, 2002. ern cancer immunotherapy. Ann Surg . Zola, H, et al: CD molecules 2005: Hu-
. Surveys & Educational Anatomic Pathol. Oncol 12(5):344, 2005. man cell differentiation molecules.
ogy Programs Hematology Glossary, Col- . Calderwood, S, et al: Recombinant Blood. 2005 Nov 1;106(9):3123-6. Epub
lege of American Pathologists, Northfield, human granulocyte colony-stimulating 2005 Jul 14.
IL, 2002. factor therapy for patients with neutrope-
. Koury, MJ, et al: In vitro maturation of nia and/or neutrophil dysfunction sec-
nascent reticulocytes to erythrocytes. ondary to glycogen storage disease type
Blood 105(5):2168, 2005. 1b. Blood 97(2):376, 2001.
Chapter
Bone Marrow
Aamir Ehsan, MD
Jennifer L. Herrick, MD
Introduction OBJECTIVES
Bone Marrow Structure At the end of this chapter, the learner should be able to:
Erythropoiesis
Granulopoiesis Le Describe the process of hematopoiesis.
Megakaryopoiesis 2. List indications for bone marrow studies.
Lymphopoiesis
Stem Cells 3. Name the most common skeletal sites for hematologic studies.
Hematogones 4. Explain the role of the clinical laboratory scientist during a bone marrow procedure.
Marrow Stromal Cells
Mast Cells 5. Describe the preparation of bone marrow aspirate for laboratory examination.
Bone-Forming Cells 6. Explain the effective use of different bone marrow preparations and specimen
Bone Marrow Function distribution.
Indications for Bone Marrow — . List complications and contraindications of the bone marrow procedure.
Studies
. Explain how to calculate the myeloid-to-erythroid (M:E) cell ratio.
Obtaining and Preparing
Bone Marrow for . State the difference between bone marrow aspirate and biopsy.
Hematologic Studies . Explain how to estimate bone marrow cellularity.
Equipment
Aspiration . List the essential components of a bone marrow report.
Preparation of Bone Marrow
Aspirate
Histologic Marrow Particle
Preparation
Bone Marrow Core Biopsy
Preparation of Trephine
Biopsy
Bone Marrow Examination
Estimation of Bone Marrow
Cellularity
Bone Marrow Differential
Count
Bone Marrow and Peripheral
Blood Interpretation
Based on Cellularity and
M:E Ratio Changes
Bone Marrow Iron Stores
Bone Marrow Report
Case Study 1
Case Study 2
Chapter 2 Bone Marrow 43
Granulopoiesis
Bone Marrow Structure
Granulopoiesis is less conspicuously oriented toward a distinct
The bone marrow is one of the body’s largest organs, repre- reticulum cell, yet may be recognized as a unit* (Fig. 2-3).
senting 3.4 to 6 percent of total body weight and averaging Early granulocytic precursors are located deep in the cords and
>” G @ _G
(BX
sinus (CV). The venous
Figure 2-1 m Graphic presentation of hematopoietic tissue. The vascular compartment consists of arteriole (A) and central
of the
sinusoids are lined by endothelial cells (End), and their wall outside is supported by adventitial-reticulum cells (Adv). Fat tissue (F) is part
(RCP), and erythro-
marrow. The compartmentalization of the hematopoiesis is represented by areas of granulopoiesis (GP), areas of erythropoiesis
poietic islands (El) with their nutrient histiocyte (Hist). The megakaryocytes protrude with small cytoplasmic projections through the vascular
the
wall (Meg). Lymphocytes (Lym) are randomly scattered among the hemapoietic cells, whereas plasma cells (Pla) are usually situated along
vascular wall.
44 Chapter 2. Bone Marrow
Megakaryopoiesis
Megakaryopoiesis occurs adjacent to the sinus endothelium.
The megakaryocytes protrude as small cytoplasmic processes
through the vascular wall, delivering platelets directly into the
sinusoidal blood.’ Megakaryocytes are situated close to marrow
sinuses and, through a mechanism that is not entirely under-
stood, shed platelets into the circulation (Fig. 2-4). Approxi-
mately 5 days are required for the megakaryocytes to produce
platelets, which are released in small groups called proplatelets.
Two-thirds of shed platelets are present in the circulation,
whereas one-third are sequestered in the spleen. The average life
span of platelets in the circulation is approximately 10 days.
Lymphopoiesis
Figure 2-2 ™ Erythropoietic island composed mainly of polychro-
matophilic normoblasts. The nutrient-histiocyte (arrow) is slightly Lymphocytes and plasma cells are minor components of nucle-
displaced off its central position by smearing of the particle. Its cyto-
ated cells in normal bone marrow. Lymphocyte production is
plasmic slender processes envelop a basophilic normoblast, establish-
ing intimate contact with the maturing red cell precursor. compartmentalized in lymphoid follicles, and lymphocytes
(Wright-Giemsa, magnification x 600) are randomly dispersed throughout the cords. The lymphoid
follicles (most often seen in elderly individuals) are unevenly
distributed and tend to influence the variability of the lympho-
around the bone trabeculae. Neutrophils in the marrow can be cyte count in aspirated bone marrow samples (Fig. 2-5).
divided into the proliferating pool and the maturation storage Plasma cells are situated along the vascular wall.
pool. The proliferating pool includes myeloblasts, promyelo-
cytes, and myelocytes. These cells, which spend 3 to 6 days in
this pool, are capable of DNA synthesis and undergo cell divi- Stem Cells
sion. The maturation storage pool consists of nondividing
metamyelocytes, bands, and segmented neutrophils. The cells The marrow stem cells have two unique biologic characteristics:
typically spend 5 to 7 days in this pool before entering into the self-renewal and multilineage differentiation. These stem
circulation. Depending on the demand, the cells from the stor- cells can be further subcategorized as pluripotential stem cells
age pool (representing 15 to 20 times as many cells as in the (which give rise to many different cell lines) and committed
blood) can be released into the peripheral blood, thus increas- stem cells. Because committed stem cells are dedicated by
ing the total white blood cell (WBC) count in minutes or lineage to one cell type and do not have the potential of self-
hours. The average life span of circulating neutrophils is 6 to renewal, they are also called progenitor cells. A progenitor
10 hours. cell is a cell committed to a single line of proliferation and
differentiation. The marrow stem cells on Wright’s stained
smears are morphologically indistinguishable from small
lymphocytes. In the presence of appropriate growth factors,
Figure 2-5 ™A lymphocytic nodule (follicle) in bone marrow as Figure 2-6 m An arrow points toward a hematogone showing high
shown here may alter very significantly the marrow differential nuclear-to-cytoplasmic ratio, homogeneous chromatin, and scant
count when aspirated and give a false impression of lymphocytic cytoplasm. It can be confused with a blast of lymphoblastic
malignancy. (H&E, magnification x200) leukemia. (Wright-Giemsa, magnification 1000)
the stem cells differentiate into myeloid and lymphoid cells Marrow Stromal Cells
that carry tissue and humoral immune functions, respec-
tively. Some lymphoid progenitor cells produced in the mar- The meshwork of stromal cells in which the hematopoietic
row mature in the thymus as T lymphocytes'®; others are cells are suspended is in a delicate semifluid state and is com-
produced and continue their maturation and differentiation posed of reticulum cells, histiocytes, fat cells, and endothelial
in bone marrow as B lymphocytes from the 12th gestational cells. The reticulum cells are associated with fibers that can be
week throughout life.'' Therefore, the bone marrow and thy- visualized after silver staining. They are adjacent to the sinus
mus are primary lymphoid organs of antigen-independent endothelial cells, forming the outer part of the wall as an adven-
progenitor lymphoid cell proliferation and differentiation, titial reticulum cell. Their fine cytoplasmic projections extend
which gives rise to new lymphocytes. These new lympho- deep into the cords, making contact with similar projections of
cytes may then populate the secondary lymphoid organs such other cells. Occasionally, the nuclear region of these cells
as lymph nodes, spleen, and lymphoid apparatus of the gas- can be seen deep in the cords surrounded by granulopoiesis.
trointestinal tract. Under appropriate stimulation, the mature Cytochemically, these cells are alkaline-phosphatase positive
lymphocytes of the peripheral lymphoid organs undergo (Fig. 2-7). Histiocytes or macrophages are seen as perisinu-
antigen-dependent effector cell proliferation, resulting in soidal cells related to the bone marrow—blood barrier, and as
cytokines and antibody production from T and B lympho- central storage macrophages of the erythropoietic islands. As
cytes, respectively. storage nutrient cells that deliver iron to the growing immature
erythroblasts, the storage macrophages send out long, slender
cytoplasmic processes that envelop the erythroid precursors.
This extensive and intimate contact with the maturing ery-
Hematogones
thropoietic cells is necessary in transferring iron from the
Hematogones are normal cellular constituents of bone marrow macrophage to the red cell precursors. As phagocytic cells,
that resemble small- to intermediate-sized lymphocytes. They the macrophages also undergo hyperplasia when there is
range in size from 10 to 20 um and have a high nuclear-to- increased destruction of hematopoietic cells. Histochemically,
cytoplasmic ratio and smooth, smudged homogeneous chro- the macrophages are acid-phosphatase positive (Fig. 2—8).
matin. Their cytoplasm is deeply basophilic and devoid of any The stromal cells produce an extracellular matrix'® com-
granules or vacuoles (Fig. 2-6). Hematogones are thought to posed of collagens, glycoproteins, proteoglycans, and other
be committed progenitor cells of lymphoid lineage. Their proteins. This extracellular matrix is essential in maintaining
numbers are increased in normal infants, older children, and normal renewal and differentiation of marrow cells. The bone
sometimes in adults (in regenerative marrow after chemother- marrow consists of red marrow (hematopoietic active) and
apy and bone marrow transplantation), as well as in marrows fatty yellow marrow (hematopoietic inactive). The marrow
of children with neuroblastoma with or without metastasis, cellularity is estimated as a percentage of red marrow to total
iron-deficiency anemia, and idiopathic thrombocytopenic marrow. The fatty yellow marrow (adipose cells) can vary
purpura.'2!5 These cells closely resemble blasts of lym- in amount according to the age of the individual patient and
phoblastic leukemia, and laboratory personnel should be the skeletal ___location from where the marrow is obtained. In
aware of the conditions in which their numbers may be young children, most of the marrow is composed of red,
increased. hematopoietic active marrow with only a few fat cells present.
46 Chapter 2. Bone Marrow
Figure 2-7 ® An alkaline phosphatase-stained reticulum cell Figure 2-9 @A string of endothelial cells aspirated from hypocellu-
extends its slender cytoplasmic projections deep in the hemopoietic lar marrow. The nuclei are elongated and slightly tapered. The
cord, maintaining an intimate contact with granulopoiesis. The cytoplasm is transparent and barely visible. (Wright-Giemsa,
background cells are stained with neutral red. (magnification x 600) magnification < 600)
The adipose tissue gradually increases after 4 years of age. In oval reticular nucleus and abundant blue-purple granules that
adults, fat cells average about 50% of the total marrow obscure the nucleus. Their granules, in addition to all other
volume in the vertebrae and flat bones of the pelvis. The substances that are present in the granules of basophils, con-
marrow fat and the extracellular matrix are dynamic tissues tain serotonin and proteolytic enzymes. The numbers of mast
similar to the hematopoietic tissue, and these may be altered cells can be increased in chronic infections, autoimmune dis-
rapidly in disease states. eases, chronic lymphoproliferative disorders, and especially
The bone marrow is a highly vascularized tissue from in systemic mastocytosis.!7'*
which endothelial cells can occasionally be aspirated.
Endothelial cells are more visible in hypoplastic marrows and
should not be mistaken for metastatic tumors (Fig. 2-9). Bone-Forming Cells
In marrow aspirates, cells are occasionally seen originating
Mast Cells from bone tissue. Osteoblasts are bone matrix—synthesizing
cells usually found in groups. They are up to 30 um in diame-
Tissue mast cells (Fig. 2-10), 6 to 12 um in diameter, are con- ter and resemble plasma cells. The osteoblast nucleus has a fine
nective tissue cells of mesenchymal origin, normally present chromatin pattern with a prominent nucleolus. A perinuclear
in the bone marrow in varying numbers. They have a round or halo, detached from the nuclear membrane with a cytoplasmic
bridge, represents the Golgi apparatus area. Osteoblasts are limitations. Bone marrow aspiration and bone marrow biopsy
alkaline-phosphatase positive. They are characteristically seen are usually performed concurrently.
in bone marrow aspirates of children and patients with meta- Although obtaining the bone marrow for examination
bolic bone diseases. carries little procedural risk for the patient, the procedure is
Osteoclasts, or bone remodeling cells, are multinucle- costly and can be quite painful. For this reason, bone marrow
ated giant cells more than 100 um in diameter, resembling studies should be performed only when clearly indicated or
megakaryocytes. The nuclei of the osteoclasts are separate whenever the physician expects a beneficial diagnostic result
from each other and may have nucleoli (compared with the for his or her patient (Table 2-1). Hematologic diseases
megakaryocyte nucleus, which is multilobed). Their cyto- affecting primarily the bone marrow and causing a decrease
plasm is well delineated and finely granular. or increase of any cellular blood elements are among the most
common indications. It is not unusual for more than one blood
element to be increased or decreased, as occurs in leukemias
Bone Marrow Function and some refractory anemias. In these situations, bone
marrow study affords specific information, and it usually
The main function of the marrow is to supply mature
precedes any other diagnostic procedure.
hematopoietic cells into the peripheral blood in a steady-
Systemic diseases may affect the bone marrow secondar-
state condition as well as to respond to increased demands. A
ily and require bone marrow studies for diagnosis or monitor-
semidormant pool of pluripotential stem cells maintains a
ing patients’ conditions. Patients having any of the solid
self-renewal property. Granulocytic, monocytic, eosinophilic,
malignant tumors may undergo bone marrow studies when
erythroid, and megakaryocyte progenitor cells are influ-
the initial diagnosis is established for evaluation of the degree
enced in their differentiation by colony-stimulating factors
of tumor spread and staging of the disease. On occasion, a
(CSFs)."°*! CSFs are produced by T lymphocytes—as
bone marrow study may result in a diagnosis of unsuspected
well as stromal cells, fibroblasts, endothelial cells, and
metastatic malignant tumor. During the course of malignant
macrophages—when stimulated by monocyte interleukin- |
disease, additional marrow studies may be performed period-
(IL-1) and tumor necrosis factor (TNF). Some CSFs, such as
ically to monitor the status of tumor burden and its therapeu-
IL-3 and granulocyte-monocyte CSF, have a broad influence
tic response.
and are required throughout proliferation and differentiation
Infections manifesting clinically as “fever of unknown
of progenitor cells. Others, which include granulocyte,
origin” may exhibit granulomas, focal necrosis, or histiocytic
monocyte, and eosinophil CSFs, are lineage specific, and
proliferations. Intracytoplasmic organisms may be seen in the
regulate division and differentiation only of correspond-
marrow. Material for morphologic studies and bacterial cultures
ing, committed progenitor cells. In addition, erythropoiesis
may be collected simultaneously during a single procedure. The
is influenced by erythropoietin produced in the kidney.****
suspected diagnoses of disseminated tuberculosis, fungal infec-
In the process of cell egression from the cords to the circula-
tions (particularly histoplasmosis and cryptococcosis), and
tion, a number of releasing factors are identified. The best
some protozoan infections are frequently confirmed through
characterized of these are granulocyte colony—stimulating fac-
such studies. Hereditary and acquired conditions occasionally
tor (G-CSF) and granulocyte—macrophage colony—stimulating
involve the bone marrow histiocytes (e.g., Gaucher’s disease as
factor (GM-CSF), but other factors, such as components of
shown in Fig. 2-11, sea blue histiocytosis, hemophagocytic
a complement system, androgenic steroids, and endotoxins,
may play a role.** The endothelial lining of the sinusoids
forms a continuous, veil-like wall through which the
mature cells migrate from extravascular sites into the cir-
culation.*> This is accomplished by close contact between
mature hematopoietic cells and endothelial cells. A tran-
sient migration pore is formed during such contact through
which the mature cells pass into the circulation without Hematologic
loss of plasma to the extravascular pool.’° It is evident that
e Anemias, polycythemia
the bone marrow is subjected to a complex regulation by
¢ Unexplained leukopenia or leukocytosis
many cellular and humoral systems of the body, and any ¢ Presence of blasts, immature, or abnormal cells in the
disease that affects these systems is likely to affect circulation
hematopoiesis. ¢ Unexplained thrombocytopenia or thrombocytosis
e Evaluation of plasma cell disorders
Systemic Diseases
Indications for Bone Marrow Studies ¢ Staging and management of solid malignant tumors arising
studies in the elsewhere in the body, such as lymphomas, carcinoma, and
In 1929, Arinkin2’ introduced bone marrow
sarcomas
diagnosis of hematopoietic disorders. Once a formidable ¢ Infections or fever of unknown origin, granulomas
task, obtaining bone marrow tissue has become, with current ¢ Hereditary or acquired metabolic disorders
improved techniques, a standard procedure. Several tech- e Systemic mast cell disease
niques have been devised, each having its own merits and
48 Chapter 2. Bone Marrow
$2 —
ifC i> Sternum
Spinal :
rocesses ([&
P a] \
Posterior i
superior Bp \
iliaccrest (/,
Anterior \\\\\
: id \Y
superior \
iliac crest
Figure 2-1 1 @ Bone marrow biopsy showing a collection of
Gaucher cells. (H&E, magnification 600)
ey; )
syndrome, and others). A simple procedure such as bone
marrow aspiration or biopsy may establish the diagnosis.
Required Materials
. 30-mL syringes
. 20-mL syringes
. 10-mL syringes
. 5-mL syringes
. 2% Lidocaine
. Prepodyne prep
. Alcohol (70%) or prep
. 23-Gauge needles
. 21-Gauge needles
. Bone marrow biopsy/aspiration needle 11-gauge X 4 in.
. Filter papers
. Buffered formalin 10% with a pH of about 6.8 or other
fixative for histologic processing of bone biopsy and
marrow particles
. Tube containing liquid EDTA anticoagulant
. One box of slides
. One slide folder
. One rubber bulb
7, Pasteur pipet Figure 2-13 ™ Adult-sized Jamshidi 11-gauge x 4-inch biopsy/
. Petri dish aspiration needle showing stylet (/eft), biopsy needle (center), and
. Sterile blades probe (right).
. Gloves (several pairs of different sizes)
. Sterile gauze and cotton balls
. Applicator sticks (Xylocaine) is infiltrated into the skin, in the intervening
. Bandage tissues between the skin and bone, and in the periosteum of the
. Culture bottles for bacterial culture. (Note: Save some bone from which the marrow is to be obtained. A cut of about
bone marrow specimen in syringe for tuberculosis and 3 mm is made through the skin with a Bard-Parker blade to
fungal cultures, when indicated.)
facilitate piercing skin and subcutaneous tissue.
. Pencil to label slides
. No. 11 Bard Parker blades The physician penetrates the bone cavity with an aspira-
tion needle, assembled with guard and stylet locked in place.
When the marrow cavity is penetrated, the stylet is removed,
a syringe is attached to the free end of the needle, and the
plunger is quickly pulled, drawing 1.0 to 1.5 mL of marrow
disease transmission, nondisposable bone marrow trays are particles and sinusoidal blood into the syringe. Because the
rarely used today. vacuum created in the syringe is important for rapid and
Several different styles of aspiration and trephine bone efficient suctioning of the cells and particles, the syringe
biopsy needles or instruments are commonly used. Most should be 10 mL or larger with a well-fitting plunger. Despite
instruments used today are patterned on the needle intro- the use of local anesthesia, the patient normally experiences
duced by Jamshidi.** These instruments are produced in discomfort during the aspiration process (aspiration pain).
several sizes for both adult and pediatric patients. An exam- Accomplishing the aspiration with a quick and continuous
ple of the Jamshidi bone marrow biopsy/aspiration needle pull on the plunger diminishes the patient’s discomfort and
for adults is shown in Figure 2-13. Modifications of the decreases the chance of clotting the specimen. A clotted
original aspiration and trephine needles have been devel- specimen is useless for smear preparation because the fibrin
oped by different companies and are manufactured as threads strip the cytoplasm off of the cells and hamper their
disposable equipment. spreading.
Keeping the volume of the initial aspirate small also pre-
vents dilution of the sample with large amounts of sinusoidal
Aspiration
blood, thus improving the quality of the aspirate. This first-
A bone marrow aspiration may be performed as an indepen- aspirated material is used immediately for preparing smears.
dent procedure or in conjunction with a bone marrow biopsy. Additional aspirate may be obtained in separate syringes if
The procedure can be performed in the outpatient setting in needed for flow cytometry, chromosome studies, bacterial
clinics or in the physician’s office. As a rule, children and very cultures, and other tests (Fig. 2-14). Once an adequate aspi-
apprehensive patients receive a mild sedative before the pro- rate is obtained, the quality of the smear depends entirely on
cedure. The site selected is shaved, if needed, and washed with the clinical laboratory scientist’s skill and speed in preparing
soap. Then an antiseptic is applied, and the area is draped with the smears and preserving the morphology of the marrow
sterile towels. A local anesthetic such as 1% to 2% lidocaine cells. Part of the first aspirate is used for the preparation of
50. Chapter 2. Bone Marrow
direct and marrow particle smears. Another portion is placed dropper or Oxford pipette and transferred to a clean glass
in an ethylene diaminetetraacetic acid (EDTA) anticoagulant- slide, which is covered gently with another slide. The two
containing tube for use as a particle preparation. If some aspi- slides are pulled in opposite and parallel directions to smear
rate still remains, it can be left to clot. The clot may be fixed the particles without crushing the cells. Some people recom-
in 10% buffered formalin or another chosen fixative and mend an alternate technique using two cover glasses. In this
processed for histologic examination. The preferred anticoag- process, the marrow particles are squashed between two
ulants for ancillary studies performed on the bone marrow cover slips, which are then gently pulled apart.
aspirate are EDTA for flow cytometry and sodium heparin for Techniques for preparing particle smears vary from person
cytogenetics. If the aspiration attempt is unsuccessful (a “dry to person and from laboratory to laboratory. The aspirate may be
tap”), an additional core biopsy may be obtained (placed in transferred into a watch glass and the particles collected with a
saline or RPMI) for flow cytometric studies. In these situa- capillary pipette or the broken end of a wooden stick applicator.
tions, touch preparations of the core biopsy are useful for With experience, one usually adapts a technique that facilitates
Wright-Giemsa stained morphologic evaluation, as well as production of high-quality slides. The clinical laboratory scien-
cytochemical and fluorescence in situ hybridization (FISH) tist should prepare an adequate number of slides of smeared
studies, if needed. marrow particles. In cases of newly diagnosed acute leukemia,
no fewer than 10 slides should be prepared. These are needed
Preparation of Bone Marrow Aspirate for histochemical stains such as myeloperoxidase, Sudan black
B, naphthyl AS-D chloroacetate esterase, alpha-naphthyl
All necessary materials, preservatives, and slides should be butyrate esterase, and others.
meticulously clean and in readiness to avoid any delay. The Marrow particle smears are used in the evaluation of cel-
aspirate in the first syringe contains mostly blood admixed lularity (usually marrow biopsy is ideal) and the relationship
with fat, marrow cells, and particles of marrow tissue, which of the cells to each other. Well-prepared smears have the
should be used for smears. Several direct smears can be pre- added advantage of excellent cell morphology, allowing
pared immediately, using the technique for blood film prepara- subtle changes in cell maturation and cytoplasmic inclusions
tion.” A small drop is placed on a glass slide, and the blood to be recognized easily
and the particles are dragged behind a spreading slide with a All direct and particle smears should be labeled at the
technique similar to that for preparing blood films. Although bedside with the patient’s name, identification number, and
this method of preparation preserves the cell morphology well, the date and then air dried.
it is inadequate for the evaluation of the cells in relationship to
each other and for the estimation of marrow cellularity. Histologic Marrow Particle Preparation
Smears of marrow particles are prepared by pouring a
small amount of the aspirate on a glass slide. The marrow The leftover marrow particles obtained during the aspiration
tissue is seen as gray particles floating in blood and fat procedure can be processed for histologic examination. The
droplets. The particles are aspirated selectively with a plastic tissue particles, admixed with blood, may be left to clot, then
Figure 2-148 Distribution of bone marrow sample. EDTA = ethylene diaminetetraacetic acid; M:E = myeloid-to-eryt
hroid ratio. (Note:
Fluorescence in situ hybridization (FISH) studies can be performed on aspirate smears, touch preparations, and all
of the tissue sections.)
Chapter 2 Bone Marrow 51
fixed in 10% buffered formalin and processed for histologic sec- Park Memorial Institute) solution. Adequate cell suspensions
tioning. However, better results are obtained if the blood and can then be made from this biopsy sample and processed for
particles are transferred to an EDTA anticoagulant—containing flow cytometric studies.
tube before clotting sets in. The blood and particles are then fil-
tered through histo-wrap filter paper, and the concentrated par-
ticles enfolded in the paper are fixed in 10% buffered formalin. Preparation of Trephine Biopsy
In the histology laboratory, these particles are collected by
TOUCH PREPARATION
scraping the paper and then embedding the particles in paraffin
The bone marrow core biopsy sample is supported lightly
for further processing.*°
without pressure between the blades of forceps and touched
several times on two or three clean slide surfaces. The biopsy
Bone Marrow Core Biopsy core sample should not be rubbed on the slide, because rub-
bing destroys the cells. The slides are air dried. The touch
A bone marrow core biopsy is especially indicated when the preparations are fixed in absolute methanol and stained with
marrow cannot be aspirated (“dry tap”), owing to pathologic Wright-Giemsa stain. In the absence of a good aspirate smear,
alterations encountered in acute leukemias, myelofibrosis, the touch preparations may be the only source for studying
hairy cell leukemia, and other disorders. A trephine bone cellular details and the maturation sequence of the bone
marrow core biopsy is also performed for the diagnosis of marrow biopsy sample. For example, in a case of hairy cell
neoplastic and granulomatous diseases. In multiple myeloma leukemia, dry taps are common and cellular morphology by
and for staging of lymphomas or solid tumors, bilateral pos- Wright-Giemsa stained touch preparation may be a useful
terior superior iliac crest biopsies are recommended, as clue to the diagnosis and allow cytochemical staining proce-
increased sampling size enhances the likelihood of capturing dures, such as tartrate-resistant acid phosphatase (TRAP)
a focal process. An adequate biopsy sample is at least 15 mm (Figs. 2-16 and 2-17). Sometimes the touch preparations
in length.*! contain enough cells to obtain differential counts and blast
When a bone marrow biopsy is performed in conjunction evaluation, and to perform histochemical studies.*”
with a marrow aspiration, customarily the biopsy sample is
obtained after the aspirate. This sequence is usually achieved by HISTOLOGIC BONE MARROW BIOPSY PREPARATION
changing the direction of the needle to avoid the aspiration arti- The biopsy specimen is immersed without delay in B-5 or
fact. However, this technique may result in an aspiration artifact 10% buffered formalin fixative. Histology laboratories may
with hemorrhage into the area of the biopsy site, leading to dif- have a choice of other preferred fixatives such as Zenker’s
ficulties in evaluating cellularity and morphology (Fig. 2-15). solution, Carnoy’s solution, and others.**** After fixation, the
Therefore, a core biopsy sample should be obtained before the biopsy specimen undergoes standard histologic processing of
aspiration or the marrow biopsy procedure is to be performed decalcification, dehydration, embedding in paraffin blocks,
through a new puncture site in the anesthetized area. In some sectioning of 2- to 3-um thick sections, and histologic staining.
cases when flow cytometry is indicated, and the aspirate is dif- The advantage of the bone marrow biopsy is that it represents
ficult to obtain (e.g., in patients with hypercellular marrow, hairy a large sample of marrow and bone structures in their natural
cell leukemia, or marrow fibrosis) an additional core marrow relationships. A variety of different stains can be used to demon-
biopsy may be obtained in normal saline or RPMI (Roswell strate marrow iron, reticulum, and collagen. However, because
Figure 2-17 ™ Tartrate-resistant acid phosphatase (TRAP) stain Figure 2-19 ™ Acid-fast stain on a bone marrow biopsy specimen
showing a strong positive reaction in neoplastic cells which is useful from the same patient as in Figure 2-18 shows acid-fast organisms,
in the diagnosis of hairy cell leukemia. (magnification x 1000) suggesting infection with Mycobacterium. (magnification x 1000)
of decalcification, the core biopsy may not be a good method for Wright-Giemsa stained aspirates or core biopsy touch pre-
studying marrow iron stores, as the processing leaches iron from parations may supply the missing morphologic details
the tissue which may be underrepresented in the iron stain. (Figs. 2-24 and 2-25). Multiple touch preparations also
Acid-fast organisms and fungi in granulomatous diseases may offer an opportunity for histochemical stains (myeloperoxi-
be detected quickly with specific stains, offering great advan- dase, Sudan black B, naphthyl AS-D chloroacetate esterase,
tages in diagnosing these infections (Figs. 2-18, 2-19, and a-naphthy] butyrate esterase, etc.), which are essential in the
2-20). For instance, mycobacterial cultures may require weeks classification of leukemias. Polymerase chain reaction
of incubation to show growth of organisms, whereas on tissue (PCR) and FISH can be performed on paraffin-embedded,
sections, the histologic and etiologic diagnosis may be made formalin-fixed marrow biopsy specimens. PCR technology
within 10 to 12 hours. When metastatic tumors and lymphomas may be used to evaluate B-cell or T-cell lymphomas, various
are found in the bone marrow, immunohistochemical stains can leukemias, and minimal residual disease.
be used on histologic sections to demonstrate specific tumor Trephine bone marrow biopsy specimens may be
markers (Figs. 2-21, 2-22, and 2-23). Thus, a very precise embedded in methyl methacrylate, a synthetic plastic
diagnosis of the origin of a tumor can be made without elabo- medium, and sectioned into | to 2um thin sections without
rate, expensive, and invasive techniques. decalcification. The morphological quality of the cells is
A disadvantage of the bone marrow biopsy is that fine extremely well preserved, and a differential count can be done
cellular details are lost in the processing; therefore, it is of on hematoxylin—-eosin (H&E) or Giemsa-stained _ slides.*
little value in the diagnosis of myelodysplastic syndromes However, this technique requires specially trained personnel,
and subtyping of acute leukemias. In these situations, the equipment, and separate handling in the histology laboratory,
Figure 2-18 ® Bone marrow biopsy specimen from an HIV-positive Figure 2-20 ™ GMS (Gomori’s methenamine silver) stain on bone
patient with tuberculosis shows a well-formed granuloma. (H&E, marrow biopsy specimen from an HIV-positive patient shows multiple
magnification x 200) budding yeasts consistent with histoplasmosis. (magnification 1000)
Chapter 2 Bone Marrow 53
see Seas
oe
eA Ie PRT LS A ;
Bone Marrow Examination
The examination of the bone marrow aspirate smears should
start at low magnification with a dry objective of 10. Scan-
ning the slide permits selection of a suitable area for exami-
nation and the differential count. “Bare nuclei” should be
avoided; such nuclei result from destruction of the marrow
cells by squashing or stripping of their cytoplasm by fibrin
threads. An area is selected in which the cells are well spread,
intact, and not diluted by sinusoidal blood. When marrow par- Figure 2-25 ™ Aspirate smear from a patient with metastatic
ticles are examined, such areas are found at the periphery of prostate carcinoma to the bone marrow. Note the cohesive crowded
groups of large neoplastic cells (A) and giandular formation (B) sug-
gesting adenocarcinoma. (Wright-Giemsa, magnification < 600)
Figure 2-22 ™ Immunohistochemical stain for prostate-specific Figure 2-24 @ H&E stained bone marrow biopsy specimen from a
antigen (PSA) performed on a marrow biopsy specimen from the patient with acute lymphoblastic leukemia shows an increased number
same patient as in Figure 2-21. The specimen shows positive stain- of blasts. Note that the blasts are not cohesive and are individually scat-
ing with PSA, thus confirming that the metastatic tumor is from tered. This is a morphological feature of hematolymphoid malignancy.
prostate. (magnification 600) Fine cellular details are lost in biopsy sections. (magnification x 600)
54 Chapter 2. Bone Marrow
Gee
Sd
Bigs
OF
Yi
sf 2 Steg)
se 52
foe
i
gets&
oad
le
da Masa
— Si ae Ve fad
Figure 2-27 ® Normal bone marrow biopsy specimen from a Figure 2-29 m Hypercellular bone marrow biopsy specimen from
50-year-old patient shows approximately 50% cellularity. (H&E, low a 60-year-old patient with 80% cellular marrow. This patient was
magnification) receiving growth factor therapy. (H&E, low magnification)
.
lymphoid aggregates and malignant lymphoma. Usually benign The fibrosis can be graded as mild, moderate, or severe. In ad-
aggregates are small and well demarcated, nonparatrabecular, dition, a description of the fibrosis as fine or coarse, and
and composed predominantly of small, round lymphocytes with focal or diffuse, may be helpful, especially when following the
plasma cells at the periphery and blood vessels present within improvement of a patient with myelofibrosis after treatment or
the aggregate (see Figs. 2-5 and 2-33). Conversely, malignant bone marrow transplantation. Trichrome stain is usually per-
follicles are usually large with ill-defined borders, paratrabecu- formed to detect any collagenous fibrosis, which if present may
lar, composed of atypical lymphocytes, and lack plasma cells at indicate irreversible fibrosis. Fibrosis and other bone marrow
the periphery (Fig. 2-34). However, a neoplastic lymphoid infil- conditions such as marked hypercellularity in acute leukemias
trate can also be interstitial, diffuse and patchy. In some cases, may result in a “dry tap” when attempting the aspiration proce-
immunohistochemical stains can be performed on the marrow dure. However, if flow cytometry is needed, it is a good practice
core biopsy to differentiate between benign lymphoid aggre- to obtain two bone marrow biopsy samples. One biopsy sample
gates and malignant lymphoma. can be put in formalin and processed for morphological exami-
Bone marrow fibrosis may be found in patients with hairy nation. The other core biopsy sample can be put in saline or
cell leukemia, in myeloproliferative and myelodysplastic syn- RPMI medium; a cell suspension is then made for flow cytomet-
dromes, sometimes in acute leukemia, after radiation, and after ric studies.
toxic injury to the marrow. On routine H&E sections, streaming
of marrow stroma and dilated sinusoids suggests marrow fibro- Bone Marrow Differential Count
sis; this finding can be confirmed and graded by performing reti-
culin and trichrome stains (Fig. 2-35). Normally, occasional A bone marrow differential count is an excellent tool for train-
reticulin-positive fibers may be seen around the blood vessels. ing a novice in bone marrow morphology and is widely used in
2 ‘a
& =
q
:
aN-- ys
Figure 2-28 @ Markedly hypocellular bone marrow biopsy speci- Figure 2-50 ® Bone marrow biopsy specimen showing normal
men from a 20-year-old patient. (H&E, low magnification) bony trabeculae. (H&E, low magnification)
56 Chapter 2 Bone Marrow
*Normal eee ranges on the agian of Texas Health ieee Center and PCa es Hospital, San Antonio, TX.
hemochromatosis, and others), then histologic sections of bone (Fig. 2-36). Hemoglobin iron and dispersed ferritin do not
marrow biopsy sample, and/or marrow aspirate, and marrow stain. Normal marrow iron is seen as fine cytoplasmic gran-
clotted particles are stained for iron. The biopsy sample and the ules within histiocytes, and 30% to 50% of marrow erythrob-
particles are a more reliable source of information, because they lasts contain iron specks within mitochondria and are called
represent a large sample of hematopoietic tissue. Bone marrow sideroblasts. Clumps of iron easily seen at scanning magnifi-
biopsy samples for iron studies should be decalcified by the cation (10) indicate increased iron storage, whereas only a
EDTA chelating method, which does not affect the storage few specks of iron found after searching several microscopic
iron.’ Rapid-acid decalcifying solutions extract iron and must fields (SOX or 100 magnification) indicates decreased iron
not be used in these cases. storage. When no stainable iron is detected on the bone
After Prussian blue staining, hemosiderin and some fer- marrow smear or tissue sections, this indicates iron storage
ritin aggregates are seen as bright blue specks and granules depletion or absence.*” The storage iron may be reported as
” ® i
8. The diagnostic conclusion. This should encompass sepa-
rate diagnoses of blood and bone marrow even where the
~~ same diagnosis is applicable to both; for example: Blood—
oe pancytopenia, Bone marrow, left posterior iliac spine aspi-
" / x * rate and biopsy—myelodysplastic syndrome, refractory
anemia with ringed sideroblasts; or Blood—acute myeloid
er. leukemia minimally differentiated (WHO classification),
‘ i}
‘ Bone marrow, left posterior iliac crest aspirate and
biopsy—acute myeloid leukemia minimally differentiated
ae
hee (WHO classification).
a % 3
ae % : *
In summary, bone marrow can provide a representative pic-
ture of disease processes and has wide application in clini-
cal medicine.*°*! Marrow examination has a significant role
Sao woe A, in the evaluation of leukemias, lymphomas, plasma cell
Figure 2-56 @ Iron stain of a bone marrow smear shows a marked disorders, myeloproliferative disorders, myelodysplastic
increase in marrow storage iron, thus ruling out the possibility of disorders, myelofibrosis, metastatic tumors, various ane-
iron-deficiency anemia. (magnification <x200) mias, granulomatous diseases, infectious diseases, meta-
bolic diseases, in evaluating the status of engraftment after
bone marrow transplantation, and in assessing chemotherapy
99 66
effects. The clinical laboratory scientist’s contribution in
“absent,” “decreased,” “adequate,” “moderately increased,”
this phase consists of preparing the optimum blood and
and “markedly increased,” or it can be given corresponding bone marrow slides and performing the differential count.
numerical values from 0 to 4, where 2 represents the normal Examination of the blood and bone marrow, correlation
or adequate iron stored in an adult. In children; however, iron
with the clinical presentation, and diagnostic conclusions
is stored mainly in ferritin and does not stain with Prussian on each specimen are the responsibility of a physician who
blue in normal iron states. Some chronic derangements in iron has adequate training and experience to integrate all of the
metabolism (i.e., myelodysplastic syndromes) may result in available clinical and laboratory information in reaching
aberrant ringing of sideroblastic iron around erythroblast the correct diagnosis.
nuclei; these cells are named ringed sideroblasts.
DIAGNOSIS
Case Study Ve tonca
Blood:
Acute lymphoblastic leukemia, precursor B-cell
Also received is bone marrow aspirate in one EDTA tube
(circulating blasts 35%)
for flow cytometry and one sodium heparin tube for cytoge-
Normochromic normocytic anemia, moderate
netic studies.
Thrombocytopenia, marked
Bone Marrow, Right Iliac Crest Aspirate Smears, Aspirate
MICROSCOPIC DESCRIPTION Particle Preparation, and Biopsy:
Peripheral Blood Acute lymphoblastic leukemia, precursor B-cell (blasts 65%,
CBC obtained from University Hospital on 02/22/06 cellularity 95%) (see comment).
reveals:
RBEP See 10Z/5 WBC: 20 X 107/L COMMENT
Hgb: 7.5 g/dL Manual differential
Flow cytometry performed on the bone marrow aspirate re-
Het: 22% Segmented neutrophils: 31%
veals a precursor B-cell phenotype. The blasts have the fol-
MCY: 94.0 fL Bands: 1%
lowing phenotype: CD19, CD10, CD34, CD22, HLA-DR
MCH: 28 pg Lymphocytes: 20%
positive; and CD20, kappa (k), lambda (A) light chain neg-
MCHC: 32 g/dL Monocytes: 2%
ative. Other T-cell and myelomonocytic markers are nega-
RDW: 19.0 Eosinophils: 1%
tive. Cytogenetic studies are pending and an amended report
Platelet count Basophils: 1%
will follow. A call was made and results were given to the
OPO 710 Metamyelocytes: 4%
patient’s physician.
Myelocytes: 3%
Promyelocytes: 2%
Blasts: 35%
Anisocytosis: moderate with occasional microcytes and few
macrocytes.
Poikilocytosis: mild with a few teardrop cells and rare schisto-
cytes.
Polychromasia: mild. NRBCs are present 2/100 WBCs Patient Name: Jane Doe
The white blood cells display a left shift with increased Pathology Services, University Health System
number of circulating blasts. The blasts are of small size, show Hospital Number: XXXXXX
minimal variation, and have high nuclear-to-cytoplasmic ratio Surgical Pathology Accession Number: SOO—00000
and fine and lacy chromatin with occasional small nucleoli. No Physician: Dr. Y
Auer rods are seen. History: 65-year-old woman with persistent pancytopenia and
The platelet count is 5.0 < 10°/L. Few giant platelets are normal vitamin B,, and folate levels.
seen. No platelet clumps are noted.
Het: 27% Segmented neutrophils: 55% progressive maturation to the neutrophil stage. Only few
MCV: 102 fL Blasts 2% blasts are present and no Auer rods are seen in their
MCH: 28 pg Lymphocytes: 30% cytoplasm.
MCHC: 32 g/dL Monocytes: 10% An iron stain performed on the aspirate smear shows
RDW: 19.0 Eosinophils: 2% increased storage iron with many ringed sideroblasts.
Platelet count Basophils: 1% Sections from the right iliac crest biopsy and right parti-
10.0 X 10°/L cle preparation reveal hypercellular marrow (cellularity
There is moderate aniso-poikilocytosis with many macro- 90%) with increased erythroid precursors. Aggregates of
cytic red cells and few target cells. Coarse basophilic stip- blasts are not seen. No lymphoid aggregates or granulomata
pling is present. are seen. Bony trabeculae are unremarkable. Reticulin stain
Polychromasia: minimal of the core biopsy did not show increased fibrosis.
The white blood cells display few circulating blasts (2%).
No hypersegmented neutrophils are seen. The platelet count DIAGNOSIS
is 10.0 x 10°/L. Few large and hypogranular platelets are
seen. No platelet clumps are noted. Blood:
Pancytopenia with macrocytic anemia (circulating
Bone Marrow blasts 2%)
Examination of the bone marrow aspirate smear reveals
hypercellular marrow. Differential count performed on aspi- Bone Marrow, Right Iliac Crest Aspirate Smears, Aspirate
rate smears: Particle Preparation, and Biopsy:
Blasts: 1% Pronormoblasts: 12% Myelodysplastic syndrome, refractory anemia with ringed
Promyelocytes: 8% Basophilic normoblasts: 25% sideroblasts (cellularity 90%; blasts 1%) (see comment).
Myelocytes: 9% Polychromatic normoblasts: 16%
Metamyelocytes Orthochromatic COMMENT
and bands: 4% normoblasts: 7%
The patient’s serum B,, and red cell/serum folate levels are
Segmented Monocytes: 4%
normal. The presence of pancytopenia in conjunction with a
neutrophils: 10% Lymphocytes: 4%
hypercellular marrow, red cell dysplasia, and many ringed
The M:E ratio is 0.51. There is an increase in erythroid pre-
sideroblasts is supportive of a diagnosis of a myelodysplastic
cursors with moderate dyserythropoesis characterized by
syndrome, refractory anemia with ringed sideroblasts subtype.
megaloblastoid changes, nuclear irregularity, binucleation,
Preliminary cytogenetic studies performed on the marrow
and nuclear fragmentation. Megakaryocytes are increased.
aspirate reveal clonal chromosomal abnormalities including
Myeloid precursors are decreased in number and show
monosomy 7, del(7q). Results called to Dr. Y at ————.
continued
Cj axenows
1. The average life spans of red cells, neutrophils, and 4. Occasionally, nonhematopoietic cells are seen in marrow
platelets in the circulation are: aspirates. Which of the following is most likely to be
a. 120 days, 7 days, and 10 days misidentified as a megakaryocyte?
b. 30 days, 7 days, and 10 days a. Reticular cell
c. 120 days, 6-10 hours, and 10 days b. Storage histiocyte
d. 30 days, 6-10 hours, and 10 hours c. Osteoclast
. Bone marrow aspirate smears are more informative than
d. Endothelial cell
tw
the bone core biopsy in the diagnosis and classification of: . If the aspirate is unsuccessful (1.e., a “dry tap”), which
a. Granulomatous diseases alternative processing steps are possible?
b. Acute leukemias a. An extra core biopsy can be obtained and placed in
c. Metastatic carcinomas saline or RPMI for flow cytometric studies.
d. Gaucher’s disease b. Touch preparations of the core biopsy can be used for
Wright—Giemsa morphologic evaluation.
3. The least reliable specimen on which to perform an iron
c. Touch preparations of the core biopsy can be used for
stain 1S:
FISH studies and/or cytochemical stains.
a. Core biopsy
d. All of the above.
b. Clot section
c. Aspirate smears
d. Particle preparation
Chapter 2. Bone Marrow
6. In early childhood up to one third of the cellularity can 9, Which is/are true for lymphoid aggregates?
be comprised of the following cells: a. Paratrabecular lymphoid aggregates are usually
a. Erythroblasts malignant.
b. Granulocytic precursors b. Benign aggregates are usually nonparatrabecular.
c. Lymphocytes c. Reactive lymphoid aggregates commonly have a
d. Histiocytes—monocytes blood vessel and plasma cells at the periphery.
. When is a bone marrow study not indicated?
d. All of the above.
a. When circulating blasts are seen on the peripheral smear 10.QO The most appropriate site for bone marrow studies in
b. For pancytopenia adults is:
c. In thrombocytopenia with no apparent cause a. Anterior superior iliac crest
d. In iron deficiency anemia b. Posterior superior iliac crest
. Which is true for hematogones? c. Sternum
a. They are malignant cells. d. Tibia
b. They belong to the granulocytic series.
c. Morphologically they mimic blasts of lymphoblastic See answers at the back of this book.
leukemia.
_d. They are most commonly seen in elderly individuals.
~} SUMMARY CHAR mw Among the most common indicators for bone marrow
studies are diseases that affect the bone marrow, causing a
decrease or increase in any of the cellular blood elements.
a The hematopoietic system consists of bone marrow, liver,
spleen, lymph nodes, and thymus. In normal adults, g In adults, the most common site for bone marrow aspira-
hematopoiesis occurs mainly in the bone marrow. tion or biopsy is the posterior superior iliac crest; in new-
borns and infants, bone marrow is obtained from the
g The bone marrow is one of the body’s largest organs, rep-
upper end of the tibia.
resenting 3.4% to 6% of total body weight and averaging
about 1500 g in adults. g Ina bone marrow aspiration, 1.0 to 1.5 mL of marrow par-
ticles and sinusoidal blood is drawn into a syringe; all
w The structure of the bone marrow consists of hematopoi-
direct smears should be prepared quickly from unclotted
etic cells (erythroid, myeloid, lymphoid, and megakary-
specimens and labeled at the bedside with the patient’s
ocytes), adipose tissue, bone and its cells (osteoblasts and
name, identification number, and date.
osteoclasts), and stroma.
# When marrow cannot be aspirated (“dry tap’’), the com-
w Erythropoiesis takes place in distinct anatomic units
mon differential diagnosis includes acute leukemia,
called erythropoietic islands.
myelofibrosis, and hairy cell leukemia.
mGranulocytic precursors are located deep in the
@ In the estimation of marrow cellularity (ratio of nucleated
hematopoietic cords and around the bone trabeculae.
hematopoietic cells to fat cells) low-power magnification
w Megakaryopoiesis occurs adjacent to the sinus endothelium; is used. Overall marrow cellularity in adults is about
megakaryocytes protrude as small cytoplasmic processes 50% + 10%.
through the vascular wall, delivering platelets directly to the
m= When performing the marrow differential count, at least
sinusoidal blood.
500 to 1000 nucleated cells are classified; the oil immer-
mu Lymphocyte production is compartmentalized in lym- sion objective is used, and megakaryocytes are not
phoid follicles, and lymphocytes are randomly dispersed included in the differential count.
throughout the hematopoietic cords.
m The normal M:E (myeloid-to-erythroid) ratio for adults is
m Hematogones are normal cellular constituents of bone 4:1; granulopoietic tissue occupies a marrow space that is
marrow that resemble small-to-intermediate sized lym- two to four times greater than that occupied by the ery-
phocytes; they range in size from 10 to 20 um, have a high thropoietic precursors.
N:C ratio and deeply basophilic cytoplasm, and are
@ The storage form of iron in the bone marrow is hemosiderin.
devoid of any granules or vacuoles.
On Wright-stained smears, iron appears as brownish-blue
m= The meshwork of stromal cells in which the hematopoi- granules. Decalcification leaches iron from the tissue and
etic cells are suspended is in a delicate semifluid state and may result in an erroneous interpretation of absent iron
is composed of reticulum cells, histiocytes, fat cells, and stores in the core biopsy.
endothelial cells.
Chapter 2 Bone Marrow 63
REFERENCES . Kobayashi, SD, et al: The transient Sul, Brynes, RK, et al: Bone marrow aspira-
i. Bloom, W, and Fawcett, DW: A Text- appearance of small blastoid cells in the tion and trephine biopsy, an approach to
marrow after bone marrow transplanta- a thorough study. Am J Clin Pathol
book of Histology, ed 10. WB Saunders,
tion. Am J Clin Pathol 96:191, 1991. LOTOS OVS:
Philadelphia, 1975, pp 204-232.
16. Gordon, MY: Annotation. Extracellular
2) By. Humphries, JE: Dry tap bone marrow
. Tavassoli, M, and Jossey, JM: Bone
matrix of the marrow microenvironment. aspiration: Clinical significance.
marrow, structure and function.
Br J Haematol 70:1, 1988. Am J Hematol 35:247, 1990.
Alan R. Liss, New York, 1978, p 43.
. Hutchinson, RM: Mastocytosis and WwWWw. Carson, FL: Histotechnology—A Self
. Lichtman, MA: The ultrastructure of the
co-existent non-Hodgkin’s lymphoma Instructional Text. ASCP Press, Chicago,
hemopoietic environment of the marrow:
and myeloproliferative disorders. Leuk 1990, pp 10, 38.
A review. Exp Hematol 9:391, 1981.
Lymphoma 7:29, 1992. . Wilkins, BS, and O’Brien, CJ: Tech-
. Tavassoli, M, and Shaklai, M: Absence of
18. Travis, WD, et al: Systemic mast cell niques for obtaining differential cell
tight junctions in endothelium of marrow
diseases. Analysis of 58 cases and litera- counts from bone marrow trephine
sinuses: Possible significance for marrow
ture review. Medicine 67:345, 1988. biopsy specimens. J Clin Pathol 41:558,
cell egress. Erythropoiesis in bone
. Clark, SC, and Kamen, R: The human 1988.
marrow. Br JHaematol 41:303, 1979.
hematopoietic colony-stimulating . Williams, WJ, and Douglas, NA: Exami-
. Aoki, M, and Tavassoli, M: Dynamics of
factors. Science 236:1229, 1987. nation of the marrow. In Beutler, E, et al
red cell egress from bone marrow after
. Neinhuis, AW: Hematopoietic growth (Eds): Williams Hematology, ed 5.
blood letting. Br JHaematol 49:337,
factors. Biologic complexity and clinical McGraw-Hill, New York, 1995, p 18.
1981.
practice. N Engl J Med 318:916, 1988. . Rosse, C, et al: Bone marrow cell popu-
. Aoki, M, and Tavassoli, M: Red cell
. Ogawa, M: Differentiation and prolifera- lation of normal infants: The predomi-
egress from bone marrow in state of
tion of hematopoietic stem cells. Blood nance of lymphocytes. J Lab Clin Med
transfusion plethora. Exp Hematol 9:231,
81:2844, 1993. SENS), WONT.
1981.
. Erslev, AJ: Humoral regulation of red ove Rubinstein, MA: Aspiration of bone
. Gulati, GL, et al: Structure and function
cell production. Blood 8:349, 1953. marrow from iliac crest comparison of
of the bone marrow and hematopoiesis. iliac crest and sternal bone marrow
. Erslev, AJ: Erythropoietin coming of
Hematol Oncol Clin North Am 2:495, age. N Engl J Med 316:101, 1987. studies. JAMA 137:1821, 1948.
1988. . Hartsock, RJ, et al: Normal variations
. Lichtman, MA, et al: Williams Hematol-
oe). Western, H, and Bainton, DF: Association
ogy, ed 7, McGraw-Hill, New York, 2006 with aging of the amount of hematopoi-
of alkaline-phosphatase-positive reticu- etic issue in bone marrow from anterior
. Becker, RP, and de Bruyn, PPH: The
lum cells in bone marrow with granulo-
transmural passage of blood cells into iliac crest. Am J Clin Pathol 43:326,
cytic precursors. J Exp Med 150:919,
myeloid sinusoids and the entry of 1965.
1979. platelets into the sinusoidal circulation. 3). Lee, GR, et al (Eds): Microcytosis and
. Lichtman, MA, et al: Parasinusoidal
A scanning electron microscopic investi- the anemias associated with impaired
___location of megakaryocytes in marrow: gation. Am J Anat 145:183, 1976. hemoglobin synthesis. In Wintrobe’s
A determinant of platelet release. 26. de Bruyn, P, et al: The migration of blood Clinical Hematology, ed 9, vol I, Lea &
Am J Hematol 4:303, 1978. cells of the bone marrow through the Febiger, Philadelphia, 1993, pp 799-800.
. Claman, HN, et al: Immunocompetence sinusoidal wall. J Morphol 133:417, 1971. A0. Hyun, BH, et al: Bone marrow examina-
of transferred thymus-marrow cell com- . Arinkin, MJ: Intravitale Unter- tion: Techniques and interpretation.
binations. J Immunol 97:828, 1966. suchungsmethodik der Knochenmarks. Hematol Oncol Clin North Am 2:513,
. Hassett, JM: Humoral immunodefi- Folia Haematol (Leipz) 38:233, 1929. 1988.
ciency: A review. Pediatr Ann 16:404, . Jamshidi, K, and Swaim, WR: Bone 4]. De Wolf Peeters, C: Bone marrow
1987. marrow biopsy with unaltered architec- trephine interpretation: Diagnostic utility
. Caldwell, CW, et al: B-cell precursors ture. A new biopsy device. J Lab and potential pitfalls. Histopathology
in normal pediatric bone marrow. Clin Med 77:335, 1971. 18:489, 1991.
Am J Clin Pathol 95:816, 1991. I), Izadi, P, et al: Comparison of buffy coat
. Longacre, TA, et al: Hematogones: A preparation to direct method for the See Bibliography at the back of this book.
multiparameter analysis of bone marrow evaluation and interpretation of bone
precursor cells. Blood 73:432, 1989. marrow aspirates. Am J Hematol
. Van den Doel, LJ, et al: Immunological 43:107, 1993.
phenotype of lymphoid cells in regenerat . Rywlin, AM, et al: A simple technique
ing marrow of children after treatment for for the preparation of bone marrow
acute lymphoblastic leukemia. smears and sections. Am J Clin Pathol
Eur J Haematol 41:170, 1988. 53:389, 1970.
Chapter |
Introduction OBJECTIVES
The Red Blood Cell At the end of this chapter, the learner should be able to:
Membrane
Red Blood Cell Membrane Le List the areas of red cell metabolism that are crucial to normal red cell survival and
Proteins function.
Deformability . Describe the chemical composition of the red cell membrane in terms of percentage
in)
Permeability of lipids, proteins, and carbohydrates.
Red Blood Cell Membrane
Lipids . List the two most important red blood cell (RBC) membrane proteins and describe
their function and the characteristics of deformability and permeability.
Hemoglobin Structure and
Function . List the abnormalities that may lead to a change in RBC structure and name the asso-
Hemoglobin Synthesis ciated RBC morphology.
Abnormal Hemoglobins of
. List the major lipid components of the red cell membrane.
Clinical Importance
Maintenance of Hemoglobin . Define the primary function of hemoglobin.
Function: Active Red Blood . List the criteria for normal hemoglobin synthesis.
Cell Metabolic Pathways
. List the structural components of normal hemoglobin.
Erythrocyte Senescence and
Hemolysis Wa
en
S). Describe hemoglobin function.
(e9
Ney
. List the proteins that carry the following components in circulation: iron, hemoelo-
bin dimers, metheme, and bilirubin.
64
Chapter 3 The Red Blood Cell: Structure and Function 65
Introduction
Three areas of red blood cell (RBC) metabolism are crucial
for normal erythrocyte survival and function: the RBC mem-
brane, hemoglobin structure and function, and metabolic
pathways (Table 3-1). Defects or problems associated with
any of these areas will result in impaired RBC survival. A
thorough working knowledge of these areas of RBC physiol-
ogy will ensure basic understanding of the various complex
erythrocyte functions.
The RBC membrane is highly elastic, responds rapidly to Figure 5-1 ™ Transmission electron microscopy (TEM) of plasma
membrane.
applied stresses of fluid forces, and is capable of undergoing
large membrane extensions without fragmentation.
The RBC membrane represents a highly complex struc- Red Blood Cell Membrane Proteins
ture consisting of a semipermeable lipid bilayer supported by
a meshlike cytoskeleton structure’ (Fig. 3-2). The RBC In early studies the RBC membrane proteins’ nomenclature was
membrane cytoskeleton is a network of proteins on the inner based on their migration rate on sodium dodecyl] sulfate (SDS)
surface of the plasma membrane that is responsible for main- gels and labeled, for example, as bands | through 8.° Today,
taining the shape, stability, and deformability or flexibility of with the use of the high-performance mass spectrometers and
the RBC.' The fluid lipid matrix contains equal amounts of sophisticated data analysis methods, a total of 340 membrane
cholesterol! and phospholipids with a mosaic of proteins proteins and 252 soluble proteins have been identified.° These
interdispersed throughout at various intervals.* The proteins proteins have been categorized on the basis of their subcellular
that extend from the outer surface and traverse the entire localization, protein family they belong to, and their function.°
membrane to the inner cytoplasmic side of the RBC are Of the 340 membrane proteins identified, 31% were
termed integral membrane proteins.* The other major class of classified as integral membrane proteins, 16% as membrane-
RBC membrane proteins, called peripheral membrane pro- associated or bound, 1% as glycosyl phosphatidylinositol
teins, is limited to the cytoplasmic surface of the membrane, (GPI)-anchored, 12% as cytoskeletal, 6% as organelle pro-
which is beneath the lipid bilayer and forms the RBC teins, 12% as cytoplasmic, and 6% as extracellular. For 16%
cytoskeleton.* Both the protein and the lipids are organized of the proteins, the subcellular localization was not available
asymmetrically within the RBC membrane.’ The chemical com- (Table 3-2).
position of the membrane mass is approximately 40% lipids, Many proteins are associated with the outer layer of the
52% proteins, and 8% carbohydrates. ! RBC membrane through a posttranslationally added GPI
anchor.’ The functional significance of this type of protein link-
age is unclear. It is known that this linkage can increase lateral
mobility of proteins migrating to the cell surface and reinsert-
Table - ‘Areas ofRedCell =
if
ing into the RBC membrane.’ It has been postulated that this
linkage may also be involved in cell signaling.’ The membrane-
: _ Metabolism Important. in
associated, GPI-anchored and cytoskeletal proteins are consid-
Normal RBC Survival and ered “peripheral” proteins. Extracellular proteins are outside
n ‘Function : e the RBC (extracellular in origin) but remain associated with the
RBC membrane. Proteins have also been recently categorized
¢ RBC membrane
according to molecular function and biological process.° The
¢ Hemoglobin structure and function
* RBC metabolic pathways majority of the proteins (34%) are involved in binding, or pos-
sess a catalytic activity (29%). Fourteen percent are transporter
66 Chapter 3. The Red Blood Cell: Structure and Function
| = integral proteins
Spectrin P = peripheral proteins
ankyrin-band 3
Phospholipids interaction
Fatty acid
chains Membrane
surface
Lipid
bilayer
Membrane
eS
>. ‘‘
L
cytoskeleton
Adducin Protein 4.1
)Spectrin dimer-dimer
Al Spectrin- | Ankyrin > interaction
Spectrin, Beta chain actin-4.1-adducin *----=-==<==
interaction
Figure 3-2 ® Schematic illustration of red blood cell membrane depicting the composition and arrangement of red cell membrane proteins.
GP-A = glycophorin A; GP-B = glycophorin B; GP-C = glycophorin C. Numbers refer to pattern of migration of sodium dodecyl-polyacrylamide
gel pattern stained with Coomassie brilliant blue. Relations of proteins to each other and to lipids are purely hypothetical positions of the
proteins relative to the inside or outside of the lipid bilayer are accurate. (Note: proteins are not drawn to scale, and are omitted.)
proteins, 9% are signal-transducer proteins, and 7% of the pro- Glycophorin is the principal RBC glycoprotein, repre-
teins are involved in structural activity.° senting approximately 20% of the total membrane protein.°
In terms of biological processes, 51% of the proteins The molecule contains approximately 60% carbohydrate and
identified are involved in cellular processes, 50% in physio- accounts for most of the membrane sialic acid, which gives the
logic, and 11% in regulatory processes. Six percent of the pro- erythrocytes their negative charge. As a result, RBCs repel
teins identified were found to be involved in development of each other as they move through the circulation. Glycophorin,
the RBC and possibly differentiation.° Regulatory mecha- similar to other integral membrane proteins, spans the entire
nisms in the RBC include regulation of cellular and physio- thickness of the lipid bilayer and appears on the external sur-
logic processes as well as enzyme activities. Because RBCs face of the RBC membrane, accounting for the ___location of
differentiate and mature during development, not all proteins many RBC antigens.*” Most of these proteins, as mentioned
are present in their active form.® Degradation of organelles previously, carry RBC antigens and are receptors (such as the
occurs during certain stages of maturation. In addition, during glycophorins) or transport proteins (such as the anion-
the 120-day life span of the mature RBC, chemical and enzy- exchange channel glycoprotein). The anion-exchange channel
matic modifications occur.® comprises approximately 30% of the RBC membrane pro-
Two of the most important protein constituents of the RBC tein.© The plasma membrane envelope is anchored to the RBC
membrane include glycophorin, an integral membrane protein, cytoskeleton network of proteins through tethering sites of in-
and spectrin, a peripheral membrane protein. Table 3—3 lists just tegral (transmembrane) proteins located in the lipid bilayer.*
a few of the integral and peripheral membrane proteins. The condensed fluid lipid bilayer plus integral (transmembrane)
proteins chemically isolates and regulates the cell interior.
The RBC cytoskeleton network of proteins provides rigid
support and stability to the lipid bilayer and is responsible for by a protein kinase present in the membrane, which is energy-
the deformability properties of the RBC membrane. It is spec- dependent, being catalyzed by adenosine triphosphate (ATP).'*
ulated that the anion-exchange channel and the glycophorins A database of all RBC proteins identified to date can be found
play a major role in anchoring the RBC membrane cytoskele- at http://proteins.biochem.mpg.de/RBC.
ton to the lipid bilayer.*'° As a result, lateral mobility of these As mentioned earlier, the normal chemical composition,
integral proteins within the lipid bilayer is relatively restricted.’ structural arrangement, and molecular interactions of the ery-
The major components of the red cell cytoskeleton throcyte membrane are crucial to normal RBC survival. In
include spectrin, ankyrin, actin, adducin, and other cytoskele- addition, they play a critical role in two important RBC char-
tal proteins.'' Spectrin is clearly the most abundant peripheral acteristics: deformability and permeability. Table 3-4 lists
protein of the RBC membrane cytoskeleton, comprising characteristics of some major components of the red cell
approximately 25% to 30% of the total membrane protein and membrane.
75% of the cytoskeleton.!”
Spectrin, a flexible rodlike molecule, is composed of a
Deformability
helix of two polypeptide chains, an alpha (a) chain (molecular
weight 240,000 d) and a beta (8) chain (molecular weight RBC membrane deformability or flexibility is critical not only
225,000 d).'° These chains intertwined side to side form het- to RBC survival as the cell travels through the microvascula-
erodimers, which link together with other aB chains to form ture, but also for its function of oxygen delivery.'° Decreased
(af), tetramers.'* Spectrin is an important factor in RBC mem- cellular deformability and red cell shape changes have been
brane integrity, because it binds with other peripheral proteins recognized as distinguishing features of a number of congen-
such as actin, ankyrin, adducin, and other cytoskeletal proteins to ital or hereditary hemolytic anemias leading to decreased
form a skeletal network of microfilaments on the inner surface of RBC survival in these disorders (see Chaps. 9 and 10).'°'*
the RBC membrane (see Fig. 3-2). These microfilaments It has already been mentioned that a loss of ATP (energy)
strengthen the membrane, protecting the cell from being broken levels leads to a decrease in the phosphorylation of spectrin
by circulatory shear forces, and also control the biconcave shape and, in turn, to a loss of membrane deformability. An accumu-
and deformability of the cell. In addition, the cytoskeletal net- lation or increase in deposition of membrane calcium also
work provides stability to the lipid bilayer interface.’ results, causing an increase in membrane rigidity and loss of
Spectrin complexes tie the RBC cytoskeleton network pliability.'? These cells are at a marked disadvantage when
together and stabilize the spectrin—actin junction.'* The they pass through the small (3- to 5-um diameter) sinusoidal
preservation of the spectrin complexes, and thus the integrity orifices of the spleen, one of the functions of which is
of the RBC membrane, requires phosphorylation of spectrin extravascular sequestration and removal of aged, damaged,
Spectrin* 0.1-um heterodimeric filamentous protein Principal structural element of RBC membrane,
consisting of a 240-kD o chain and a which plays a major role in the RBC cytoskeleton
225-kD B chain constitutes 25%-30% of membrane organization
the mass of membrane proteins
Ankyrin* 210-kD globular protein composed of Primary determinant of mechanical coupling of
1879 amino acids the lipid bilayer to the membrane skeleton
Adducin* Protein doublet of approximately 97 and 103 kD Promotes the association of spectrin with
F-actin in a manner similar to band 4.1
Band 4.1* Composed of 622 amino acids, constitutes Dual functions: promotes high-affinity
5% of the mass of membrane proteins association between spectrin and F-actin and
may link the skeleton to the membrane by virtue of
its associations with glycophorin and band 3
Major RBC transmembrane protein and Functions: catalyzes chloride—bicarbonate exchange;
major integral protein that has two distinct contains binding sites for ankyrin, band 4.1, band 4.2,
domains: the transmembrane and cytoplasmic; and several glycolytic enzymes; plays a role in
911-amino acid protein, comprising 15%-20% recognition and removal of normal senescent RBCs
of the total membrane protein
Band 4.2 Known to associate with the cytoplasmic Actual function unknown: however, a deficiency
___domain of band 3 of the protein is associated with several types of
inherited hemolytic anemias
Hemoglobin Synthesis body iron supply is bound to heme in the hemoglobin mole-
cule (see Chap. 6 for a discussion of iron kinetics).
Normal hemoglobin production is dependent on three processes
(Fig. 3-9): SYNTHESIS OF PROTOPORPHYRINS
1. Adequate iron delivery and supply Protoporphyrin synthesis begins in the mitochondria with the
2. Adequate synthesis of protoporphyrins (the precursor of formation of delta-aminolevulinic acid (SALA) from the amino
heme) and acid, glycine, and succinyl coenzyme A (CoA), which is the
3. Adequate globin synthesis major rate-limiting step in heme biosynthesis (Fig. 3—10).*! The
mitochondrial enzyme, 5ALA synthetase, which mediates this
IRON DELIVERY AND SUPPLY reaction, is influenced by erythropoietin and requires the pres-
Iron, in the ferric state (Fe**), is delivered to the membrane of ence of the cofactor pyridoxal phosphate (vitamin B,).*!
the RBC precursor by the protein carrier transferrin. Most of Porphyrinogens, not porphyrins, are the intermediates of
the iron that crosses the membrane and enters the cytoplasm heme synthesis. Porphyrinogens are unstable tetrapyrroles
of the cell is committed to hemoglobin synthesis and thereby that are readily and irreversibly oxidized to form porphyrins.
proceeds to the mitochondria (where ferric iron is reduced to In contrast, porphyrins are highly stable resonating molecules
ferrous [Fe**] iron for insertion into the protoporphyrin ring to that are normally found in small quantities in the urine as a
form heme). Excess iron in the cytoplasm aggregates as fer- result of normal RBC catabolism.*°
ritin, the amount of which depends on the ratio between the Excessive formation of porphyrins can occur if any one
level of plasma iron and the amount of iron required by the of the normal enzymatic steps in heme synthesis is blocked
erythrocyte for hemoglobin synthesis. Two-thirds of the total and can result in one of a number of metabolic disorders col-
lectively called the porphyrias.**-™ (See Chap. 6.)
GLOBIN SYNTHESIS
Globin chain synthesis occurs on RBC-specific cytoplas-
mic ribosomes, which are initiated from the inheritance of
Vicenaea various structural genes. Each gene results in the formation
of a specific polypeptide chain. Each somatic diploid cell,
Abnormality Be Morpholosy. including the RBC, contains four alpha (a), two zeta (oy
Cholesterol Re nntaton in the RBC” Target cells two beta (B), two delta (5), two epsilon (€), and four gamma
membrane (i.e., liver disease) (y) genes.'° The a and ¢ genes are located on chromosome
Abetalipoproteinemia with Acanthocytes 16, and the B, 5, €, and y genes on chromosome 11
cholesterol accumulation (Fig. 3-11). The resulting gene products formed have been
LCAT deficiency with Hemolysis with red
called a, C, B, 8, € and y globin chains. Throughout embry-
cholesterol accumulation cell fragmentation
Decreased phosphorylated Bite cells and onic and fetal development, activation of the globin genes
spectrin or altered spectrin spherocytes progresses from the ¢ to the a gene and from the € to the y,
5, and B genes.*>
Chapter 3 The Red Blood Cell: Structure and Function 7\
Polyribosomes
Hemoglobin (oB)2
Globin
», Porphobilinogen->
URO > COPRO
Transferrin
Figure 3-9 = Hemoglobin synthesis in the reticulocyte. 8-ALA = delta-aminolevulinic acid; URO = uroporphyrinogen; COPRO = copropor-
phyrinogen; PROTO = protoporphyrin.
HEMOPROTEINS
Tricarboxylic
acid cycle
Succinyl-CoA
+
Glycine
BgPO, ALA-
synthase
i
HOOC-CH,—CH,-C=CH,NH>
6-Aminolevulinate 5 Proto IX
Porphobilinogen CYTOSOL
synthase
COOH IX |p
HOOC CH,
HC CH A HC Koon 5
Uro’gen Uro’gen Uro’gen
eee SE SET ETTTSESS Sa ea ae eee
synthase cosynthase Pp P decarboxylase Pp
ye H HC CH
HoN
P A P M
Uro’gen III Copro’gen III
Figure 3-10 m Synthesis of heme. The heme biosynthetic pathway, showing the distribution of enzymes between the mitochondria and the
cytoplasm. Condensation of glycine and succinyl coenzyme A yields SALA, which is irreversible; two molecules of ALA undergo condensation
by the enzyme ALA dehydrase to yield porphobilinogen (PBG). In the presence of uroporphyrinogen III cosynthase and uroporphyrinogen |
synthase, PBG yields uroporphyrinogen Ill. Uroporphyrinogen Ill undergoes four decarboxylation steps, catalyzed by the enzyme uropor-
phyrinogen decarboxylase, to yield coproporphyrinogen Ill. Coproporphyrinogen Ill is transported from the cytosol into the mitochondria,
where the enzyme coproporphyrinogen oxidase acts on the propionic acid side chains to yield protoporphyrinogen IX. Catalyzed by protopor-
phyrinogen IX oxidase, protoporphyrinogen IX is oxidized to protoporphyrin IX. Protoporphyrin IX combines with ferrous iron to yield heme
(catalyzed by heme synthase). (Intermediates between uroporphyrinogen and coproporphyrinogen, designated by X, remain unidentified.)
BPO, = pyridoxal phosphate. (From Tietz, MW: Textbook of Clinical Chemistry. WB Saunders, Philadelphia, 1986, with permission.)
72 Chapter 3. The Red Blood Cell: Structure and Function
Infant
Time (months)
Oo Gyp
10) Ay2 Figure 3-12 = Changes in globin chain synthesis during fetal devel-
Hb F opment, birth, and infancy. (From Hillman, RF, and Finch, CA: Red
Cell Manual, ed 7. FA Davis, Philadelphia, 1996, p 11, with
Oy So permission.)
Hb A,
On Bo
Hb A In the cytoplasm, each synthesized globin chain links with
heme (ferroprotoporphyrin IX) to form hemoglobin, which
Chromosome 16 Chromosome 11 primarily consists of two a chains, two 6 chains, and four
heme groups (see Fig. 3-8). The structure of the chains and the
Figure 5-1 1 @ Genetic control and formation of human
hemoglobins. resulting hemoglobin molecule is described in the following
four steps:
The € and ¢ chains normally appear only during embry- 1. The primary structure relates to the number and sequence of
onic development (Table 3-6). These two chains, plus the a amino acids constituting each globin chain. The a chains
and y chains, are constituents of embryonic hemoglobins: Hb have 141 amino acids; the non-a chains have 146 amino
Gower | (¢,€,), Hb Gower 2 (a,€,), and Hb Portland (C,7,).*° acids. The sequence of amino acids is different in each chain.
The € and ¢ chains are produced up to approximately 3 months 2. The secondary structure occurs with the twisting of the
after conception. The a chain is always present. Production amino acid chain around an axis in a helical conformation.
of y chains is active from the third month of fetal development 3. The tertiary structure consists of bending the twisted amino
until 1 year postnatally.*° In the fetus, the major hemoglobin is acid chain into a three-dimensional shape resembling an
ayy, (hemoglobin F). The y chains occur as a mixture of two “irregular pretzel.” The polar groups are oriented outward,
types of chains, differing only by one amino acid at position and the nonpolar groups are interior. The heme molecule is
136. G-gamma (“y) contains glycine, whereas A-gamma (“y) nestled in a nonpolar pocket and attached to a proximal his-
has alanine at that position.” The ratio of Sy to “y is approxi- tidine residue.
mately 3:1 at birth and 2:3 by | year of age.'’ By the age of 4. The quaternary structure is the assembling of the four
2 years, hemoglobin F comprises less than 2% of the total three-dimensional chains with their respected heme groups.
hemoglobin. Production of 8 chains rise gradually prenatally The result is a complete, functional hemoglobin molecule.
and reaches adult percentages between 3 and 6 months postna- (Review Fig. 3-8.)
tally.*° Figure 3-12 depicts the time sequence of globin chain
Table 3-6 shows the composition of hemoglobin found dur-
synthesis during fetal development, birth, and infancy.
ing normal human development. The precise order of amino
All normal adult hemoglobins are formed as tetramers
acids is critical to the structure and function of the hemoglo-
consisting of two a chains plus two (non-a) globin chains. Nor-
bin molecule.
mal adult RBCs contain the following types of hemoglobin:
The rate of globin synthesis is directly related to the rate
° 95% to 97% of the hemoglobin is HbA, which consists of of porphyrin synthesis, and vice versa: protoporphyrin synthe-
a8, chains sis is reduced when globin synthesis is impaired. There is, how-
° 2% to 3% of the hemoglobin is HbA,, which consists of a6, ever, no such relationship with iron uptake when either globin
chains or protoporphyrin synthesis is impaired; iron accumulates in
° 1% to 2% of the hemoglobin is HbF (fetal hemoglobin), the RBC cytoplasm as ferritin aggregates. The iron-laden,
which consists of ay, chains nucleated RBC is termed a sideroblast, and the anucleated
Chapter 3. The Red Blood Cell: Structure and Function WES
OXYHEMOGLOBIN
OXYHEMOGLOBIN —>
N oO
~—-- 25% Oo RELEASED
Mean venous
l Oo tension
OooO
|
Pso | Normal Ps9 = 26 to 30 mmHg
Saturation
(%) |
25 DEOXYHEMOGLOBIN
25 { 40 50 TS 100
POs (mmHg)
DEOXYHEMOGLOBIN
TISSUE VENOUS OXYGEN TENSION
Figure 3-14 ® Normal hemoglobin-oxygen dissociation curve. (From Hillman, RF, and Finch, CA: Red Cell Manual, ed 7. FA Davis,
Philadelphia, 1996, p 18, with permission.)
Hemoglobin—oxygen affinity also can be expressed by In addition to the reasons listed previously for shifts in
Ps) values, which designate the PO, at which hemoglobin the curve, inherited abnormalities of the hemoglobin mole-
is 50% saturated with oxygen under standard in vitro con- cule can result in either situation. These abnormalities are
ditions of temperature and pH.” The P;, of normal blood is described by the P;, measurements. Abnormalities in hemo-
26 to 30 mmHg." An increase in P;, represents a decrease globin structure or function can therefore have profound
in hemoglobin—oxygen affinity, or a shift to the right of the effects on the ability of RBCs to provide oxygen to the
oxygen dissociation curve. A decrease in Ps) represents an tissues.
increase in the hemoglobin—oxygen affinity, or a shift to
the left of the hemoglobin—oxygen dissociation curve.
Abnormal Hemoglobins of Clinical
Importance
The hemoglobins previously described, oxyhemoglobin and
ee << Normal reduced hemoglobin, are physiologic hemoglobins because
“Left-shifted” (2 they function in the transport and delivery of oxygen within
~“Right-shifted” the circulation. Abnormal hemoglobins of clinical significance
Abn Hb ‘—
i iat
that are unable to transport or deliver oxygen include the
70 TpH following:
60 Temp
P50 TTemp ° Carboxyhemoglobin
50 4-5---->f- TPs0 ¢ Methemoglobin
P50 ¢ Sulfhemoglobin
PHOSPHOGLUCONATE
PATHWAY
(oxidative)
EMBDEN-MEYERHOF
PATHWAY GSH GSSG
(non-oxidative)
Glucose
ATP
ADP [HK]
Glucose 6-P
NADP NADPH
6-P-Gluconate
[ce]}
G-6-PD
2
[Prk]
Fructose 1,6-diP
METHEMOGLOBIN
REDUCTASE [All
Glyceraldehyde <> DHAP
PATHWAY LUEBERING-RAPAPORT
a Hemoglobin ‘
Methemoglobin [A] (yaon> —[earo]|1,3-diP-Glycerate
PATHWAY
HK Hexokinase
[rex]
Glucose-6-phosphate isomerase ADP 2,3-diP-Glycerate
PFK Phosphofructokinase ATP
A Aldolase 3-P-Glycerate
[row]}
TPI Triose phosphate isomerase
GAPD Glyceraldehyde-3-phosphate dehydrogenase
PGM Phosphoglycerate mutase
2-P-Glycerate
E Enolase
PK
LDH
Pyruvate kinase
Lactic dehydrogenase
DPGM __ Diphosphoglyceromutase
fel!
P-Enolpyruvate
DPGP _ Diphosphoglycerate phosphatase ADP
G-6-PD
6-PGD
Glucose-6-phosphate dehydrogenase
6-Phosphogiuconate dehydrogenase
ATP
[x] Pyruvate
foal
GR Glutathione reductase
GP Glutathione peroxidase NADH
DHAP _ Dihydroxyacetone-P NAD
PGK Phosphoglycerate kinase Lactate
R NADH-methemoglobin reductase
Figure 3-16 @ Red cell metabolism. (From Hillman, RF, and Finch, CA: Red Cell Manual, ed 7. FA Davis, Philadelphia, 1996, p 15, with
permission.)
sulfhydryl groups of hemoglobin, membrane protein, and 2,3-DPG, which is important because of its profound effect
enzymes subsequent to oxidation.) on the affinity of hemoglobin for oxygen.“ Stores of this
The methemoglobin reductase pathway is another impor- organic phosphate can serve as a reserve for additional ATP
tant component of RBC metabolism. Two methemoglobin generation.
reductase systems are important in maintaining heme iron in
the reduced (Fe**, ferrous) functional state.“° Both pathways Erythrocyte Senescence and Hemolysis
depend on the regeneration of reduced pyridine nucleotide and
are referred to as the NADH and NADPH methemoglobin The RBC, a 6- to 8-um biconcave disc (Fig. 3-17), travels
reductase pathways. In the absence of the enzyme methemo- 200 to 300 miles during its 120-day life span. During this
globin reductase and the reducing action of the pyridine time, circulating RBCs undergo the process of senescence or
nucleotide NADH, there is an accumulation of methemoglobin, aging. Various metabolic and physical changes associated
resulting from the conversion of the ferrous iron of heme to the with the aging of RBCs are listed in Table 3-9. Each day
ferric form (Fe**). Methemoglobin is a nonfunctional form of 1% of the old RBCs in circulation are taken out by a system
hemoglobin, having lost oxygen transport capabilities, as the of fixed macrophages in the body known as the reticuloen-
metheme portion cannot combine with oxygen. Normal effi- dothelial system (RES) or the mononuclear phagocytic system
ciency of the methemoglobin reductase pathway is exemplified (MPS). Modifications of the RBC membrane proteins that
by the fact that usually no more than 1% of RBC hemoglobin normally occur during RBC senescence play a role in enhanc-
exists as methemoglobin in the RBCs of healthy individuals." ing the recognition of aging red cells and their removal by
Another important pathway that is crucial to RBC func- phagocytic cells of the MPS. These RBCs are replaced by the
tion is the Leubering—Rapaport shunt. This pathway causes an daily release of 1% of the younger RBCs (reticulocytes) from
extraordinary accumulation of the RBC organic phosphate the bone marrow storage pool. As erythrocytes become older,
Chapter 3. The Red Blood Cell: Structure and Function 77
RETICULOENDOTHELIAL SYSTEM
(spleen and bone marrow) NORMAL
Bilirubin + albumin |
Liver
General
circulation
Figure 3-17 @ Scanning electron micrograph (SEM) of a normal
red cell. Small
intestine
ity. At a certain critical point, the RBCs are no longer able bilirubin
to traverse the microvasculature and are phagocytized by Conjugated = normal
the RES cells. Although RES célls are located in various 0-0.2 mg/100 mL
Unconjugated= normal , Large
organs and throughout the body, those of the spleen, 0.2—0.8 mg/100 mL a intestine
called littoral cells, are the most sensitive detectors of RBC
abnormalities.*’ Urine f j
Several enzyme activities Only 5% to 10% of normal RBC destruction occurs through
ae toned 1G
Density Sialic acid intravascular hemolysis (Fig. 3—19).'° During this process,
Spheroidal shape Deformability RBC breakdown occurs within the lumen of the blood ves-
MCHC MCV sels. The RBC ruptures, releasing hemoglobin directly into
Internal viscosity Phospholipid the bloodstream. The hemoglobin molecule dissociates into
Agglutinability Cholesterol
af dimers and is picked up by the protein carrier, haptoglo-
Na* Kt
Methemoglobin Protoporphyrin bin.** The haptoglobin—hemoglobin complex prevents renal
Oxygen affinity excretion of the hemoglobin and carries the dimers to the
liver cell for further catabolism. The hepatocyte uptake
MCHC= mean cell earecastnconcentration;1 MCV = mean cell
volume.
and processing are identical at this point to the process
Source: Garratty, G: Basic mechanisms of in vivo cell destruction. previously described for extravascular hemolysis (see
In Beil, C (Ed): A Seminar in Immune-Mediated Cell Destruction. Fig. 3-18). Haptoglobin levels, therefore, fall in plasma as
American Association of Blood Banks, Bethesda, MD, 1981, with
permission.
haptoglobin is removed by the hemoglobin—haptoglobin
complex formation. It is estimated that as little as 1 to 2 mL
78 Chapter 3 The Red Blood Cell: Structure and Function
of the RBC intravascular hemolysis can totally deplete sequent degradation. As a result, the methemalbumin circu-
the amount of plasma haptoglobin.** Normally, 50 to lates until additional hemopexin is produced by the liver to
200 mg/dL of plasma haptoglobin is available and repre- serve as the protein carrier. Table 3-10 lists the various
sents the hemoglobin-dimer binding capacity.** As hapto- plasma proteins and the substances they carry. It is this circu-
globin is depleted, unbound hemoglobin dimers appear in lating methemalbumin that imparts a brown tinge to the
the plasma (hemoglobinemia) and are filtered through the plasma or blood. Intravascular hemolysis that occurs as a
kidneys and reabsorbed by the renal tubular cells. The renal result of RBC senescence is so minimal that it is limited to the
tubular uptake capacity is approximately 5 g per day of fil- involvement of only haptoglobin, which is rarely depleted.
tered hemoglobin.** Beyond this level, free hemoglobin Hemoglobinemia and hemoglobinuria, as well as the other
appears in the urine (hemoglobinuria). processes discussed, come into play only with excessive
Hemoglobinuria is always associated with hemoglobine- intravascular hemolysis, which can occur in patients having
mia. A normal plasma hemoglobin level is approximately 2 to various hemolytic anemias (see Chaps. 9 through 14).
5 mg/dL, which is released as a result of intravascular hemoly- This chapter has outlined and described three important
sis.** Depending on the amount of hemolysis and type of hemo- areas of RBC structure and metabolism: the red cell mem-
globin, the plasma may be pink, red, or brown. Likewise, in brane, hemoglobin structure and function, and red cell meta-
hemoglobinuria the urine also may be pink, red, brown, or bolic pathways. An understanding of these aspects of the
black. Two hemoglobin pigments, oxyhemoglobin and methe- RBC is important to appreciating the development and patho-
moglobin, are produced by auto-oxidation of the hemoglobin in genesis of the many forms of inherited and acquired RBC
the urinary tract when the urine is acidic.** Oxyhemoglobin is defects that result in hemolytic anemias.
bright red, and methemoglobin is dark brown. The color of the
urine, therefore, depends on the amount of hemolysis and the
concentration and relative proportions of these two pigments.
Oxyhemoglobin predominates in alkaline urine, and methemo-
globin predominates in acidic urine.**
Hemoglobin that is neither processed by the kidneys nor
bound to haptoglobin is oxidized to methemoglobin, which is
further disassembled as metheme groups are released and glo-
bin degraded. Free metheme is quickly bound by another
transport protein, hemopexin, and is carried to the liver cell to
be catabolized, as previously described. The heme-binding
capacity of hemopexin is approximately 50 to 100 mg/dL;
when this is exceeded, the metheme groups combine with
albumin to form methemalbumin.** Albumin cannot transfer
the metheme across the membrane of the hepatocyte for sub-
INTRAVASCULAR HEMOLYSIS
Globin
Methemalbumin
Hemosiderin Protein
<< Fecal urobilinogen Substance Carried
Hemoglobin
Methemoglobin Transferrin Iron
Haptoglobin Hemoglobin dimers
Figure 3-19 m@ Intravascular hemolysis. (From Hillman, RF, and Hemopexin Metheme
Finch, CA: Red Cell Manual, ed 7. FA Davis, Philadelphia, 1996, Albumin Bilirubin
p 23, with permission.)
Chapter 3. The Red Blood Cell: Structure and Function 79
REFERENCES oo . Tellen, MJ: Erythrocyte adhesion recep- Ny. Lichtman, MA: Williams Hematology,
tors: Blood group antigens and related ed 7. McGraw-Hill, New York, 2006.
fond. Wintrobe, MM: Wintrobe’s Clinical
molecules. Transfus Med Rev 19:32, . Antonelou, MH, et al: Organization of
Hematology, ed 11. Lippincott, Williams
2005. the erythroid cell membrane in a novel
& Wilkins, Philadelphia, 2004.
. Van Kim, CL, et al: Rh proteins: Key case of congenital anemia. Blood Cells
i). Bennett, V: Spectrin-based membrane
structural and functional components of Mol Dis 30(1):43, 2003.
skeleton: A multipotential adaptor
the red cell membrane. Blood Rev . Eber, S, and Lux, SE: Hereditary sphero-
between plasma membrane and
20(2):93, 2006. cytosis—defects in proteins that connect
cytoplasm. Physiol Rev 70:1029, 1990.
. Helms, JB, and Zurzolo, C: Lipids as tar- the membrane skeleton to the lipid
eS). Somerharju, P, et al: Lateral organization
geting signals: Lipid rafts and intracellu- bilayer. Semin Hematol 41(2):118, 2004.
of membrane lipids. The superlattice
lar trafficking. Traffic 5(4):247, 2004. 18. Iolascon, A, et al: Red blood cell mem-
view. Biochem Biophys Acta
. An, X, et al: Identification and functional brane defects. Rev Clin Exp Hematol
1440(1):32, 1999.
characterization of protein 4.1R and Gy) 22. 008%
. Bruce, LJ, et al: A band 3-based macro-
actin-binding sites in erythrocyte beta IS). Nunomura, W, and Takakuwa, Y: Regu-
complex of integral and peripheral pro-
spectrin: Regulation of the interactions lation of protein 4.1R interactions with
teins in the RBC membrane. Blood
by phosphatidylinositol-4,5-bisphos- membrane proteins by Ca** and calmod-
101:4180, 2003.
phate. Biochemistry 44(31):10681, 2005. ulin. Front Biosci 11:1522, 2006.
Nn. Fairbanks, G, et al: Electrophoretic
tO . Kakhniashvili, DG, et al: The human 20. Heinrich, V, et al: Elastic thickness com-
analysis of the major polypeptides of the
erythrocyte proteome: Analysis by ion pressibility of the red cell membrane.
human erythrocytes membrane.
trap mass spectrometry. Mol Cell Biophys J 81(3):1452, 2001.
Biochemistry 10:2606, 1971.
Proteomics 3:501, 2004. . Murray, RK, et al: Harper’s Illustrated
. Pasini, EM, et al: In-depth analysis of
— WW. Murray, RK, et al: Harper’s Illustrated Biochemistry, ed 26. McGraw-Hill, New
the membrane and cytosolic proteome of
Biochemistry, ed 27. McGraw-Hill, New York, 2003.
red blood cells. Blood 108:791, 2006.
York, 2006. . Kargol, A, et al: A study of porous
. Kooyman, DL, et al: In vivo transfer of
14. Mandal, D, et al: Mapping of a spectrin- structure of cellular membranes in
GPI-linked complement restriction
binding ___domain of human erythrocyte human erythrocytes. Cryobiology
factors from erythrocytes to the endothe-
membrane protein 4.2. Biochem J 364 50(3):332, 2005.
lium. Science 269(5220):89, 1995.
(Pt 3):841, 2002.
Chapter 3. The Red Blood Cell: Structure and Function 8]
Zs Wong, P: The behavior of the human ery- SY, Sassa, S: The porphyrias. Photodermatol erythrocytes. Mol Cell Biochem
throcyte as an imperfect osmometer: A Photoimmunol Photomed 18(2):56, 2002. 232(1-2):81, 2002.
hypothesis. J Theor Biol 238(1):167, 2006. oD) Sassa, S, et al: Late-onset porphyrias: 42. Li, DJ, et al: Potential of peroxynitrite to
24. Wandersee, NJ, et al: Erythrocyte adhe- What are they? Cell Mol Biol (Noisy- promote the conversion of oxyhemoglo-
sion is modified by alterations in cellular le-grand) 48(1):97, 2002. bin to methemoglobin. Acta Biochim
tonicity and volume. Br J Haematol . Robbins, SL, and Cotran, RS: Pathologic Biophys Sin (Shanghai) 36(2):87, 2004.
131(3):366, 2005. Basis of Disease, ed 7. WB Saunders, 4 (eS). Gopalachar, AS, et al: Phenazopyridine-
OB, Pal, C, et al: An integrated view of protein Philadelphia, 2006. induced sulfhemoglobinemia. Ann Phar-
evolution. Nat Rev Genet 7(5):337, 2006. . de Alarcon, PA, and Werner, EJ (Eds): macother 39(6):1128, 2005.
26. Nicolas, V, et al: Functional interaction Neonatal Hematology. Cambridge 44. van Wijk, R, and van Solinge, WW: The
between Rh proteins and the spectrin- University Press, Cambridge, UK, 2005. energy-less red blood cell is lost:
based skeleton in erythroid and epithelial . Wahr, JA, et al: Allosteric modification Erythrocyte enzyme abnormalities of
cells. Transfus Clin Biol 13(1-2): of oxygen delivery by hemoglobin. glycolysis. Blood 106(13):4034, 2005.
23-82006. Anesth Analg 92:615, 2001. 45. Jiang, W, et al: Structure and function of
2ie Di Leo, E, et al: Mutations in MTP gene . Squires, JE: Artificial blood. Science glucose-6-phosphate dehydrogenase-
in abeta- and hypobeta-lipoproteinemia. 295(5557):1002, 2002. deficient variants in Chinese population.
Atherosclerosis 180(2):311, 2005. 38. Stowell, CP: Hemoglobin-based oxygen Hum Genet 119(5):463, 2006.
28. Vickaryous, NK, et al: Deletion of carriers: transfusion medicine. Curr Opin 46. Akintowa, DA: Theoretical mechanistic
N-terminal amino acids from human Hematol 9(6):537, 2002. basis of oxidants of methaemoglobin for-
lecithin:cholesterol acyltransferase differen- 39) Dubey, ML, et al: Decreased level of mation. Med Hypotheses 54:312, 2000.
tially affects enzyme activity toward alpha- 2,3-diphosphoglycerate and alteration of 47. Arese, P, et al: Band 3/complement-
and beta-substrate lipoproteins. Biochim structural integrity in erythrocytes mediated recognition and removal of
Biophys Acta 1646(1—2):164, 2003. infected with Plasmodium falciparum in normally senescent and pathological hu-
vag). Leung, GK, et al: A deficiency of micro- vitro. In Molecular and Cellular man erythrocytes. Cell Physiol Biochem
somal triglyceride transfer protein Biochemistry, vol. 246, pp 137-141. 16:133, 2005.
reduces apolipoprotein B secretion. Springer, Netherlands, 2003. 48. Burtis, CA, et al: Tietz Textbook of
J Biol Chem 275(11):7515, 2000. 40. Bradberry, SM: Occupational methaemo- Clinical Chemistry and Molecular
30. Sperelakis, N (Ed): Cell Physiology globinaemia. Mechanisms of production, Diagnostics, ed. 4. Elsevier Saunders,
Sourcebook: A Molecular Approach, ed features, diagnosis and management Philadelphia, 2006.
3. Academic Press, San Diego, 2001. including the use of methylene blue.
3 ome. Akagi, R, et al: Gene defects involving Toxicol Rev 22(1):13, 2003.
delta-aminolaevulinate dehydratase and 4}. Srivastava, S, et al: Methemoglobin re-
coproporphyrinogen oxidase in a por- ductase activity and in vitro sensitivity
phyria patient. Br J Haematol 132(2):237, towards oxidant induced methemoglo-
2006. binemia in Swiss mice and beagle dogs
Chapter
Anemia
Diagnosis and Clinical Considerations
Armand B. Glassman, MD
Classification of Anemia . Calculate the significance of red blood cell indices as related to the diagnosis of anemia.
Hemoglobin and Hematocrit
. Describe the appearance of the peripheral blood smear in various anemias.
Red Blood Cell Indices
Red Blood Cell Indices and . Explain the diagnostic value of the reticulocyte count.
Other Tests
MN
=)
oO
So. List factors to be evaluated in the interpretation of a bone marrow aspirate smear.
OOO
Overview of the Treatment
of Anemias
Other Factors Involved in
Red Blood Cell Production
Tests in the Diagnosis of
Anemia
Hemoglobin
Hematocrit
Red Blood Cell Indices
Peripheral Blood Smear
Reticulocyte Count
Bone Marrow Smear and
Biopsy
Patient Studies of Anemia
Definition of Anemia There are varied types and causes of anemias. Anemia is
often caused by or associated with an underlying disease.
Anemia in its broadest sense is the inability of the circulat- Determining the specific cause of and characterizing an
ing blood pool to supply the tissue with adequate oxygen anemia in an individual is important to the physician so
for proper metabolic function.' Clinically the diagnosis of that he or she can base the appropriate therapy and assess
anemia is made by patient history, physical examination, the prognosis of the disease in that patient. Anemia is usu-
signs and symptoms, and hematologic laboratory findings. ally associated with decreased levels of hemoglobin or a
82
Chapter 4 Anemia: Diagnosis and Clinical Considerations 83
decreased packed red blood cell (RBC) volume, also the geriatric age group, the difference between hemoglobin
known as the hematocrit (Hct). Under rare circumstances, levels of men and women narrows. Hemoglobin levels of
certain abnormal hemoglobins have very strong oxygen- geriatric men usually decrease slightly, and those of post-
binding capacities or oxygen is not released normally menopausal women approach those of men. The reference
to tissue, resulting in all the clinical signs and symptoms ranges used by each laboratory should be obtained, because
of anemia even though the hemoglobin or hematocrit value they best reflect the patient population served.
is normal or even raised. From a practical laboratory stand- Other factors, including geographic elevation, influ-
point, however, the usual diagnostic criterion for anemia ence individual “normal” hemoglobin levels. Persons
is a decreased hemoglobin (Hgb), hematocrit, or RBC living at elevations above 8000 feet may have persistently
count.* increased hemoglobin values secondary to decreased oxy-
Because most patients with anemia have lowered hemo- gen saturation in the ambient atmosphere. Lung diseases
globin levels, the anemia may be classified arbitrarily as may alter oxygen diffusion across the lung alveolar mem-
either moderate (7 to 10 g Hgb/dL) or severe (less than 7 g branes. A compensatory sequel to chronic lung disease
Hgb/dL).* Moderate anemias do not usually produce clinically may result in increased hemoglobin levels (secondary
evident signs or symptoms, especially if the onset is slow. polycythemia).
However, depending on the patient’s age or cardiovascular con- Various diseases and disorders are associated with lower
dition, even moderate amounts of anemia may be associated than usual hemoglobin levels; these include nutritional defi-
with exertional dyspnea (difficulty breathing), light-headedness, ciencies, external or internal blood loss, accelerated destruc-
vertigo, muscle weakness, headache, or general lethargy. tion of RBCs, ineffective or decreased production of RBCs,
Anemia of rapid onset, such as that resulting from gastrointesti- abnormal hemoglobin synthesis, bone marrow replacement
nal hemorrhage, may be associated with significant clinical by infection or tumor, and bone marrow suppression by
symptoms, such as hypotension, tachycardia, and dyspnea. toxins, chemicals, or radiation.*
These symptoms are usually associated with the precipitous
loss of intravascular volume as well as the oxygen-carrying
capacity of the RBCs. Another example of acute massive blood Causesof Anemia
loss is hemorrhage from extensive trauma.
Anemia has many causes (Table 4-2), which can be broadly
classified as nutritional deficiency (e.g., folate or vitamin
Considerations by Age, Sex, and Other B,, deficiencies); blood loss (hemorrhage); accelerated
Factors destruction of RBCs (immune and nonimmune hemolysis);
bone marrow replacement (e.g., by cancer); infection; toxi-
Newborn infants (younger than | week old) have a hemoglo- city; hematopoietic stem cell arrest or damage; and heredi-
bin value of 18 + 4 g/dL as a reference range. At approxi- tary or acquired defect of hemoglobin, RBC enzymes or
mately 6 months of age, the reference range is 12.5 + 1.5 other metabolic abnormalities. Categories may be simplified
g/dL. Childhood levels from the ages of | to 15 years have a to include conditions of increased RBC destruction, abnor-
reference range of approximately 13.0 = 2 g/dL. Adult mal or decreased RBC production, or some combination
hemoglobin reference ranges are approximately 16.0 + 2 thereof. There are many causes and a variety of classifica-
g/dL for men and 14.0 + 2 g/dL for women (Table 4—1). In tions of anemia.
cr
Table 4-1.Reference Range 'Values
for Hemoglobin nee
ee BUY Hemoglobin (g/dL)
- eea2aac r fA
Infants
Blood loss (hemorrhage, surgery, menses)
Newborns (< | week old) 14.0-22.0 Accelerated destruction of RBCs (immune and nonimmune
6 mo old 11.0-14.0 hemolytic)
Children (1-15 years old) 11.0-15.0 Nutritional deficiency (folate, vitamin B,;, iron)
Bone marrow replacement (cancer, infection)
Adults
Infection (viral, bacterial, microbacterial, mycobacterial,
14.0-18.0 parasitic)
WV Ona ae 10;9 Toxicity (hydrocarbons, medications, radiation)
12.0-16.0 Hematopoietic stem cell arrest or damage (aging, cytokines)
WHO 13.3a=) Hereditary or acquired defect (G6PD deficiency,
spherocytosis)
WHO = World Healthh Onnaion Unknown/idiopathic
84 Chapter 4 Anemia: Diagnosis and Clinical Considerations
Significance of Anemia and of new RBCs in response to proper nutrients, vitamins, and
other factors may be evaluated via the reticulocyte count.
Compensatory Mechanisms
Red Blood Cell and Hemoglobin Production
Clinical Diagnosis of Anemia
In a healthy ambulatory person, approximately 1% of the
senescent circulating RBCs are lost daily. Normally, the bone The clinical diagnosis of anemia is made by a combination of
marrow continues to produce RBCs. A laboratory measure of factors, including patient history, physical signs, and changes
this replacement is the reticulocyte count. In healthy people, in the hematologic profile. The signs and symptoms of anemia
reticulocytes, early circulating RBCs containing residual are generally nonspecific, such as fatigue and weakness, and
RNA, account for 0.5% to 2.0% of the circulating RBCs in the may include gastrointestinal symptoms such as nausea, con-
adult. Replacement of RBCs requires a bone marrow with stipation, or diarrhea. The patient may complain of dyspnea
adequate functioning stem cells, normal RBC maturation after a level of exertion that previously had not caused any
processes, and the ability to release mature RBCs from the problems.°°
bone marrow. Proper hemoglobin and RBC production require A patient example may be useful here. A man who had
a variety of nutritional factors, including iron, vitamin B,,, and been able to climb three flights of stairs without difficulty or
folic acid, and normal pathways of hemoglobin synthesis.* significant shortness of breath might report that now he must
Decreased production of hemoglobin and/or RBCs results in a stop after climbing one flight of stairs and is then very short of
hypoproliferative anemia. The role of hemoglobin synthesis in breath. Subsequent information indicates that the patient has
anemias is covered in greater detail in Chapter 6. passed very dark stools (melena) over the past week. Measure-
In severe anemias (less than 7 g Hgb/dL), symptoms of ment of his hemoglobin reveals a level of 8 g/dL. The initial
functional impairment of a number of organ systems may diagnostic impression from the clinical information is that the
be evident. With minimal exercise, the patient’s cardiac and patient’s anemia is secondary to gastrointestinal bleeding.
respiratory rates may increase dramatically. If the anemia is Physical signs of anemia are usually not specific for the
secondary to blood loss and decreased intravascular vol- underlying causative disease. Occasionally, however, the
ume, the patient’s blood pressure may drop significantly underlying diagnosis may be suspected from certain physical
when he or she is raised from the reclining to a sitting or findings. One example would be signs of malnutrition and
standing position. The heart rate will increase in order to neurologic changes with loss of proprioception (position
elevate the cardiac output to keep pace with peripheral sense) and vibration awareness in a patient with vitamin B,,
tissue oxygen demands in the face of a decreased oxygen- deficiency. Another example would be severe pallor, smooth
carrying capacity of the lowered hemoglobin level. Respi- tongue, and an esophageal web in a patient with severe iron-
ratory symptoms, including dyspnea on exertion, may also deficiency anemia.’ Light-skinned patients who are anemic
occur with anemia. may appear to have pale coloration of mucosal membranes,
An interesting compensatory mechanism in response nail beds, and skin. Occasionally, the temperature may be
to anemia is an increase in the 2,3-diphosphoglycerate slightly elevated, particularly in patients having certain types
(2,3-DPG) levels. This compound is a remarkable physiologic of hemolytic anemia. In the presence of anemia, heart mur-
regulator of normal hemoglobin oxygen-carrying capacity murs may be heard; these are sometimes secondary to the
and tissue oxygen delivery. In the presence of 2,3-DPG, cause of the anemia and sometimes related to the increased
hemoglobin can more readily release the oxygen it is carrying cardiac workload required to bring oxygen to the tissues.
to peripheral tissues. This enhanced release occurs regardless Patients with bacterial endocarditis have fever, heart mur-
of pH or blood arterial oxygen level. murs, and anemia. Bacterial endocarditis is a clinical example
A normal individual responds to anemia with elevated in which the damaged infected myocardial valve and heart
levels of erythropoietin (Epo) (see Chap. 1). The Epo level is murmur are related etiologically to the anemia. Prosthetic
sometimes used as an ancillary diagnostic aid in the differen- heart valves, arterial grafts, or disseminated intravascular
tial diagnosis of anemia. Epo is a hormone with molecular coagulation (DIC) can cause a form of mechanical hemolytic
mass of approximately 31,000 daltons. It has a plasma half- anemia known as microangiopathic hemolytic anemia.
life of between 6 and 9 hours and is produced by the peritubu- A broad category of anemias includes those termed
lar complex of the kidney. Epo levels vary as a result of aplastic. Aplastic anemia is defined as pancytopenia result-
altered oxygen tension in the tissues of the kidney. Increased ing from bone marrow production failure. Hypoprolifera-
Epo production occurs when there is a decreased hemoglobin tive anemia can be solely RBC associated. Additional infor-
level, a hemoglobin structural problem in which oxygen is not mation concerning aplastic anemia is found in Chapter 8.
released, or low ambient oxygen tension at high altitude. On In general, there are two types of aplastic anemia. The
the other hand, high oxygen levels in the kidney result in a first type includes congenital and so-called idiopathic
decrease in Epo production. Recombinant Epo is now avail- acquired forms. The second type is secondary or acquired.
able for treatment of certain types of anemias, particularly Causes of secondary aplastic anemia include exposure to
end-stage renal disease, anemia associated with human chemicals (benzene and some other fluorocarbons), therapeu-
immunodeficiency virus, and anemia associated with cancer tic agents (especially cancer chemotherapy agents), infection
and certain other chronic disorders.* Bone marrow production (some types of hepatitis and parvovirus infections), and
Chapter 4 Anemia: Diagnosis and Clinical Considerations 85
ionizing radiation. This chapter deals primarily with the de- concentration (MCHC).'° The MCV is used as an estima-
crease in the RBC and oxyhemoglobin carrying capacity. It tion of the average size of the RBC. It may be calculated by
should be noted that aplastic anemia is a pancytopenia involv- dividing the hematocrit by the number of RBCs or mea-
ing red cells, white cells, and platelets. sured directly using automated cell counters. If the MCV is
within the reference range, the RBCs are considered nor-
mocytic; if less than normal, microcytic; and if greater than
Classification of Anemia normal, macrocytic. Both MCH and MCHC values are used
The individual types of anemias can be classified according to determine the content of hemoglobin in RBCs. Most
to several different criteria. A bone marrow dynamic classifica- automated hematology instruments also provide an RBC
tion is hypoproliferative or accelerated destruction (hemolytic), distribution width (RDW) value. The RDW is an index
or a combination of the two (sometimes called ineffective of size variation and has been used to quantitate the amount
hematopoiesis). Anemias are often classified clinically accord- of anisocytosis seen on a peripheral blood smear. The refer-
ing to their associated causes, such as blood loss, iron deficiency, ence range for RDW is 11.5% to 14.5% for both men
hemolysis, infection, metastatic bone marrow replacement, or and women. The MCV is not dependable when RBCs
nutritional deficiency. Anemias can also be categorized quanti- vary markedly in size. If MCHC is normal, the RBCs are
tatively by their hematocrit, hemoglobin level, blood cell referred to as normochromic; hypochromic RBCs have a
indices (e.g., normochromic, hypochromic [microcytic] or less than normal MCHC; and there are no truly hyper-
“hyperchromic” [macrocytic]), or reticulocyte count.” The chromic RBCs.
clinical laboratory scientist is frequently involved in the quan-
titative measurements that lead to these classifications and in Red Blood Cell Indices and Other Tests
subsequent evaluations.
The RBC indices are accurately calculated by the auto-
mated hematology instrumentation. These instruments pro-
Hemoglobin and Hematocrit vide precise numeric values for hemoglobin levels, the
numbers of RBCs, and the MCV (see Chap. 31). Although
Measurement of the hemoglobin level and packed cell volume
less precise, careful microscopic examination of a periph-
is the usual method ‘of determining whether a patient has
eral blood smear can tell the examiner whether the RBCs
anemia. As discussed earlier, reference ranges may vary by age
are normocytic, microcytic, macrocytic, normochromic, or
and sex (see Table 4-1) as well as state of hydration, patient
hypochromic. A proper specimen is required to obtain accu-
positioning, and local laboratory patient population determina-
rate answers.'!
tions. Hemoglobin assays (see Chap. 31) are based on the spec-
RBC index calculations and reference ranges are:
trophotometric absorbance readings of cyanmethemoglobin
MCV equals Hct (in %) multiplied by 10, divided by
compared with known amounts (a standard curve). Several
RBC count (in millions/uL); reference range: 90 + 10 fL.
companies manufacture automated instruments that include
these determinations as part of a hematologic profile (see Chap. Hct (%) X 10
MCV =
31). The hematocrit, or packed red blood cell volume (PCV), 1s RBC count (millions/uL)
determined by centrifugation of blood of either capillary or
MCH equals Hgb (in g/dL) multiplied by 10, divided by
venous origin or can be calculated on some automated instru-
RBC count (in millions/uL); reference range: 29 + 2 pg.
ments. The reference PCV for adult men varies by institution
(i.e., 47% + 5%). For adult women during the reproductive Nias Hgb (g/dL) x 10
years, the PCV reference range is 42% + 5%. On the basis of RBC count (millions/uL)
hemoglobin or PCV values and the duration of onset, anemias
MCHC equals Hgb (in g/dL) multiplied by 100, divided
may be classified as mild, moderate, or severe; and as either
by Hct (as a percentage); reference range: 34 + 2%.
acute or chronic. The approximate relationship of the hemoglo-
bin level to hematocrit is 1:3, a ratio that may vary with the MCHC —Hgb (g/dL) x 100
cause of the anemia and the effect of that cause on the RBC Het (%)
indices, particularly the mean corpuscular volume (MCV).
Use of the RBC indices in the differential diagnosis of
Microscopic examination of a properly prepared periph-
anemia can provide a general idea as to what is occurring
eral blood smear is a requirement for the clinical and labora-
clinically (Table 4-3). A normocytic normochromic anemia
tory evaluation of anemia. This technique is discussed later may be the result of bone marrow failure, hemolytic anemia,
under the tests in the diagnosis of anemia. Histologic exami- or some subset of either of these conditions.
nations of the bone marrow smear and aspirate are adjuncts to Making the differential diagnosis of bone marrow failure
elucidate the cause of the anemia further.
requires information about RBC production. This information
can be obtained from the reticulocyte count, which indicates
whether there is bone marrow capacity for increased RBC
Red Blood Cell Indices
production. Because RBC destruction may exceed produc-
The RBC indices are the mean corpuscular volume (MCV), tion, the reticulocyte count, in fact, measures effective RBC
mean cell hemoglobin (MCH), and mean cell hemoglobin production. Hemolytic anemia occurs when there is decreased
86 Chapter 4 Anemia: Diagnosis and Clinical Considerations
HEMOGLOBIN / HEMATOCRIT
¢ History of acute
blood loss
¢ Autoimmune
hemolytic anemia
¢ Anemia of chronic
disease
¢ Dyserythropoiesis Normal or high
¢ Anemia of infection
¢ Aplastic anemia
Low iron Normal iron ¢ Congenital
¢ Pernicious ¢ Folate ¢ Myeloproliferative
dyserythropoiesis
anemia malnutrition disease
anemia (CDA)
¢ Severe * GI problem Liver disease
* lron deficiency ¢ Hemoglobin ¢ Bone marrow malnutrition ¢ Liver disease ¢CDA
¢ Anemia of chronic electrophoresis examination for ¢ Some Gl
disease for thalassemias sideroblastic problem
¢ Renal disease anemias
of anticoagulant to blood owing to improper amount of blood RNA, which may be associated with lead poisoning and some
drawn, and reading error—particularly for hematocrit values malignancies. Howell-Jolly bodies (see Chap. 5), which
determined after centrifugation. In our laboratory, the coeffi- are small, round, blue inclusions seen in RBCs, are the result
cient of variation for hematocrit, within the reference range, is of leftover fragments of DNA. Howell—Jolly bodies are often
approximately 2%. With centrifuge hematocrit techniques, the seen in hyposplenism or asplenism, pernicious anemia,
lower hematocrit values are associated with the higher coeffi- and some hemoglobinopathies—-particularly thalassemia.'*"'°
cients of variation. Pappenheimer bodies, which are iron-containing or siderotic
granules, appear as purplish-blue granules with Wright’s stain
and as coarse blue granules with Prussian blue iron stain. The
Red Blood Cell Indices clinical disorders associated with Pappenheimer bodies
include sideroblastic anemia, alcoholism, thalassemia, and
RBC indices were introduced earlier. The MCV is the average
some myelodysplastic syndromes. Nucleated RBCs with iron
volume, expressed in femtoliters (fL), of RBCs, and is mea-
granules are known as sideroblasts, and RBCs containing iron
sured directly or calculated from the hematocrit and RBC
granules but without a nucleus are referred to as siderocytes.
count. The MCH is the content of hemoglobin in the average
RBC. MCH is calculated from the hemoglobin concentration Ringed sideroblasts are those in which more than five granules
and the RBC count. MCHC, the average concentration of
are arranged in a ring around the nucleus of an orthochromatic
normoblast (Fig. 4-3). Ringed sideroblasts are indicative of
hemoglobin in a volume of packed RBCs, is calculated from
the hemoglobin concentration and the hematocrit.'° ineffective erythropoiesis.
RBC indices are readily available from the automated A threadlike blue ring contained entirely within an abnor-
hematology counting devices. In devices in which the MCV mal RBC, which may or may not have a “figure-of-8” and a
is derived from the voltage changes formed during the RBC round or oval configuration, is known as a Cabot ring (see
count and the hemoglobin is measured by spectrophotometric Chap. 5). This is a remnant of the nuclear membrane. This
determination of the cyanmethemoglobin, the values are cal- infrequent finding may be seen in several clinical disorders,
culated as follows: the hematocrit equals the MCV times the including pernicious anemia, lead poisoning, and other severe
anemias. Heinz bodies (Fig. 44) are small, rounded, angular
RBC count, the MCH equals the hemoglobin divided by the
RBC count, and the MCHC equals the hemoglobin divided by inclusions about | um in diameter that are aggregates of dena-
the hematocrit. The reference range for MCV is 80 to 100 fL; tured hemoglobin and are negative when stained with Prussian
for MCH it is 27 to 31 pg; and for MCHC it is 32% to 36%. blue or other iron stains. Heinz bodies can be demonstrated only
In various anemic states, the indices may be altered as by using supravital stains (e.g., crystal violet) and are not visible
follows: in microcytic anemia, from an MCV of less than with the usual Wright’s stain. The clinical disorders that have
80 fL down to a low of approximately 50 fL; from an MCH been associated with Heinz bodies include glucose-6-phosphate
of less than 25 pg to approximately 15 pg; and from an dehydrogenase (G6PD) deficiency after exposure to oxidizing
MCHC of less than 30% to 22%. In the macrocytic anemias, drugs, a variety of unstable hemoglobinopathies, and alpha-
MCV values are usually greater than 100 fL and may be as thalassemia; they can also be seen after splenectomy.
high as, and sometimes even higher, than 120 fL; the MCHC
may be normal or decreased. The MCHC may be increased
only in the presence of spherocytosis, if at all. Reticulocyte Count
The reticulocyte count is useful in determining the response
and potential of the bone marrow. Reticulocytes are
Peripheral Blood Smear
Much information concerning the cause of an anemia can be
determined from a peripheral blood smear. Coexistent neutrope-
nia, thrombocytopenia, and anemia may indicate bone marrow
failure or a lack of a nutritional substance to provide adequate
bone marrow production. The size and shape of the RBCs can
be noted. Alteration in size of the RBCs results in anisocytosis;
alterations in their shape result in poikilocytosis. The hemoglo-
bin (chromatic) content of the RBCs can be inspected visually
on the peripheral smear. In addition, cytologic details on the
peripheral smear may provide clues to the etiology of the ane-
mia, the bone marrow response, or both. The white cells may be
evaluated. For example, excess lobulations of the polymor-
phonuclear leukocytes are seen in the hypersegmented granulo-
cytes of macrocytic anemias (see Chap. 7).
Basophilic stippling in the RBCs may suggest the pres-
ence of increased bone marrow production and reticulocytosis
(Fig. 4-2). It may also indicate that there are remnants of Figure 4-2 @ Reticulocytosis (new methylene blue stain).
Chapter 4 Anemia: Diagnosis and Clinical Considerations 89
Figure 4-5 @ Ringed sideroblast (center) and siderocytes (surround- Figure 4-4™ Heinz bodies. (From Bell, A: Hematology. Listen, Look
ing cells). (From Bell, A: Hematology, Listen, Look and Learn. and Learn, Health and Education Resources, Inc., Bethesda, MD,
Health and Education Resources, Inc., Bethesda, MD, with with permission.)
permission.)
non-nucleated RBCs that still contain RNA. Reticulocytes Bone Marrow Smear and Biopsy
may be visualized after incubation with a variety of so-called
supravital dyes, including crystal violet or brilliant cresyl blue Bone marrow aspiration and biopsy are important diagnostic
(see Chap. 31). RNA is precipitated as a dye-protein complex. tools in the determination of anemia. Bone marrow interpre-
Reticulocytes under normal circumstances lose their RNA a tation and evaluation are covered in Chapter 2. Factors to be
day or so after reaching the bloodstream from the marrow. evaluated in interpretation of a bone marrow aspirate smear
Reticulocyte activity can be expressed as an absolute count, a and biopsy include maturation of the red and white cell series,
production index, or a percentage. The reference values for presence of megakaryocytes, ratio of myeloid to erythroid
reticulocytes range from 0.5% to 2.0% as a percentage of all cells, abundance of iron stores, presence or absence of granu-
RBCs. Because anemia should be accompanied by increased lomas, tumor cells, and overall estimate of bone marrow
bone marrow activity, the reticulocytes should be expected cellularity.
to increase. One may correct the reticulocyte count for the Interpretation of the bone marrow requires a differential
following formula: count of the myeloid, lymphoid, and erythroid series; an iron
stain; and other appropriate techniques, such as immunohisto-
Patient Hct % X Reticulocyte %
Corrected reticulocyte % = Re(crenee ice means) chemical stains if a differential diagnosis of lymphoprolifera-
tive or myeloproliferative disorders is being considered.
Manual reticulocyte counting is associated with poor Other appropriate specific stains may be indicated if metasta-
reproducibility. In the reference range, the coefficient of tic tumor or infection is suspected or being evaluated.'7"'’ The
variation is said to be as great as 50%. Automated reticulocyte other tests that may be performed in the diagnosis of anemias
counting using fluorescent compounds and automated instru- and the chapters in which these tests are discussed are listed
ments results in better reproducibility.”'° in Table 4—S.
Interpretation of the reticulocyte count must take into Anemia has physiologic, functional, and quantitative
account the age and nutritional status of the patient. Nor- parameters that may be related to hemoglobin or hematocrit
mal adults have a reticulocyte count between 0.5% and levels. The differential diagnosis of anemia requires careful
2.0%, or from 24 to 100 X 10° reticulocytes per liter. The consideration of a wide variety of marrow, extramedullary,
newborn infant has a higher reticulocyte count, which falls and interrelating disease states. A large armamentarium of
to the adult range usually by the second or third week of tests is available to aid in the differential diagnosis of the mul-
life. Sources of error in the reticulocyte count include sam- tiple types of anemia. Successful studies of causes of anemia
pling error resulting from counting relatively few reticulo- require broad knowledge of clinical laboratory techniques and
cytes in a large number of erythrocytes. The 95% confi- medicine.
dence level when counting 100 RBCs, where the true
reticulocyte count is 1%, ranges from 0.4% to 1.6%; obvi-
Patient Studies of Anemia
ously, there is a very high coefficient of variation unless
large numbers of RBCs are counted. Usually 1000 cells are Individual patient studies of anemia are addressed in Chapters
counted for a manual reticulocyte count which is in con- 6 to 14.
trast to the automated method that counts 30,000 cells.
90. Chapter 4 Anemia: Diagnosis and Clinical Considerations
*Primary use.
G6PD = glucose-6-phosphate dehydrogenase; PK = pyruvate kinase.
Questions
_1. Which of the following laboratory results would not bea 5 Homi hematocrit measured on automated lemololosy
usual criterion for making a diagnosis of anemia? instruments?
a. Decreased hemoglobin level a. Centrifugation
b. Decreased hematocrit level b. Photometrically
c. Decreased platelet count c. Calculation (MCV X RBC count)
d. Decreased RBC count d. Calculation (MCH x Hgb)
i) . What condition is not a cause of anemia? . A patient has the following results: Hct 26%; Hgb 8
a. Dietary deficiency g/dL; and RBC count 3.5 X 10°/uL. Calculate the RBC
b. Moderate exercise indices—MCV, MCH, and MCHC—and determine the
c. Decreased RBC production classification of the anemia.
d. Increased RBC destruction or loss a. MCV 88 fL; MCH 30 pg; MCHC 33 g/dL; normocytic,
3. Which response represents the most complete and correct normochromic
listing of the most common clinical signs of anemia? b. MCV 101 fL; MCH 33 pg; MCHC 35 g/dL;
a. Fatigue, weakness, dyspnea, pallor macrocytic, normochromic
b. Urticaria, hypertension, inflammation, nausea c. MCV 74 fL; MCH 22 pg; MCHC 31 g/dL; microcytic,
c. Nausea, hypertension, temperature elevation, melena hypochromic
d. Rapid pulse, inflammation, temperature elevation, d. MCV 70 fL; MCH 22 pg; MCHC 38 g/dL; microcytic,
dehydration hyperchromic
4. What is the most commonly accepted method for . Which of the following would not be characteristically
measuring hemoglobin? found on a peripheral blood smear in a case of
a. Conversion of hemoglobin to oxyhemoglobin, anemia?
followed by spectrophotometric measurement a. Anisocytosis and/or poikilocytosis
b. Iron content measured by radioimmunoassay b. Basophilic stippling, Howell—Jolly bodies, and Pap-
technique penheimer bodies
c. Copper sulfate measured by specific gravity c. Cabot rings and Heinz bodies
d. Conversion of hemoglobin to cyanmethemoglobin, d. Dohle bodies and toxic granules
followed by spectrophotometric measurement
Chapter 4 Anemia: Diagnosis and Clinica! Considerations 9]
8. What is the diagnostic value of the reticulocyte count in 9. Which of the following is a lesser factor to be considered
the evaluation of anemia? in the interpretation of a bone marrow aspirate smear?
a. Determines response and potential of the bone a. Maturation of red and white blood cell series
marrow b. M:E ratio
b. Determines compensation mechanisms for anemia c. Type and amount of hemoglobin
c. Determines the corrected RBC count after d. Estimate of bone marrow activity
calculation
d. Determines the potential sampling error for RBC See answers at the back of this book.
count
@ Anemia is usually characterized by decreased RBC count, g The hematocrit (Hct), or packed red cell volume, is deter-
hemoglobin, and/or hematocrit levels. mined by centrifugation of blood of either capillary or
venous origin; it can be calculated by the mean corpuscu-
mw Moderate anemias (7 to 10 g Hgb/dL) that develop slowly
lar volume (MCV) multiplied by the RBC count.
may have few clinical symptoms.
g The ratio between the hemoglobin and hematocrit values
m Severe anemias or those that develop rapidly (e.g., fol-
Sees
lowing acute blood loss) may be associated with dyspnea,
general lethargy, and orthostatic hypotension. mw The hematocrit reference range of adult males is 47% +
5%: for women, it is 42% + 5%
m Hemoglobin levels vary with age and sex. Reference
ranges should be determined and reported for infants, g The MCV is used as an estimation of the average size of
children, and adult men and women. the RBC, and is calculated by the hematocrit multiplied
by 10, divided by the RBC count (reference range: 80 to
m There are many causes of anemia. They include nutri- 100 fL).
tional deficiencies, blood loss, increased destruction or
decreased production of RBCs, infections, toxicity, hered- g The mean corpuscular hemoglobin (MCH) is a measure of
ity, and acquired defects. hemoglobin content and is calculated by the hemoglobin
multiplied by 10, divided by the RBC count (reference
m= Chronic anemia is usually compensated for by increased range: 27 to 31 pg).
levels of 2,3-DPG, which enables enhanced release of
oxygen from hemoglobin. m The mean corpuscular hemoglobin concentration
(MCHC) is derived by hemoglobin multiplied by 100,
m Reference range values for red cell indices can be used to divided by the hematocrit (reference range: 32% to 36%).
aid in the classification and diagnosis of anemias.
w The red cell distribution width (RDW) is an index of size
m Treatment of anemia varies with the cause. Generally, the variation and has been used to quantitate the amount of
causes should be determined before a treatment is instituted. anisocytosis seen on the peripheral blood smear (refer-
gw The cyanmethemoglobin method is the reference method ence range: 11.5% to 14.5%).
for measuring hemoglobin. mAnemias may be classified as normocytic and _nor-
m= Morphological examinations of the peripheral blood mochromic, microcytic and hypochromic, or macrocytic
smear and bone marrow preparations are valuable aids in and normochromic.
the diagnosis of anemias. m The reticulocyte count is useful in determining the
m Clinical symptoms associated with anemia may include response and potential of the bone marrow. Staining tech-
vertigo, light-headedness, muscle weakness, headache, or niques include new methylene blue or brilliant cresyl
dyspnea. blue. Normal adult values are 0.5% to 2.0%. The count
can be determined by automated or manual methods.
m Adult hemoglobin reference ranges are approximately
14 to 18 g/dL for men and 12 to 16 g/dL for women. w The corrected reticulocyte count is determined by patient
packed corpuscular volume (PCV) multiplied by the retic-
m Persons living at elevations above 8000 feet may have
ulocyte count (%), divided by the reference PCV.
persistently increased hemoglobin values secondary to
decreased oxygen saturation in ambient atmosphere.
92 Chapter 4 Anemia: Diagnosis and Clinical Considerations
REFERENCES 8. Kushner, JP: Hypochromic anemias. In IS). Nienhuis, AW: The thalassemias. In
Wyngaarden, JB, et al (Eds): Cecil Text- Wyngaarden, JB, et al (Eds): Cecil Text-
—_ . Beck, WS: Hematology, ed 3. MIT
book of Medicine, ed 19, vol 1. WB Saun- book of Medicine, ed 19, vol 1. WB
Press, Cambridge, MA, 1981, p 16.
ders, Philadelphia, 1992, pp 839-846. Saunders, Philadelphia, 1992,
ie). Reiss, RF: Laboratory diagnosis of
Allen, RH: Megaloblastic anemias. In pp 883-888.
erythroid disorders. In Tilton, RC, et al
Wyngaarden, JB, et al (Eds): Cecil Text- . Forget, BG: Sickle cell anemia and asso-
(Eds): Clinical Laboratory Medicine.
book of Medicine, ed 10, vol 1. WB Saun- ciated hemoglobinopathies. In Wyngaar-
Mosby- Year Book, St. Louis, 1992,
ders, Philadelphia, 1992, pp 846-856. den, JB, et al (Eds): Cecil Textbook of
pp 898-937.
. Savage, RA, et al: The red cell indices: Medicine, ed 19, vol 1. WB Saunders,
. Hoffbrand, AV, et al: Erythropoiesis and
Yesterday, today, and tomorrow, Routine Philadelphia, 1992, pp 888-893.
general aspects of anaemia. In Hoffbrand,
Hematologic Testing. Clin Lab Med . Brunning, RD, and McKenna, RW: Atlas
AY, and Pettit, JE (Eds): Essential
EWS, IWS. of Tumor Pathology. Tumors of the Bone
Haematology, ed 3. Blackwell Scientific,
. Jones, BA, et al: Complete blood count Marrow. Armed Forces Institute of
Oxford, 1993, pp 12-35.
specimen acceptability. A College of Pathology, Washington, DC, 1994,
. Woodson, RD, et al: Introduction to
American Pathologists Q-probes study of p 496,
hemopoiesis. In MacKinney, AA, Jr (Ed):
703 laboratories. Arch Pathol Lab Med . Young, NS: Aplastic anemia and related
Pathophysiology of Blood. John Wiley &
119:203, 1995. bone marrow failure syndromes. In
Sons, New York, 1984, pp 12-15.
2. Benz, EJ: Structure, function, and synthe- Wyngaarden, JB, et al (Eds): Cecil
Nn. Nathan, DG: Hematologic disease. In
sis of the human hemoglobins. In Wyn- Textbook of Medicine, ed 19, vol 1.
Wyngaarden, JB, et al (Eds): Cecil Text-
gaarden, JB, et al (Eds): Cecil Textbook WB Saunders, Philadelphia, 1992,
book of Medicine, ed 19, vol 1. WB
of Medicine, ed 19, vol 1. WB Saunders, pp 831-837.
Saunders, Philadelphia, 1992,
Philadelphia, 1992, pp 872-877. IIS) Ross, DW: Laboratory evaluation of the
pp 817-822.
. Benz, EJ: Classification and basic patho- patient with hematologic disease. In Bick,
ON. Lindenbaum, J: An approach to the ane-
physiology of the hemoglobinopathies. In RL, et al (Eds): Hematology. Clinical and
mias. In Wyngaarden, JB, et al (Eds):
Wyngaarden, JB, et al (Eds): Cecil Text- Laboratory Practice, vol 1. Mosby-Year
Cecil Textbook of Medicine, ed 19, vol
book of Medicine, ed 19, vol 1. WB Saun- Book, St. Louis, 1993, pp 7-16.
1. WB Saunders, Philadelphia, 1992,
ders, Philadelphia, 1992, pp 877-879.
pp 822-831.
. Benz, EJ: Hemoglobinopathies with
. Kushner, JP: Normochromic, normocytic
altered solubility or oxygen affinity. In
anemias. In Wyngaarden, JB, et al (Eds):
Wyngaarden, JB, et al (Eds): Cecil Text-
Cecil Textbook of Medicine, ed 19,
book of Medicine, ed 19, vol 1.
vol 1. WB Saunders, Philadelphia, 1992,
WB Saunders, Philadelphia, 1992,
pp 837-839.
pp 879-883.
Chapter |
Evaluation of Cell
Morphology and
Introduction to Platelet
and White Blood Cell
Morphology
Kathy W. Jones, MS, MT(ASCP), CLS(NCA)
Introduction OBJECTIVES
Examination of the Peripheral At the end of the chapter, the learner should be able to:
Blood Smear
A Define the terms flagged and reflex test as they pertain to automated hematology
The Normal Red Blood Cell
results.
Assessment of Red Cell
Abnormality . Describe the importance of maintaining competency in morphological identification.
Variations in Red Cell 3. List justifications for performing a manual morphology review.
Distribution . List the steps in the performance of a peripheral blood smear examination.
Normal Distribution
Abnormal Distribution . Identify normal red blood cell morphology on a peripheral smear.
Examination of the Peripheral Blood 3. Find an optimal area for the detailed examination and
Smear enumeration of cells.
¢ The RBCs should not quite touch each other.
A blood smear examination may be performed for a variety ¢ There should not be areas containing large amounts of
of reasons. It may be requested by the physician in response broken cells or precipitated stain.
to perceived clinical features or to an abnormality discovered ¢ The RBCs should have a graduated central pallor.
in a previous CBC. The examination may also be initiated by
High-Power (40) Scan
the technologist as a result of an abnormality in the CBC or
in response to a flagged result reported from the hematology — . Determine the WBC estimate.
analyzer. A flagged result is an indication that a particular ° The WBC estimate is performed under high power (400
result has not met established laboratory criteria and must be magnification). WBCs are counted in ten fields and aver-
reviewed. All laboratories should have a documented proto- aged. The estimate is reported according to the values
col for the examination of a laboratory-initiated blood smear given in Table 5-1.
examination. This protocol may be derived from studies per- * The WBC estimate can also be performed using a factor
formed in the facility or may be based on consensus stan- which is based on the fact that each WBC seen in 400x
dards published by nationally recognized organizations. magnification (high power field) is equivalent to
The microscopic examination of a peripheral blood approximately 2000 cells per uL of blood. For example,
smear provides a wealth of information to the clinician. It is if the average number of WBCs counted per high power
used to detect or verify abnormalities and subsequently may field was 5, the WBC estimate would be 5 2000 or
provide the physician with information from which they may 10,000/uL.
be able to make a differential diagnosis. Various forms of 2. Correlate the WBC estimate with the WBC counts per mm?
anemia may actually be diagnosed from abnormal red cell from the automated instruments.
morphology reported on a blood smear examination. The 3. Evaluate the morphology of the WBCs and record
report of abnormal white cell morphology may in fact indi- any abnormalities, such as toxic granulation or Dodhle
cate what additional testing may be required. Abnormal bodies
platelet morphology may detect a platelet function deficiency
Oil Immersion (100) Examination
even when sufficient numbers of platelets have been reported
from the analyzer. = . Perform a 100 WBC differential count.
The examination of the blood smear should include eval- ° Counting should be performed by moving in a zig-zag
uation of the red cell, white cell, and platelet morphology. To manner on the smear (Fig. 5—1).
evaluate the smear thoroughly the technologist should review ¢ All WBCs are to be included until a total of 100 have been
at least 8 to 10 oil immersion fields (OIF). The red cell mor- counted.
phology evaluation should include examination for deviations 2. Evaluate the RBCs for anisocytosis, poikilocytosis,
in size, shape, distribution, concentration of hemoglobin, hypochromasia, polychromasia, and inclusions.
color, and the appearance of inclusions. The white cell mor- 3. Perform a platelet estimate and evaluate platelet morphology.
phology evaluation should consist of differentiation of the ¢ Count the number of platelets in 10 OIFs.
white blood cells and their overall appearance including ¢ Divide by 10.
nuclear abnormalities, cytoplasmic abnormalities, and the ¢ Multiply by 15,000/mm‘* if the slide was prepared by an
presence of abnormal inclusions that may denote a disease automatic slide spinner; multiply by 20,000/mm+ for all
process. Platelet counts should be verified, and in addition the other blood smear preparations.
smear should be reviewed for platelet shape and size abnor- 4. Correct any total WBC count per mm* that has greater than
malities and for clumping. 10 nucleated red blood cells (NRBCs) per 100 WBCs.
When abnormal morphology is identified on the smear, ¢ When performing the WBC differential, do not include
the technologist must determine if the abnormality is possibly NRBCs in your count, but report them as the number of
artifactual and not pathological. For instance, refractile arti- NRBCs per 100 WBCs.
facts may be the result of water contamination and should not ¢ Use the following formula to correct a WBC count:
be confused with red cell inclusions. Echinocytes or crenated
WBC/mm? X 100
cells may also be artifacts if practically every cell in the thin Corrected WBCs/mm? =
100 + No. of NRBCs/100 WBCs
portion of the film has a uniformly spiculed membrane.
The following describes the necessary steps in the exam- The examination of the peripheral blood smear is per-
ination of the peripheral blood smear. formed as part of the hematologic laboratory workup called
Low-Power (10) Scan the CBE.
4000-7000
7000-10,000
10,000-13,000
13,000-18,000
should be in the thin portion of the smear where the red cells
are slightly separated from one another or at most, barely
touching, with no overlap. The thin area should represent at
least one-third of the entire film.* The reviewer should avoid
the thicker portion of the slide where cells are overlapping
and the edges of smear where cells may be artifactually dis-
Percentage of Cells that Differ in Size or Shape from Normal torted in size, shape, and color. An exception is to be made
RBCs when scanning for platelet clumping.
Normal 5%
Slight S%-10%
Se 10%-25%
Abnormal Distribution
2+ 25%-S50%
AGGLUTINATION
338 50%-15%
4+ >715% Agglutination is an aggregation of red cells into random
clusters or masses. Agglutination is the result of an antigen—
Sample Situations antibody reaction within the body, and in cases of autoag-
2+ Microcytes Few schistocytes glutination the reaction is actually with the patient’s own
1+ Macrocytes Few burr cells cells and the patient’s serum or plasma. Such is the case with
1+ Target cells cold antibody syndromes, for example, cold hemagglutina-
3+ Anisocytosis 2+ Poikilocytosis tion disease and paroxysmal cold hemoglobinuria (PCH).
Agglutination occurs at room temperature during sample
preparation and appears as interspersed areas of clumping
Included in this chapter are flowcharts that correlate the throughout the peripheral smear (Fig. 5—4). The use of saline
abnormal morphology with a possible pathology. This scheme will not disperse these agglutinated areas; however, warm-
should enable the learner to more easily associate an abnor- ing the sample to 37°C helps to break up the agglutinins,
mal morphology with the clinical condition. allowing for the possibility of normal slide preparation for
morphology review. The MCHC and MCV from these speci-
mens are usually falsely elevated in response to the agglutinin
Variations in Red Cell Distribution formation. Other forms of autoagglutination may also occur
Normal Distribution spontaneously but are more likely to be seen in connection
with certain hemolytic anemias, atypical pneumonia, staphy-
The area of smear that is reviewed for morphologic abnormal- lococcal infections, and trypanosomiasis. Agglutination is not
ities is of the utmost importance. The area to be reviewed to be confused with rouleaux which is described next.
‘e
(siderotic granules)
1
1
1
Schistocyte Basophilic stippling Rouleaux
1 (fragmented cell) (coarse)
1
9 »-
1
1
1
1
‘
1
4
Oval macrocyte Stomatocyte 1 Tear drop Howell-Jolly
1
1
'
'
'
'
'
'
'
Hypochromic Tr
aN
1
1
'
1
1
1
(Reticulocyte) 1
Variations in Size
Anisocytosis
Any significant variation is size is known as anisocytosis. This
size variation is frequently found in the leukemias and in most
forms of anemia. The severity of the variation should also
correspond to an increased RDW. Anisocytosis results from
abnormal cell development, and typically results from a defi-
ciency in the raw materials (i.e., iron, vitamin B,,, folic acid)
needed to manufacture them or by a congenital defect in the
Figure 5-4 m™ Note the agglutination on the smear from a patient cell’s structure. Cell size may deviate, from measuring smaller
with cold hemagglutinin disease.
than the normal 7 um, to being larger than normal. The terms
ROULEAUX used to describe these abnormalities are microcyte (<6 um) and
Rouleaux is a condition in which red cells appear as stacks of macrocyte (29 wm). These terms are used in conjunction with
coins on the peripheral smear. The stacks may be short or the terms microcytosis and macrocytosis and should also corre-
long, but regardless of the length, the red cells appear stacked late with the red cell indice results. Anisocytosis is graded in
on one another. These stacks are rather evenly dispersed most facilities as 1+ to 4+ (see Table 5—2). When reporting
throughout the smear. Rouleaux formation is the result of anisocytosis, it is important to describe the morphology picture
elevated globulins or fibrinogen in the plasma where the red in terms of microcytosis or macrocytosis, or in cases of a dimor-
cells have been more or less “bathed” in this abnormal plasma phic population there may be the appearance of both (Fig. 5-6).
which gives them a sticky consistency. This lowers the zeta (C)
potential, thus facilitating the stacking effect (Fig. 5-5). The
Normocytes
use of a saline dilution of the serum disperses rouleaux.
Rouleaux formation correlates well with a high erythrocyte The average size of the erythrocyte is indicated by the mea-
sedimentation rate. surement of the MCV, a result generated by the automated
Rouleaux is seen in patients with hyperproteinemias hematology analyzer. The MCV is considered an integral part
such as multiple myeloma and Waldenstrom’s macroglobu- of a CBC. Observation of red cell morphology on the blood
linemia. It may also be seen in chronic inflammatory disor- smear provides a quality control check on the electronic
ders, and some lymphomas. It is important to note that in MCV, as well as the other two red cell indices, mean corpus-
cases of severe rouleaux it may be impossible to evaluate cell cular hemoglobin (MCH) and mean corpuscular hemoglobin
size or shape. concentration (MCHC).’ A “normal” MCV would correspond
Peripheral smears reviewed in the thick portions of the to the MCV reference range (80 to 100 fL for adults). Subse-
smear and entire smears made too thick may appear to exhibit quent review of the blood smear should yield no significant
Pigure 5-6 m Note the different size (anisocytosis) and shape (poikilo-
Figure 5-5 @ Peripheral blood showing marked rouleaux formation. cytosis) of the red cells. Compare the largest (macrocytic) cell below
Note the “stacked coin” appearance of the red cells. the arrow in the center of the field with the smaller (microcytic) cells.
Chapter 5 Evaluation of Cell Morphology and Introduction to Platelet and White Blood Cell Morphology 99
size variation from the normal 7- to 8-um red cell. This sce- excess plasma cholesterol may be taken up by the red cell
nario is referred to as normocytic and the red cells are referred which subsequently leads to an increase in the surface area of
to as normocytes. This information would prove useful to the the cell. However, this last mechanism may not be reflective of
physician in the diagnosis of anemia. In the case of a normal a “true” macrocytosis (obstructive liver disease). Macrocytes
MCV and a high RDW (normal RDW is 11.5% to 14.5%), the should be evaluated for shape (oval versus round) as shown in
reviewer would expect to see a mixture of large and small Figure 5—6, color (red versus blue), pallor (if present), and the
cells.* This scenario is referred to as a dimorphic population presence or absence of inclusions. The conditions in which
and may be the result of a recent blood transfusion, or possi- macrocytes may be seen are listed in Figure 5—7.
bly the patient may be in the recovery stages of anemia.
Patient history plays an important role in this situation.
Microcytes
Macrocytes A microcyte is a small cell having a diameter of less than 7 um
and an MCV of less than 80 fL. Anemias associated with
Macrocytes are cells that are approximately 9 um or larger in microcytes are said to be microcytic. The hemoglobin content
diameter, having an MCV of greater than 100 fL.* Anemias of these cells may be normal to decreased. A consequence of
associated with these cells are referred to as macrocytic. These any defect that results in impaired hemoglobin synthesis may
cells may appear in the peripheral circulation by several mech- produce a microcytic, hypochromic (MCHC <32% and cells
anisms. One mechanism is impaired deoxyribonucleic acid with increased central pallor) blood picture. When erythroid
(DNA) synthesis, which results in megaloblastic erythro- cells are deprived of any of the essential elements in hemoglo-
poiesis leading to a decreased number of cellular divisions, bin synthesis (see Chap. 6), the result is an increase in cellular
and consequently a larger cell. This form of erythropoiesis divisions and consequently a smaller cell in the peripheral
produces a megaloblastic anemia and may be the result of B,, blood. This form of abnormal hemoglobin synthesis is seen in
or folate deficiency, chemotherapy, or any process producing a iron deficiency, deficiency of heme synthesis (sideroblastic
nuclear maturation defect. Macrocytes with an oval shape anemia), deficiency of globin synthesis (thalassemia), and
(macroovalocytes), neutrophilic hypersegmentation, as well as chronic disease states. In the case of iron deficiency, microcy-
MCV values exceeding 120 fL are typically seen in this type tosis will not be visually apparent until iron stores in the body
of anemia. have been completely exhausted and iron deficient erythro-
The most common cause of nonmegaloblastic macrocyto- poiesis takes place, as in iron deficiency anemia (IDA). Iron
sis is accelerated erythropoiesis which results from conditions deficiency is the most common cause of anemia, affecting
such as acute blood loss or alcoholism. The cells are released some 30% of the world population and accounting for up to
prematurely from the marrow, are non-nucleated, and appear 500 million cases worldwide, which also makes it the most
larger than a mature erythrocyte. On a Wright-stained smear the common microcytic/hypochromic anemia in the world.° It is
cells will appear as round polychromatophilic macrocytes and especially common in women of childbearing age. The causes
on a supravitally (i.e., new methylene blue) stained smear they of this deficiency may vary depending on the age and sex of the
appear as reticulocytes. Neutrophilic hypersegmentation is not patient and it is important to determine what instigated the
typically seen in this form of macrocytosis.° deficiency before treatment begins.
Macrocytosis may result from other conditions, such as Decreased or defective globin synthesis also presents
hypothyroidism and various bone marrow disorders, as well as as a microcytic/hypochromic anemia, but in most cases this
occur in neonatal blood, postsplenectomy, and in cases where results from a genetic abnormality producing a hereditary
MORPHOLOGY - MACROCYTE
Hemoglobin Content—Color Variations Figure 5-9® Note the large central pallor in many of the red cells
depicting hypochromia.
Normochromia
The term normochromic indicates the red cell is essentially red cells with increased central pallor or hypochromia. A lower
normal in color. A normochromic erythrocyte has a well MCHC result typically correlates with a larger central pallor in
hemoglobinized cytoplasm with a small but distinct zone of the affected red cells. In general, this is very reliable; however,
central pallor. The area of pallor does not exceed 3 um when it does not take into account the situation in which a true
measured linearly. In a well stained peripheral smear, the nor- hypochromia is observed in the presence of a normal MCHC.
mal red cells will appear reddish-orange in color. The term In many cases, the MCHC will not be concordant with what is
normochromic is used to describe an anemia with a normal observed on the peripheral smear. The morphologist should not
MCHC and MCH. When used in conjunction with a normal be unduly influenced by the RBC indices in the evaluation of
MCV, the anemia would be described as a normochromic/ hypochromia. True hypochromia will appear as a delicate
normocytic anemia (see Fig. 5—2). shaded area of pallor as opposed to pseudohypochromia (the
water artifact), in which the area of pallor is distinctly outlined.
Hypochromia It is important to note that not all hypochromic cells are micro-
cytic. Target cells possess some degree of hypochromia, and
Any RBC having a central area of pallor of greater than 3 um is there are macrocytes and normocytes that can be distinctly
said to be hypochromic. There is a direct relationship between hypochromic.
the amount of hemoglobin deposited in the red cell and the The most common condition manifesting hypochromia
appearance of the red cell when properly stained. The term is IDA. In severe cases of IDA, red cells exhibit an inordi-
hypochromia literally means “low color” and indicates that the nately thin band of hemoglobin. Patients with iron defi-
cells have less than the normal amount of hemoglobin. Typically ciency may have many hypochromic cells, depending on the
any irregularity in hemoglobin synthesis will lead to some magnitude of the deficiency. In addition to large numbers of
degree of hypochromia (Fig. 5—9). hypochromic cells, there may be large numbers of micro-
Most clinicians choose to assess hypochromia based on cytes as well. Iron deficiency anemia is commonly referred
the mean corpuscular hemoglobin concentration (MCHC), to as a microcytic/hypochromic anemia. Hypochromia in the
which by definition measures hemoglobin content in a given alpha (a) and beta (8) trait thalassemia syndromes is much
volume of red cells (100 mL). When the MCHC is <32% the less pronounced. However red cells in the a—-thalassemias
anemic process is described as being hypochromic and the slide and $-—thalassemia homozygous states show significant
reviewer should scan the peripheral smear for the presence of amounts of pallor.’ Sideroblastic anemias show a prominent
Possible pathology
Slight 1%
Te 3%
2+ 5%
3+ 10%
4+ >11%
Figure 5-10 @ Note polychromasia in the cell with the arrow.
102 Chapter 5 Evaluation of Cell Morphology and Introduction to Platelet and White Blood Cell Morphology
Target Cells (Codocytes) (HS). This is an inherited, autosomal dominant condition and
is due to a deficiency of, or a dysfunction in, the membrane
Target cells appear on the peripheral blood as a result of an proteins spectrin, ankyrin, band 3 and/or protein 4.2.° The
increase in RBC surface membrane. They are artificially membrane cytoskeleton is dependent on these particular pro-
induced on the smear and their true circulating form, as seen teins to maintain the shape, deformability, and elasticity of
with an electron microscope, is a bell-shaped cell. The name the red cell. The deficiency and/or dysfunction of any one
codocyte is from the Greek word kodon meaning bell. In air- these membrane components will destabilize the cytoskele-
dried smears, however, they appear as “targets,” with a large ton, resulting in abnormal red cell morphology and a shorter
portion of hemoglobin displayed at the rim of the cell and a lifespan for the affected red cells in circulation.* Spherocytes
portion of hemoglobin that is central, eccentric, or banded are typically seen in large numbers in peripheral smears from
(Fig. 5-11). As the name implies, the cell actually resembles these patients. Premature destruction of these abnormal
a target and is sometimes referred to as a “bull’s eye” cell as erythrocytes in the spleen may produce a mild to severe
well as a “Mexican hat’ cell. hemolytic anemia depending on the severity of the abnormal-
The mechanism of targeting is related to excess mem- ity. Erythrocytes from patients with hereditary spherocytosis
brane cholesterol and phospholipid and decreased cellular he- have a mean influx of sodium twice that of normal cells.
moglobin. This is well documented in patients with liver dis- Because these spherocytes have increased ability to metabo-
ease, in whom the cholesterol/phospholipid ratio is altered. lize glucose, they can handle the excessive intracellular
Mature red cells are unable to synthesize cholesterol and sodium while in the plasma, but when they reach the microen-
phospholipid independently. As cholesterol accumulates in vironment of the spleen, the active—passive transport system is
the plasma, as seen in liver dysfunction, the red cell is ex- unbalanced with increased sodium and decreased glucose
panded by increased membrane lipid, resulting in increased resulting in swelling and hemolysis of these cells (see Chap. 9).
surface area. Consequently, the osmotic fragility is also de- In more than one-half of these patients, the MCHC is greater
creased (see Chap. 31). Target cells are seen in many types of than 36%. Yet, for individuals not exhibiting an increased
anemia (Fig. 5—12); however, they are most prominent in the MCHC, a careful observation of their peripheral smear is the
hemoglobinopathies, thalassemias and liver disease. key to diagnosis.’ Figure 5-13 depicts a blood smear from a
patient with hereditary spherocytosis.
The acquired forms of spherocytosis share the mutual
Spherocytes
defect with HS in that there is a loss of membrane. In the nor-
Spherocytes have a reduced surface-to-volume ratio that results mal aging process of red cells they gradually lose their func-
in a cell with no central pallor. Because of their density tionality through loss of cellular lipids, proteins, etc.; thus,
(intense color) and smaller size, they are easily distinguished spherocytes are produced as a final stage before senescent red
in a peripheral smear. Their shape change is irreversible and cells are detained in the spleen and trapped by the reticuloen-
may also be seen as microspherocytes. They are considered dothelial system. This natural process does not typically
the most common form of the erythrocyte morphological dis- result in anemia. Another mechanism of producing sperocytes
orders stemming from an abnormality of the cell membrane. that may result in a mild to severe anemia is autoimmune
This abnormality may be hereditary or acquired and may be hemolytic anemia. The coating of the red cells with antibodies
produced by a variety of mechanisms affecting the red cell and the detrimental effect of complement activation
membrane. Perhaps the most detailed mechanism for sphering results in the membrane loss of cholesterol accompanied by a
is the congenital condition known as hereditary spherocytosis loss of surface area without hemoglobin loss producing sphero-
cytes. The reduced surface-to-volume ratio of all spherocytes
renders them abnormally susceptible to osmotic lysis; conse-
quently, they have an increased osmotic fragility. Hemolysis is
known to result from membrane abnormalities; therefore, other
hemolytic processes may also produce spherocytes. They may
also be seen as microspherocytes in the peripheral smears of
burn patients. Figure 5—14 lists the more common pathologic
conditions in which spherocytes are seen.
Stomatocytes
The word stomatocyte is derived from the Greek word stoma,
which means mouth. They have a central pallor which is said
to be slit-like or mouth-like on peripheral blood smears. These
red cells are of normal size, but are not biconcave, and in wet
preparations appear bowl-shaped (Fig. 5-15). The abnormal
eo morphology resulting in the stomatocyte is thought to be the
result of a membrane defect. Stomatocytosis is associated with
Figure 5-11 ™ Note the target cell at the arrow.
abnormalities in red cell cation permeability that lead to
Chapter 5 Evaluation of Cell Morphology and Introduction to Platelet and White Blood Cell Morphology 103
changes in red cell volume, which may be either increased (hy- condition or at most a mild normochromic/normocytic
drocytosis) or decreased (xerocytosis), or is some cases, near anemia), hemolytic anemia, alcoholic cirrhosis, and acute
normal.’ Hydrocytosis and xerocytosis represent the alcoholism. Stomatocytosis is also present on peripheral
extremes of a spectrum of red cell permeability defects.? The blood smears of patients with Rh deficiency syndrome, also
exact physiologic mechanism of stomatocytic shape is poorly known as Rh null disease, in which erythrocytes from these
understood and the molecular basis of this disorder is rare individuals have either absent (Rh,,,) or markedly
unknown. Stomatocytosis may be acquired or congenital. As reduced (Rh,,.g)Rh antigen expression. This may result in a
with hereditary spherocytosis, stomatocytes are seen in sig- mild to moderate hemolytic anemia, and mutations in the Rh30
nificant numbers in the hereditary form known as hereditary and RhAG genes have been associated with this syndrome.”
stomatocytosis and in smaller numbers in the acquired form.
Many chemical agents can induce stomatocytosis in vitro
(phenothiazine and chlorpromazine); however, these changes Ovalocytes and Elliptocytes
are reversible.'° Stomatocytes are known to have an increased Many investigators consider the terms ovalocyte and ellipto-
permeability to sodium; consequently, their osmotic fragility cyte to be interchangeable; however, for the purposes of this
is increased. discussion, they are viewed as distinct and separate. This
Stomatocytes are more often artifactual than a true man- morphological abnormality is thought to be the result of a
ifestation of a particular pathophysiologic process. The arti- mechanical weakness or fragility of the membrane skeleton
factual stomatocyte has a distinct slit like area of central and may be acquired or congenital. The pathogenesis of the
pallor, whereas the area of pallor in the genuine stomatocyte formation of either of these cells is unknown. Ovalocytes may
appears shaded. Several of the associated disease states in be considered as more egg-shaped and have a greater ten-
which stomatocytes may be found are hereditary spherocyto- dency to vary in their hemoglobin content. They can appear
sis (the stomatospherocyte is best viewed in wet prepara- normochromic or hypochromic, normocytic or macrocytic.
tions); hereditary stomatocytosis (which is usually a benign Megaloblastic anemia is characterized by oval macrocytes
(macroovalocytes) that may be 9 um or more in diameter and
lack central pallor (Fig. 5—16).*
MORPHOLOGY - SPHEROCYTES
Figure 5-13 m Note the spherocyte at arrow in a blood smear from Figure 5-14 @ Correlation of spherocytes to pathologic processes.
a patient with hereditary spherocytosis.
104 Chapter 5 Evaluation of Cell Morphology and Introduction to Platelet and White Blood Cell Morphology
MORPHOLOGY - OVALOCYTE/ELLIPTOCYTE
Fragmented Cells (Schistocytes, Helmet Schistocytes are the extreme form of red cell fragmen-
Cells, Keratocytes) tation (Fig. 5-22). Whole pieces of red cell membrane
appear to be missing, and bizarre red cells are apparent.
Schistocytes are split, cut, or cloven cells resulting from some Schistocytes may occur in patients with microangiopathic
form of trauma to the cell membrane. It is recognized that not hemolytic anemia, disseminated intravascular coagulation
all membrane alterations occur pathologically. However, (DIC), heart valve surgery, hemolytic uremic syndrome,
there are certain triggering events in disease that invariably thrombotic thrombocytopenic purpura,'* renal graft rejec-
lead to fragmentation such as alteration of normal fluid circu- tion, vasculitis, in severe burn cases, as well as march hemo-
lation. Examples of fluid alterations are the development of globinuria (a form of hemoglobinuria seen in soldiers and
fibrin strands, damaged endothelium, or a damaged heart long-distance runners).
valve prosthesis. The flow of blood in the circulation may Keratocytes are red cells that have been caught on fibrin
actually sweep the erythrocytes through the fibrin strands, strands in circulation, and rather than splitting, the cell hangs
splitting the red cell. The shapes of these cells vary based on over the fibrin fusing two sides of the cell together, creating a
the shear forces and presentation of the red cells as they are vacuole. Once the cell escapes from the fibrin strand it
cut by the fibrin. Intrinsic defects of the red cell make it less appears in the peripheral blood as a red cell with a vacuole in
deformable and, therefore, more likely to be fragmented as it one end resembling a blister and is called a blister cell. It also
traverses the microvasculature of the spleen. Examples such is said to resemble a women’s handbag and may be called a
as antibody-altered red cells and red cells containing inclu- pocket-book cell (see Fig. 5—22). Once the vacuole ruptures,
sions have significant alterations that increase their likelihood the resulting cell appears to have two horns. This “horned”
of being fragmented, consequently decreasing their survival cell also resembles a helmet and is sometimes reported as
time. such, but is actually a keratocyte (Greek for keras, horn).* The
Figure 5-19 m Irreversibly sickled cells. Figure 5-20@ Reversible, oat-shaped sickle cell.
106 Chapter 5 Evaluation of Cell Morphology and Introduction to Platelet and White Blood Cell Morphology
Possible pathology
A helmet cell and a bite cell are, therefore, one and the same.
The helmet cells may also be seen in patients with pulmonary
emboli, myeloid metaplasia and DIC. All fragmented red cells
le es are considered fragile and their survival time is diminished sig-
Zins
MORPHOLOGY
- FRAGMENTED CELLS a >
yy
Figure 5-24# Correlation of fragmented cells to pathologic processes. HA = hemolytic anemia; DIC = disseminated intravascular coagula-
tion; HUS = hemolytic uremic syndrome; TTP = thrombotic thrombocytopenic purpura.
Acanthocytes (Thorn Cells, Spur Cells) lecithin-cholesterol acyltransferase, which has been well doc-
umented in patients with severe hepatic disease. This enzyme
An acanthocyte is defined as a cell of normal or slightly reduced is synthesized by the liver and is directly responsible for
size, possessing 3 to 12 spicules of uneven length distributed esterifying free cholesterol; when this enzyme is deficient,
along the periphery of the cell membrane. The uneven projec- cholesterol is increased in the plasma. Acanthocytes may also
tions of the acanthocyte are blunt rather than pointed, and the be seen in myeloproliferative disorders, microangiopathic
acanthocyte can easily be distinguished from the peripheral hemolytic anemia (MAHA), and autoimmune hemolytic ane-
smear background because it appears to be saturated with hemo- mias. The presence of acanthocytosis in peripheral blood
globin. It appears essentially as a spherocyte with thorns. The smears remains the hallmark of the clinical diagnosis of most
MCHC is, however, always in the normal range (Fig. 5—25). neuroacanthocytosis syndromes, such as chorea-acanthocytosis
Specific mechanisms relating to the formation of acan- (ChAc) and McLeod syndrome."
thocytes are unknown; however, some details about these The red cell responds to this excess cholesterol in one of
peculiar cells are of interest. Acanthocytes contain an excess two ways, depending on the balance of other lipids in the mem-
of cholesterol and have an increased cholesterol-to-phospho- brane. It will become a target cell or an acanthocyte. Once an
lipid ratio; consequently their surface area is increased. acanthocyte is formed, it is very liable to splenic sequestration
The lecithin content of acanthocytes is decreased. The only and fragmentation, and the fluidity of the membrane is directly
inherited condition in which acanthocytes are seen in high affected. The most prominent pathologies in which acantho-
numbers is the rare condition abetalipoproteinemia. Most cytes may be observed are listed in Figure 5—26.
cases of acanthocytosis are acquired, such as the deficiency of
MORPHOLOGY - ACANTHOCYTES gh
deficiency, and in conditions in which inclusion bodies are with thalassemic syndromes, sickle cell anemia as well as
formed. They may also be seen in megaloblastic processes as other hemolytic anemias, and in megaloblastic anemias.
large tear-shaped cells (macroteardrops). Refer to Figure 5—3
for a composite of abnormal red cell morphology. Basophilic Stippling
Red cells that contain ribosomes can potentially form stippled
Red Gell Inclusions cells; however, it is thought that the actual stippling is the result
Howell-Jolly Bodies of the drying of cells in preparation for microscopic examina-
tion. Coarse, diffuse, or punctate basophilic stippling may occur
Howell—Jolly bodies (Fig. 5-28) are nuclear remnants con- and consist of ribonucleoprotein and mitochondrial remnants
taining DNA. They are | to 2 jm in size and may appear (Fig. 5-29). These aggregates of ribosomes result from an alter-
singly or doubly in an eccentric position on the periphery of ation in the biosynthesis of hemoglobin.
the cell membrane. They are thought to develop in periods of Diffuse basophilic stippling appears as a fine blue dusting,
accelerated or abnormal erythropoiesis. They may be seen in whereas coarse stippling is much more clearly outlined and eas-
Romanowsky, i.e., Wright’s, Giemsa, or supravitally stained ily distinguished. Punctate basophilic stippling is a coalescing of
peripheral smears. smaller forms and is very prominent and easily identifiable.
A fragment of the chromosome becomes detached and is Stippling may be found in any condition showing defective
left floating in the cytoplasm after the nucleus has been or accelerated heme synthesis, such as alcoholism, thalassemia
extruded. Under ordinary circumstances, the spleen effec- syndromes, megaloblastic anemias, and arsenic intoxication. It
tively pits these nondeformable bodies from the cell. How- is also considered a characteristic feature in the diagnosis of lead
ever, during periods of erythroid stress, the pitting mechanism poisoning. Basophilic stippling may be seen on a Romanowsky
cannot keep pace with inclusion formation. or supravitally stained peripheral smear. It is important for the
Howell-Jolly bodies may be seen after surgical splenec- reviewer not to confuse stippling with Pappenheimer bodies.
tomy, congenital absence of the spleen, or splenic atrophy The primary differentiation factors are that stippling appears
after multiple infarctions. They may also be seen in patients
Figure 5-27 @ Teardrop cells (peripheral blood). Figure 5-28 ™ Howell-Jolly body.
Chapter 5 Evaluation of Cell Morphology and Introduction to Platelet and White Blood Cell Morphology 109
z »
® *
_ 2 a
°
‘ ‘s
= 4
Hemoglobin CC Crystals
Hemoglobin (Hb) C crystals may be found in hemoglobin CC
disease. HbC disease is a mild chronic hemolytic anemia in
which the patient is homozygous for the abnormal hemoglobin
C.'5 HbCC crystals are formed by the crystallization of the
abnormal hemoglobin into one end of the red cell membrane.
The crystal forms in a hexagonal shape with blunt ends, leaving
the remainder of the cell with the appearance of being empty.
These crystals tend to stain dark red and are said to resemble a
“bar of gold” and may be referred to as such (Fig. 5—33).
HbCC crystals may not always be demonstrated in HbC
Figure 5-32 ™ Note the appearance of a Cabot’s ring in the cell at
disease, but their appearance has been found to increase after
the arrow.
splenectomy. HbCC crystals are not seen in HbC trait (HbAC).
Spherocytes
° G6PD deficiency
¢ Pulmonary emboli
¢ Hemolytic anemias
Schistocytes
* Posttransfusion
¢ Hereditary spherocytosis * Disseminated intravascular coagulopathy (DIC)
Elliptocytes * Thrombotic thrombocytopenic purpura (TTP)
¢ Hemolytic uremic syndrome
* Hereditary elliptocytosis
¢ Iron-deficiency anemia Teardrop Cells
¢ Thalassemias ¢ Severe anemias
Stomatocyte ¢ Myeloproliferative disorders
¢ Pernicious anemia
* Acute alcoholism
¢ Malignancies
Chapter 5 Evaluation of Cell Morphology and Introduction to Platelet and White Blood Cell Morphology 11]
continued
Soe
Ru extort sesh
L. A prominent morphologic clue when suspecting lead “5, Morphological abnormalities found in
cases of
poisoning is the presence of which of the following on a severe burns, microangiopathic hemolytic anemias,
peripheral smear? and disseminated intravascular coagulation
a. Heinz bodies (DIC) are:
b. Target cells a. Schistocytes
c. Siderotic granules b. Crenated cells
d. Basophilic stippling c. Ovalocytes
i) . In which of the following disease states would you d. Stomatocytes
expect to find oval macrocytes on the peripheral smear? . Oat-shaped cells may be associated with:
a. Iron deficiency anemia a. Myelofibrosis
b. Lead poisoning b. Hereditary spherocytosis
c. Megaloblastic anemia c. Burns
d. Hereditary spherocytosis d. Sickle cell anemia
. All but one of the following are possible mechanisms for . How would a cell be classified that has a diameter of
the production of macrocytes: 9 um and an MCV of 104 fL?
a. Liver disease a. Macrocytic
b. Postsplenectomy status b. Microcytic
c. Pernicious anemia c. Normal
d. Thalassemia minor d. Either normal or slightly microcytic
. An abnormal erythrocyte seen in liver disease and hemo- . Abnormal platelet morphology may be observed most
globinopathies and thalassemias and is characterized by prominently in:
the “bull’s eye” area is known as a: a. Idiopathic myelofibrosis
a. Stomatocyte b. Anemia of chronic disorders
b. Target cell c. Hereditary spherocytosis
c. Schistocyte d. Septic shock
d. Hypochromic cell
Chapter 5 Evaluation of Cell Morphology and Introduction to Platelet and White Blood Cell Morphology 115
. Which type of red cell inclusion is a DNA remnant? The following answer pool is used for items 15 to 29
a. Heinz body (Match)
b. Howell—Jolly body
aa. Anisocytosis f. Polychromasia k. Schistocytes
c. Pappenheimer body
d. Cabot ring b. Poikilocytosis | g. Microspherocytes —_1. Rouleaux
. Which of the following are considered microcytic/ c . Hypochromasia_h. Target cells m. Acanthocytes
hypochromic anemias? d. Microcytic i. Stomatocytes n. Dacrocytes
a. Autoimmune hemolytic anemia
oO . Depranocyte J. Blister cells o. Echinocytes
b. Pernicious anemia
c. Iron deficiency anemia lhaks RBCs with a large area of central pallor
d. Megaloblastic anemia 16. Variation in the size of the red blood cells
11. A hypersegmented neutrophil may be seen in which of ____17. Also known as codacytes
the following anemias?
eS: RBCs appearing stacked on each other
a. Iron deficiency
b. Megaloblastic pe NS: MCV of 65%
c. Autoimmune hemolytic anemia eee): RBCs with a mouthlike central pallor
d. Anemia of chronic disorders
21 . RBCs without an area of central pallor
12. Precipitates of denatured hemoglobin found primarily in
22. RBCs with evenly distributed spicules on the
patients with hemolytic anemia resulting from oxidant
membrane
stress describe:
a. Howell—Jolly bodies 23. RBC fragments
b. Heinz bodies . RBCs appearing bluish in color
c. Basophilic stippling
. Variation in the shape of the RBCs
d. Pappenheimer bodies
. Congenital abetalipoproteinemia
. Pappenheimer inclusions are formed from:
a. Excess a-chains 27. The formation of a vacuole in an RBC “trapped”
b. Excess B-chains by fibrin
c. Excess iron . Formed when an RBC with an inclusion squeezes
d. Oxidant stress out of a tight space
14. RBC inclusions resulting from an acceleration in hemo- . Seen in HbS disease
globin biosynthesis and consists of RNA:
a. Howell—Jolly bodies See answers at the back of this book.
b. Heinz bodies
c. Basophilic stippling
_d. Pappenheimer bodies
g Any regenerative red cell process will result in inclusions w Heinz bodies cannot be seen on a Wright-stained periph-
such as Howell—Jolly bodies, basophilic stippling, and eral smear.
Pappenheimer bodies. w Abnormal platelet morphology includes lack of granula-
= Howell—Jolly bodies, Pappenheimer bodies, and basophilic tion, giant platelets, and megakaryocytic fragments.
stippling may be seen in peripheral smears stained w Abnormal white cell morphology includes the presence or
with both Romanowsky type stain (i.e., Wright’s, absence of cytoplasmic granulation, or presence of cyto-
May-—Grumwald) and supravital stain (i.e., new methylene plasmic vacuolizaton, as well the appearance of the cellu-
blue, brilliant cresyl blue) lar nucleus.
w Siderotic granules and Pappenheimer bodies are basically a Hyposegmentation describes decreased neutrophil seg-
the same inclusion. The differentiating factor is that on an mentation (<2 lobes).
iron stain the inclusions are known as siderotic granules w Hypersegmentation describes increased neutrophil seg-
where on Wright’s stain they are known as Pappenheimer
mentation (25 lobes).
bodies.
REFERENCES Is Orkin, SH, and Nathan, DG: The tha- Approach. Williams & Wilkins, Balti-
lassemias. In Nathan and Oski’s Hema- more, 1996, p 77.
| . Bain, B: Current concepts: Diagnosis
tology of Infancy and Childhood, ed 5. . Womack, EP: Treating thrombotic
from the blood smear. N Eng] J Med
WB Saunders, Philadelphia, 1998, p 818. thrombocytopenic purpura with plasma
353(5):498, 2005.
8. Bolton-Maggs, P, et al: Guidelines for exchange. Lab Med 30:276, 1999.
. Stiene-Martin, E, et al: Clinical Hematol-
the diagnosis and management of heredi- . Storch, A, et al: Testing for acanthocyto-
ogy Principles, Procedures, Correlations.
tary spherocytosis. Br JHaematol sis. J Neuro 1252(1):84, 2005.
ed. 2. Lippincott-Raven, Philadelphia,
126:455, 2004. . Lawrence, C, et al: The unique red cell
1998.
\O. Gallager, P: Red cell membrane disor- heterogeneity of SC disease: Crystal for-
WwW. Shojana, AM: Protein synthesis in mega-
ders. ASH Education Book, 2005, cited mation, dense reticulocytes, and unusual
loblastic disorders. In Gross, S, and
from, http://www.asheducationbook.org/ morphology. Blood 78(8):2104, 1991.
Roath, S (Eds): Hematology: A Problem
cgi/content/full/2005/1/13. pp 1-11. . Kaplan, L, et al: Clinical Chemistry The-
Oriented Approach. Williams & Wilkins,
Bicorct 100Gh a7 10. Chen, J, and Huestis, W: Role of mem- ory, Analysis, Correlation, ed 2. Mosby,
Bere One brane lipid distribution in chlorpromazine- St. Louis, 2003, p 686.
. Glassey, E (Ed): Color Atlas of Hematol- induced shape change of human erythro-
ogy. College of American Pathologists, cytes. Biochim Biophys Re eae
See Bibliography at the back of this book
Hematology and Clinical Microscopy 1323(2):299, 1997
Ae eae Nowafield IU; 11. Mohandas, N: A cell by any other name.
.
Ake
Davenport,
:
J: Macrocytic
;
anemia. Am 12
Blood 106(13):4017, 2005.
. Camilo, N, and Ravindranath, Y: Hemo-
Fam Physician 53(1):155(8), 1996.
globinopathies: Abnormal structure in
. Provan, D, and Weatherall, D: Red cells
hematology. In Gross, S, and Roath, S
II: Acquired anaemias and poly-
(Eds): Hematology: A Problem Oriented
cythaemia. Lancet 355:1260, 2000.
PART 2
ANEMIAS
Chapter
Iron Metabolism
and Hypochromic
Anemias
Kathleen Finnegan, MS, MT(ASCP)SH
Introduction OBJECTIVES
Normal Iron Metabolism At the end of this chapter, the learner should be able to:
Distribution and
Requirements . List the components of the hemoglobin molecule.
Daily Iron Requirements and . State the function of iron in relation to hemoglobin.
Absorption
lron Transport . Discuss the absorption and transport of iron into the protoporphyrin ring during
Ww
Oo
Laboratory Evaluation of . Describe the physiologic factors that affect absorption and the amount of iron needed
Iron Status by the body.
Serum Iron
. Evaluate laboratory tests that are used to differentiate and identify categories of
Total lron-Binding Capacity
different types of anemia.
Transferrin Saturation
Ferritin . Discuss the pathophysiology of iron deficiency anemia, anemia of chronic disease,
Transferrin Receptor sideroblastic anemia, and the porphyrias.
Free Erythrocyte . List and compare the laboratory findings in iron deficiency anemia, anemia of
Protoporphyrin and Zinc
chronic disease, and sideroblastic anemia.
Protoporphyrin
. Define the porphyrias.
lron-Deficiency Anemia
Etiology . Compare and contrast the hypochromic anemias.
Pathophysiology
. Describe iron overload and hereditary hemochromatosis.
Clinical Features
Laboratory Findings . List the causes of iron overload.
Treatment
. List the characteristics of iron overload and hereditary hemochromatosis.
Anemia of Chronic Disease
Pathophysiology
Clinical Features
Laboratory Findings
Treatment
Sideroblastic Anemia
Pathophysiology
Laboratory Findings
Treatment
The Porphyrias
118 Chapter 6 Iron Metabolism and Hypochromic Anemias
Iron Overload
and Hemochromatosis
Hereditary
Hemochromatosis
Pathophysiology
Clinical Features
Laboratory Findings
Treatment
Secondary
Hemochromatosis
African Iron Overload
Case Study 1
Case Study 2
Case Study 3
menstruation. In pregnant and lactating women, the MDR iron ingested in the diet, only 5% to 10% is absorbed. The
increases to 3.0 mg per day. In the second and third trimesters, foods that increase and decrease iron absorption are listed in
the daily requirement of iron can increase to as high as 5 to Table 6-3.
6 mg. At delivery, there is a loss of 600 to 700 mL of blood. Regulation of iron absorption occurs within the intestinal
In total, during pregnancy approximately 1000 mg of iron is mucosa of the small bowel. The vast majority of iron is
utilized. This equals and at times exceeds the amount of stor- absorbed in the duodenum and the first portion of the jejunum.
age iron in an average woman of childbearing age. Lactation Iron molecules within the diet and iron complexes within the
and breast feeding also contribute to the loss of iron associ-
ated with pregnancy. Because milk is a poor source of iron,
many baby formulas are supplemented with iron. Infants who
are fed only mothers’ breast milk are at a significant risk of
developing IDA.
During periods of growth, such as infancy and adoles-
cence, iron requirements are substantially increased. During Individual MDR (mg)
the first year of life, absorption of 150 to 175 mg of iron is
required to maintain an appropriate hemoglobin concentra- Infant 1.0
Child 0.5
tion. The MDRs for iron for different individual groups are
Menstruating woman 2.0
listed in Table 6-1. Pregnant or lactating woman 3.0
Many foods are rich in iron, including meats, legumes, Adult man or nonmenstruating woman 1.0
green vegetables, cereals, and prunes (Table 6-2). Of the
120 Chapter 6 Iron Metabolism and Hypochromic Anemias
Iron Storage
Iron that is not used for erythropoiesis is stored in the
mononuclear phagocytic system (MPS) or reticuloendothelial
Amount and type of iron accessible from food (RE) cells of the bone marrow, liver, and spleen. Iron taken in
Functional state of gastrointestinal mucosa and pancreas
Current iron stores excess is stored in two forms, ferritin and hemosiderin. RE
Erythropoietic needs cells ingest old red cells and catabolize the hemoglobin to
recycle the iron. The major form of storage iron is ferritin.
Ferritin is water soluble and is easily mobilized by the body
for utilization.
protein interacts with the TfR to regulate iron uptake. Trans- The second storage form of iron is hemosiderin. Hemo-
ferrin receptor is capable of binding two molecules of trans- siderin is not water soluble. The iron in hemosiderin is
ferrin, each carrying ene or two molecules of iron. The iron released more slowly than that from ferritin and is less read-
content of the cell is regulated by control of the iron uptake ily available for utilization. Hemosiderin represents aggre-
and storage capacity. As cellular iron levels fall, the levels of gates of iron and can be visualized in tissue with the use of a
ferritin decrease and the transferrin receptors increase. When Prussian blue stain for iron. Hemosiderin appears as granules
cellular iron increases, ferritin increases, and TfRs fall. Proteins and aggregates. Ferritin is seen only via electron microscopy.
involved in iron metabolism are summarized in Table 6-6. Hemosiderin does become available in iron-deficient patients.
Transferrin receptor Transmembrane glycoprotein diamer with Receptor mediated ferric transferrin
two transferrin binding sites
HFE MHC class I glycoprotein complexes HFE binds with transferrin receptor
with B,-microglobulin reducing the affinity for transferrin
“FE = eho gene protein;eMac = major Aistcectapateility Rpt. DMT1 = Hpac metal transporter 1;
GI= gastrointestinal.
122 Chapter 6 Iron Metabolism and Hypochromic Anemias
* Please note that nt Oates vary by;institution, patient ei tea and testing oaoaayen
Laboratory Evaluation of Iron Status of iron depletion. It is increased in iron overload. Ferritin is an
acute phase reactant protein and is increased in inflammatory
The laboratory assessment of iron includes the measurement states, malignancy, infections as well as liver disease.’
of serum iron, total iron-binding capacity (TIBC), transferrin,
percent saturation of transferrin, serum transferrin receptors,
Transferrin Receptor
and ferritin. Zinc protoporphyrin (ZPP) is an indirect assess-
ment of iron availability. Reference ranges for iron status are Measurement of serum transferrin receptors (sTfRs) is
listed in Table 6—7. another tool for the assessment of iron status. The sTfRs are
»
inversely proportional to the amount of body iron. The con-
Serum Iron centration is increased in iron deficiency, but only when the
iron stores are depleted. The reason is that, with the lack of
Serum iron is a measure of transferrin-bound iron. Normal intracellular iron, the regulatory proteins directly increase the
serum iron concentration for males is 65 to 170 wg/dL. In synthesis of transferrin receptors.* TfRs do not increase with
women it is usually lower. The concentration of iron fluctu- anemia of chronic disease.
ates in individuals and is not always used for investigation of
iron metabolism without other laboratory tests. Early morning
Free Erythrocyte Protoporphyrin
specimens are preferred because of diurnal variation.
and Zinc Protoporphyrin
Total Iron-Binding Capacity Free erythrocyte protoporphyrin (FEP) is a heme precursor
that incorporates iron into the hemoglobin molecule. When
Total iron-binding capacity (TIBC) is the total amount of iron iron is not available to be incorporated into the protoporphyrin
that can be bound by transferrin in the plasma or serum. Each ring to form heme, excess protoporphyrins form. These excess
gram of transferrin will bind 1.4 mg of iron. The normal range rings complex with zinc to form zinc protoporphyrin (ZPP).
is 250 to 450 pg/dL. The binding capacity is normally one- ZPP is increased in iron-deficient erythropoiesis. The ZPP
third saturated. The measurement of TIBC is effectively an value correlates inversely with the ferritin level.
indirect measurement of transferrin concentration.
Transferrin Saturation
lron-Deficiency Anemia
Iron deficiency is described as a microcytic hypochromic
Percent saturation of transferrin is functionally measured as
anemia. The red blood cells are small in size and have an
the maximum amount of iron that is bound in plasma or
increase in central pallor, since they are lacking hemoglobin.
serum. Serum iron and TIBC are used to calculate the percent
The World Health Organization states that iron-deficiency
saturation. A transferrin saturation value below 16% is an in-
anemia (IDA) is the most common anemia* and is estimated
dicator of iron deficiency. Transferrin saturation is consis-
to affect approximately 2 billion people worldwide.’ IDA
tently increased in iron overload. The percent saturation is
results when there is a lack of adequate iron stores in the body
calculated as follows:
to meet physiological needs for the production of red blood
% saturation = S&UuMiron y iq9% cells. About 3% of children younger than 2 years of age, up to
DBE
3% of adolescent females, and fewer than 1% of adolescent
males have IDA.'° For high-risk groups see Table 6-8.
Ferritin
Etiology
Ferritin is directly proportional to the amount of iron stored.
Ferritin is a much better measurement than serum iron and IDA may occur because there is an increased demand for iron,
TIBC for the assessment of body iron stores. Normal range for or abnormal utilization for iron, poor diet, increased blood
ferritin is 20 to 300 g/dL. When low, ferritin is a good index loss, or malabsorption. IDA can develop slowly after the
Chapter 6 Iron Metabolism and Hypochromic Anemias 123
i Pinan
6-9 Causes of Iron-Deficiency
Prenatal/Neonatal
Premature birth
Inadequate Absorption
Low birth weight
Inflammatory bowel disease
Anemia during pregnancy
Resection of the bowel
Low iron formula
Celiac disease
Lack of iron supplements after 6 months in breastfed infants
Antacid therapy
Infancy/Childhood High gastric pH
Restricted diets Loss or dysfunction of absorptive enterocytes
Lack of iron supplements Poor bioavailability
Growth spurts Excess bran
Chronic infection
Decreased Iron Intake
Chronic or acute blood loss
Meat-poor diet
Adolescent/Adult Cereal-rich diet
Fad diets Malabsorption
Menstruation Elderly
Excessive weight gain Increased [ron Utilization
Pregnancy _
Growth spurts
Lactation and breast feeding Pregnancy
Elderly
Improper diet Loss of Iron
Chronic bleeding Gastrointestinal blood loss
Gastrointestinal bleeding Ulcer
Gastritis
Social/Economic Epistaxis
Low socioeconomic groups Hemorrhoids
Recent immigration from developing countries Menorrhagia
Pulmonary blood loss
Malignancy or cancer
Trauma
Excess phlebotomy
oie ke
Stage 2: Iron
Stage 1: Iron Deficient
Depletion Erythropoiesis
(IDA without (IDA with Stage 3: IDA
Laboratory Features Normal* Anemia) Mild Anemia) (Severe Anemia)
the RBC morphology is normal. This stage creates no overt (ZPP). The hemoglobin and hematocrit values are reduced and
effect on erythropoiesis. The red cell distribution width the red blood cells may appear microcytic. Ferritin levels
(RDW), however, is usually increased, which sometimes is are decreased, TIBC increases, and TfRs increase on the sur-
the first indication of an anemia developing. It is estimated face of the red blood cells. Other iron-dependent tissues may be
that nearly 50% of U.S. infants are in stage | of IDA at some- affected.
time during development.**
STAGE 3: IRON-DEFICIENCY ANEMIA
STAGE 2: IRON-DEFICIENT ERYTHROPOIESIS The final stage of anemia develops when the red blood cells
The second stage is iron-deficient erythropoiesis and is subclin- are severely deficient in iron. The advanced stage will show
ical. Plasma iron levels drop. When there is a decrease in iron a marked decrease in hemoglobin and hematocrit and hemo-
that is needed for heme synthesis, excess protoporphyrin accu- globin formation is delayed with the formation of red cells
mulates and complexes with zinc to form zinc protoporphyrin that are hypochromic and microcytic (Fig. 6-4). There is
ineffective erythropoiesis as a result of depleted storage iron
and diminished transport iron. The bone marrow shows
decreased hemoglobinization and ragged cytoplasm in red
blood cell precursors (Fig. 6-5). At this stage there is a
severe deficiency in total body iron. Most patients are not
diagnosed until this stage appears. Erythropoietin levels
increase, producing a slight reticulocytosis.
Clinical Features
PERIPHERAL BLOOD
In patients with severe IDA, a microcytic hypochromic anemia
Another manifestation that may be associated with IDA is seen on the peripheral blood smear (see Fig. 6-4). The mean
is pica.'* Pica is defined as the persistent eating or craving of corpuscular volume (MCV) can range from 55 to 74 fL; the
nonfood substances such as clay, dirt (geophagia), or ice mean corpuscular hemoglobin concentration (MCHC) can
(pagophagia). range from 22 to 31 g/dl; and the mean corpuscular hemoglobin
Infants with IDA are at risk for developmental, behavior, (MCH) can range from 14 to 26 pg. Microcytes, anisocytosis,
and motor difficulties. Symptoms include irritability, loss of and an increased RDW are the first morphological signs. The
memory, and difficulties in learning. They also have increased peripheral blood smear usually contains increased poikilocyto-
susceptibility to infection and poor growth.’ sis with the presence of a few codocytes (target cells), ellipto-
cytes or ovalocytes, and dacrocytes (teardrop cells) in addition
Laboratory Findings to microcytes. The reticulocyte count, which measures the abil-
ity for the bone marrow to produce new red blood cells, is
The features and indices associated with iron-deficiency ane- decreased in relation to the severity of the anemia as a result of
mia are summarized in Table 6-11. ineffective erythropoiesis. The white blood cell count is usually
Morphological
Usually hypochromic, microcytic RBCs
Mild to moderate anisopoikilocytosis
Decreased storage iron
Decreased sideroblasts
Absence of ringed sideroblasts
Laboratory
Decreased serum iron Figure 6-8 @ Peripheral blood of a patient with iron-deficiency
Decreased serum ferritin anemia after therapy. Note the two populations of red blood cells:
Decreased % transferrin saturation microcytic hypochromic and normal cells.
Increased total iron-binding capacity (TIBC)
Increased free erythrocyte protoporphyrin (FEP)
Increased serum soluble transferrin receptor ievels include ferrous gluconate and ferrous fumerate. In some cases
where intestinal absorption of iron is impaired, parenteral
administration of iron dextrans is used, which may also be
B
given for patients who cannot tolerate iron supplements. Par-
normal. The platelet count may be normal, increased, or enteral iron therapy may be given intravenously or intramuscu-
decreased. larly. Blood transfusions are used only if hemoglobin drops to
dangerously low levels. Each hospital will determine its own
IRON STUDIES critical level, but generally patients are transfused when their
Serum iron is decreased as a result of the depletion of iron hemoglobin level is less than 8 g/dL. After therapy, reticulo-
stores, TIBC is increased, percent transferrin is decreased, and cytes begin to rise within 3 to 5 days, and reach a maximum
TfRs are increased. Serum ferritin, the storage form of iron, is level within 8 to 10 days.'? Hemoglobin levels will increase in
decreased in all stages of IDA and is usually the first indication about 2 to 3 weeks and usually return to normal within 6 weeks.
of an IDA developing. Serum ferritin is a sensitive marker of The normal cell population will slowly predominate in about
iron storage and is an important laboratory test to help differen- 6 to 10 weeks. Microcytosis may take up to 4 months to
tiate IDA from other microcytic hypochromic anemias. The resolve. Restoration of the patient’s iron stores usually takes
sTfR is useful in detecting IDA. Patients with IDA have a mean about 6 months.'° A response to iron therapy is defined as an
sTfR level greater than two times the mean of the normal range. increase of | g of hemoglobin in | month (Fig. 6-8).
BONE MARROW
Usually a bone marrow assessment is not indicated for an Anemia of Chronic Disease
uncomplicated case of IDA. The bone marrow shows a mild
Anemia of chronic disease (ACD) or anemia of inflammation
to moderate erythroid hyperplasia with a decreased M:E ratio.
(AOI) is a common hematological disorder. ACD is the second
A common finding is the presence of poorly hemoglobinized
most prevalent anemia after IDA and the most commonly
noromoblasts with a scanty cytoplasm. Nuclear fragments,
found anemia in hospitalized patients.'°'* ACD is multifactor-
karyorrhexis, budding, and multinuclearity are also found.
ial in nature and is associated with chronic infections, autoim-
Iron content is decreased and nuclear cytoplasmic asynchrony
mune disease, chronic inflammation, and malignant neoplasms
is also evident during maturation. Cytoplasm maturation lags
(Table 6-12). ACD usually presents | to 3 months after the
behind nuclear maturation (see Fig. 6-5).
onset of a chronic disease. The anemia is characterized by
decreased serum iron levels, decreased TIBC, and decreased
Treatment saturation of transferrin. Serum ferritin levels, however, are
usually increased as a result of the iron being trapped in the
The first choice in the treatment for IDA is correction of the RES cells of the bone marrow. This iron can be visualized in
primary disease state. The next step is oral dietary supple- bone marrow aspirates stained with Prussian blue.
ments, which are necessary to replenish the body stores. Oral
supplements of ferrous sulfate are the standard treatment or Pathophysiology
treatment of choice.”'* The recommended dose of treatment is
3 tablets per day containing 60 mg of elemental iron'*'> The The pathogenesis of ACD is unclear. It has been suggested
major obstacle with oral supplements is the side effect of that chronic disease states block the transfer of storage iron to
nausea and stomach discomfort. Other oral supplements erythroid precursors within the bone marrow.
Chapter 6 Iron Metabolism and Hypochromic Anemias 127
Clinical Features
So: Se?C008
PP$9.0 °0.00% BPO
69%c008%e> 0,© 22 ooo 40,8
, | Sock Anemia present for several months following development
@ a of a chronic disease
GS) ®
Fe%e® ©.8 6°@°8° i
© ee.
; ® Oo” 00 9@ Vay
Morphological
Usually normocytic RBCs (normal MCV)
00 Ox ay
6 fea© OYSen 500.
(i
Wee®
@ @ 0 j op @
May be hypochromic or normochromic
0%~98S.
»D80,O56 00 90,900
O90 SD ot
92,0 §eef O0542,” %o ©& 09,0 5° 06°! Normal number of bone marrow erythrocytic precursors
Increased storage iron
; Decreased sideroblasts
e@c® @
eo “e050 ©,% 6
9&5 Rare to absent ringed sideroblasts
Laboratory
Decreased serum iron
Decreased total iron-binding capacity (TIBC)
Decreased % transferrin saturation
Normal to increased serum ferritin levels
Figure 6-9 ™ Normochromic, normocytic red blood cells in a Normal serum soluble transferrin receptor levels
patient with anemia of chronic disease. (Wright's stain, x 200)
128 Chapter 6 Iron Metabolism and Hypochromic Anemias
BONE MARROW
In the bone marrow there is an adequate number of erythroid
precursors, and the M:E ratio may be increased because of
decreased erythropoiesis. Sideroblasts are decreased but the
macrophages appear to have an increased amount of storage Inherited
Congenital sideroblastic anemia, sex-linked
iron. Hemosiderin is the long-term storage of iron and appears Autosomal recessive sideroblastic anemia
as very coarse aggregates of iron (Fig. 6-10). In contrast,
hemosiderin is absent in IDA. Acquired
Primary or idiopathic
Myelodysplasia (RARS)
Secondary
Treatment
Lead
When possible, treatment for ACD is to treat the underlying Alcohol
Drugs, including isoniazid and chloramphenicol
disease first. In cases in which treating the underlying disease
is not feasible, other options are necessary. Blood transfusions
are given for a rapid and effective intervention. Transfusions
are helpful when there is a severe or life-threatening anemia.
For patients with ACD associated with chronic infection Pathophysiology
or malignancy, supplements of iron should be avoided.* Ery- Sideroblastic anemias involve abnormalities of the enzymes
thropoietic agents for patients with ACD are currently regulating heme synthesis. The ringed sideroblast is a charac-
approved for use by patients with cancer who are undergoing teristic of this group of anemias. Several investigators have
chemotherapy. The therapeutic effect involves counteracting identified enzyme deficiencies, including deficiencies of delta
the antiproliferative effects of cytokines, along with the stimu- 5-aminolevulinic acid synthetase and uroporphyrinogen
lation of iron uptake and heme biosynthesis in the erythroid decarboxylase, in patients with sideroblastic anemia. No one
cells2 defect, however, can account for all inherited and acquired
disorders. The ringed sideroblasts are sideroblasts in which
iron is accumulated in the mitochondria that surround the
Sideroblastic Anemia
nucleus (Fig. 6-11). These siderotic granules are visible with
The sideroblastic anemias are a group of disorders character- Prussian blue stain.
ized by a hypochromic anemia, ineffective erythropoiesis, an The hereditary sideroblastic anemias are rare. The con-
increase in serum and tissue iron, and the presence of ringed genital sex-linked type is more common than the autosomal
sideroblasts in the bone marrow. It is a very diverse group of recessive variant. The anemias typically appear early, usually
anemias and can be inherited or acquired. The inherited sider- within the first few months or years of life. The molecular
oblastic anemias include sex-linked congenital sideroblastic defect involves the enzyme 5-aminolevulinic synthetase.*°
anemia and autosomal recessive sideroblastic anemia. The This enzyme initiates the heme synthetic pathway. Heme syn-
acquired sideroblastic anemias can be primary (also referred thesis is impaired as iron enters the erythroid precursor which
to as idiopathic) or secondary (Table 6-14). The secondary cannot be incorporated in the heme molecule because the pro-
sideroblastic anemias are typically the result of toxins or toporphyrin ring cannot be formed. Iron then accumulates in
drugs, such as chloramphenicol. the mitochondria.
Figure 6-10 @ Increased reticuloendothelial iron in a patient with Figure 6-11 @ Ringed sideroblast as detected by Prussian blue
anemia of chronic disease. (Prussian blue stain, x 200) staining of a bone marrow aspirate. (Prussian blue stain, x 1000)
Chapter 6 Iron Metabolism and Hypochromic Anemias 129
Laboratory Findings
The principal clinical feature is a refractory or progressive
anemia with the characteristic ringed sideroblasts in the bone
marrow aspirates (Table 6-15).
PERIPHERAL BLOOD
The anemia is moderate to severe. The red blood cells are
dimorphic, ranging from microcytes to normocytes (Fig. 6-13).
The MCV, MCH, and MCHC are usually normal. The RDW is
increased, representing the dual population of red blood cells.
Other abnormalities of the red cells include anisocytosis, poik-
ilocytosis, target cells, Pappenheimer bodies (iron deposits in
the red cell), and basophilic stippling. The WBC and platelet : @e® ©
Treatment
ifferential Diag!
d Bone Marrow
The differential diagnosis comparing RBC indices and resulting from excess iron. In this disorder of iron storage,
peripheral blood and bone marrow features of IDA, ACD, and there is an inappropriate increase in intestinal iron absorption
sideroblastic anemia is presented in Table 6-16. that leads to excess iron in the tissues. Iron overload may be
primary or inherited or secondary to the chronic anemias and
their treatment. The excess iron is stored in your liver, heart,
The Porphyrias
The porphyrias are a group of rare inherited disorders that
involve a block in porphyrin synthesis that is due to a
defect in the enzymes in the pathway of heme synthesis
(Fig. 6-14). This causes porphyrin heme precursors to
accumulate in tissues and large amounts are excreted in
urine and feces. The porphyrias are classified as acute or
nonacute according to their clinical presentation, and as
erythropoietic or hepatic, depending on the site of abnormal
metabolism.
The porphyrias are associated with neurovisceral
attacks’! which include photosensitivity, motor dysfunction,
sensory loss, mental disturbances, and sometimes abdominal
pain. The classification and characteristics of the porphyrias
are summarized in Table 6-17. The most common is acute
intermittent porphyria characterized by colicky abdominal
pain, vomiting, diarrhea, constipation, and central nervous
system involvement.
Erythroid/
Disease Dehcient oe Course Inheritance Hepatic Symptoms
and pancreas** and damages these organs. The different types The third stage is iron overload with early symptoms that
of hemochromatosis are summarized in Table 6-18. include lethargy and arthralgia. The last stage is iron overload
with organ damage and cirrhosis. The liver organ is com-
monly implicated and hepatomegaly is present in 95% of
Hereditary Hemochromatosis Cases ge
Hereditary hemochromatosis (HH) is a recessive genetic disor- The involvement of hepcidin in hemochromatosis has
der and is one of the most frequent genetic diseases in Caucasian also been studied. Hepcidin is a peptide synthesized in the
populations, affecting approximately | in 300 people.'*?* Preva- liver and is involved with iron homeostasis. The rate of iron
lence is especially high in the Irish, Scottish, and Welsh popula- influx into the plasma depends on the activity of hepcidin.*°78
tions. In 1996, a mutant gene linked to the HLA-A locus on the When plasma iron levels are increased, the synthesis of hep-
cidin increases, which diminishes the release of iron from the
short arm of chromosome 6, called the HFE gene, was first
described in patients with hereditary hemochromatosis.’*** This
mutation is a single-base change that results in the substitution
of tyrosine for cysteine at position 282 of the HFE protein
(C282Y). The C282Y mutation alters the conformation of the
Table 6-18 Classification Of
HFE protein and interferes with its function of regulation of iron _Hemochromatosis sean
absorption.
Hereditary Hemochromatosis
Classical hemochromatosis type |
Juvenile type 2
Pathophysiology Transferrin receptor type 3
African overload type 4
The disease is caused by a gradual accumulation of iron in
the tissues which leads to chronic liver disease, arthritis, Secondary Hemochromatosis
diabetes, pituitary damage, congestive cardiac failure, Hereditary disorders
and cardiac arrhythmias (Fig. 6-15). Patients with HH Thalassemia
Sickle cell anemia
absorb two to three times as much dietary iron as normal
Sideroblastic anemia
individuals.2> The end result is intestinal absorption of Enzyme-deficiency anemia
iron that generally exceeds iron loss by approximately Hereditary spherocytosis
3 mg per day.7 25
Acquired Disorders
Four stages of the disorder have been described.” The Anemia not due to blood loss in which multiple
first stage is a genetic predisposition with no abnormality transfusions are required
other than increased serum transferrin saturation. The second Dyserythropoietic anemia
stage involves 2 to 5 g of iron overload without symptoms.
132. Chapter 6 Iron Metabolism and Hypochromic Anemias
Laboratory Findings
Although iron metabolism is abnormal, erythropoiesis is
normal and hematologic abnormalities are usually not seen.
Other laboratory abnormalities include increased liver function
enzyme tests, particularly alanine and aspartate aminotransam-
inases (ALT, AST).
IRON STUDIES
In patients with HH, serum iron, serum ferritin and serum
Figure 6-15 @ Liver biopsy from a patient with idiopathic
hemochromatosis and cirrhosis. Note the excess deposits of iron. transferrin levels are increased. TIBC is usually within nor-
(Ferric ferricyanide stain) mal limits. Increased percent transferrin saturation is an
important hallmark of the disease. The laboratory criteria for
diagnosing HH include transferrin saturation greater than
mucosal cells and macrophages. When plasma iron drops, the 50% for females and transferrin saturation greater than 60%
synthesis of hepcidin is decreased, allowing these cells to for males.* Diagnosis can be confirmed by direct analysis of
release more iron. Hepcidin expression is increased in iron the HFE gene.
overload and decreased in iron deficiency.™*
Other polymorphisms have been found, including
mutation H63D. H63D is a histidine to aspartic acid substi-
Treatment
tution at position 63. C282Y and H63D account for the The goal of treatment of iron storage disease is to remove the
majority of the cases of HH. The roles of hepcidin and HFE excess amount of iron from the body. Patients who have
protein are related. symptoms of HH require therapeutic phlebotomy. Removal of
500 mL of blood once or twice a week is performed until
Clinical Features there is a marked decrease in serum transferrin percent satu-
ration and serum ferritin. The goal is to have the serum fer-
Signs and symptoms of HH usually occur in midlife. The ritin less than 50 g/L and transferrin saturation below 30%.'*
most common complaint is joint pain. Chronic arthritis of the Once this is achieved, phlebotomy frequency is reduced to
second and third metacarpophalangeal joints is highly sugges- two to four times per year. In the event that therapeutic phle-
tive of the disease. Early symptoms are usually nonspecific. botomy is not appropriate, desferrioxamine, a chelating agent,
They may include fatigue, arthralgia, bronze discoloration of is used to reduce iron stores. For patients with hemoglobins
the skin, and erectile dysfunction. As the disease progresses, greater than 10 g/dL, phlebotomy is the choice of treatment;
en Mle
D = decrease; I = increase; N = normal; V = variable; TIBC = total iron-binding capacity; ZPP = zinc protoporphyrin.
Chapter 6 Iron Metabolism and Hypochromic Anemias 133
for patients with hemoglobins less than 10 g/dL, desferriox- African Iron Overload
amine is the treatment of choice.
A distinct iron-loading disorder is prevalent in Africa,
affecting up to 10% of some rural populations.**?? These
Secondary Hemochromatosis individuals have a predisposition to iron overload as a
result of excessive dietary iron intake. African iron over-
Secondary hemochromatosis can be acquired or secondary to
load is not due to mutations of the HFE gene and is not
other inherited hemolytic anemias. The common characteristics
linked to the HLA locus, but is thought to be related to
of secondary hemochromatosis are anemia, ineffective erythro-
cooking in iron pots.
poiesis, and iron overload. Secondary hemochromatosis patients
A summary and comparison of iron studies in the
are transfused repeatedly. This leads to increased iron storage
microcytic hypochromic anemias and hemochromatosis are
because there is no mechanism for iron excretion. Iron overload
provided in Table 6-19.
from transfusion therapy is treated with chelation therapy.
c. 2 mg/day
Case Study I d. | mg/day
5. Prussian blue staining of the bone marrow in this patient
A mother brought her 17-month-old female child to the would reveal:
pediatrician for a general check-up. The child was recently a. Increased storage and increased sideroblastic iron
adopted from Central America. On physical examination, b. Increased storage and decreased sideroblastic iron
the child appeared pale and listless. The laboratory tests c. Decreased storage and decreased sideroblastic iron
included WBC count = 16.5 X 10°/L; RBC count = 2.97 x d. Increased ringed sideroblasts
10'°/L: HGB = 4.2 g/dL; Het = 15.8%; MCV = 53.3 fL;
MCH = 14.1 pg; MCHC = 26.5 g/dl; RDW = 23.2%; PLT ANSWERS
count = 496 x 10°/L. Examination of the peripheral blood
smear revealed microcytic, hypochromic red blood cells
with marked anisocytosis and poikilocytosis.
QUESTIONS
AR
WN Oy
5es
So
a
©
QUESTIONS
Case Study ete ra
il, What is the most likely cause of this patient’s disorder?
2. If serum iron studies were performed on this patient, a. Iron deficiency anemia
they would reveal: b. Anemia of chronic disease
a. Decreased serum iron, decreased serum ferritin, and
c. Acquired sideroblastic anemia
increased TIBC
d. Hereditary hemochromatosis
b. Decreased serum iron, normal to increased serum fer-
. The serum iron studies in this patient would reveal:
ritin, and decreased TIBC a. Increased serum iron, increased serum ferritin, and
c. Increased serum iron, increased serum ferritin, and increased percent transferrin saturation
decreased or normal TIBC b. Decreased serum iron, normal serum ferritin, and
d. Increased serum iron, increased serum ferritin, and decreased percent transferrin saturation
increased TIBC c. Decreased serum iron, decreased serum ferritin, and
3. Prussian blue staining of a bone marrow aspirate speci- decreased percent transferrin saturation
men from this patient would reveal: d. Increased serum iron, decreased serum ferritin, and
a. Decreased stainable bone marrow iron and decreased increased percent transferrin saturation
ringed sideroblasts . What additional test would be most useful in confirming
b. Decreased stainable bone marrow iron and increased the diagnosis?
ringed sideroblasts a. Hemoglobin electrophoresis
c. Increased stainable bone marrow iron and decreased b. Vitamin B,, and folate levels
marrow sideroblasts c. Reticulocyte count
d. Increased stainable bone marrow iron and increased d. Measurement of transferrin saturation
ringed sideroblasts . What is the appropriate therapy or treatment for this
disorder?
a. Blood transfusion
ANSWERS
b. Chelation therapy
c. Phlebotomy
d. Iron supplements
OO
WN
ANSWERS
Case Study 3 gs
CS)tek
(Sr
ety
ey
Questions
: 1. The average adult has a total body iron content of: c _ 6 In the older patients, iron deficiency most often develops
a. 25 mg from:
b. 100 mg a. Impaired absorption of iron
c. 250 mg b. Poor diet
(4.8500 mg c. Chronic gastrointestinal bleeding
d. The aging process
2. Two-thirds of the total body iron is present as:
a. Hemosiderin . Microcytosis of red blood cells is reflected by:
b. Ferritin a. Increased MCV
c. Transferrin b. Decreased MCV
@ Hemoglobin iron c. Increased hemoglobin concentration
d. Decreased hemoglobin concentration
3. One milligram of blood contains:
a. 2 mg of iron iil . Anemia of chronic disease is commonly associated with
b. | mg of iron each of the following except:
.) mg of iron : a. Infections
d. 10 mg of iron b. Malignant neoplasms
c. Autoimmune disorders
4. On average, what percentage of the ingested iron is ab- d. A congenital defect
sorbed each day?
25% to 35% . Each of the following are causes of secondary siderob-
5% to 10% lastic anemias except:
c. 80% to 90% a. Alcohol
d. 100% b. Lead
c. Medications
5. Dietary iron is absorbed predominantly in the: d. Radiation therapy
a. Stomach
uodenum and first part of the jejunum 13. A common feature of sideroblastic anemia is which of
c. Transverse colon the following?
d. Sigmoid colon: a. Ringed sideroblasts
b. Decreased iron
6. The protein responsible for transporting
iron in the c. Decreased ferritin
bloodstream is: d. Macrocytes
a. Hemoglobin
b. Hemosiderin . Which of the following is the most sensitive assay in
ransferrin screening for hereditary hemochromatosis?
d. Ferritin a. Serum iron
_b. Serum ferritin
7. The hypochromic anemias represent a related group of c. Transferrin saturation
disorders with: d. Total iron-binding capacity
a. A quantitative defect in hemoglobin synthesis
b. A qualitative defect in globin protein chains ile The most often cause of secondary hemochromatosis is:
a. Drugs
c. Excess hemoglobin synthesis
d. Vitamin B,, and folate deficiency
b. Diet
c. Repeated blood transfusions
8. The most common cause of hypochromic anemia 1s: d. Malabsorption
a. Sideroblastic anemia
b. Megaloblastic anemia See answers at the back of this book.
c. Iron deficiency anemia
d. Lead poisoning
6 Chapter 6 |ron Metabolism and Hypochromic Anemias
@ Ferrous iron combines with protoporphyrin in the mito- w Hereditary hemochromatosis is an inherited iron overload
chondria of the red blood cell to form heme. disorder.
m@ Hemoglobin is formed from two a and two B globin pro- w Increased transferrin saturation is an important hallmark
tein chains, four heme groups, and four oxygen mole- of HH.
cules. mSecondary hemochromatosis can be hereditary or
mw The transferrin receptor is a tool for the assessment of acquired and is most commonly caused by repeated trans-
iron. The TfR level is inversely proportional to the fusions.
amount of iron in the body. mwThe most common gene mutation found in hereditary
g@ Serum ferritin levels are an indirect measure of iron stores hemochromatosis is C282Y.
in the body.
REFERENCES hemochromatosis. Nat Genet 13:399, WebMD ACP Medicine Online 2004.
1996. Accessed 5/17/06.
Sy. Brittenham, GM: Red blood cell function
. Lesperance, L, et al: Putting a dent in . Rose, EA, et al: Pica: Common but com-
and disorders of iron metabolism: Iron
iron deficiency. Contemp Pediatrics monly missed. J Am Board Fam Pract
metabolism. ACP Medicine Online 2002.
19(7):60, 2002. 13(5):353, 2000.
Accessed 5/17/06.
. Centers for Disease Control (CDC)
co . Beutler, E, et al: Iron deficiency and
tO. Andrews, NC: Disorders of iron metabo-
MMWR Weekly Iron deficiency-United overload. Am Soc Hem (1):40, 2003.
lism. N Engl J Med 341(26):1986, 1999.
States 1999-2000. . Loil, JG: Anemia. In Fox, JA (Ed); Pri-
1S) . Riedl, HD, et al: Characterization and
. Carley, A: Anemia: When is it iron mary Health Care of Infants, Children and
partial purification of ferrireductase from
deficiency? Pediatr Nurs 29(2):127, Adolescents, ed. 2. Mosby, St. Louis,
human duodenal microvillus membranes.
2003. 2002, pp. 471480.
Biochem J 309:745, 1995.
. Tender, J, and Cheng, TL: Iron defi- . Weiss, G, and Goodnough, LT: Anemia
. Brittenham, GM, et al: Clinical conse-
ciency anemia. In Burg, FD, et al (Eds): of chronic disease. N Engl J Med
quences of new insights in the patho-
Gellis and Kagan’s Current 352(10):1011, 2005.
physiology of disorders or iron and heme
Pediatric Therapy. W.B. Saunders, . Wians, FH, et al: Discriminating between
metabolism. Am Soc Hematol 1:39,
Philadelphia, 2002, pp. 633-637. iron deficiency anemia and anemia of
2000.
Wil. Joosten, E, et al: Upper and lower gas- chronic disease using traditional indices
Nn . Gunshin, H, et al: Cloning and character-
trointestinal evaluation of elderly inpa- of iron status vs transferring receptor con-
ization of a mammalian proton-coupled
tients who are iron deficient. Am J Med centration. Am J Clin Pathol 115(1):112,
metal-ion transporter gene. Nat Genet
107(1):24, 1999. 2001.
16:383, 1997.
. Brittenham, GM: Red blood cell function 18. Means, RT, and Krantz, SB: Progress in
. Feder, JN, et al: A novel NHC class I-like
and disorders of iron metabolism. understanding the pathogenesis of the
gene is mutated in patients with hereditary
Chapter 6 Iron Metabolism and Hypochromic Anemias 137
anemia of chronic disease. Blood 22 . O'Neil, J, and Powell, L: Clinical 27. Fleming, RE, and Sly, WS: Hepcidin: A
80(7):1639, 1992. aspects of hemochromatosis. Semin putative iron regulatory hormone rele-
19. Means, RT: Recent developments in the Liver Dis 25(4):381, 2005. vant to hereditary hemochromatosis and
anemia of chronic disease. Curr Hematol 3. Hash, RB: Hereditary hemochromatosis. the anemia of chronic disease. PNAS
Rep 2(2):116, 2003. J Am Board Fam Pract 14(4):266, 2001. 98(15):8160, 2001.
20. Schrier, SL: Anemia production defects: . Meeker, J, and Miller, S: Hereditary . Barton, JC, et al: Management of
Production defects with marrow ery- hemochromatosis. MLO 37(10):10, 2005. hemochromatosis. Ann Intern Med
throid hyperplasia and ineffective ery- . Pietrangelo, A: Hereditary hemochro- 129:932, 1998.
thropoiesis. ACP Medline Online 2002. matosis-a new look at an old disease. 29. Gordeuk, V, et al: Iron overload in
Accessed on 5/17/06. N Engl J Med 350(23):2383, 2004. Africa: Interaction between a gene and
BM. Sassa, S, and Kappas, A: The porphyrias: . Kohli, M, et al: Long term survival dietary iron content. N Engl J Med
Clinical presentation of the porphyries. in patients with hereditary hemochro- 82605m L992:
ACP Medline Online 2002. Accessed matosis. Gastroenterology 110:1107,
5/17/06. 1996.
Chapter /
| Megaloblastic
Anemias
Mitra Taghizadeh, MS, MT(ASCP), CLS(NCA)
Introduction OBJECTIVES
Biochemical Aspects At the end of this chapter, the learner should be able to:
Clinical Manifestations of
| . Define megaloblastic anemia.
Megaloblastic Anemia
Z . Compare and contrast the morphological characteristics of megaloblasts and
Hematologic Features
normoblasts.
Ineffective Hematopoiesis
Bone Marrow Morphology . Characterize the peripheral blood morphology of megaloblastic anemia.
Peripheral Blood
. Identify the bone marrow morphology of megaloblastic anemia.
Morphology
Etiology of Megaloblastic . Describe pernicious anemia, including the pathophysiology and clinical and
Anemia laboratory findings.
Vitamin B,, Deficiency . Describe the Schilling test as a diagnostic tool for pernicious anemia.
Folic Acid Deficiency
. List the causes of vitamin B,, and folate deficiencies.
Laboratory Diagnosis of
Megaloblastic Anemia . Evaluate laboratory tests used for the diagnosis of megaloblastic anemia.
Laboratory Tests for the
. Compare and contrast the treatment for vitamin B,, and folate deficiencies.
Diagnosis of Vitamin B,,
and Folic Acid Deficiencies . Using the peripheral blood findings, differentiate the anemia of liver disease from a
Other Laboratory Tests megaloblastic anemia caused by vitamin B,, or folate deficiency.
Treatment 1] . List other causes of macrocytic nonmegalobiastic anemias.
Vitamin B,, Deficiency
Folic Acid Deficiency De, Evaluate laboratory tests used for the diagnosis of macrocytic anemias.
Response to Therapy
Vitamin-Independent
Megaloblastic Changes
Inherited
Acquired
Drug and Toxin Induced
Macrocytic
Nonmegaloblastic Anemias
Causes of Macrocytic
Nonmegaloblastic
Anemias
Case Study
138
Chapter 7 Megaloblastic Anemias 139
Thymidylate
— synthetase
Figure 7-1 ™ Thymidine synthesis pathway from uridine nucleotide. Uracil is incorporated into DNA in the absence of thymine. UDP =
uridine diphosphate; (UDP = deoxyuridine diphosphate; dUTP = deoxyuridine triphosphate; (UMP = deoxyuridine monophosphate;
dTMP = deoxythymidine monophosphate; dTDP = deoxythymidine diphosphate; dTTP = deoxythymidine triphosphate: CH, THF =
methylene tetrahydrofolate.
140 Chapter 7 Megaloblastic Anemias
et
Figure 7-5 @ Howell-folly body in an RBC in pernicious anemia Figure 7-7 ™ Neutrophil hypersegmentation in pernicious anemia.
(arrow). (From Bell, A: Hematology. In: Listen, Look and Learn.
Health and Education Resources, Inc., Bethesda, MD, with
permission.)
The diagnosis of megaloblastic anemia is usually based on
the morphological characteristics of the peripheral blood and
the results of other biochemical tests. Bone marrow examina-
transfused to a patient with severe and untreated megaloblas- tion is generally not required. Bone marrow aspirates are per-
tic anemia, which is indicative of extracorpuscular hemolysis.° formed for diagnosis of other disorders with a megaloblastic
Multilobed neutrophils, termed hypersegmented neu- picture, such as myelodysplastic syndrome or erythroleukemia.
trophils, are seen in the peripheral smear in 98% of cases? The clinical features of megaloblastic anemia are summarized
(Fig. 7-7). Hypersegmented neutrophils refer to one or more in Table 7-1.
neutrophils with six lobes, or five or more neutrophils with
five or more lobes.’ They are larger than normal mature neu- Etiology of Megaloblastic Anemia
trophils. The number of hypersegmented neutrophils counted
per 100 white cells as determined by the differential should be The major causes of the megaloblastic anemias are vitamin
reported. Although hypersegmented neutrophils appear early B,, deficiency, folic acid deficiency, or a combination of both.
in the peripheral blood, they are the last morphological fea- Megaloblastic anemia can also be present in non-vitamin-
ture to disappear. Their absence does not rule out the diagno- deficient diseases, such as myelodysplastic syndromes and
sis of megaloblastic anemia. acute leukemias. Drug-induced megaloblastic anemia is
Other notable abnormal laboratory tests are elevated lac- caused by drugs that interfere with the metabolism of either
tate dehydrogenase (LD), indirect bilirubin and urobilinogen, vitamin B,, or folic acid. Examples of these drugs are
decreased haptoglobin, increased serum iron and ferritin, and discussed later in the chapter.
increased erythropoietin.*°'°
Bone Marrow
Hypercellular
Low ME ratio
Megaloblasts
Giant bands and metamyelocytes
Peripheral Blood
Pancytopenia
Macro-ovalocytes
Hypersegmented neutrophils
Biochemical Changes
Decreased haptoglobin
Elevated LD
Elevated indirect and direct bilirubin
Increased serum iron and ferritin
Increased erythropoietin
Figure 7-6 ™ Cabot ring in pernicious anemia (arrow).
142 Chapter 7 Megaloblastic Anemias
Ce y |&
Cobamide
5,6-Dimethyl-
benzimidazole
Bio
+TC ll
Deoxyuridine Thymidine
‘Table 7-2Causes ofVitamin Be |
| Deficiency
Dietary Deficiency
Strict vegans
synthetase
Malabsorption
NADPH +H Pernicious anemia
Gastrectomy (total or partial)
5, 10-CHo THF DHF < DHF reductase Blind loop syndrome
Fish tapeworm (Diphyllobothrium latum)
NADP + Diseases of ileum
Wale Chronic pancreatic disease
CH3 THF
Figure 7-10 The role of vitamin B,, and folate in DNA synthesis. DIETARY VITAMIN B,, DEFICIENCY Nutritional vitamin B,,
CH,THF = methylene tetrahydrofolate; THF = tetrahydrofolate; deficiency is uncommon in western countries and is limited to
DHF = dihydrofolate; CH;THF = methyl tetrahydrofolate; (UMP = strict vegetarians. In this group, the decrease in vitamin B,, is
deoxyuridine monophosphate; dTMP = deoxythymidine
accompanied by an increased plasma folate level.
monophosphate.
It is worth noting that children born to a vegan mother (one
who consumes no animal food) or to a mother who has an
untreated vitamin B,, deficiency are vitamin B,, deficient, espe-
Vitamin B,, plays an important role in two key reactions
cially if they are breast-fed.’ Untreated infants are severely
in the body. First, it is necessary in the synthesis of methionine
megaloblastic, with retarded growth and psychomotor develop-
from homocysteine. In this biochemical reaction, both vitamin
ment. Neurologic complications, such as irritability, anorexia,
B,, and folic acid are involved. Vitamin B,, acts as a coenzyme,
and failure to thrive, have been reported in severely vitamin B,-
methylcobalamin (MeCb), for the enzyme methyltransferase
deficient infants.”!°
(Fig. 7-10). Second, vitamin B,, is important in conversion of
The major cause of vitamin B,, deficiency is malabsorp-
methylmalonyl CoA, a Krebs cycle intermediate, to succinyl
tion. The most common form of intestinal malabsorption is
CoA. In this reaction vitamin B,, also acts as a coenzyme,
pernicious anemia. Other causes of vitamin B,, deficiency are
adenosylcobalamin (AdoCb), for the enzyme methylmalonyl
summarized in Table 7—2 and discussed later.
CoA mutase. Adenosylcobalamin acts as a hydrogen (H) car-
rier, taking the H from methylmalonyl CoA to make succinyl
PERNICIOUS ANEMIA
CoA (Fig. 7-11).
DEFINITION Pernicious anemia is the most common cause
of vitamin B,, deficiency. It is a chronic disease caused by the
CAUSES OF VITAMIN B,, DEFICIENCY
deficiency of IF. The cause for a decrease in IF is gastric parietal
Vitamin B,, deficiency progresses through four stages: stage I,
cell atrophy, which is associated with a concomitant decrease in
negative vitamin B,, balance; stage II, vitamin B,, depletion;
other gastric juices. Lack of IF leads to defective vitamin B,5
stage III, vitamin B,,-deficient erythropoiesis; and stage IV,
absorption and, consequently, megaloblastic anemia.
vitamin B,,-deficient anemia. Stages | and II are referred to as
the depletion stages. Stages III and IV are referred to as the
deficient stages.'*
COR
Sa CON CO oe Con
Ado Co
CH3 — CH2>—CO — S—CoA——*»CH3—CO—COOH ~—® CH2—CH2— COOH
Propionyl CoA Methylmalonyl CoA Succinyl CoA
Pernicious anemia is more common in people of the cobalamin-binding site of IF and is found more often. Type
Scandinavian, English, and Irish descent, with a prevalence in Il, or binding antibody, is detected as a complex with IF.°"°
women (female-to-male ratio of 4:1).° Pernicious anemia 1s Thyroid antibodies have often been found in the serum of
more common after age 50. In blacks, the disease may start patients with pernicious anemia or their relatives.°
earlier.'° Pernicious anemia occurs rarely in children, and, if Lymphocytotoxic antibodies have also been detected in
it occurs, it may be in the congenital form. Congenital perni- one-third of patients with pernicious anemia. A decrease in
cious anemia is characterized by the total absence of IF and suppressor T cells and an increase in the CD4:CD8 ratio sup-
normal secretion of other gastric juices. There are no antibod- port the presence of cell-mediated immunity in patients with
ies present against IF or the parietal cells. Juvenile pernicious pernicious anemia.°
anemia is similar to adult pernicious anemia, with the age of An association between pernicious anemia and other
onset in the second decade.°® autoimmune diseases, such as thyroid disease, diabetes melli-
tus, and rheumatoid arthritis, has also been noted.®° The posi-
PATHOPHYSIOLOGY The main cause of pernicious anemia is tive response to steroids in some patients with pernicious
atrophic gastritis characterized by atrophy of the gastric mucosa anemia supports the autoimmune mechanism.°
with decrease of gastric secretions and IF. The cause of gastric The Helicobacter pylori microorganism has been identi-
atrophy is probably autoimmune, with poorly defined genetic fied as a major cause of gastritis and peptic ulcers. This
predisposition.® IF is essential for absorption of vitamin B,,. In microorganism induces autoantibodies against the gastric pro-
the absence of IF, only a small amount of vitamin B,, is ton pump H+, K+-ATPase in 20% to 30% of infected patients.
absorbed, causing a gradual deficiency in vitamin B)>. These antibodies are associated with gastritis, increased
atrophy, and apoptosis in the corpus mucosa. Patients who
Genetic Factors develop these antibodies present clinical features similar to
The congenital form of pernicious anemia is inherited as an those of patients with autoimmune gastritis.'*
autosomal-recessive trait and is primarily seen in children
before age two.*!” The genetic contribution to the adult form of CLINICAL MANIFESTATIONS OF VITAMIN B,,
pernicious anemia is supported by: DEFICIENCY
The onset of pernicious anemia is generally insidious. Patients
|. The concordant presence of pernicious anemia in identical
with pernicious anemia and other vitamin B,, deficiencies have
twins
all the signs and symptoms of megaloblastic anemia mentioned
2. The increased risk in relatives of patients with pernicious
earlier. Fever is usually present in severe anemia. Loss of
anemia
appetite is a common complaint. Glossitis (sore tongue) 1s
3. The presence of achlorhydria with or without malabsorp-
reported in 50% of patients.° Although the initial presentations
tion in relatives of patients with pernicious anemia
may vary among patients with vitamin B,, deficiency, the clas-
4. The findings that relatives of patients with pernicious ane-
sic symptoms include weakness, glossitis, and paresthesias.°
mia may produce antibody to gastric parietal cells (24%, as
The bone marrow morphology of patients with vitamin B,5
compared with 3.8% in patients with a negative family
deficiency is megaloblastic, and the peripheral smear contains
history).'°!?
macro-ovalocytes. In addition to hematologic abnormalities,
The exact cause of the genetic predisposition of pernicious vitamin B,, deficiency is associated with gastrointestinal, throm-
anemia is not yet clear. A weak association has been made botic, psychiatric, and neurologic complications.
between pernicious anemia and the human leukocyte antigen
(HLA), but it is not conclusive. An association between HLA- NEUROLOGIC MANIFESTATIONS Neurologic problems are
B7 and pernicious anemia has been reported in whites. The more common in pernicious anemia than in other types of
association of HLA-D antigens Dw2, Dw5, and DR2 with vitamin B,, deficiencies. The degree of neurologic involvement
pernicious anemia is more significant than that of other HLA is not directly related to the degree of anemia. Neurologic man-
antigens.°'* IF antibodies are associated with HLA-Dw2.° ifestations in the absence of hematologic abnormalities have
been reported in many cases.'°
Immunologic Factors The neurologic abnormalities may be mild, moderate, or
The serum of patients with pernicious anemia contains autoan- severe and may involve degeneration of peripheral nerves,
tibodies to parietal cells, to IF, and to thyroid tissue. The pari- posterior columns of the spinal cord, and posterior and lateral
etal cell antibodies are found in the serum of approximately columns of the spinal cord (Table 7-3). Because multiple neu-
80% to 90% of all patients with pernicious anemia. The anti- ropathies are involved, the terms subacute combined degener-
bodies are also present in the gastric juices of 75% of patients ation (SCD) and combined system disease may be used.°”°
with pernicious anemia. This antibody is specific for the pari- In the earlier stage of pernicious anemia, the peripheral
etal cells only, and does not show any cross-reactivity.° nerves are affected, causing paresthesia and areflexia.2°?! The
Antibodies to IF are demonstrated in the serum of patient often experiences symmetric tingling or “pins-and-
approximately 50% to 70% of patients with pernicious ane- needle” sensations in the toes and later in all four limbs. Less
mia. These antibodies are more specific for diagnosis of ane- often, the patient may complain of numbness. At the later
mia than antiparietal antibodies. Two types of IF antibodies stage, the posterior spinal columns may be involved. At this
have been reported. Type I, or blocking antibody, attaches to stage, the patient may complain of clumsiness and have an
Chapter 7 Megaloblastic Anemias 145
Pteroyl residue
OH 5 6
N H.| H
NEO Shs
4
eae
Hon’ SS} : las ae 75
CH2
COOH
Pteridine residue p-Aminobenzoic acid L-Glutamic
residue acid residue
A. Folic Acid
Figure 7-12 ™ Structure of folic acid and its derivative. A. Folic acid (pteroylglutamic acid). The three components are defined by vertical
lines. B. Tetrapteroyltriglutamic acid (tetrahydrofolate triglutamate), the active form of folate present in the tissues.
Serum folate is in the form of CH,THF and enters all CAUSES OF FOLIC ACID DEFICIENCY
tissue cells in this form. DIETARY DEFICIENCY The main cause of folic acid defi-
ciency is decreased dietary intake. Other causes are malab-
ABSORPTION AND METABOLISM
sorption, increased requirement, and drug-induced folate
Dietary folic acid is in the form of polyglutamic acid (many deficiencies (Table 7-4). Nutritional folate deficiency is usu-
glutamic acid residues). Once in the intestinal lumen, it is
ally a consequence of poverty, old age, alcoholism, preg-
acted on by the enzyme folate deconjugase, which is present nancy, and chronic diseases. In the United States, folic acid
in the epithelial cells of intestine, to form monoglutamic acid has been added to cereal grains, rice, and milled flour to
(single glutamic acid residue) (Figs. 7-13 and 7-14). Monog- increase the average adult’s intake by 100 ug.?! The U.S. gov-
lutamic acid is then reduced and methylated to CH;THEF." ernment urges women planning to become pregnant and in
When CH,THE is absorbed from the circulation into the tis- early pregnancy to eat a diet rich in folic acid or take vitamin
sue cells, it transfers its methyl group to homocysteine to supplements to prevent the adverse effects of folic acid defi-
form methionine and THF. The THF formed reconjugates ciency on their fetus.
with additional glutamic acid residues to form the cellular
THF (see Figs. 7-12 and 7-13). The THF is then methylated MALABSORPTION The most common causes of folate
to form coenzyme methylene THF (N°N'°CH,THEF) necessary malabsorption are tropical sprue and_ gluten-sensitive
for the formation of thymidine monophosphate from uridine enteropathy. Tropical sprue is an infection that causes
monophosphate. This is the key reaction for DNA synthesis intestinal atrophy with clinical manifestations of weakness,
(see Fig. 7-10). weight loss, and steatorrhea. Tropical sprue affects the
Chapter 7 Megaloblastic Anemias 147
Polyglutamic acid ———» Polyglutamate Le——> CH; THE ————> CH; THF
CH3
TH
oye|O4
Glu
aseBniuooep
Folate
reductase Homocysteine THF Methionine
Monoglutamate
Glu
THF
Figure 7-13 @ Absorption and metabolism of folic acid. CH;THF = methyl tetrahydrofolate; Glu = glutamic acid; THF = tetrahydrofolate.
entire intestine and, therefore, Causes a wide variety of CLINICAL MANIFESTATIONS OF FOLIC ACID
nutritional deficiencies, including vitamin B,, deficiency.° DEFICIENCY
Affected individuals respond well to antibiotics. Clinical manifestations of folate deficiency are the same as
Gluten-sensitive enteropathy has the same clinical those for vitamin B,, deficiency, mentioned earlier. The
manifestations as those mentioned for tropical sprue. It onset of anemia is insidious, with the distinct morphology
includes both nontropical sprue and childhood celiac dis- characteristic of megaloblastic anemia in the bone marrow
ease. Affected individuals cannot digest gluten, a protein and in the peripheral blood. Although neuropathy is mainly
found in wheat and other grains. Lesions are most severe in characteristic of vitamin B,, deficiency, several cases of
the proximal intestiné. Childhood celiac disease is a malab- neurologic abnormalities, such as depression, dementia,
sorption syndrome resulting in anemia caused by iron defi- and peripheral neuropathy associated with folic acid defi-
ciency and, to a lesser degree, by vitamin B,, and folate ciency, have been reported.*° Some of these neuropathies,
deficiencies. in particular depression, have responded favorably to treat-
The requirement for folic acid also increases during ment with folate.*°***4
rapid cellular proliferation in hematologic diseases such as Folic acid and vitamin B,, are both necessary cofactors of
sickle cell anemia, thalassemia, hereditary spherocytosis, and the enzyme methionine synthase, which converts homocysteine
autoimmune hemolytic anemia. to methionine. The deficiency of these vitamins causes an
increased level of plasma homocysteine. Hyperhomocysteine- It is worth noting that to date, there is no standard algo-
mia is a risk factor for thrombosis.*!*° An association between rithm for investigation of vitamin B,, deficiency. Different
folate deficiency and development of leukemia in high-risk laboratories choose different algorithms based on an individ-
patients has been reported.*° ual patient, availability of the procedure, and the cost.
The most specific diagnostic tests used for vitamin B,, and SERUM AND RED CELL FOLATE
folate deficiencies are serum B,, and serum and red cell folate Serum and red cell folate are decreased in patients with
levels. Red cell folate is a better indicator of tissue folate levels folate deficiency. The normal range for serum folate levels
(Table 7-5). Serum cobalamin is usually measured via radioiso- is 5 to 16 ng/mL. The minimal folate level for a normal
tope dilution, or enzyme-linked immunosorbent assays which DNA synthesis is about 4 ng/mL.*’ Serum folate is sensitive
are normally automated. Serum folate may be measured by to dietary folate intake. False low serum folate levels have
microbiological assays, or by radioassay, or via an enzyme- been reported in patients taking antibiotics or certain cyto-
linked immunosorbence assay.” toxic drugs.**’° Red cell folate is a valuable test reflecting
the body folate stored. It is less affected by recent diet than
is the serum assay. In normal adults, the red cell folate con-
yEvaluation = centration ranges from 160 to 640 ug/L of packed red cells.”
In patients with a severe B,, deficiency, the serum folate
level is increased while the red cell folate level is decreased.
In this case, the increased serum folate level is caused
Screening Tests by increased CH;THF in the serum (the methyl trap
Complete blood count hypothesis), resulting from the lack of vitamin B,, necessary
WBC differential/RBC morphology
for conversion of homocysteine to methionine and methyl-
Reticulocyte index
THF to THF (see Fig. 7-10).
Serum lactate dehydrogenase (LD)
Iron studies
Bone marrow aspirate
Other Laboratory Tests
Vitamin B,, Deficiency
Serum B,, level Other laboratory tests that may support a diagnosis of vitamin
Gastric autoantibodies B,, are antibodies to IF, blood test for gastric atrophy, serum
Blood test for gastric atrophy vitamin B,, binding proteins, the Schilling test, methylmalonic
Serum cobalamin binding proteins
Serum and urine methylmalonic acid
acid (MMA) and total homocysteine assays, and the deoxyuri-
Total homocysteine dine (dU) suppression test. These laboratory tests are useful
when the other laboratory tests are inconclusive. Macrocytic
Folic Acid Deficiency anemia due to vitamin B,, and or folate deficiency should
Serum and red cell folate levels
Serum homocysteine be distinguished from macrocytosis present in patients with
myelodysplastic syndrome, hemolytic anemia, and those
Other Assays exposed to chemotherapeutic drugs (see Table 7-5).
Schilling test
dU suppression test
GASTRIC AUTOANTIBODIES
dU = deoxyuridine. Antibodies to IF are present in the serum of about 50% to
70% of patients with pernicious anemia.° Although the IF
Chapter 7 Megaloblastic Anemias 149
antibody test is not a sensitive test, its specificity for the diag-
nosis of pernicious anemia is 95%.° Decreased serum B,, and
the presence of the antibody to IF are indicative of pernicious
anemia. Parietal cell antibody is present in 80% to 90% of
patients with pernicious anemia, however, its diagnostic value
for pernicious anemia is limited because this antibody is spe-
cific for immune gastritis.°
SERUM HOMOCYSTEINE parenteral treatment. Several studies have shown that oral
Serum homocysteine levels are elevated in patients with both vitamin therapy can be as effective and efficient as par-
vitamin B,, and folate deficiencies, because both vitamins are enteral therapy in conducting hematologic and neurologic
necessary in the conversion of homocysteine to methionine. remissions and in maintaining normal serum B,,.°*'*° In
Serum homocysteine is also increased in a short-term folate- patients with pernicious anemia, about 1% of the oral dose
restricted diet and in patients with congenital homocystinuria is absorbed.'“° Therefore, high doses of oral vitamin (1000
and vitamin B, inhibitor.2°? to 2000 ug/day) are sufficient to replace the parenteral ther-
apy. The efficiency of the treatment should be checked by
DEOXYURIDINE SUPPRESSION TEST occasional measurement of serum B,, in patients undergoing
The dU suppression test measures the level of 5,10-CH, THF. the oral therapy.'*°
It is an indirect measurement of thymidylate synthesis in vitro Vitamin B,, is injected intramuscularly or subcuta-
and is abnormal in both vitamin B,, and folate deficiencies neously. Although the treatment protocol varies, the initial
(see Fig. 7-10). The principle of this test is based on the fact dose administered is higher in order to saturate the body stor-
that B,,- or folate-deficient cells cannot convert deoxyuridine age. Vitamin B,, may be given as 100 to 1000 ug/day for
to deoxythymidine, and therefore the radioactive-labeled 2 weeks, then weekly until hematologic values are normal-
thymidine will be incorporated into DNA. In patients with nor- ized and then monthly for life.' Vitamin B,, therapy can be
mal levels of B,, or folate, deoxyuridine is converted to thymi- monitored by reticulocyte counts.
dine, which suppresses the labeled thymidine incorporation
into the DNA.?* Specificity of the deficient vitamin can be
Folic Acid Deficiency
determined by addition of vitamin B,, or folate to the test sys-
tem and a correction of the original abnormal result. This test The recommended therapeutic dose to treat folate deficiency is
is relatively time consuming and therefore, despite its sensitiv- | to S mg/day for 2 to 3 weeks.'° Folic acid vitamin is water sol-
ity, 1s not used as a diagnostic test. uble and is given orally. Lifelong therapy is not required
The diagnosis of vitamin B,, deficiency may be com- because it is usually possible to treat folate deficiency within a
plex and inconclusive in the absence of hematologic and short period of time.'’° Folic acid may be given along with
neurologic abnormalities.** Many studies have shown that vitamin B,, when both vitamins are deficient or in a therapeutic
some patients with vitamin B,, deficiency may seek med- trial when the exact cause of megaloblastic anemia is not
ical help because of complications other than the classical known. In this case, folate therapy will correct the hematologic
features of megaloblastic anemia. Examples of these com- abnormalities in vitamin B,, patients, but the neurologic mani-
plications are retinopathy, neuropathy, vascular disorders, festations will progress.
infertility, and physical and mental growth retardations in Folic acid given as prophylaxis (0.25 to 0.5 mg/day) is
infancy.'>*°?'404445 However, these conditions have been recommended during pregnancy and dialysis and may be
reversed on early treatment with vitamin B,,. However, required in patients with hemolytic anemia and in patients
vitamin B,, deficiency should be included in the differential who are on antifolate drugs.'!?*'*? Folic acid can be injected
diagnosis in patients with neurologic and neuropsychiatric to hospitalized patients and in those who cannot take the med-
problems.** To prevent vitamin B,,-associated complica- ication by mouth.
tions, numerous attempts have been made at early diagno-
sis of vitamin B,, and folate deficiencies.
Response to Therapy
The initial sign of a positive response to therapy is an increase
Treatment
in the reticulocyte count. The number of circulatory reticulo-
Transfusion is rarely required in patients with megaloblastic cytes increases 3 to 5 days after therapy, with a peak at about
anemia unless the anemia is severe (hematocrit of less than 4 to 10 days.'* The reticulocyte count may increase to 50% to
15%) or is associated with congestive heart failure. In these 70% initially.’ The megaloblastic morphology of the bone mar-
cases packed red cells should be administered slowly to pre- row disappears within 24 to 48 hours after therapy. The hemat-
vent pulmonary edema! ocrit rises in about 5 to 7 days after therapy, reaching normal
levels in 4 to 8 weeks. Giant metamyelocytes and hyperseg-
mented neutrophils disappear within 2 weeks. The entire thera-
Vitamin B,, Deficiency peutic response process may take only 3 to 6 weeks, depending
on the severity of the disease.*!!
Most people with a vitamin B,, deficiency require lifelong
vitamin therapy. Cyanocobalamin and hydroxocobalamin are
the two therapeutic forms of vitamin B,,. Hydroxocobalamin Vitamin-Independent Megaloblastic
is preferred by some physicians because it has a longer half-
Changes
life. Cyanocobalamin is less expensive and converts to the
physiologic form. This group of disorders is characterized by the presence
Vitamin B,, can be administered orally to patients with of megaloblastic changes in the bone marrow and in the
dietary vitamin deficiency or to those who cannot tolerate peripheral blood that are refractory to vitamin B,, and
Chapter 7 Megaloblastic Anemias 151]
N = normal range.
Chapter 7 Megaloblastic Anemias 153
th ophysiology of megaloblastic anemia is: 7. The glycoprotein necessary for absorption of vitamin B,, is:
a. Defective RNA synthesis and abnormal cytoplasm a. Albumin
maturation b. Transcobalamin II
b. Defective DNA synthesis and abnormal nuclear matu- c. Haptoglobin
ration d. Intrinsic factor
c. Defective RNA synthesis and abnormal nuclear matu-
. Which of the following are clinical manifestations of
ration
both B,, deficiency and folate deficiency?
d. Defective DNA synthesis and abnormal cytoplasm
a. Anemia and jaundice
maturation
b. Thrombocytosis
. Which of the following laboratory findings coincide with c. Hemoglobinemia
megaloblastic anemia? d. Hemoglobinuria
a. Increased serum iron and serum bilirubin
. Which of the following Schilling test results corresponds
b. Decreased serum iron and serum bilirubin
to a diagnosis of pernicious anemia?
c. Decreased serum muramidase
a. Part I abnormal, part II not corrected
d. Increased haptoglobin b. Part I abnormal, part II corrected
. Megaloblastic anemia is associated with: c. Part [ and part IT abnormal
a. Ineffective erythropoiesis and increased reticulocytes d. Part I normal, part IH corrected
b. Ineffective erythropoiesis and decreased reticulocytes 10. Which of the following is not a cause of vitamin B,,
c. Ineffective erythropoiesis and decreased LDH deficiency?
d. Ineffective erythropoiesis and decreased erythropoietin a. Atrophic gastritis
. According to the morphological classification of ane- b. Total gastrectomy
mias, megaloblastic anemia is a: c. Blind loop syndrome
a. Macrocytic, hypochromic anemia d. Chronic glossitis
b. Macrocytic, hyperchromic anemia . Macrocytosis associated with acute blood loss is charac-
c. Macrocytic, normochromic anemia terized by:
d. Normocytic, normochromic anemia a. Decreased reticulocyte count
. Which of the following are not seen on the peripheral b. Increased reticulocyte count
smear of a patient with megaloblastic anemia? c. Pancytopenia
a. Macro-ovalocytes d. Macro-ovalocytes
b. Hypersegmented neutrophils . Which of the following is associated with pernicious
c. Hyposegmented neutrophils anemia and not macrocytic anemia due to liver disease?
d. Howell—Jolly bodies a. Increased LD
. Which of the following are the characteristic findings b. Increased bilirubin
of the bone marrow in a patient with megaloblastic c. Increased MCV
anemia? d. Hypersegmented neutrophils
a. Hypocellular with low M:E ratio
b. Hypercellular with high M:E ratio See answers at the back of this book.
c. Hypocellular with high M:E ratio
d. Hypercellular with low M:E ratio
154. Chapter 7 Megaloblastic Anemias
mw Vitamin B,, and folic acid in the form of cofactors are w Vitamin-independent megaloblastic anemias can be inher-
essential for two key reactions in the body. ited or acquired.
@ Intrinsic factor enhances vitamin B,, absorption through gs Macrocytic nonmegaloblastic anemias are characterized
the receptors present on the brush borders of the ileum. by a high MCV, macrocytes in the peripheral blood, and
w Transcobalamin II transfers vitamin B,, to the tissues. normocellular or hypercellular bone marrow with ery-
throid hyperplasia.
w The main cause of vitamin B,, deficiency is pernicious
anemia. a The most common causes of macrocytic anemia are liver
disease and alcoholism.
gw Pernicious anemia is the lack of intrinsic factor.
Clinical manifestations that are often associated with
vitamin B,, deficiency are anemia, fever, glossitis, and
neurologic symptoms.
Chapter 7 Megaloblastic Anemias = 155
REFERENCES the autosomal recessive megaloblastic ieS)Cn. Ballas, SK, and Saidi, P: Thrombosis,
anemia (MGA1) region. Blood 91:3593 megaloblastic anemia, and sickle cell
— . Lichtman MA. et al: Williams Hematol- )
Aplastic Anemia
Including Pure Red Cell
Aplasia, Congenital
Dyserythropoietic
Anemia, and Paroxysmal
Nocturnal
Hemoglobinuria
Sherrie L. Perkins, MD, PhD
Definition OBJECTIVES
Pathogenesis At the end of this chapter the learner should be able to:
Etiology
1. Define aplastic anemia.
Acquired Aplastic Anemia
2. Understand the etiologic subclassification of aplastic anemia.
Idiopathic or Primary
Secondary Causes 3 . List four causes of acquired aplastic anemia and identify the most common cause.
Congenital Aplastic Anemia 4. Name the most common congenital and acquired disorders associated with the
Fanconi’s Anemia development of aplastic anemia.
Clinical Manifestations of
Nn . Describe the clinical and laboratory features associated with aplastic anemia.
Aplastic Anemia
Laboratory Evaluation 6. Describe the bone marrow findings in aplastic anemia.
Treatment, Clinical Course, 7. List the treatment modalities used in aplastic anemia and identify the best therapy for
and Prognosis younger patients.
Related Disorders 8. List clinical and laboratory features associated with pure red cell aplasia.
Pure Red Cell Aplasia
9. Describe the common characteristics seen in congenital dyserythropoietic anemias
Congenital
(CDAs).
Dyserythropoietic
Anemias 10. Describe the clinical and laboratory features associated with paroxysmal nocturnal
Paroxysmal Nocturnal hemoglobinuria (PNH).
Hemoglobinuria
Laboratory Evaluation of
PNH
Case Study 1
Case Study 2
156
Chapter 8 Aplastic Anemia = 157
Definition
“ ‘Table é
e-1 Postulated Pathogenic
Aplastic anemia is a bone marrow failure disorder (or group 3 _ Mechanisms in 3
of disorders) characterized by cellular depletion and fatty Laan of Aplastic
replacement of the bone marrow. The concomitant decreases
Anemia — ‘
in hematopoietic progenitors lead to diminished production of
erythrocytes, leukocytes, and platelets and development Tee or Unknown
of peripheral blood cytopenias or pancytopenia. The loss Direct Bone Marrow Toxic Effects
Radiation
of functional bone marrow may occur following a variety of
Drugs (i.e., chemotherapy drugs, other drugs)
bone marrow insults that include drugs, chemicals, irradia- Benzene
tion, infections, and immune dysfunction. Though the inciting Toxins or chemicals
mechanisms vary, all lead to the loss of bone marrow precur- Starvation
sor cells or damage of the bone marrow microenvironment Some bone marrow infections
required to sustain bone marrow cell growth and differentia- Immune-Mediated Bone Marrow Damage
tion. Thus, the hematopoietic progenitor cells that give rise to Drug-induced autoantibodies
the various peripheral blood elements lose their ability to self Autoimmune disorders
renew and produce progeny. This leads to a loss of bone Pregnancy
Some bone marrow infections
marrow cellular mass and bone marrow failure. Clinical crite-
ria that have been used to define aplastic anemia include Congenital Bone Marrow Defects
(1) marrow of less than 25% normal cellularity and (2) at least Fanconi’s anemia
two blood cytopenias defined as neutrophil count less than Dyskeratosis congenita
500 per microliter or platelets less than 20,000 per microliter
or anemia with corrected reticulocyte count of less than 1%.'
BONE MARROW
Self-renewal
1
Agents That Regularly Produce Bone Marrow Hypoplasia with Sufficient Doses
Ionizing radiation
Benzene and benzene derivatives
Chemotherapeutic agents (e.g., busulfan, vincristine)
drug received, and drug-induced antibodies have been identi- incidence and predictability of bone marrow suppression vary
fied in only a few patients. Thus, the association between the with the type of drug (see Table 8-4). For example,
drug and development of aplastic anemia is dependent on epi- chemotherapeutic agents are well known to regularly cause
demiologic data and a temporal relationship to drug ingestion bone marrow hypoplasia in a dose-related manner. Other
and development of bone marrow failure.'* The mechanism of drugs (antibiotics, anticonvulsants, analgesics) are much less
drug-induced bone marrow failure suppression is usually predictable. Knowledge of potential bone marrow side effects
unknown, and it is impossible to identify which patients will must be kept in mind when using these drugs and appropriate
react adversely to a drug. Luckily, such idiosyncratic reac- monitoring of peripheral blood indices performed. Often,
tions to drugs are relatively rare. It is estimated that 1 person drug-induced bone marrow hypoplasia is fully reversible on
in 20,000 to 30,000 may have an idiosyncratic reaction to removal of the drug. A small minority of patients may develop
chloramphenicol, which is about 10 times the incidence of irreversible damage.*°"*
developing aplastic anemia for the general population not
taking chloramphenicol.'°
Chloramphenicol has been shown to cause two types
of bone marrow effects.'° The most common reaction is a
reversible bone marrow suppression that occurs while the
patient is receiving the drug and is associated with vacuoliza-
tion of bone marrow precursor cells (Fig. 8—2) and increased
serum iron levels, reflecting ineffective erythropoiesis. The
second reaction seen is development of an irreversible aplas-
tic anemia that occurs weeks to months after drug exposure.
This more severe reaction is not predictable on the basis of
the dose, duration, or route of administration of the drug.
Because of the strong association with development of aplas-
tic anemia, chloramphenicol use has decreased. Currently the
drug is administered only for specific indications when no
other reasonable alternative exists.'>'°
A wide variety of other drugs have been implicated as Figure 8-2 @ Vacuolization of bone marrow hematopoietic
direct suppressors of hematopoiesis and are occasionally precursor cells indicating toxicity in a patient being treated with
associated with development of bone marrow aplasia. The chloramphenicol. (Wright-Giemsa stain, X 1000 magnification)
160 Chapter 8 Aplastic Anemia
being the most notable (Table 8—6). The hemoglobin concen- Table 8-6 Characteristic Abnormal
tration is usually 70 g/L or lower, and the anemia is usually
CBC Values Seenin
normochromic and normocytic, although the cells may
occasionally be macrocytic with moderate anisocytosis and
Severe Aplastic Anemia
poikilocytosis. The corrected reticulocyte count is character- Red Blood Cells
istically low (less than 1%, or less than 25 Xx 10°/L absolute Hematocrit = 0.20-0.25 (L/L) or 20%-25%
reticulocyte count), reflecting the lack of bone marrow regen- Hemoglobin concentration = 70 g/L
erative activity. The white cells, in particular the myeloid and Absolute reticulocyte count = 25 = 10°/L
Corrected reticulocyte count < 1%
monocytic cells, as well as platelets are decreased. Lympho-
cytes may be normal or decreased in number. White Blood Cells
There is no specific morphological abnormality of the Total leukocyte count S$ 1.5 x 10°/L
Absolute neutrophil count = 0.5 x 10°/L
blood smear that is diagnostic of aplastic anemia. Examina-
tion of the blood smear confirms a normochromic, normo- Platelets
cytic anemia with little or no evidence of regeneration such as Platelet count = 20-60 x 10°/L
polychromatophilic cells, basophilic stippling, or nucleated
162 Chapter 8 Aplastic Anemia
Figure 8-7 © Focal area of bone marrow hyperplasia adjacent to a Related Disorders
hypoplastic area in early aplastic anemia. (H & E stain, 100
magnification) Pure Red Cell Aplasia
Pure red cell aplasia is an uncommon disorder in which the
erythroid cells in the bone marrow are selectively destroyed.
Treatment, Clinical Course, This gives rise to an anemia without other associated cytope-
nias. More than 900 cases have been reported in the world’s
and Prognosis literature. This may be an acquired or congenital process
Untreated aplastic anemia has an extremely poor prognosis, as (Table 8-8). Pure red cell aplasia is characterized by severe,
the patients undergo progressive decreases in blood counts and chronic, normocytic to slightly macrocytic anemia. Reticulo-
subsequent lethal infection or bleeding.'*°*° However, some cytes are decreased and may be absent. There is no evidence
patients recover spontaneously. Until the early 1970s, patients of hemolysis or hemorrhage. White cells and platelets are
with severe aplastic anemia were treated via supportive trans- normal. A bone marrow biopsy usually demonstrates normal
fusions, possible treatment with androgens or anabolic steroids cellularity with a notable absence of erythroid precursor cells.
to stimulate hematopoiesis, and an extensive search for the Erythropoietin levels are often markedly increased as the
possible etiologic agent to prevent further exposure. Even so, body attempts to compensate for the profound anemia,
the course was usually progressive, with a 5-year mortality although cases arising secondary to development of anti-
rate of about 70% and only 10% of patients fully recovering.”’ erythropoietin antibodies (discussed further later in this chap-
Currently, the treatment of choice for aplastic anemia in ter) may have low or absent erythropoietin levels.****
patients younger than 50 years of age is allogeneic bone Acquired causes of pure red cell aplasia are most
marrow or peripheral stem cell transplantation. This therapy common. These may be short-lived acute illnesses or have a
is optimal if bone marrow from an HLA-matched sibling is more prolonged chronic course. Viral illnesses have been
used, although unrelated donors are becoming more readily associated with development of a transient cessation of red
164 Chapter 8 Aplastic Anemia
ong niitalDyse16
onge etic Anem jas
rythropcDie
Typeii
Inheritance cn Autosomal recessive Autosomal recessive Familial: autosomal dominant
Sporadic: variable
Red cells Macrocytic Normocytic Macrocytic
Anemia Mild to severe Mild to moderate Mild to moderate
Light microscopy-marrow Intranuclear chromatin bridges Binucleation Giant erythroblasts
Electron microscopy “Swiss cheese” nuclei Peripheral cisternae No specific findings
Acidified serum lysis Negative Positive Negative
Genetic localization 15q15. -15.3 20q11.2 Familial: 15q22
Sporadic: variable
Postulated biochemical defect Unknown Underglycosylation of band 3 Unknown
Associated abnormalities Skeletal defects, skin None Familial: visual defects
hypopigmentation monoclonal gammopathy
Sporadic: association with
lymphoma, mental
retardation
166 Chapter 8 Aplastic Anemia
red cell membrane. This causes the red cells to be highly sen- marrow hypoplasias.**° Usually patients presenting with
sitive to complement-mediated hemolysis. As this is a stem hemolytic type PNH have small base pair deletions (1 to 14 base
cell disorder, abnormalities are seen in leukocytes and pairs) or insertions (1 to 8 base pairs) or substitutions that lead
platelets, as well as in red cells. PNH is characterized by to DNA reading frameshifts. These may occur throughout the
recurrent, episodic intravascular hemolysis, hemoglobinuria, PIG-A_ gene.’ PNH arising in association with aplastic
and venous thrombosis. Hemolysis may be worse at night due anemia will have similar mutations but may also demonstrate
to decreased plasma pH, or may be more chronic in nature.°?*? tandem duplications and insertions that are significantly
PNH is also strongly associated with aplastic anemia. In larger in size than those seen in PNH.***° Patients with
aplastic anemia atypical red cell clones with a PNH pheno- combined aplastic anemia and PNH are more likely to have
type may develop and many cases of PNH may evolve into multiple mutations at many sites and a variety of different
aplastic anemia or bone marrow failure.°°**° mutational types.°°°’ Within the bone marrow, the cells con-
PNH arises secondary to a somatic mutation in the bone taining the P/G-A mutation may be a minority of bone
marrow pluripotential stem cell that gives rise to complete or marrow cells or the mutation may be expressed in nearly all
partial deficiencies of cell surface glycophosphatidylinositol hematopoietic cells.°*°
(GPI) anchor proteins. The mutated gene is phosphatidylinos- The GPI anchoring defect seen in PNH causes variable
itolglycan A (PIG-A), which is located on the X chromosome. deficiencies in at least 17 cell surface proteins that may affect
Similar mutations are seen in patients with bone marrow the function of hematopoietic cells (Table 8-11). Included in
failure syndromes, such as aplastic anemia, or other bone these proteins are two to three membrane glycoproteins that nor-
mally regulate complement fixation and activation on the cell
surface. These proteins include decay-accelerating factor (DAF
or CD55), which regulates the activity of C3 convertase, homol-
ogous restriction factor (HRF), which regulates C9 binding and
activity; and membrane inhibitor of reactive lysis (MIRL or
CD59), which modulates complement mediated red blood cell
lysis. A deficiency in one or more of these proteins leads to
enhanced formation of membrane complement lytic com-
plexes on the cell surface and increased complement-mediated
hemolysis (Fig. 8-11). White blood cells and platelets also
demonstrate abnormal cell surface GPI anchoring protein
levels; however, it is the red cell deficiency that gives rise to the
classic episodic intravascular hemolysis. In all cell types there
may be variable expression of the abnormal phenotypes, per-
haps reflecting genetic mosaicism. Thus some cells express
normal levels of the GPI anchoring proteins and other cells
express lesser levels of these proteins or they may be completely
absent. This has led to description of three different subtypes of
PNH cells (Table 8—12), and the relative numbers of each PNH
cell type will correlate with disease severity. A patient may have
some or all of these cellular subtypes present and the numbers
of each subtype may shift over the course of the disease.5?4
Clinically PNH has an estimated prevalence of 1 to
10 cases per million population. It is seen primarily in adults but
Figure 8-10 @ Binucleated erythroid precursor with chromatin has also been described in children and adolescents.>25* The dis-
bridge seen in a patient with type | congenital dyserythropoietic ease displays a wide spectrum of symptomatology and usually
anemia. (Wright-Giemsa stain x 1000 magnification) begins as an insidious onset of anemia. Virtually all patients are
Chapter 8 Aplastic Anemia 167
Panel A Panel B
cg
complement
complex
Other Receptors
Urokinase receptor
EONS sis Figure 8-11 @ A. The normal cell is protected from C3b binding
Enzymes and complement-mediated hemolysis because the PIG-A anchored
Alkaline phosphatase proteins CD59 and CD55 are present and thus complement fixation
Acetylcholinesterase cannot occur. B. The PNH cell has decreased CD55 and CD59 pro-
5’-Ectonucleotidase tein on the cell surface due to lack of GP! anchoring proteins,
allowing for C3b binding and complement fixation that leads to red
Other Proteins cell lysis. (Adapted from Kinoshita, T, and Inoue, N: Relationship
CD24 between aplastic anemia and paroxysmal nocturnal hemoglobinuria.
CD48 Int |Hematol 75:117, 2002.)
CD52 (Campath-1)
CD66c 2
CD67 anemic, often with severe anemia. The classically described pat-
JMH-bearing protein tern of episodic hemolysis at night causing dark urine upon
awakening is frequently not seen. Hemolysis is more likely to
occur in an irregular fashion, precipitated by infection, surgery,
and transfusion. Hemolysis may also be occurring chronically
throughout the day. Hemoglobinuria or dark urine may be pre-
sent or absent. Chronic urinary iron loss or hemosiderinuria is a
constant feature and may lead to an iron-deficiency anemia.
Chronic hemolysis may also result in chronic renal failure due erythrocytes present. Thus hemoglobin levels may vary from
to renal tubular damage.**°° normal to <6 g/dL. Morphologically the red cells appear
Abnormal platelet function in PNH patients is frequently normochromic and normocytic without significant abnormal-
associated with venous thromboses. These are a common ities. Occasionally slight macrocytosis and polychromasia
cause of death in this patient population, and may affect up to may occur owing to increased reticulocytes or red cells may
one third of patients in a significant manner. Thrombotic be hypochromic and microcytic due to iron deficiency sec-
events may cause severe abdominal or back pain or severe ondary to hemosiderinuria or hemoglobinuria (Fig. 8-12).
refractory headaches. Development of Budd-Chiari syndrome The increased reticulocytosis noted in association with PNH
(hepatic vein thrombosis), mesenteric vein, or cerebral vein is in direct contrast to the hypoproliferative response seen
thromboses are most common. Thrombophlebitis may also with aplastic anemia and reflects the marrow response to
occur, leading to pulmonary thromboembolism. Arterial hemolysis. However, although the reticulocyte count is usu-
thromboses are rare. Rare patients may experience bleeding ally elevated (5% to 10%), the absolute reticulocyte count
due to poor platelet function.**°* may be low for the degree of anemia, reflecting the abnormal
Patients are often leukopenic at some time during the hematopoietic state. Occasional nucleated red blood cells
course of their disease. This will lead to increased suscepti- may be seen, particularly in episodes of severe hemolysis.
bility to infection and sepsis. Often leukocytes will show Spherocytes are typically absent, although occasional schisto-
decreased leukocyte alkaline phosphatase (LAP) activity. In cytes may be seen on the smear, particularly in the presence
addition to functional defects in the leukocytes, patients of intravascular thrombosis.°**!
also develop leukopenia in addition to thrombocytopenia White blood cells, in particular granulocytes, and platelets
and anemia. Treatment with cytokines or transfusions is have the same membrane defects seen in the red cells that will
usually indicated. render them more sensitive to complement lysis. Granulocytes
As noted in the preceding text, aplastic anemia may pro- appear morphologically normal. However, neutropenia is often
ceed or coexist with PNH.'°*°S Patients with aplastic anemia observed and the neutrophils will have decreased leukocyte
often develop abnormal hematopoietic cell clones that have a alkaline phosphatase activity. Platelet counts also vary in
PNH phenotype and are associated with mutations in the P/G- PNH with mild to moderate thrombocytopenia and platelet
A gene. It should be noted that small numbers of abnormal, counts ranging from 50 to 100 x 10°/L commonly observed.
PIG-A-deficient clones may also be seen in normal marrows. Although thrombocytopenia is often present, the most common
This suggests the possibility of a survival advantage for these platelet dysfunction is abnormal clotting with increased venous
abnormal hematopoietic cells in a patient with aplastic thromboses.°**!
anemia. It has been hypothesized that the PNH phenotype The bone marrow often shows erythroid hyperplasia in
may allow the hematopoietic precursor to escape immune- response to chronic hemolysis (Fig. 8-13). As noted in the
mediated destruction. In contrast, the P/G-A—deficient clones preceding text, some patients with PNH will develop
do not have a survival advantage over normal hematopoietic hypoplastic or aplastic marrows. Often adequate numbers of
cells, resulting in the mosaicism characteristically seen in myeloid and megakaryocytic precursors are present. Bone
PNH. A recent study showed up to 89% of patients with aplas- marrow iron stains usually demonstrate decreased iron.°!
tic anemia had some number of PNH-phenotype granulocytes Diagnostic testing for PNH includes demonstration of
present at the time of diagnosis.°’ Similar other studies have hemosiderin in the patient’s urine by staining with Prussian
shown 50% to 60% involvement by the abnormal PNH-like
clone in newly diagnosed aplastic anemia.°»*>>
In addition to the linkage between aplastic anemia and
PNH, there also appears to be some overlap with large gran-
ular lymphocytic leukemias. Often PNH patients will show
clonal large granular lymphocyte expansions, and this may
reflect the abnormal immune system that is associated in this
disorder. Large granular lymphocyte expansions are also seen
in aplastic anemia, and it has been recently hypothesized that
the expansion of the GPI deficient clones may be due to an
attempt by the marrow progenitor cell to escape antigen
driven immune attack.°’° Thus, the pathophysiologic under-
pinnings of aplastic anemia, PNH, and other bone marrow
failure syndromes appear to be closely related and may have
similar or overlapping pathophysiologic features.°°!
Laboratory Evaluation of PNH Figure 8-12 ® Peripheral blood smear from a patient with PNH
(Wright-Giemsa stain, x600 magnification) demonstrating
Characteristic laboratory findings in PNH include anemia,
normochromic, normocytic red cells, as well as occasional
leukopenia, and thrombocytopenia. The anemia may be mild hypochromic, microcytic, and polychromatophilic cells. A nucleated
to severe depending on the number and subtype of PNH type red cell is also seen.
Chapter 8 Aplastic Anemia 169
Figure 8-13 @ Bone marrow aspirate smear from a patient with 1. Hemolysis occurs with the patient’s cells and not with con-
paroxysmal nocturnal hemoglobinuria demonstrating erythroid trol cells.
hyperplasia. (Wright-Giemsa stain, x 500 magnification) NO. Hemolysis is enhanced by acidified serum and does not
occur with heat-inactivated serum (heating to 56°C for
30 minutes to inactivate complement) (Table 8-13 and
blue. Hemoglobinuria, when present, will be detected by dip-
Fig. 8—15).°*°!
stick. This must be coupled with microscopic examination to
determine that intact red cells are absent. Occasionally patients A very specific test for the diagnosis of PNH is the use of flow
will show hemoglobin casts, and this is often associated with cytometry to assess the presence or absence of GPI-anchoring
renal failure. Because of ongoing hemolysis, indirect bilirubin proteins. This method is very sensitive and specific, utilizing
will often be increased, as will plasma hemoglobin, while hap- monoclonal antibodies against CD55 and CD59 proteins that
toglobin is decreased. The direct anti-globulin test (Coombs’ modulate complement activity. Detection of decreased or
test) will be negative.*°! absent expression of these proteins by flow cytometric analy-
Several diagnostic tests have been developed for the sis 18 diagnostic of PNH. CD59 is usually present in high lev-
determination of PNH. Initial screening is usually done with a els on the red cell membrane with somewhat lower levels of
sugar water test (sucrose hemolysis test) when a diagnosis of CD55. The patient’s red blood cells can be stained with
PNH is considered. Placing blood into a sucrose-containing fluorochrome-labeled anti-CD59 or anti-CD55 for flow cyto-
medium of low ionic strength will promote the binding of metric analysis. Patients with PNH will show variably
complement, especially C3, to the red cell membrane. This decreased to absent expression of CD59 and/or CD55, which
low ionic strength solution will activate the classic or alterna- is manifested by a shift in the peak channel of fluorescence.
tive complement lysis pathways leading to red cell hemolysis. In most patients, a population of normal cells is often com-
Normal cells should be unaffected (Fig. 8-14). Most monly seen, reflecting phenotypic mosaicism (Fig. 8-16).
patients with PNH will show 10% to 80% red cell lysis. Less The degree of CD55 or CD59 deficiency is often associated
Patient’s serum
0.2 N HCl
CD55 CD59
300 300
240 240
S 180 Sa 100
PNH 8 8
S 120 | M1 8 120 | M1
60 60
0 0
100 7) 10%. "O02 9 "102% ~ 104 10° 101 102 103 104
A Log fluorescence B Log fluorescence
300 300
240 240
S 180 § 180
Normal & 8
S 120 M1 S 120 M1
60 60
0 0
10° 10! 10? 103 102 10° 10! 10° 108 104
C Log fluorescence D Log fluorescence
Figure 8-16 @ Flow cytometry diagnosis of PNH. The flow cytometric histograms from a patient with PNH (A, B) show decreased expression
of CDSS (A, arrow) and CD59 (B, arrow) in addition to populations of red cells containing relatively normal levels of CD55 and CD59. For
comparison, a normal control showing single red cell populations for both CD55 (C) and CD59 (D). (From Smith, LJ: Paroxysmal nocturnal
hemoglobinuria. Clin Lab Sci 17:172, 2004, with permission.)
Chapter 8 Aplastic Anemia ‘1171
Case Study 2
was no history of recent exposure to drugs, toxins, or radia-
tion. There was no history of any other illness, and she had
been in good health until the past 2 weeks.
Laboratory data revealed a normochromic, normocytic A 43-year-old man presented to his physician with com-
anemia with a hematocrit of 20%, WBC of 20 x 10°/L, and plaints of lower back pain, fatigue, easy bruising, and a sud-
platelets of 20 X 10°/L. The peripheral blood showed den onset of dark urine on arising in the morning. Laboratory
9% neutrophils, 90% lymphocytes, and 1% monocytes. The studies and a bone marrow aspirate were ordered by his
corrected reticulocyte count was 0.5%. A bone marrow aspi- physician. The CBC revealed an anemia (hematocrit, 28%),
rate and biopsy specimen was markedly hypocellular, with leukopenia (white blood cell [WBC] count 33 x 10°/L), and
less than 5% cellularity composed of lymphocytes and thrombocytopenia (platelets, 76 * 10°/L). The reticulocyte
plasma cells admixed with a rare myeloid or erythroid precur- count was 5.1%, (3.2% corrected). The RBCs showed mod-
sor. No megakaryocytes were noted. No dysplastic changes erate polychromasia, slight anisocytosis, and poikilocytosis,
were noted in the few hematopoietic cells seen, and there was and | nucleated RBC per 100 WBCs. The chemistry profile
no increase in the number of blasts. Further studies failed to was normal except for an increased lactate dehydrogenase
identify an underlying etiology for the patient’s pancytopenia. value and an increased indirect bilirubin. The bone marrow
The patient was followed for the next 3 months with no reso- analysis revealed a hypercellular marrow with relative ery-
lution of her symptoms and no improvement in her blood throid hyperplasia. There were adequate numbers of myeloid
count. She received two transfusions of red blood cells as a and platelet precursors.
result of worsening anemia. Further studies were performed after initial test results
The patient’s family was tested, and she was found to were evaluated. The urinary hemosiderin, sugar water test,
have a sister who was HLA compatible. The sister had and the Ham’s test were all positive. Flow cytometry
a collection of peripheral stem cells for an allogeneic trans- showed a population of red cells with decreased expression
plantation. The stem cells were infused into the patient of CD55 and CD59.
continued continued
172 Chapter 8 Aplastic Anemia
QUESTIONS
te What is the most likely diagnosis?
2 . What type of hemolysis is evident in this patient?
3: If the granulocytes in this patient were scored for LAP
activity, what would you expect the result to be?
4. What is the clinical outcome in this patient?
ANSWERS
. Paroxysmal nocturnal hemoglobinuria.
. Intravascular hemolysis.
. LAP would be expected to be decreased.
. Clinical outcome is variable but patients often have sig-
BRWN
Questions
k How isaplastic anemia best defined? ae Always causes fatal, severe aplastic anemia
a. A condition in which bone marrow production of = d. May cause a spectrum of disease that may not manifest
cells, white blood cells, and platelets has failed until several years following the benzene exposure
b. A condition in which severe anemia is seen
6. Which of the following statements about chloramphenicol-
c. A condition in which platelets are decreased
induced aplastic anemia is true?
d. A condition in which there is pancytopenia with a
a. The onset of the bone marrow aplasia is not pre-
hypercellular bone marrow
dictable by the dose or the duration of drug exposure
. Which is the most common cause of aplastic anemia? b. Low doses of chloramphenicol never lead to aplastic
a. Drug ingestion anemias
b. Toxin exposure c. People who receive chloramphenicol are 500 times
c. Idiopathic or unknown more likely to develop aplastic anemia than the gen-
d. lonizing radiation eral population
. Which of the following represents the most complete list d. The longer a patient receives chloramphenicol, the more
of etiologies causing aplastic anemia? likely it is that he or she will develop aplastic anemia
a. Secondary and congenital . Which of the following drugs is not associated with the
b. Idiopathic and congenital development of aplastic anemia?
c. Secondary and idiopathic a. Chloramphenicol
d. Secondary, idiopathic, and congenital b. Phenylbutazone
. Which of the following has not been associated with an c. Aspirin
acquired type of aplastic anemia? d. Chemotherapeutic agents
a. lonizing radiation . lonizing radiation causes aplastic anemia by which of
b. Increased chromosomal breakage the following mechanisms?
c. Chemical agents a. A dose-dependent destruction of bone marrow stem
d. Drugs cells
. The aplastic anemia associated with benzene exposure is b. An idiosyncratic delayed development of bone
characterized by which of the following statements? marrow aplasia
a. Always occurs while the exposure to benzene is c. Disruption of chemical bonds to form free radicals
occurring that damage bone marrow cells
b. Is always irreversible d. All of the above
Chapter 8 Aplastic Anemia 173
. What is the most common congenital disorder associated 16. Which statement best describes paroxysmal nocturnal
with aplastic anemia? hemoglobinuria?
a. Fanconi’s anemia a. Acquired hemolytic anemia associated with cellular
b. Thrombocytopenia-absent radius (TAR) syndrome membrane abnormalities
c. Congenital dyserythropoietic anemia, type | b. Congenital hemolytic anemia associated with the
d. Diamond—Blackfan anemia inflammatory response
10. Which of the following is not seen in the peripheral c. A premalignant condition that almost always results in
blood of patients with aplastic anemia? development of acute leukemia.
a. Normochromic, normocytic anemia d. A common disorder that frequently resolves with time.
b. Increased reticulocyte count . What causes the red cell defect of PNH?
c. Relative lymphocytosis a. Rare red cell antigens
d. Decreased neutrophils b. Lack of GPI-anchored proteins on the erythrocyte
ke What is the appearance of the bone marrow in aplastic membrane
anemia? c. Excessive amounts of complement components
a. Hypercellular Ca 10. C9
b. Normocellular d. Glucose-6-phosphate dehydrogenase enzyme
c. Hypocellular deficiency
d. Fibrotic - 18. Which of the following is a correct description of the
|W, The differential diagnostic considerations for bone mar- sugar water test (sucrose hemolysis test)?
row hypoplasia do not include which of the following a. PNH cells are lysed by complement after exposure to
disorders? low-ionic-strength sugar water.
a. Severe malnutrition b. PNH cells are lysed by antibody and complement
b. Myeloproliferative disorder after heating to 56°C in sugar water solution (5%).
c. Aplastic anemia c. Patient’s serum is acidified to enhance complement
d. Recent chemotherapy binding and lysis of patient cells.
d. Patient’s serum is heat-inactivated and treated with
is What is the treatment of choice for severe aplastic ane- HCl; complement is added; patient cell lysis occurs.
mia in patients who are younger than age 50?
a. Multiple transfusions 19) What is a correct description of Ham’s test (acidified
b. Androgens serum lysis test)?
c. Bone marrow transplantation a. PNH cells are lysed by complement after exposure to
d. Erythropoietin therapy low-ionic-strength sugar water.
b. PNH cells are lysed by antibody and complement
14. What is the definition of pure red cell aplasia? after heating to 56°C in sugar water solution (5%).
a. Lack of hematopoietic precursors in the bone marrow c. Patient’s serum is acidified to enhance complement
b. Abnormal, giant normoblasts in the bone marrow binding and lysis of patient cells.
c. Lack of erythroid precursors with normal white blood d. Patient’s serum is heat inactivated and treated with
cell and megakaryocytic precursors HCI; complement is added; patient cell lysis occurs.
d. Dysplastic red cell precursors with normal white cell
. The basis of the flow cytometric test for diagnosis of
and megakaryocytic precursors
PNH is which of the following?
LS: What features do the congenital dyserythropoietic ane- a. PNH will have increased amounts of complement
mias (CDAs) have in common? detected on the cell surface.
a. Anemia, microcytosis, erythroid hyperplasia, and b. PNH cells are easily lysed and will show decreased
abnormal erythroblasts number when analyzed.
b. Anemia, erythroid hyperplasia with abnormal c. An affected patient will show decreased levels of
erythroblasts, and indirect hyperbilirubinemia CD55 and CD59 binding in a subset of cells.
c. Anemia, lysis in acidified serum, and indirect hyper- d. All of the patient cells will show decreased levels of
bilirubinemia GPI-anchored proteins on the erythrocyte membrane.
d. Anemia, macrocytosis, erythroid hyperplasia with
abnormal erythroblasts, and indirect hyperbilirubinemia See answers at the back of this book.
174. Chapter 8 Aplastic Anemia
REFERENCES . Keohane, EM: Acquired aplastic anemia. 10. Goldstein, BD: Benzene toxicity. Occup
: } F Clin Lab Sci 17:165, 2004. Med 3:541, 1988.
L. nek oe ops ane mada . Nebert, DW, et al: NAD(P)H:quinone 11. Farris, GM, et al: Benzene-induced
P Macieienstl IP ea une a oxidoreductase (NQO1) polymorphism, hematotoxicity and bone marrow com-
cn Roar a ae exposure to benzene, and predisposition pensation in B6C3F1 mice. Fund Appl
SOG i area a ae to disease: A HuGE review. Genet Med Toxicol 36:119, 1997. -
3K ga Ss: Piri acoiz a n sais
See 4:62, 2002. 12. Watanabe, , KH, KH, et al:: B Benzene toxicoki
icoki-
Rieter aut Sie ones . Rangan, U, and Snyder, R: Scientific netics in humans: exposure of bone mar-
eS oueri P update on benzene. Ann NY Acad Sci row to metabolites. Occup Environ Med
nt JHematol 68:19, 1998. 837:105. 1997 51:414. 1994
4. Marsh,
arsh,
JC:
JC: Bone ‘failure
marrow failure syn
syn- Pao we
. Santesson, G: Uber chronische Set cae
P
dromes. Clin Med 5:332, 2005. 13. Smith, MT, et al: Increased transloca-
vergiftungen mit steinkolen benzin. tions and aneusomy in chromosomes
Nn . Dokal, I: Inherited aplastic anaemia.
Vier todes falle. Arch Hygiene 31:336, 8 and 21 among workers exposed to ben-
Hematol J 4:3, 2003.
1897. zene. Cancer Res 58:2176, 1998.
Chapter 8 Aplastic Anemia 175
. Malkin, D, et al: Drug-induced aplastic faces in the crowd. Genes Dev Sl. Heimpel, H: Congenital dyserythropoi-
anemia: Pathogenesis and clinical IES), PAOD). etic anemias: Epidemiology, clinical sig-
aspects. Am J Pediatr Hematol Oncol 34, Slavin, S, et al: Reduced-intensity condi- nificance, and progress in understanding
12:402, 1990. tioning for the treatment of malignant their pathogenesis. Ann Hematol 83:613,
. Flegggg, P, et al: Chloramphenicol. Are and life-threatening non-malignant disor- 2004.
concerns about aplastic anaemia justi- ders. Clin Transpl 275, 2003. . Smith, LJ: Paroxysmal nocturnal hemo-
fied? Drug Saf 7:167, 1992. 35). Walne, AJ, et al: Dyskeratosis congenita: globinuria. Clin Lab Sci 17:172, 2004.
. Yunis, AA: Chloramphenicol toxicity: A disorder of defective telomere mainte- . Perkins, SL: Paroxysmal nocturnal
25 years of research. Am J Med 87:44N, nance? Int J Hematol 82:184, 2005. hemoglobinuria, ed 4. ASCP Press,
1989. 36. Locasciulli, A: Acquired aplastic anemia Chicago, 2006, pp 141-148.
. Chen, J: Animal models for acquired in children: Incidence, prognosis and treat- . Young, NS, et al: The relationship of
bone marrow failure syndromes. Clin ment options. Paediatr Drugs 4:761, 2002. aplastic anemia and PNH. Int J Hematol
Med Res 3:102, 2005. 37 . Geissler, K: Pathophysiology and treat- 76 (Suppl 2):168, 2002.
. Amrtland, H: Occupational poisoning in ment of aplastic anemia. Wien Klin 5) Gordon-Smith, EC, et al: Views on the
manufacture of luminous watch dials. Wochenschr 115:444, 2003. pathophysiology of aplastic anaemia. Int
JAMA 92:466, 1929. 38. Kurre, P, et al: Diagnosis and treatment J Hematol 76(Suppl 2):163, 2002.
. Lavin, MF: Radiation-induced cell death of children with aplastic anemia. Pediatr 56. Kinoshita, T, and Inoue, N: Relationship
and its implications in human disease. Blood Cancer 45:770, 2005. between aplastic anemia and paroxysmal
Results Probl Cell Differ 24:213, 1998. . Deeg, HJ, et al: Long-term outcome after nocturnal hemoglobinuria. Int J Hematol
. Cronkite, EP: Radiation-induced aplastic matrow transplantation for severe aplas- TRAN, BOWL.
anemia. Semin Hematol 4:273, 1967. tic anemia. Blood 91:3637, 1998. Dh Socie, G, et al: Late clonal diseases of
. Brown, KE, et al: Hepatitis-associated AO. Gillio, AP, et al: Comparison of long-term treated aplastic anemia. Semin Hematol
aplastic anemia. N Engl J Med outcome of children with severe aplastic 37:91, 2000.
SOMOS, MIN anemia treated with immunosuppression 58. van den Heuvel-Eibrink, MM, et al:
. Baranski, B, et al: Epstein-Barr virus in versus bone marrow transplantation. Biol Childhood paroxysmal nocturnal haemo-
the bone marrow of patients with aplastic Blood Marrow Transplant 3:18, 1997. globinuria (PNH), a report of 11 cases in
anemia. Ann Intern Med 109:695, 1988. 4 mk. Kumar, M, and Alter, BP: Hematopoietic the Netherlands. Br J Haematol 128:571,
. Young, N, and Mortimer, P: Viruses and growth factors for the treatment of aplastic 2005.
bone marrow failure. Blood 63:729, 1984. anemia. Curr Opin Hematol 5:226, 1998. Se): Wang, H, et al: Relative increase of
24. Torok-Storb, B, et al: Increased prevalence 42. Perkins, SL: Pediatric red cell disorders granulocytes with a paroxysmal noctur-
of CMV gB3 in marrow of patients with and pure red cell aplasia. Am J Clin nal haemoglobinuria phenotype in aplas-
aplastic anemia. Blood 98:891, 2001. Pathol 22(Suppl):S70, 2004. tic anaemia patients: The high preva-
Ue Knobel, B, et al: Pancytopenia—a rare . Lim, LC: Acquired red cell aplasia in lence at diagnosis. Eur J Haematol
complication of miliary tuberculosis. Isr association with the use of recombinant 66:200, 2001.
J Med Sci 19:555, 1983. erythropoietin in chronic renal failure. 60. Risitano, AM, et al: Large granular lym-
. Nakao, S, et al: Immune pathophysiology Hematology 10:255, 2005. phocyte (LGL)-like clonal expansions in
of aplastic anemia. Int J Hematol 82:196, 44. Chisaka, H, et al: Parvovirus B19 and paroxysmal nocturnal hemoglobinuria
2005. the pathogenesis of anaemia. Rev Med (PNH) patients. Leukemia 19:217, 2005.
Mie Choudhry, VP, et al: Pregnancy associated Virol 13:347, 2003. 61. Marsh, JC, and Elebute, MO: Stem cells
aplastic anemia—a series of 10 cases with 45, Thompson, DF, and Gales, MA: Drug- in paroxysmal nocturnal haemoglobin-
review of literature. Hematology 7:238, induced pure red cell aplasia. Pharma- uria and aplastic anaemia: Increasing
2002. cotherapy 16:1002, 1996. evidence for overlap of haemopoietic
28. Sanders, JE: Chronic graft-versus-host 46. Masuda, M, et al: Pure red cell aplasia defect. Transfus Med 13:377, 2003.
disease and late effects after hematopoi- with thymona: evidence of T-cell clonal 62. Krauss, JS: Laboratory diagnosis of
etic stem cell transplantation. Int J disorder. Am J Hematol 54:324, 1997. paroxysmal nocturnal hemoglobinuria.
Hematol 76(Suppl 2):15, 2002. 47. Ward, JH: Autoimmunity in chronic lym- Ann Clin Lab Sci 33:401, 2003.
. Comerci, GD: Medical complications of phocytic leukemia. Curr Treat Options 63. Parker, C, et al: Diagnosis and manage-
anorexia nervosa and bulimia nervosa. Oncol 2:253, 2001. ment of paroxysmal nocturnal hemoglo-
Med Clin North Am 74:1293, 1990. A8. Go, RS, et al: Aplastic anemia and pure binuria. Blood 106:3699, 2005.
. Smith, OP: Shwachman-Diamond syn- red cell aplasia associated with large 64. Meyers, G, and Parker, CJ: Management
drome. Semin Hematol 39:95, 2002. granular lymphocyte leukemia. Semin issues in paroxysmal nocturnal hemoglo-
. Tischkowitz, MD, and Hodgson, SV: Hematol 40:196, 2003. binuria. Int JHematol 77:125, 2003.
Fanconi anaemia. J Med Genet 40:1, 2003. 49. Willig, TN, et al: Diamond-Blackfan 65. Leach, A, and Gaumer, H: Diagnosis and
. Collins, N, and Kupfer, GM: Molecular anemia. Curr Opin Hematol 7:85, 2000. detection of PNH using GPI anchored
pathogenesis of Fanconi anemia. Int J . Wickramasinghe, SN, and Wood, WG: proteins. Clin Lab Sci 9:91, 1996.
Hematol 82:176, 2005. Advances in the understanding of the
. Kennedy, RD, and D’ Andrea, AD: The congenital dyserythropoietic anaemias.
Fanconi anemia/BRCA pathway: New Br JHaematol 131:431, 2005.
Chapter ‘
Se
Introduction to
Hemolytic Anemias
Intracorpuscular Defects:
I. Hereditary Defects of the Red Cell
Membrane
Théresa L. Coetzer, PhD
Stan Zail, MBBCh, MD, FRCPath
Hereditary Defects of the . Name laboratory tests that help to classify the cause of red cell hemolysis.
Red Cell Membrane
. Identify the red cell membrane abnormalities associated with hereditary spherocytosis.
Red Cell Membrane
Structure . Recognize abnormal laboratory results associated with hereditary spherocytosis.
Classification
. Name the functional abnormalities affecting membrane skeleton proteins in
Aa
oy
6b
Go
—
Hereditary Spherocytosis
hereditary elliptocytosis.
Case Study 1
Hereditary Elliptocytosis 8. Recall laboratory findings associated with hereditary elliptocytosis.
Clinical Phenotypes 9. Identify the abnormalities that cause the severe fragmentation and microspherocytosis
Case Study 2 characteristic of hereditary pyropoikilocytosis.
176
Chapter 9 Hemolytic Anemias: Intracorpuscular Defects: I. Hereditary Defects of the Red Cell Membrane —-177
R.E. CELL
Serum
Fe transferrin
Hb Globin Amino acid
Bilirubin pool
Hb
Bilirubin-albumin
Hb-dimer Met-Hb Hb-haptoglobin (unconjugated)
Ferri-heme + globin
Heme-
>
hemopexin
Methem-albumin ——~>
|
|
Hemoglobinuria
Hemosiderinuria
Urobilinogenuria
Figure 9-1 Diagrammatic representation of the degradation of hemoglobin after intravascular or extravascular destruction of red cells.
Fe = iron; Hb = hemoglobin; RBC = red blood cell; R.E. = reticuloendothelial cell.
Some of the free plasma hemoglobin may be oxidized to normal maturation time of | day, again leading to a falsely
methemoglobin with subsequent dissociation of ferriheme, elevated reticulocyte count. These cells (so-called shift retic-
which combines with albumin to form methemalbumin. ulocytes) are recognizable as large bluish-gray erythrocytes
Methemalbumin can be detected spectroscopically by the on Romanowsky (Wright’s, Giemsa) stains.
Schumm’s test. This test is relatively insensitive and is seldom The reticulocyte production index (RPI) corrects the
positive in mild hemolytic states. In routine practice, determi- hematocrit to a normal value of 45% and takes into account
nation of red cell survival using Cr°'-labeled red cells is seldom the maturation time of the reticulocyte at a particular hemat-
required to document an increased rate of red cell destruction. ocrit (approximately 1.0 day at a hematocrit of 45%, 1.5 days
The fate of hemoglobin when processed intravascularly or at 35%, 2.0 days at 25%, and 2.5 days at 15%).!
extravascularly is shown diagrammatically in Figure 9-1.
RPI = % Reticulocytes xX Hematocrit
Reticulocyte maturation time 45
TESTS REFLECTING INCREASED RED CELL
PRODUCTION For example, an RPI of 5.3 is calculated for a patient
The compensatory bone marrow response to hemolysis suspected of having a hemolytic state with the following
results in the delivery of young red cells in the form of retic- indices: hemoglobin, 12.0 g/dL; hematocrit, 36%; reticulo-
ulocytes into the circulation. These young cells contain RNA, cyte count, 10%; shift cells present.
which stains supravitally with dyes such as new methylene An RPI of greater than 2.5 to 3.0 is generally regarded as
blue or brilliant cresyl blue. The normal reticulocyte count indicative of a hemolytic state, but it is very important to
has a range of 0.5% to 2.0%, reflecting the fact that each day exclude the presence of hemorrhage in a particular patient, as
approximately 1% of the red cell mass is destroyed and this too may lead to an elevated RPI. Although the RPI is
replaced by young red cells from the bone marrow, because probably the single most useful test to detect a hemolytic
red cell survival is approximately 120 days. The reticulocyte state, a cautionary note is in order, as the test may not be sen-
count is always elevated in a hemolytic state in which there is sitive enough to detect mild hemolytic states (see Chap. 31).
a normal compensatory bone marrow response. However, a
more accurate assessment of red cell production is required,
Establishing the Cause of Hemolysis
because the percentage of reticulocytes may be “spuriously”
elevated as the reticulocytes may be diluted into a lesser Once having documented the presence of hemolysis, it is our
number of total circulating red cells. In addition, in response experience that the approach followed by Lux and Glader? in
to the anemia, reticulocytes may leave the bone marrow pre- establishing the cause of hemolysis is pragmatic and logical,
maturely and mature in the circulation for longer than the and this is the technique that is followed in this chapter and is
Chapter 9 Hemolytic Anemias: Intracorpuscular Defects: |. Hereditary Defects of the Red Cell Membrane yas
" Box 9-1 Predominant Red Cell Morphology Commonly Associated with Nonimmune
Hemolytic Disorders
j
function by regulating HCO;/Cl exchange and facilitating proteins, as well as to myosin, and the N-terminal 30-kD
the transfer of carbon dioxide from tissues to lungs. It is divided ___domain interacts with glycophorin C, p55, and band 3. The pro-
into two structurally and functionally distinct domains (see tein 4.1 mRNA is subject to extensive alternative splicing, yield-
Fig. 9-4B). The 43-kD N-terminal cytoplasmic ___domain contains ing tissue- and development-specific isoforms.”' Posttranslational
binding sites for ankyrin and proteins 4.1 and 4.2, and also binds deamidation of asparagine 502 occurs during development
glycolytic enzymes and hemoglobin. The 52-kD C-terminal of reticulocytes into mature red cells, and these two forms are
region has 14 transmembrane segments intercalated in the lipid designated protein 4.1a and b.
bilayer that form the anion-exchange channel.'” The genes coding for the major membrane proteins
Ankyrin is the major connecting protein that links the have been cloned and sequenced and their chromosomal
membrane skeleton to the bilayer.*’ It is a pyramid-shaped localization identified (Table 9-1). Mutations in any of
protein with an N-terminal 89 kD band 3-binding ___domain, these genes that alter the amount or function of the
formed by 24 tandem repeats containing 33 amino acids each expressed proteins compromise the integrity of the mem-
(see Fig. 9-4C). Spectrin binds to the central 62-kD ___domain brane and manifest in altered red cell morphology. These
of ankyrin, whereas the 55-kD C-terminal portion is a regula- cells are unable to survive passage through the spleen,
tory ___domain that is alternatively spliced at the mRNA level, resulting in hemolytic anemia.
yielding ankyrin isoforms, which modulate the interaction
with spectrin and band 3.
Classification
Protein 4.1 is a globular protein with four structural
domains as depicted in Figure 94D.' A 10-kD ___domain The hereditary hemolytic anemias due to red cell mem-
enhances the spectrin—actin interaction by binding to both brane protein defects may be classified according to the
Chapter 9 Hemolytic Anemias: Intracorpuscular Defects: |. Hereditary Defects of the Red Cell Membrane 18]
Figure 9-5 @ Transmission electron micrographs of negatively stained red blood cell membrane skeletons. A. An area of spread skeleton net-
work. B, C. The hexagonal lattice made up of spectrin tetramers (Sp4), hexamers (Sp6), or double tetramers (2Sp4). Cross-linking
junctional complexes contain short F-actin filaments and protein 4.1. Globular ankyrin structures are bound to spectrin filaments about
80 nm from their distal ends. (From Liu, SC, et al: Visualization of the hexagonal lattice in the erythrocyte membrane skeleton. | Cell Biol
104:527, 1987, with permission.)
Molecular
Mass* Chromosomal
Protein Localization Diseases
Skeletal Proteins
a-Spectrin 1q21 > q23 HS, HE, HPP
6-Spectrin 14q23 > q24.2 jay, JENE, Nae
Protein 4.1 ; 1p33 > p34.2 HE
Integral Proteins
Band 3 17q21 > q22 HS, SAO
Glycophorin C 2q14 > q21 HE
Linker Proteins
Ankyrin : 206 8p11.2
Protein 4.2 : 77 15q15 > q21
*Proteins were separated via SDS-PAGE and stained with Coomassie blue or periodic acid—Schiff reagent (PAS).
‘Molecular weight is calculated from the amino acid sequence and expressed as kilodaltons (kD).
HS = hereditary spherocytosis; HE = hereditary elliptocytosis; HPP = hereditary pyropoikilocytosis;
SAO = Southeast Asian ovalocytosis.
182 Chapter 9 Hemolytic Anemias: Intracorpuscular Defects: |. Hereditary Defects of the Red Cell Membrane
!
a Spectrin
21
| EF hand
Spectrin self-association
B Spectrin
17
[
pi2eKo|— BILGSkD pils3kD |
Spectrin Ankyrin Protein 4.1
self-association Actin
Adducin
A
Band 3
14 Transmembrane
Cytoplasmic ___domain segments Cytoplasm
NH» 43 kD 52 kD COOH
Ankyrin
NH> 89 kD 62 kD 55 kD COOH
Cc Band 3 Spectrin
Protein 4.1
NH> 30 kD
Glycophorin C Spectrin
Band 3 Actin
D p55 Myosin
Figure 9-4 @ Schematic representation of the structure of four of the red blood cell membrane proteins involved in hereditary hemolytic
anemias. The molecular masses of the various domains are given in kilodaltons (kD). A. Spectrin. The subunit structure of a- and B-spectrin,
showing the antiparallel arrangement of the N- and C-terminals, the homologous triple helical spectrin repeat units (indicated above the a
and chains), and the trypsin-resistant domains (open squares). a-Spectrin has five domains (al to aV) and 21 repeats. B-Spectrin has four
domains (8! to BIV) and 17 repeats. Proteins interacting with spectrin are indicated below the relevant domains. B. Band 3. The 52-kD mem-
brane ___domain contains 14 transmembrane segments and is responsible for anion exchange. The 43-kD N-terminal cytoplasmic ___domain and
the proteins binding to this ___domain are shown. C. Ankyrin. The N-terminal 89-kD ___domain consists of 24 ankyrin repeat units, which bind
to band 3. The 62-kD ___domain interacts with spectrin and the C-terminal 55-kD regulatory ___domain modulates the activity of the protein.
D. Protein 4.1. The four structural domains of the protein are depicted as open rectangles and the proteins binding to each ___domain are
shown below.
interactions
Vertical
in
SpD-D interaction
HYPOTHESIS:
e a = pn as =
HPP
Figure 9-5 ® Diagrammatic illustration of the vertical and horizontal interactions between the red cell membrane components (top). The bot-
tom section illustrates the pathophysiology of the red cell lesion in hereditary spherocytosis (HS), hereditary elliptocytosis (HE), and hereditary py-
ropoikilocytosis (HPP). A defect in a vertical interaction resulting in spherocytes and HS is illustrated at the bottom left. A defect in a horizontal in-
teraction resulting in elliptocytes and poikilocytes (HE and HPP) is illustrated at the bottom right. (From Palek, J: Disorders of red cell membrane
skeleton: An overview. In Kruckeberg, WL, et al (Eds): Erythrocyte Membranes 3: Recent Clinical and Experimental Advances. Alan R. Liss,
New York, 1984, p 177, with permission.)
Hereditary Spherocytosis
MODE OF INHERITANCE
HS is characterized by osmotically fragile, spherical red cells,
and is the most common hereditary hemolytic anemia in people
of northern European origin (Fig. 9-6). It has been documented
in other race groups, such as the Japanese and South African
blacks, but with a lower prevalence. In at least 75% of cases it
follows a classic autosomal dominant pattern of inheritance, but
in the remaining families both parents are clinically normal,
suggesting autosomal recessive inheritance or variable pene-
trance of a dominant gene or a de novo mutation.
Figure 9-6 @ Photomicrograph of peripheral blood smear from a
patient with hereditary spherocytosis (HS). Note the microsphero-
MOLECULAR DEFECTS cytes (small condensed spherocytes with no central pallor), indi-
The underlying molecular defects in HS are heterogeneous cated by the black arrow and the pincered (mushroom-shaped)
and several genetic loci have been implicated.”*** In the vast cell, indicated by the white arrow.
184 Chapter 9 Hemolytic Anemias: Intracorpuscular Defects: |. Hereditary Defects of the Red Cell Membrane
QUANTITATIVE RED BLOOD CELL both before and after splenectomy but can be low, normal, or
MEMBRANE PROTEIN DEFECT high. The mean corpuscular hemoglobin (MCH) tends to paral-
lel the MCV. Although the MCV is usually normal, because of
the red cell’s spherical shape, the diameter of some cells is sub-
stantially decreased and these appear as dark, rounded micros-
pherocytes on the peripheral smear (see Fig. 9-6). The mean
corpuscular hemoglobin concentration (MCHC) is elevated
(higher than 36%) in about 50% of cases and probably reflects
Din eete sth,
mild cellular dehydration, particularly of cells that have under-
gone splenic conditioning and that have low levels of cell water
and potassium (see the earlier section on Pathophysiology).
Automated blood counters using laser light scattering or
profiles obtained by aperture impedance analysers are very
useful and have a high degree of accuracy in diagnosing HS.
These detect the right shift in the distribution of red cell he-
moglobin concentration and the presence of a subpopulation
Splenic sequestration
Erythrostasis of microspherocytes. An example is shown in Figure 9-8.
cells are often, but not exclusively, found in band 3-deficient have abnormal osmotic fragility of red cells that have been
patients. An interesting observation is the presence of acantho- stressed by prior sterile incubation for 24 hours. During
cytes on the peripheral blood smears of HS patients with the 24-hour incubation, HS cells have greater loss of mem-
8-spectrin defects. brane surface because of the relative membrane instability (see
Fig. 9-9). A corollary of the use of the incubated osmotic
SPECIAL LABORATORY TESTS
fragility test is that if the test result is normal, it is highly
OSMOTIC FRAGILITY TEST This test is essentially a mea- unlikely that one is dealing with a patient with HS. Increased
sure of the surface-to-volume ratio of the red cell. If the test osmotic fragility is however independent of the cause of spher-
is performed on fresh red cells, it is then also a measure of the ical cells; for example, it may also be found in HPP (see section
proportion of cells that have undergone splenic conditioning. later in this chapter), autoimmune hemolytic anemia, and burns.
When red cells are placed in a series of graded hypotonic salt
solutions, water rapidly enters the cells and osmotic equilib- AUTOHEMOLYSIS TEST This relatively sensitive test in the
rium is achieved. The cells swell and become spherical, and diagnosis of HS measures the structural and metabolic integrity
eventually a critical volume is reached, at which point the cel- of the HS red cell membrane under conditions of erythrostasis
lular contents (hemoglobin) leak out and ultimately the cell and relative glucose depletion, that is, sterile incubation of red
may burst. Red cells of patients with HS, because of their cells in their own plasma for 48 hours at 37°C. The HS red cell
decreased surface area-to-volume ratio, can tolerate less is leaky to sodium. To “keep its head above water,” the cell uti-
swelling than normal cells and lyse at higher concentrations lizes adenosine triphosphate (ATP) and glucose to drive the
of salt than do normal cells (Fig. 9-9). About 25% of HS cation pump to a greater extent than normal. Associated with
patients have normal osmotic fragility of fresh red cells, par- the increased activity of the pump, there is a greater turnover of
ticularly in the very group that is mildly affected and is diffi- membrane phospholipids and associated membrane fragmenta-
cult to diagnose on morphological grounds. Generally, patients tion with a decrease in surface-to-volume ratio until the critical
in the latter group, as well as patients with more typical cases, hemolytic volume is reached and autohemolysis occurs. The
usual range of autohemolysis in HS cells is variable and is
about 10% to 50%, compared with control values of 0.2% to
100 2.0%. However, a minority of patients show only minimally
elevated autohemolysis or may even be within the normal
range. In most HS patients, addition of glucose markedly
diminishes autohemolysis but not usually to within the normal
range of samples incubated with glucose (0% to 1.0%). A
oljo) minority of patients show no correction of autohemolysis with
glucose, a finding also obtained with many patients with sphe-
Haemolysis
% rocytosis associated with autoimmune hemolytic anemia. It
should be noted that many laboratories do not use this test rou-
tinely (see Chap. 31 for a description of the procedure).
2 “eGy
x 28
a
CDSS
Ine osmotic fragility are markedly increased, and autohemolysis is
increased and unaffected by glucose. Pre- and postincubation
osmotic fragility are uniformly increased in spherocytic HE, and
autohemolysis is characteristically increased but corrected by
glucose. Children with HE and infantile poikilocytosis have
increased osmotic fragility and autohemolysis in the early
neonatal period that revert to normal with the development of
more prominent elliptocytosis.
QUESTIONS
1. How can the anemia presented here be classified?
2. What parameter(s) is (are) suggestive of effective ery-
thropoiesis?
3. Do the laboratory values presented here indicate hemo-
lysis in this patient?
4. What is the significance of the RPI value?
has been mapped to chromosome |6q23-qter.” Xerocytes have with hereditary xerocytosis do not benefit from splenec-
an increased surface-to-volume ratio, an increased MCHC, and tomy, presumably because of more generalized sequestra-
presumably increased cell viscosity, which make them less tion of these cells.
deformable and liable to sequestration in the reticuloendothelial
system. The red cells are not specifically sequestered in the
spleen, so that splenectomy does not have a beneficial effect.
Ou axtions
1. What happens when normal donor red cells are transfused hematocrit, 23%: reticulocyte count, 8%; polymorphs on
into a patient with an intracorpuscular red cell defect? peripheral smear. What is the RPI?
a. Donor cells are destroyed. a. 8
@) Donor cells have normal survival. b. 4
c. Depends on the severity of the defect.
d. Depends on the severity of the anemia.
2. Which of the following tests is not used to determine 4. What tests are useful in the classification of the cause of
increased red cell destruction? ted cell hemolysis?
a. Unconjugated (indirect) bilirubin ( aCoombs test
b. Serum haptoglobin b. Hemoglobin level and electrophoresis
c. Schumm’s test c. Reticulocyte count
Fa .
fd) Reticulocyte count d. Red cell enzyme studies and iron-binding
3. An anemic patient investigated for a hemolytic state has capacity
the following laboratory findings: hemoglobin, 8 g/dL;
Chapter 9 Hemolytic Anemias: Intracorpuscular Defects: |. Hereditary Defects of the Red Cell Membrane 15
5. Which of the following red cell membrane protein defi- a. Susceptibility of spectrin to thermal denaturation
ciencies does not cause hereditary spherocytosis? .)Defective membrane spectrin tetramer assembly and
a. Ankyrin spectrin deficiency
“b.\Protein 4.1 c. Unstable membrane lipids
c. Protein 4.2 d. Membrane ankyrin deficiency
d. Band 3
9. Which of the following findings are not typical of South-
6. Which of the following laboratory tests would not be east Asian ovalocytosis?
typical of hereditary spherocytosis? a. A deletion of nine amino acids in band 3
a. Increased osmotic fragility b. Resistance to infection by malaria parasite
b,Spherocytes on peripheral smear €)a deficiency of band 3
c. Decreased MCHC d. Rigid spoon-shaped ovalocytes
d. Increased RPI
10. Which disorders are classified as disorders of membrane
7. Which is the most frequent functional abnormality cation permeability?
affecting membrane skeleton proteins in common A. Hereditary stomatocytosis and hereditary xerocytosis
hereditary elliptocytosis? b. Sideroblastic anemia and myelofibrosis
a. Defective binding of spectrin to ankyrin c. Autoimmune hemolytic anemia and microangiopathic
@ Defective spectrin tetramer assembly hemolytic anemia
c. Defective binding of ankyrin to protein 3 d. Ehlers—Danlos syndrome and Bernard—Soulier syn-
d. Deficiency of protein 4.1 drome
8. Which of the following abnormalities is (are) thought to
cause the severe fragmentation and microspherocytes See answers at the back of this book.
_ characteristic of hereditary pyropoikilocytosis? %
~) SUMMARY m The most frequently used test reflecting increased red cell
destruction is the serum unconjugated (indirect) bilirubin
and serum haptoglobin determinations. Other tests, if
mw Anemia may occur with a moderate shortening of the red destruction is primarily intravascular, are those that test for
cell life span if there is an associated depression of bone the presence of hemoglobinemia, hemoglobinuria, and
marrow function, which may occur with certain systemic hemosiderinuria.
diseases or exposure to chemicals.
g The reticulocyte production index (RPI) is calculated by
ms Hemolytic anemias may be classified as follows: the following equation:
Intracorpuscular RPI = % Reticulocytes y Hematocrit
¢ Hereditary defects Reticulocyte maturation time 45
* Defects in the red cell membrane mg The red blood cell membrane skeleton consists of struc-
¢ Enzyme defects tural proteins, «- and ®-spectrin, actin, and protein 4.1.
¢ Hemoglobinopathies w The major integral red blood cell membrane proteins are
band 3 and the glycophorins.
¢ Thalassemia syndromes
w Hereditary hemolytic anemias due to red blood cell mem-
e Acquired defects
brane protein defects are classified into four main groups:
¢ Paroxysmal nocturnal hemoglobinuria
¢ Hereditary spherocytosis (HS)
Extracorpuscular
¢ Hereditary elliptocytosis (HE) including hereditary
¢ Immune hemolytic anemias pyropoikilocytosis (HPP) and Southeast Asian Ovalocy-
¢ Infections tosis (SAO)
REFERENCES . Speicher, DW, et al: Properties of human reflection of erythrocyte spectrin defi-
red cell spectrin heterodimer (side-to- ciency in hereditary spherocytosis.
1. Hillman, RS, and Finch, CA: Red Cell
side) assembly and identification of an N Engl J Med 315:1579, 1986.
Manual, ed 7. FA Davis, Philadelphia,
essential nucleation site. J Biol Chem 26. Wichterle, H, et al: Combination of two
1996.
ZOTAA TTS, 1992: mutant a spectrin alleles underlies a
2. Lux, SE, and Glader, BE: Disorders of
. Speicher, DW, et al: Location of the severe spherocytic hemolytic anemia.
the red cell membrane. In: Nathan, DG,
human red cell spectrin tetramer binding J Clin Invest 98:2300, 1996.
and Oski, FA (Eds): Hematology of
site and detection of a related “closed” PN Coetzer, TL, et al: Partial ankyrin and
Infancy and Childhood, ed 2. WB
hairpin loop dimer using proteolytic spectrin deficiency in severe hereditary
Saunders, Philadelphia, 1981.
footprinting. J Biol Chem 268:4227, spherocytosis. N Engl J Med 318:230,
ca Liu, SC, and Derick, LH: Molecular
1993. 1988.
anatomy of the red blood cell membrane
. Hanspal, M, and Palek, J: Biogenesis of 28. Hanspal, M, et al: Molecular basis of
skeleton: Structure-function relation-
normal and abnormal red blood cell spectrin and ankyrin deficiencies in
ships. Semin Hematol 29:231, 1992.
membrane skeleton. Semin Hematol severe hereditary spherocytosis: Evi-
4. Liu, SC, et al: Visualization of the hexag-
29:305, 1992. dence implicating a primary defect of
onal lattice in the erythrocyte membrane
. Woods, CM, and Lazarides, E: Degrada- ankyrin. Blood 77:165, 1991.
skeleton. J Cell Biol 104:527, 1987.
tion of unassembled alpha and beta spec- 29) Lux, SE, et al: Hereditary spherocytosis
Nn . Gilligan, DM, and Bennett, V: The junc-
trin by distinct intracellular pathways: associated with deletion of the human
tional complex of the membrane skele-
Regulation of spectrin topogenesis by ankyrin gene on chromosome 8. Nature
ton. Semin Hematol 30:74, 1993.
beta spectrin degradation. Cell 40:959, 345:736, 1990.
6. Davis, LH, and Bennett, V: Mapping the
1985. 30. Bass, EB, et al: Further evidence for
binding sites of human erythrocyte
. Tanner, MJ: Band 3 anion exchanger and localization of the spherocytosis gene on
ankyrin for the anion exchanger and
its involvement in erythrocyte and kid- chromosome 8. Ann Intern Med 99:192,
spectrin. J Biol Chem 265:10589, 1990.
ney disorders. Curr Opin Hematol 9:133, 1983.
~~ . Pasternack, GR, et al: Interactions
2002. eS) . Jarolim, P, et al: Mutations of conserved
between protein 4.1 and band 3: An
. Peters, LL, and Lux, SE: Ankyrins: arginines in the membrane ___domain of
alternative binding site for an element of
Structure and function in normal cells erythroid band 3 lead to a decrease in
the membrane skeleton. J Biol Chem
and hereditary spherocytes. Semin membrane-associated band 3 and to the
260:3676, 1985.
Hematol 30:85, 1993. phenotype of hereditary spherocytosis.
ee). Marfatia, SM, et al: In vitro binding
. Conboy, JG: Structure, function, and Blood 85:634, 1995.
studies suggest a membrane-associated
molecular genetics of erythroid mem- a2 Bouhassira, EE, et al: An alanine to thre-
complex between erythroid p55, protein
brane skeletal protein 4.1 in normal and onine substitution in protein 4.2 cDNA
4.1, and glycophorin C. J Biol Chem
abnormal red blood cells. Semin Hema- associated with a Japanese form of
269:8631, 1994.
tol 30:58, 1993. hereditary hemolytic anemia (protein 4.2
\O. Gallagher, PG, and Forget, BG: Spectrin
Wh, Palek, J: Disorders of red cell membrane Nippon). Blood 79:1846, 1992.
genes in health and disease. Semin
Hematol 30:4, 1993. skeleton: An overview. In: Kruckeberg, 83: Eber, SW, et al: Prevalence of increased
WL, et al (Eds): Erythrocyte Membranes osmotic fragility of erythrocytes in
10. Sahr, KE, et al: The complete cDNA and
3: Recent Clinical and Experimental German blood donors: Screening using a
polypeptide sequences of human erythroid
Advances. Alan R. Liss, New York, modified glycerol lysis test. Ann Hema-
a spectrin. J Biol Chem 265:4434, 1990.
1984, p 177. tol 64:88, 1992.
— . Winkelmann, JC, et al: Full length sequence
. Walensky, DL, et al: Disorders of the red 34. Coetzer, TL, et al: Southeast Asian Oval-
of the cDNA for human erythroid 8 spec-
blood cell membrane. In: Handin, RL, ocytosis (SAO) in a South African kin-
trin. J Biol Chem 265:11827, 1990.
i) . Speicher, DW, et al: A structural model
et al (Eds): Blood: Principles and Prac- dred with hemolytic anemia. Blood
tice of Hematology, ed 2. Lippincott, 87:1656, 1996.
of human spectrin: Alignment of chemi-
cal and functional domains. J Biol Chem Philadelphia, 2003, Chapter 51. 3D Coetzer, TL, et al: Molecular determi-
24. Gallagher, PG: Disorders of the red nants of clinical expression of hereditary
257:9093, 1982.
13. Speicher, DW, and Marchesi, VT: blood cell membrane: Hereditary sphero- elliptocytosis and pyropoikilocytosis.
Erythrocyte spectrin is comprised of cytosis, elliptocytosis and related disor- Blood 70:766, 1987.
ders. In: Lichtman, MA, et al (Eds): 36. Coetzer, TL, et al: Four different muta-
many homologous triple helical
segments. Nature 311:177, 1984. Williams Hematology, ed 7. McGraw- tions in codon 28 of « spectrin are asso-
14. Yan, Y, et al: Crystal structure of the
Hill, New York, 2006, p 571. ciated with structurally and functionally
25. Agre, P, et al: Inheritance pattern and abnormal spectrin «1/74 in hereditary
repetitive segments of spectrin. Science
262:2027, 1993. clinical response to splenectomy as a elliptocytosis. J Clin Invest 88:743, 1991.
Chapter 9 Hemolytic Anemias: Intracorpuscular Defects: |. Hereditary Defects of the Red Cell Membrane 195
Sik Sahr, KE, et al: Sequence and exon-in- Al. Coetzer, TL, and Palek, J: Partial spec- 47. Liu, S-C, et al: Molecular defect of the
tron organization of the DNA encoding trin deficiency in hereditary pyropoikilo- band 3 protein in Southeast Asian ovalo-
the al ___domain of human spectrin. Appli- cytosis. Blood 67:919, 1986. cytosis. N Engl J Med 323:1530, 1990.
cation to the study of mutations causing 42. Hanspal, M, et al: Molecular basis of 48. Schofield, AE, et al: Defective anion
hereditary elliptocytosis. J Clin Invest spectrin deficiency in hereditary transport activity of the abnormal band
84:1243, 1989. pyropoikilocytosis. Blood 82:1652, 1993. 3 in hereditary ovalocytic red blood
38. Roux, AF, et al: Molecular basis of 43. Wilmotte, R, et al: Low expression allele cells. Nature 355:836, 1992.
Spal/65 hereditary elliptocytosis in a LELY of red cell spectrin is associated 49. Narla, M, et al: Molecular basis for
North Africa: Insertion of a TTG triplet with mutations in exon 40 (aV/41 poly- membrane rigidity of hereditary ovalocy-
between codons 147 and 149 in the a morphism) and intron 45 and with partial tosis. A novel mechanism involving the
spectrin gene from five unrelated fami- skipping of exon 46. J Clin Invest cytoplasmic ___domain of band 3. J Clin
lies. Blood 73:2196, 1989. SEZ OSE OB: Invest 89:686, 1992.
319) Hassoun, H, et al: A novel mobile ele- 44. Williams, TN: Red blood cell defects . Carella, M, et al: Genome-wide search
ment inserted in the a spectrin gene: and malaria. Mol Biochem Parasitol for dehydrated hereditary stomatocytosis
Spectrin Dayton. A truncated « spectrin 149:121, 2006. (hereditary xerocytosis): Mapping of
associated with hereditary elliptocytosis. 4S, Tanner, MJA, et al: Melanesian heredi- locus to chromosome 16(16q23-qter).
J Clin Invest 94:643, 1994. tary ovalocytes have a deletion in red Am J Hum Genet 63:810, 1998.
40. Burke, JPWG, et al: Reduced spectrin cell band 3. Blood 78:2785, 1991.
ankyrin binding in a South African 46. Jarolim, P, et al: Deletion in erythrocyte
hereditary elliptocytosis kindred band 3 gene in malaria-resistant South-
homozygous for Spectrin St Claude. east Asian ovalocytosis. Proc Natl Acad
Blood 92:2591, 1998. Sci USA 88:11022, 1991.
Chapter
Hemolytic Anemias
Intracorpuscular Defects:
Il. Hereditary Enzyme Deficiencies
Armand B. Glassman, MD
Denise M. Harmening, PhD, MT(ASCP), CLS(NCA)
History OBJECTIVES
Enzyme Deficiencies: Hexose At the end of this chapter, the learner should be able to:
Monophosphate Pathway
Glucose-6-Phosphate 1. Name the most common glycolytic enzyme deficiency associated with the hexose
monophosphate shunt or pentose phosphate pathway.
Dehydrogenase
Deficiency 2 _Name the most common glycolytic enzyme deficiency associated with the
Mode of Inheritance Embden—Meyerhof pathway.
Pathogenesis
Clinical Manifestations 3. Identify the red blood cell cytoplasmic inclusions associated with oxidative
Laboratory Testing denaturation of hemoglobin.
Enzyme Deficiencies: 4. List laboratory test results that would suggest a deficiency of glucose-6-phosphate
Glycolytic Pathway dehydrogenase (G6PD).
Pyruvate Kinase Deficiency
5. Identify a laboratory test result that would indicate a pyruvate kinase (PK) deficiency.
Other Enzyme Deficiencies
of the Glycolytic Pathway 6. Name the deficiency that causes hemoglobin to be oxidized from the ferrous to the
awa bs ferric state.
Enzyme Deficiencies: :
Methemoglobin Reductase
Pathway
Methemoglobin Reductase
Deficiency
Case Study
History group, which was later found to be associated with red cell
enzyme abnormalities. Biochemical and molecular studies
In 1926, 72 plantation workers in Panama suffered acute hemol- rapidly advanced the further characterization of these anemias.
ysis after receiving the antimalarial drug 8-aminoquinoline. The most common anemia in this group 1s caused by the
Subsequent reports from widely scattered geographic locations deficiency of glucose-6-phosphate dehydrogenase (G6PD),
added credence to the relationship of hemolysis, cyanosis, and an enzyme in the hexose monophosphate, shunt, or phospho-
methemoglobinemia with the ingestion of certain antimalarial gluconate pathway. The second most frequent enzyme defi-
drugs. In 1953 Dacie and associates! evaluated apparently het- ciency is that of pyruvate kinase (PK), an essential enzyme in
erogeneous cases of congenital hemolytic anemias that had sev- the Embden—Meyerhof pathway. Many other enzyme defi-
eral common characteristics. There was no detectable abnormal ciencies of these pathways have also been identified. They are
hemoglobin, the antiglobulin test result was negative, the variably associated with HNSHA. Laboratory testing is
osmotic fragility test was normal, and there were no spherocytes directed toward identification of the specific enzyme defi-
seen on the peripheral smear. The term hereditary nonsphero- ciency. Treatment is generally supportive, although experi-
cytic hemolytic anemia (HNSHA) was used to describe the mental gene therapy may have a future role.
196
Chapter 10 Hemolytic Anemias: Intracorpuscular Defects: Il. Hereditary Enzyme Deficiencies 197
Enzyme Deficiencies: Hexose gene, there is a reduced activity variety designated Gd A—,
Monophosphate Pathway which can be demonstrated in 10% to 15% of the men. Approx-
imately 20% of African American women are heterozygous for
Glucose-6-Phosphate Dehydrogenase the Gd A— gene. Gd A— is the prototype of the mild form of
Deficiency G6PD deficiency.
Among whites, G6PD Mediterranean (G6PD Med) is the
G6PD deficiency, one of the most common genetic abnormali- most common abnormal variant, although the overall preva-
ties known, is thought to affect more than 400 million people lence is low. Among Kurdish Jews, however, the incidence of
worldwide. Carson and associates? identified the enzyme G6PD G6PD Med may be as high as 50% to 60%. G6PD Med
deficiency in 1956 in an individual who developed hemolytic (also known as G6PD B-—) is the prototype of a more severe
anemia after the administration of the antimalarial drug pri- enzyme deficiency associated with acute hemolytic anemia,
maquine (8-aminoquinoline). Yoshida’ first purified the enzyme including favism. The variant Gd Canton is more commonly
from human red cells in 1966. Further progress characterized found in native people of Southeast Asia and China. There is a
the diverse variants of G6PD by sequencing of amino acids, high frequency of G6PD deficiency in Taiwan and southern
cloning of cDNA, and sequencing of nucleotides. China. Approximately 20% to 40% of neonatal jaundice in these
More than 400 variants of G6PD enzyme have been areas is related to G6PD deficiency, whereas neonatal jaundice
described on the basis of biochemical and genetic analyses.*° is rarely attributed to G6PD deficiency in the United States.'”
Recent advances in molecular biology have enabled classifi- The type of G6PD variant found in selected populations 1s listed
cation of the variants into approximately 50 gene mutation in Table 10-1.
groups.*’ Nearly all of these variants are the results of point The variants have been generally designated by geo-
mutations* that produce structurally abnormal or functionally graphic names. With the use of modern techniques of molec-
defective enzymes, or both. ular biology, these variants have been reclassified in terms of
the exact sites of nucleotide substitutions. Using this nomen-
Mode of Inheritance clature, Gd A+ would be designated as G6PD A?’°S to indi-
cate the presence of guanine at nucleotide 376."
G6PD deficiency is transmitted by a mutant gene located on
the X chromosome.? The gene encoding G6PD has been
Pathogenesis
mapped to the Xq28 region in humans.'° The disorder is fully
expressed in men who are hemizygous when they inherit the G6PD catalyzes the first step in the hexose monophosphate
mutant gene from their mothers, who are usually silent carri- shunt (or pentose phosphate) aerobic glycolytic pathway.
ers. In women, full expression of the disorder occurs when Oxidative catabolism of glucose is accompanied by reduction
two mutant genes (homozygous) are inherited. The heterozy- of nicotinamide adenine dinucleotide phosphate (NADP) to
gous woman has a mosaic of one red blood cell population NADPH (Fig. 10-1), which is subsequently required to reduce
with normal enzyme activity and another with deficient glutathione. Reduced glutathione (GSH) is an important source
enzyme activity.'' The expression of G6PD deficiency varies of reducing potential that protects hemoglobin from oxidative
markedly among heterozygotes, which is explained in part by denaturation. The sequence of biochemical reactions shown in
the X-inactivation hypothesis.'? In females, one of the two X Figure 10-2 occur within the normal RBC with adequate lev-
chromosomes (maternally or paternally derived) becomes els of appropriate enzymes and substrate to prevent the accu-
randomly inactivated in each cell of the early embryo. Thus, mulation of intracellular oxidants.
each somatic cell in a heterozygote expresses either one or the The activity of G6PD is highest in young erythrocytes and
other Gd allele. The ratio of the two cell types may vary decreases with cell aging. Under normal conditions, the individ-
widely, not only in different individuals but also among dif- ual with G6PD deficiency compensates for the shortened life
ferent tissues, even within the same individual." span of the erythrocytes by producing more early red cells
Distribution of the mutant gene for G6PD deficiency is
worldwide; however, the highest incidence occurs in the darkly
pigmented racial and ethnic groups. In fact, most of the studies
of G6PD variants have been performed on samples from Table 10-.
African Americans, people of Mediterranean ancestry, and
Asians. The prevalence of G6PD deficiency varies remarkably,
Enzyme Type Population Usually Associated
dependent upon the racial and ethnic group. Normally active
G6PD, type Gd B, is the most common form of the enzyme in Gd B (normal variant) All
all populations and exists in 99% of whites in the United States. Gd Med (also known Whites (Mediterranean
as Gd B—) area)
Another variety of the G6PD enzyme, Gd A+, is commonly
Gd A+ (normal variant) Blacks (~ 16% of African
found in Africans, has normal activity, but differs from Gd B by Americans)
a single amino acid substitution that alters its electrophoretic Gd A-— Africans
mobility.'? The Gd A+ variant is found in about 20% of African Gd Canton Asians
men.'>'6 Among African Americans who possess the Gd A+
198 Chapter 10 Hemolytic Anemias: Intracorpuscular Defects: Il. Hereditary Enzyme Deficiencies
PHOSPHOGLUCONATE
PATHWAY
(oxidative)
EMBDEN-MEYERHOF :
PATHWAY GSH
(non-oxidative)
Glucose
ee
Glucose 6-P 6-P-Gluconate
[eri]!
Fructose 6-P Pentose-P
[Pre]
Fructose 1,6-diP
METHEMOGLOBIN |
REDUCTASE
Glyceraldehyde <>» DHAP
PATHWAY LUEBERING-RAPAPORT
H lobi NAD PATHWAY
Fy tlemoaionin [ll |(Nap [caro |
1,3-diP-Glycerate
arp? —[Pek]|
2,3-diP-Glycerate
3-P-Glycerate
[row] 2-P-Glycerate
[el]
P-Enolpyruvate
are» [PK]
Pyruvate
NADH
NAD [-ou]{
Lactate
Figure 10-1 @ Red cell metabolic pathways. The nucleated red cell depends almost exclusively on the breakdown of glucose for energy
requirements. The Embden—Meyerhof (nonoxidative or anaerobic) pathway is responsible for most of the glucose utilization and generation of
ATP. In addition, this pathway plays an essential role in maintaining pyridine nucleotides in a reduced state to support methemoglobin
reduction (the methemoglobin reductase pathway) and 2,3-diphosphoglycerate synthesis (the Luebering—Rapaport pathway). The phospho-
gluconate pathway couples oxidative metabolism with pyridine nucleotide and glutathione reduction. It serves to protect red cells from
environmental oxidants. (Adapted from Hillman, RS, and Finch, C: Red Cell Manual, ed 7. FA Davis, Philadelphia, 1996, p 15, with permission.)
(reticulocytosis). Oxidative stress, however, can lead to a mild fail to maintain adequate levels of GSH.*!” 21,22 The resulting oxida-
to severe hemolytic episode. A deficiency of GSH results in tion of hemoglobin leads to progressive precipitation of
oxidative destruction of certain erythrocyte components, includ- irreversibly denatured hemoglobin (Heinz bodies) (Fig. 10-3).
ing sulfhydryl groups of globin chains and the cell mem- The cells lack normal deformability when sulfhydryl groups
brane.'?*' In addition, more than 50 chemical agents may are oxidized and consequently encounter difficulties passing
induce hemolysis in G6PD-deficient erythrocytes. The drugs through the microcirculation. Premature destruction of the cells
that have more commonly been reported to induce hemolysis in results when they undergo intravascular lysis or are sequestered
individuals with G6PD deficiency are listed in Table 10-2. The and destroyed in the liver and spleen. This early destruction may
hemolytic episode results when G6PD-deficient erythrocytes sometimes be detected in the peripheral blood smear with the
lem
ency
bee aca
Acetanilide Pamaquine
Chloramphenicol Pentaquine
Dapsone Phenylhydrazine
Daunorubicin Primaquine
Doxorubicin Sulfacetamide
Methylene blue Sulfamethoxazole
(Gantanol)
Nalidixic acid Sulfanilamide
Naphthalene Sulfapyridine
Niridazole (Ambilhar) Thiazolesulfone
Nitrofurantoin (Furadantoin) Toluidine blue
Trinitrotoluene (TNT) Figure 10-4 ® Peripheral blood smear from a patient with a G6PD
deficiency. Note the small, condensed “bite” or “helmet” cells.
Source: Modified from Beutler, E, and Yoshida, A: Genetic variation
of glucose-6-phosphate dehydrogenase: A catalog and future
prospects. Medicine 67:311, 1988. *
However, newborns with this intrinsic defect and adults who
take certain therapeutic drugs or develop infections may suf-
fer various degrees of hemolysis from these challenges to the
formation of small, condensed bite- or helmet-shaped red cells G6oPD-deficient erythrocytes.
(Fig. 10-4). Symptoms of the disorder are related to the severity of the
Certain G6PD-deficient individuals also exhibit sensitiv- hemolytic episode. Two to three days after the administration of
ity to the fava bean (favism) (Fig. 10-5). These individuals the offending drug, the erythrocyte count decreases, along with
develop severe hemolysis after ingesting the fava bean or the hemoglobin content. The anemia appears normochromic
even after inhaling the plant’s pollen. Favism is found in some and normocytic, and there is an increase in reticulocytes. The
individuals with G6PD deficiency of the Mediterranean and patient may experience back pain. Hemoglobinuria and jaun-
Canton types. Some chemicals isolated from fava beans dice may also be evidence of the hemolytic process. The clini-
destroy red cell glutathione, leading to symptomatic hemoly- cal features of the two most common variants are compared in
sis in susceptible G6PD-deficient individuals. Table 10-3.
The hemolytic episode in Gd A— is usually self-limiting.
Clinical Manifestations Young cells that are produced in response to the anemia have
levels of G6PD that are nearly normal’ and have better sur-
The majority of G6PD-deficient persons are asymptomatic vival characteristics. Hemolysis associated with G6PD Med
most of the time and go through life without ever being aware is more easily induced, usually more severe, and has been
of their genetic trait. G6PD enzymatic activity that is 20% of reported to result in death on occasion. Red blood cell trans-
normal or even slightly less is sufficient for normal red fusions may be indicated for hemolytic episodes in patients
cell function and survival under ordinary circumstances. with G6PD Med.
Figure 10-3 ™ Heinz bodies, using peripheral blood from a patient Figure 10-5 @ Fava beans.
with G6PD deficiency, stained with the supravital stain, crystal violet.
200 Chapter 10 Hemolytic Anemias: Intracorpuscular Defects: Il. Hereditary Enzyme Deficiencies
Gd Med (GdiB= vais ote to convert NADP to NADPH (i.e., are deficient in G6PD) will
lack fluorescence. The quantitative assay of G6PD is based on
Clinical Feature Gd A-— Gd Med the measurement of the rate of reduction of NADP to NADPH
measured at 340 nm.2’ G6PD variants can also be identified
Cells affected Aging All erythrocytes
via electrophoretic methods.
by defect erythrocytes
Hemolysis with drugs Unusual Common The diagnosis of G6PD deficiency during an acute
Hemolysis with Common Common hemolytic episode may be difficult. The deficiency may be
infection obscured by the younger erythrocyte population (which has
Favism No Occasionally
more G6PD) as the older G6PD-deficient erythrocytes are
Degree of hemolysis | Moderate Severe
Transfusions required No Occasionally destroyed.
Chronic hemolysis No No
Hemolytic disease Rare Occasionally
of newborn Enzyme Deficiencies: Glycolytic
Pathway
A number of hereditary enzyme deficiencies have been
Laboratory Testing described in the RBC glycolytic pathway (Embden—Meyerhof
pathway). Most of these glycolytic enzyme deficiencies are
G6PD deficiency should be suspected after a clinical episode
inherited autosomal recessive and do not demonstrate any
of acute hemolysis after administration of chemical or thera-
abnormal RBC morphology. This differentiates them from RBC
peutic agénts known to cause the reaction in patients with the
membrane defects and hemoglobinopathies, which do have spe-
disorder. Laboratory changes of the nonspecific type include:
cific abnormal RBC morphology associated with the specific
a fall in hemoglobin (and hematocrit), hemoglobinuria (urine
disease (see Chaps. 9 and 11). These inherited glycolytic
can turn brown to almost black secondary to the presence of
enzyme deficiencies generally cause chronic nonspherocytic
hemoglobin), Heinz bodies in the erythrocytes, evidence of
hemolytic anemia (CNSHA), which vary in the degree of sever-
hemolysis in the serum, elevated serum bilirubin levels, and
ity. The ability to compensate for anemia caused by the hemol-
markedly decreased or absent haptoglobin levels. Generally
ysis is reflected in the increased production of RBCs in the bone
there are no significant alterations in leukocyte or platelet
marrow and reticulocytosis. Reticulocytes still have cytoplas-
counts or function.
mic organelles and are capable of protein synthesis and oxida-
Laboratory investigation of hemolytic anemia when there
tive phosphorylation leading to ATP production. Several
is evidence (family history or drug sensitivity, or both) of
enzymes including pyruvate kinase (PK), hexokinase (HK), and
G6PD deficiency may include several screening procedures.
aldolase have a much higher activity in reticulocytes and are
Oxidative denaturation of hemoglobin results in formation of
often termed the “age-related” enzymes.
Heinz bodies. These small particles of precipitated hemoglo-
bin can be visualized by supravital staining using certain basic
dyes such as crystal violet (see Fig. 10-3). Heinz bodies will Pyruvate Kinase Deficiency
appear as small (1- to 4-um) purple inclusions, usually on the
cell periphery. They are not seen with Romanowsky stains In 1960 DeGruchy and associates**”’ reported that some patients
such as Wright’s stain. Although Heinz bodies may be seen in with hereditary nonspherocytic hemolytic anemia (HNSHA)
some other enzyme deficiencies, they are not seen in PK defi- had elevated red blood concentrations of 2,3-diphosphoglycer-
ciency, which is the second most common RBC enzyme defi- ate (2,3-DPG). This elevation suggested a block in anaerobic
ciency. Some unstable hemoglobins also form Heinz bodies glycolysis further down the pathway (see Fig. 10-1). The
after incubation of erythrocytes at 37°C for 48 hours. enzyme was identified in 1961, when a severe deficiency of red
Other test procedures that may be used to screen for blood cell PK was found in three patients with HNSHA.*° PK
G6PD deficiency include the methemoglobin reduction test™* catalyzes one of the reaction steps in the Embden—Meyerhof
and the ascorbate-cyanide test.*° In the methemoglobin reduc- pathway of anaerobic glycolysis. Because mature red blood
tion test, which is a simple and sensitive screening procedure, cells lack mitochondria, they are dependent on anaerobic gly-
G6PD-deficient erythrocytes fail to reduce methemoglobin in colysis for the generation of adenosine 5’-triphosphate (ATP).
the presence of methylene blue. The ascorbate-cyanide test, The diminished capacity to generate ATP in PK-deficient red
which measures perioxidative denaturation of hemoglobin, is blood cells results in cell membrane fragility and a hemolytic
not specific for G6PD deficiency, because it will yield moder- anemia.
ately positive results if the patient has PK deficiency or cer- Since its discovery in 1961, more than 300 cases of PK
tain unstable hemoglobins. deficiency have been reported, and many of these were cases
The fluorescent spot test and the specific G6PD assay are of variant enzymes with different biochemical characteris-
positive only with G6PD deficiency.*° When a mixture of tics.*'*? The nucleotide sequence of cDNA for the human
glucose-6-phosphate, NADP, saponin, and buffer is mixed PK gene and sequence of several of the mutations that cause
Chapter 10 Hemolytic Anemias: Intracorpuscular Defects: II. Hereditary Enzyme Deficiencies 201
HNSHA have been described.***4 PK deficiency is the most Interestingly, these patients may tolerate exercise to a
common enzymatic disease involving the anaerobic gly- greater degree than might be expected from the extent of their
colytic pathway of the red blood cell. Together, G6PD defi- anemia. Red blood cell concentrations of 2,3-DPG are increased
ciency and PK deficiency constitute most cases of HNSHA up to three times the normal levels in patients with PK defi-
arising from red blood cell enzyme deficiencies. ciency because of the enzyme block* (see Fig. 10-1). The
increase of 2,3-DPG decreases the affinity for O,, which is more
MODE OF INHERITANCE readily released to the tissues where it is needed. For this reason,
PK deficiency is inherited as an autosomal recessive trait, transfusion therapy should be based on the patient’s tolerance of
but true homozygotes are rare and are restricted to children the anemia. Removal of the spleen benefits some patients
of consanguineous parents. The most common mode of because it increases the life span of the altered red blood cells.
inheritance is that of double heterozygosity; that is, when
two mutant variants of the PK enzyme are simultaneously LABORATORY TESTING
inherited from each parent.*?*>° To date, approximately 20 The peripheral blood smears of patients with PK deficiency
different mutations of the PK gene are known to produce typically show a normochromic, normocytic anemia with
hemolytic anemia.*’ Thus, the clinical symptoms of PK varying degrees of reticulocytosis. Accelerated erythropoiesis
deficiency are observed both in true homozygotes and in may result in polychromasia, poikilocytosis, anisocytosis, and
double heterozygotes for the PK gene. Both sexes appear to nucleated red blood cells. Both the hemoglobin and the hema-
be affected equally. There is increasing evidence that PK tocrit levels are decreased from normal. The serum usually
deficiency is worldwide in distribution, with most of the has a moderate increase in unconjugated bilirubin, and the
cases reported to date in northern Europe, the United States, haptoglobin level is decreased or absent.*°-*”
and Japan. Other cases have been reported in Australia, Several screening tests may be used to distinguish the
Canada, China, Costa Rica, Hong Kong, Italy, Mexico, the nonspherocytic anemia of PK deficiency from the anemias of
Near East, New Zealand, the Philippines, Saudi Arabia, hereditary spherocytosis and the unstable hemoglobinopathies.
Spain, and Venezuela.*°*3*! These tests are nonspecific and serve only as a mechanism for
The Pennsylvania Amish have a high frequency of PK classifying the type of anemia. Diagnosis is made on the basis
deficiency, which has been traced back to a single immigrant of specific testing for the PK enzyme.
couple. In affected families, consanguinity is common. Thus Screening tests may include the osmotic fragility test and
the PK deficiency in the Amish population is the result of a the autohemolysis test (see Chap. 31), as well as the antiglobu-
true homozygote condition.” lin test and red blood cell survival tests. Erythrocytes that are
PK-deficient have osmotic fragility which is nearly normal
PATHOGENESIS when the test is performed on freshly drawn blood. If the blood
PK deficiency results in a decreased capacity to generate ATP is incubated for a few hours, some patients exhibit an increase
(see Fig. 10-1 and Chap. 3). The ATP-requiring membrane in osmotic fragility.'” Sterile defibrinated blood is used to per-
pumps that maintain the proper electrochemical gradients form the test for autohemolysis. When normal erythrocytes are
begin to fail with decreasing concentrations of ATP. This incubated in their own serum at 37°C, they gradually lyse, show-
results in cell water loss with cell shrinkage, distortion of cell ing up to 3.5% lysis after 48 hours. Erythrocytes from patients
shape, and increased membrane rigidity.**** These membrane with nonspherocytic anemias, as well as those with hereditary
abnormalities lead to premature destruction of the red blood spherocytosis, demonstrate an increased amount of autohemol-
cells in the spleen and liver with consequent anemia. It has ysis. When glucose is added before incubation, erythrocytes
been shown that PK-deficient reticulocytes consume six to from the patient with hereditary spherocytosis demonstrate a
seven times more oxygen than normal reticulocytes.** In most decreased amount of autohemolysis. The addition of glucose
cells, the drop in ATP regeneration because of a block in the does not correct the increased autohemolysis of PK-deficient
glycolytic pathway would be compensated for by oxidative erythrocytes. The antiglobulin test in PK deficiency is negative,
phosphorylation, but that capacity is lost in red blood cells as and the red blood cell survival is decreased.
they mature and they lose mitochondria. There is a fluorescence screening test, which is relatively
simple and sensitive. It is used for the presumptive diagnosis
CLINICAL MANIFESTATIONS of PK deficiency. The test is based on the following coupled
The severity of the hemolytic disease associated with PK defi- enzyme assay:
ciency varies from mild to severe, depending on the properties
PEP +2 ADP + Ms** PK enzyme
Pyruvate + ATP
of the mutant enzymes.***>“° True homozygotes are anemic
and jaundiced at birth and may require repeated transfusions
Pyruvate + NADH + Ht —##“» Lactate + NAD*
during life. Less severely affected patients may come to clini-
cal attention later in childhood or early adulthood because of (UV fluorescence) (No fluorescence)
anemia, jaundice, or an enlarged spleen. The hemolytic ane-
mia is often more pronounced during periods of infection or This assay takes advantage of the fact that NADH fluo-
other stresses. There is an increased incidence of pigmented resces when it is illuminated with long-wave ultraviolet (UV)
gallstone formation in these patients, as is true with all chronic light, whereas NAD does not fluoresce. Phosphoenolpyruvate
hemolytic disorders. (PEP), NADH, adenosine diphosphate (ADP), Mg’*, and
202 Chapter 10 Hemolytic Anemias: Intracorpuscular Defects: Il. Hereditary Enzyme Deficiencies
lactate dehydrogenase (LDH) are added to a patient sample of phosphate isomerase (GPI) is the third most commonly
blood, which is spotted on filter paper and examined with a identified enzyme deficiency in the glycolytic pathway.
UV light. If the blood lacks PK enzyme, NADH will not be Although there have been reports of other enzyme deficien-
oxidized and the fluorescence will persist for 45 minutes to cies (glycolytic and nonglycolytic), not all such deficiencies
an hour. If the blood is normal and has the PK enzyme, the flu- have been associated with hemolytic anemia. The other
orescence will disappear in 15 minutes, because NAD* does reported glycolytic enzyme deficiencies are summarized in
not fluoresce.***’ It should be noted that leukocytes contain a Table 10-4, and the prevalence and characteristics listed.”
PK isoenzyme that will also catalyze the same reaction. There- Laboratory tests are available to assay many of the spe-
fore, blood must be centrifuged and plasma and buffy coat cific enzymes. Some of these tests may be available only
removed prior to testing the erythrocytes. In addition, patients through reference laboratories. Most laboratories, however,
who have recently been transfused may have enough donor will be able to screen patients with a suspected hemolytic ane-
cells remaining in circulation to give erroneous test results. mia caused by enzyme deficiency. The antiglobulin, erythro-
Any abnormal fluorescence spot test should be followed cyte survival, autohemolysis, osmotic fragility, and Heinz
with a confirmatory quantitative PK enzyme assay. This body tests can all be useful in distinguishing the enzyme
involves the same coupled reaction mechanisms as previously deficiencies from hereditary spherocytosis and the unstable
described, but the conversion of NADH to NAD* is measured hemoglobinopathies.
spectrophotometrically at 340 nm under standard conditions.
Most PK-deficient individuals have 5% to 25% of normal
activity.°°47 Enzyme Deficiencies: Methemoglobin
Reductase Pathway
Other Enzyme Deficiencies Methemoglobin Reductase Deficiency
of the Glycolytic Pathway
Hemoglobin that is oxidized from the ferrous to the ferric state is
Except for the deficiencies of PK, reports of hereditary called methemoglobin. Normally, about 1% of the circulating
enzyme deficiencies are relatively rare. The prevalence of PK hemoglobin is in the form of methemoglobin. The balance
deficiencies has been reported to be 51 cases per million between methemoglobin formation and reduction is maintained
white population based on gene frequency studies.** Glucose by the NADH-methemoglobin reductase (also called diaphorase)
Qu esevens
1. What is the most common glycolytic enzyme deficiency a. Increased formation of Heinz bodies
associated with the pentose phosphate pathway (aerobic b. Lack of fluorescence in the fluorescent spot test
pathway)? c. Failure to reduce methemoglobin in the presence of
a. PK deficiency methylene blue
(Dy G6PD deficiency ()AII of the above
c. Hexokinase deficiency 5. Which laboratory test result would indicate a patient who
d. Glutathione reductase deficiency is PK-deficient?
2. What is the most common glycolytic enzyme deficiency @) Abnormal rate of hemolysis that is independent of the
associated with the Embden—Meyerhof pathway (anaero- presence or absence of glucose in the incubation
bic pathway)? medium of the autohemolysis test
(@)PK deficiency b. Lack of fluorescence in the fluorescent spot test
b. G6PD deficiency c. A change in the indicator from red to yellow in the
c. Hexokinase deficiency orthocresol red test
d. Glutathione reductase deficiency d. Increase in osmotic fragility
3. Oxidative denaturation of hemoglobin results in formation 6. What deficiency causes hemoglobin to be oxidized from
of small particles that are visualized with supravital stain- the ferrous to the ferric state’?
ing. What is the term for these particles? a. G6PD deficiency
a. Basophilic stippling b. PK deficiency
b. Howell—Jolly bodies NADH-methemoglobin reductase deficiency
c. Pappenheimer bodies d. Lactate dehydrogenase deficiency
@ Heinz bodies
See answers at the back of this book.
4. In the evaluation of a patient for G6PD deficiency, which
of the following test results would indicate a deficiency of
the enzyme?
nny
“-') SUMMARY CHAF w Patients with G6PD abnormalities are usually asympto-
matic until exposed to conditions of lowered oxygen ten-
sion, certain chemicals or substances (including fava
w Hereditary nonspherocytic hemolytic anemia (HNSHA)
beans), and some medications.
encompasses a group of disorders associated with red
blood cell (RBC) abnormalities. mw GOPD is diagnosed by clinical symptoms and laboratory
determinations. Laboratory testing includes finding the
m Glucose-6-phosphate dehydrogenase (G6PD) enzyme
presence of Heinz bodies in erythrocytes (not specific),
abnormalities are the most common cause of HNSHA.
electrophoresis of G6PD, and, where indicated, molecular
m= GOPD is an enzyme in the hexose monophosphate (or genetic studies.
pentose phosphate) shunt pathway.
@ PK deficiency, which was first identified in 1961, is asso-
mw GOPD enzyme variants are noted for their association ciated with a diminished capacity to generate ATP, result-
with particular racial and ethnic backgrounds. ing in fragile red blood cells and a hemolytic anemia.
m Pyruvate kinase, an essential enzyme of the Embden— @ Laboratory diagnosis of PK deficiency requires specific
Meyerhof pathway, is the second most common enzyme testing of PK activity. A fluorescent screening test is used.
abnormality associated with HNSHA.
@ Other red cell enzyme deficiencies include methemoglo-
m There are more than 400 G6PD variants. The gene for bin reductase and glucose phosphate isomerase deficien-
G6PD is mapped to chromosome Xq28. cies. Almost any enzyme of the aerobic and anaerobic
m Clinical expression of G6PD deficiency is more often metabolic pathways has been implicated in HNSHA,
evident in men because it is a sex-linked (X chromosome) although the levels of hemolysis are quite variable and
abnormality. hemolysis may be entirely absent.
m Deficient G6PD activity results in reduced glutathione
levels, which cause increased oxidative denaturation of
hemoglobin and subsequent hemolysis.
Chapter 10 Hemolytic Anemias: Intracorpuscular Defects: Il. Hereditary Enzyme Deficiencies 205
REFERENCES 16. Beutler, E: The molecular biology of . Miwa, S, and Fujii, H: Pyruvate kinase
G6PD variant and other red cell enzyme deficiency. Clin Biochem 23:155,
— . Dacie, JR, et al: Atypical congenital
defects. Annu Rev Med 43:47, 1992. 1990.
haemolytic anemia. Q J Med 22:79,
We Chiu, DTY, et al: Molecular characteri- . Miwa, S, et al: Concise review:
I)sy3}.
i). Carson, PE, et al: Enzymatic deficiency
zation of glucose-6-phosphate dehydro- Pyruvate kinase deficiency: Historical
genase (G6PD) deficiency in patients perspective and recent progress of mol-
in primaquine sensitive erythrocytes.
of Chinese descent and identification ecular genetics. Am J Hematol 42:31,
Science 124:484, 1956.
of new base substitution in the IPDS.
. Yoshida, A: Glucose-6-phosphate dehy-
human G6PD gene. Blood 81:2150, is)ies). Tani, K, et al: Human liver type pyru-
drogenase of human erythrocytes.
OB. vate kinase: Complete amino acid
I. Purification and characterization of
. Beutler, E: Glucose-6-phosphate sequence and the expression in mam-
normal (B+) enzyme. J Biol Chem
dehydrogenase: New perspectives. Blood malian cells. Proc Natl Acad Sci USA
241:4966, 1966.
PENSE, WIS). SolOZ OS ss
Beutler, E, and Yoshida, A: Genetic vari-
12). Lichtman, M, et al: William’s Hematol- 34. Baronciani, L, and Beutler, E: Analysis
ation of glucose-6-phosphate dehydroge-
ogy, ed 7. McGraw-Hill, Columbus, OH, of pyruvate kinase-deficiency mutations
nase: A catalog and future prospects.
2000. that produce nonspherocytic hemolytic
Medicine 67:311, 1988.
20. Arese, P, and De Flora, A: Pathophysiol- anemia. Proc Natl Acad Sci USA
Nn . Beutler, F: The genetics of glucose-
ogy of hemolysis in glucose-6-phosphate 90:4324, 1993.
6-phosphate dehydrogenase deficiency.
dehydrogenase deficiency. Semin Hema- 3))e Wintrobe, MM: Greer JP (Ed):
Semin Hematol 27:137, 1990.
tol 27:1, 1990. Wintrobe’s Clinical Hematology, ed 11.
. Hirono, A, and Miwa, S: Human
ie)pat. Johnson, RM, et al: Oxidant damage Lippincott Williams & Wilkins,
glucose-6-phosphate dehydrogenase:
to erythrocyte membrane in glucose-6- Philadelphia, 2004.
Structure and function of normal and
phosphate dehydrogenase deficiency: 36. Lakomek, M, et al: Erythrocyte pyruvate
variant enzymes. Haematologia 25:85,
Correlation with in vivo reduced kinase deficiency: A kinetic method for
1993.
glutathione concentration and mem- differentiation between heterozygosity
~— . Vulliamy, TJ, et al: Diverse point muta-
brane protein oxidation. Blood 83:1117, and compound-heterozygosity. Am J
tions in the human glucose-6-phosphate
1994. Hematol 31:225, 1989.
dehydrogenase gene cause enzyme defi-
. Beutler, E: Glucose-6-phosphate dehy- oi Baronciani, L, and Beutler, E: Prenatal
ciency and mild or severe hemolytic ane-
drogenase deficiency. In Stanbury, JB, diagnosis of pyruvate kinase deficiency.
mia. Proc Natl Acad Sci USA 85:5171,
et al (Eds): The Metabolic Basis of Blood 84:2354, 1994.
1988.
Inherited Disease. McGraw-Hill, New 38. Feng, CS, et al: Prevalence of pyruvate
ee Kletzien, RF, et al: Glucose-6-phosphate
York, 1978, p 1430. kinase deficiency among the Chinese:
dehydrogenase: A “housekeeping” enzyme
. Beutler, E, et al: The hemolytic effect of Determination by the quantitative assay.
subject to tissue-specific regulation by
primaquine. IV. The relationship of cell Am J Hematol 43:271, 1993.
hormones, nutrients, and oxidant stress. age to hemolysis. J Lab Clin Med 3), de Medicis, E, et al: Hereditary nonsphe-
FASEB J 8:174, 1994. 44:439, 1954. rocytic hemolytic anemia due to pyru-
\O. Desorges, JF: Genetic implications of 24. Grewer, GJ, et al: The methemoglobin vate kinase deficiency: A prevalence
G-6PD deficiency. N Engl J Med reduction test for primaquine-type sensi- study in Quebec, Canada. Hum Hered
294:1438, 1976. tivity of erythrocytes: A simplified A42:179, 1992.
10. Pai, GS, et al: Localization of loei procedure for detecting a specific hyper- 40. Fonella, A, et al: Iron status in red-cell
for hypoxanthine phosphoriboxyltrans- susceptibility to drug hemolysis. JAMA pyruvate kinase deficiency: Study of
ferase and glucose-6-phosphate dehydro- 180:386, 1962. Italian cases. Br J Haematol 83:485,
genase and biochemical evidence of . Jacob, HS, and Jandl, JH: A simple NOE).
nonrandom X-chromosome expression visual screening test for glucose-6- 4A]. Wei, DC, et al: Homozygous pyruvate
from studies of a human X-autosome phosphate dehydrogenase deficiency kinase deficiency in Hong Kong ethnic
translocation. Proc Natl Acad Sci USA employing ascorbate and cyanide. minorities, J Paediatr Child Health
77:2810, 1980. N Engl J Med 274:1162, 1966. 28:334, 1992.
— se . Beutler, E, et al: The normal human . Perkins, S. Glucose-6-phosphate dehy- . Kanno, G, et al: Molecular abnormality
female as a mosaic of X-chromosome drogenase deficiency. In Kjeldsberg, CR of erythrocyte pyruvate kinase defi-
activity. Studies using the gene for (ed): Practical Diagnosis of Hematologic ciency in the Amish. Blood 83:2311,
G-6-PD deficiency as a marker. Proc Disorders 1 Benign Disorders. American 1994.
Natl Acad Sci USA 48:9, 1962. Society for Clinical Pathology, Chicago, . Keith, AS: Pyruvate kinase deficiency
i). Beutler, E: Biochemical abnormalities 2006, pp. 113-122. and related disorders of red cell glycoly-
associated with hemolytic states. In 27. Beutler, E: Red Cell Metabolism. sis. Am J Med 41:762, 1966.
Weinstein, IM, and Beutler, E (Eds): A Manual of Biochemical Methods, . Mentzer, WC, Jr, et al: Selective reticu-
Mechanisms of Anemia in Man. ed 2. Grune & Stratton, New York, 1975. locyte destruction in erythrocyte pyru-
McGraw-Hill, New York, 1962, p 195. . De Gruchy, GC, et al: Non-spherocytic vate kinase deficiency. J Clin Invest
. Luzzatto, L: Glucose-6-phosphate dehy- congenital hemolytic anemia. Blood 50:688, 1971.
drogenase: Genetic and haematological 1371, 1960. . Lakomek, M, et al: Erythrocyte pyruvate
aspects. Cell Biochem Funct 5:101, 1987. . Robinson, MA, et al: Red cell metabo- kinase deficiency: Relations of residual
. Takizawa, T, et al: A single nucleotide lism in non-spherocytic congenital enzyme activity, altered regulation of
base transition is the basis of the com- haemolytic anaemia. Br J Haematol defective enzymes and concentrations of
mon human glucose-6-phosphate dehy- W327, Uo. high-energy phosphates with the severity
drogenase variant A(+). Genomics . Valentine, WN, et al: A specific gly- of clinical manifestations. Eur J Haema-
15228, 1987. colytic enzyme defect (pyruvate kinase) tol 49:82, 1992.
. Beutler, E: Glucose-6-phosphate dehy- in three subjects with congenital non- 46. Rapaport, SI: Introduction to Hematol-
drogenase deficiency. N Engl J Med spherocytic hemolytic anemia. Trans ogy, ed 2. JB Lippincott, Philadelphia,
324:169, 1991. Assoc Am Physicians 74:100, 1961. 1987, Chaps 6 and 7.
206 Chapter 10 Hemolytic Anemias: Intracorpuscular Defects: Il. Hereditary Enzyme Deficiencies
47. Perkins, S: Hereditary erythrocyte disor- 48. Beutler, E, and Gelbart, T: Estimating the 50. Jaffe, ER: Hereditary methemoglobine-
ders due to deficiencies of the glycolytic prevalence of pyruvate kinase deficiency mias associated with abnormalities in the
pathway. In Kjeldsberg, CR (Ed): from the gene frequency in the general metabolism of erythrocytes. Am J Med
Practical Diagnosis of Hematologic white population. Blood 95:1, 2000. 41:786, 1966.
Disorders | Benign Disorders. American . Van Wijk, R, and van Solinge, WW: The 51. Evelyn, KA, and Maloy, HT: Micro deter-
Society for Clinical Pathology, Chicago, energy-less red blood cell is lost: Ery- mination of oxyhemoglobin, methemoglo-
2006, pp. 105-112. throcyte enzyme abnormalities of glycol- bin and sulfhemoglobin in a single sample
ysis. Blood 106:13, 2005. of blood. J Biol Chem 126:655, 1938.
Chapter
Hemolytic Anemias
Intracorpuscular Defects:
Ill. The Hemoglobinopathies
Denise M. Harmening, PhD, MT(ASCP), CLS(NCA)
David Yang, MD
Hallye Zeringer, MT(ASCP)SH
207
208 Chapter 11 Hemolytic Anemias: Intracorpuscular Defects: III. The Hemoglobinopathies
spp = species.
214 Chapter 11 Hemolytic Anemias: Intracorpuscular Defects: III. The Hemoglobinopathies
renal papilla, ultimately causing renal papillary necrosis, presents itself in the second decade of life. The third decade in
which frequently develops along with the pyelonephritis (see HbSS disease is characterized by interstitial nephritis, papillary
the following discussion of Sickle Cell Nephropathies). necrosis, pyelonephritis, and the nephrotic syndrome, to name
The multiple factors responsible for the increased sus- a few of the renal disorders that may develop. In the fourth
ceptibility to infection in patients with HbSS disease are out- decade and beyond, end-stage renal disease develops as one or
lined in Table 11-8. more of the sickle cell nephropathies results in chronic renal
The relationship between the incidence of the malarial falurce2
parasite and the frequency of the abnormal HbS gene requires It should be noted that hyperuricemia and gross hema-
further explanation. Malaria, caused by a parasite of the turia occur commonly in patients with HbSS disease. Hyper-
Plasmodium species, is still a serious disease in tropical areas, uricemia occurs in approximately 15% of children and 40%
with Plasmodium falciparum being responsible for the most of adults with HbSS disease because of the increased urate
life-threatening situations. The original geographic distribu- production associated with the accelerated erythropoietic rate
tion of sickle cell disease overlaps with that of malaria. As in and decreased renal clearance of urate.'* Gross hematuria
thalassemias, these hemoglobinopathies are believed to pro- occurs commonly not only in sickle cell anemia but also in
vide some kind of selective advantage for malaria. This is sickle cell trait because of the sickling, stasis, ischemia, and
applicable only to subjects carrying one abnormal HbS gene extravasation of blood in the kidney.!*!7"*
(sickle cell trait). The precise mechanism of this protective
effect from malaria is not known; however, cells carrying
STROKE Stroke occurs in up to 22% of patients with HbSS
HbS, when parasitized by P. falciparum, may sickle more
disease.”'*!*-'°7° Stroke represents an array of neurologic
quickly than will nonparasitized cells, leading to preferential
complications caused by an ischemic or hemorrhagic lesion in
destruction of the parasitized cells.'° The sickling could affect
a specific cerebral vessel. The neurologic manifestations may
the cycle of the parasite in one of two ways: directly, by
be focal, such as hemiparesis, or more generalized, such as
killing the parasite: or indirectly, by causing the parasitized
coma or seizure. Recurrent episodes of stroke cause progres-
sickle cells to be sequestered in the spleen. The fact that per-
sively greater impairment and increased mortality. The most
sons homozygous for the HbS gene often lack splenic func-
common cause of stroke in children is cerebral infarction.'*'°
tion by the time they reach adulthood (autosplenectomy or
With age, subarachnoid and intracerebral hemorrhage become
functional asplenia) may be one reason why malaria is excep-
increasingly common. A distinguishing pathologic feature of
tionally severe, and often fatal, in these cases. Thus, HbS con-
arterial vessels involved in stroke is the presence of intimal
fers a relative degree of protection from malaria only in the
and medial proliferation, which results in severe restriction of
heterozygous (trait) state. It is balanced by the decreased fit-
blood flow and is found in approximately 80% of angiograms
ness of the homozygous state and the gene frequency eventu-
of patients with sickle cell anemia and stroke.”° Most likely
ally reaches an equilibrium in the population.
this is owing to the interaction of sickled cells with endothe-
lium, which results in endothelial damage, inflammation, and
SICKLE CELL NEPHROPATHIES In the kidney, intravascu-
the local release of growth factors that stimulate proliferation
lar sickling occurs more rapidly than in any other organ owing
of subintimal and medial cells. In the later stages of this vas-
to deoxygenation of HbS in the acidic and hyperosmolar envi-
culopathy, chronic ischemia leads to the formation of a deli-
ronment of this organ.*'* The combination of hypoxia, hyper-
cate subcortical network of collateral vessels that are prone to
tonicity, and acidosis in the kidney causes sickling, stasis, and
rupture, likely accounting for the increased incidence of hem-
ischemia of the renal medulla and papillary tip, leading to pro-
orrhagic stroke with increasing age.” HbSS disease patients
gressive renal events. Eventually, over time, a number of sickle
with hemorrhagic stroke (intracerebral or subarachnoid hem-
cell nephropathies develop. Hyposthenuria, the inability of the
orrhage) have a high mortality rate during the acute stage
kidney to concentrate the urine, is the earliest and most com-
(may be as high as 50%).°7"'8
mon nephropathy in sickle cell disease, occurring usually in the
Patients with a characteristic abnormality of cerebral
first decade.'*'’ Progressive renal pathology occurs in patients
blood flow (higher than normal velocity of flow on transcra-
with HbSS disease as renal tubular dysfunction and atrophy
nial Doppler ultrasonography) have been shown to be at much
greater risk to develop stroke.'° This risk can be significantly
reduced by maintaining these patients on a regular program of
‘Table 11-8s Factors Responsible for
blood transfusion designed to suppress erythropoiesis such
the Increased that no more than 30% of the circulating red cells are the
_ Susceptibility of” = patients own.'? A follow-up study demonstrated that the
Patients with Sickle Cell high risk of stroke returned after transfusion therapy was
Anemia to Infections discontinued.?!
sickle globin gene (8°). As a result, individuals with sickle cell falling reticulocyte count may herald the onset of such a cri-
trait produce both normal HbA and HbS, with a predominance sis. There may be a neutrophilic leukocytosis with a shift to
of HbA in an approximate ratio of 60:40.3° The structural the left and thrombocytosis. The bone marrow reflects marked
formula is a8,8,°C"". The frequency of this heterozygous erythroid hyperplasia, except during an aplastic crisis.
condition in American blacks is approximately 8%.*° In vitro In individuals with the trait, sickled cells are not typically
experiments demonstrate cells that are homozygous for hemo- present on the peripheral blood smear. On rare occasions, how-
globin S sickle when the oxygen level is decreased to 4% to 6% ever, sickled cells may be observed in the peripheral blood
whereas cells heterozygous for hemoglobin § (sickle cell trait) smear during a crisis episode.
sickle when the oxygen level is decreased to 2%.” Individuals
with HbS trait are usually asymptomatic, but occasionally LABORATORY SCREENING FOR SICKLE CELL DISEASE
episodes of hematuria and hyposthenuria occur because of sick- According to the Clinical Practice Guideline on Sickle Cell
ling in the kidney. The potential for sickling exists, therefore, Disease published by the U.S. Department of Health and
and drastic lowering of pH or reduction in oxygen tension can Human Services in April 1993, all newborns, regardless of
precipitate a crisis. Causes for these include severe respiratory race or ethnic background, should be screened for the pres-
infections, air travel in unpressurized aircraft, anesthesia, and ence of HbS (Table 11—9).°
congestive heart failure. Even excessive exercise can lead to a Newborn screening for sickle cell disease began in the
significant buildup of lactic acid, resulting in sickling and sub- United States in the early 1970s. The initial screening pro-
sequent infarction. Several deaths of American black soldiers grams grew out of the recognition that sickle cell anemia was
with sickle cell trait have been reported as a result of rigorous associated with significant morbidity and mortality. Today
basic training at altitudes greater than 4000 feet, which led to a newborn hemoglobinopathy screening is universally required
buildup of lactic acid, followed by acidosis and subsequent by Law or Rule in the 50 U.S. states, the District of Columbia.
organ infarction.** Puerto Rico, and the Virgin Islands.”
The majority of screening programs currently use iso-
LABORATORY DIAGNOSIS electric focusing (IEF) of an eluate from dried blood spots to
The following laboratory tests should be performed to diag- characterize hemoglobin and a few programs use HPLC or
nose sickle cell disease: the complete blood count, a reticulo- cellulose acetate electrophoresis as the initial screening
cyte count, evaluation of the peripheral smear, hemoglobin method.'*’° Most programs retest abnormal screening speci-
electrophoresis or isoelectric focusing (IEF), and measure- mens using a second, complementary electrophoretic tech-
ment of hemoglobins A, and F by high-performance liquid nique, HPLC, immunologic tests, orDNA-based assays.'* IEF
chromatography (HPLC).°'!** is an equilibrium process in which hemoglobin migrates in a
The chronic anemia of HbSS typically is quite severe, pH gradient to a position of 0 net charge. The formation of
with hemoglobins ranging between 6 and 8 g/dL. The RBC discrete, sharp bands via IEF allows for more precise and
indices are normochromic and normocytic. The peripheral red accurate quantification than standard electrophoresis. HPLC
blood picture can be striking, with numerous target cells, is emerging as the method of choice for the initial screening
fragmented red cells, polychromasia, nucleated red cells, and of hemoglobin variants and quantification of HbA, and HbF.
usually sickled cells (see Fig. 11-5). Siderotic granules and Although a greater number of samples can be processed daily
Howell—Jolly bodies may be seen in the red cells as a result using IEF, the advantages of HPLC are that it is generally eas-
of rapid RBC turnover and “stressed” erythropoiesis. The ier to interpret, less prone to inconsistencies related to sample
average reticulocyte count is between 5% and 20%. This preparation, application, and staining techniques; and 1s fully
count decreases, however, during an aplastic crisis; indeed, a automated, thereby reducing the chance for clerical error.*°
Mass spectrometry (MS) is emerging as a possible screening
methodology with the introduction of robust and simple-to-
use electrospray tandem MS instruments capable of greater
throughput with comparable sensitivity and specificity of IEF
and HPLC, in preliminary studies.”
The definitive test for HbS is hemoglobin electrophore-
sis on cellulose acetate at alkaline pH, followed if necessary
by electrophoresis on citrate agar at acid pH (Fig. 11-10).
Electrophoresis separates different hemoglobins by electrical
charge, and hemoglobin separation by citrate agar elec-
trophoresis depends on the combination of charge and the
ability of the hemoglobin to combine with components in the
agar gel mixture. The patient with sickle cell anemia produces
no normal B chains; therefore, there will be no HbA on elec-
trophoresis (unless the patient has been recently transfused).
HbS constitutes 80% or more of hemoglobin, with HbF rang-
ing from 1% to 20%.°*'°'* When HbF levels are higher than
Figure 1 1-9 m Sickle trait (peripheral blood). Note the normal
appearing smear. 20%, there is a decrease in the severity of the disease.'° High
216 Chapter 11 Hemolytic Anemias: Intracorpuscular Defects: Ill. The Hemoglobinopathies
levels of HbF are seen transiently in newborns and in heredi- chains to form af dimers compared with normal 6 globin. For
tary persistence of fetal hemoglobin (HPFH). In sickle cell this reason, HbS comprises less than 50% of the hemoglobin in
anemia, the HbA, level may be slightly increased, with a heterozygotes, and a further reduction is seen in HbC trait.
mean of 3.4%.*» Hemoglobins with similar charges have sim- Two previously used screening tests should not be used
ilar migration patterns during electrophoresis, especially on for newborn screening. These are (1) the sickle cell prepara-
cellulose acetate. Hemoglobins D and G both migrate to the tion using sodium metabisulfite and (2) solubility tests using a
same position as HbS at alkaline pH. Hemoglobin E (HbE) concentrated phosphate buffer, a hemolyzing agent, and
and hemoglobin O,,.., (HbO,,..,) migrate in the same position sodium dithionate. These tube solubility tests either isolate
as HbC. Citrate agar electrophoresis 1s very useful, because it HbS at an interface or cause the abnormal hemoglobin to pre-
clearly separates HbS from HbG and HbD, and HbC from cipitate (Fig. 11-11). Both tests depend on the concentration of
HbE and HbO,,,,, at acid pH.*' In sickle cell trait, hemoglobin HbS in the red cell or hemolysate. These tests are inappropri-
electrophoresis at alkaline pH shows 60% HbA, 40% HbS, ate screening tests because they are not sufficiently sensitive to
and usually elevated HbA, (mean is 3.6%).° At acid pH, one detect low levels of HbS and do not distinguish between sickle
band is present in the A position (HbA + HbA,), whereas the trait and different forms of sickle cell disease.'*°
other band migrates to the S position (see Fig. 11—10).° The predominance of HbF and low level of HbS in cells
It should be noted that most hemoglobin variant traits, of the neonate is believed to explain the unreliability of these
without coexistent conditions such as iron deficiency, or tha- tests during the newborn period. Although sickle cell disease
lassemia, have alkaline electrophoretic patterns with an approx- should not be diagnosed from either a sickle cell preparation
imate 60:40 ratio of normal to abnormal hemoglobin. This is or solubility test, because neither of these tests will reliably
because of the effect of charge on assembly of globin chains: distinguish sickle cell trait from sickle cell anemia, many
relatively positively charged globins, such as S (+1) and C hospitals still perform these tests on adult patients. It is impor-
(+2) have a slight disadvantage in assembling with the a tant to note that some rare hemoglobins also sickle, giving
A2/C S ze as C S A/Ap F
Normal
Control
AS (trait)
S disease
C disease
SC disease
Figure | 1-10 @ Electrophoretic patterns of hemoglobin on (A) cellulose acetate, run at pH 8.4, and (8B) citrate agar, run at pH 6.0 to 6.5.
apl = point of application.
Chapter 11 Hemolytic Anemias: Intracorpuscular Defects: III. The Hemoglobinopathies DAG
HDC yariem
Hb Cg cactows
H Ziquinchor
HDS Mempnis
HbSpravis
1D Mesaers
Dpono-Atesre
Consideration y indicanons
To improve the oxygen-carrying transport Severely anemic as reflected byee Asst fivporension
in red cells by simple transfusions high-output cardiac failure, angina, or cerebral dysfunction
Sudden fall in hemoglobin or hematocrit levels during acute
splenic or hepatic sequestration crisis
Hemoglobin level < 5.0 g/dL or hematocrit of 15% in patients
who exhibit fatigue and dyspnea along with erythroid
hypoplasia or aplasia
To improve microvascular perfusion by decreasing the Life-threatening events, such as cerebrovascular accidents
number of red cells containing HbS by partial exchange including stroke and transient ischemic attacks (TIAs)
transfusion Arterial hypoxia syndrome (fat embolization)
Acute progressive lung disease
Unresponsive acute priapism
Eye surgery when performed under local anesthesia and in the
nonanemic * patient
Source: Chance, S, et alal (Eds):pvenr
1 Peteandhen of Sickle Cell Disease. US pene of Health fed Human
Services, Public Health Service, National Institutes of Health. NIH Pub 91:2117, August 1991.
donor availability, a narrow application to the youngest cells, and transplantation of the hematopoietic stem cells back
patients in good clinical condition, and rates of transplant- into the diseased mouse to cure immunodeficiency.** Theoreti-
related morbidity and mortality that are somewhat fixed.* cally, a similar approach can be applied to treat sickle cell ane-
Nevertheless, as the experience of transplantation for sickle mia, and successful phenotypic correction of the sickle cell
cell disease has expanded, there has been a transition from mutation by gene replacement has already been demonstrated
using this modality as an experimental intervention in mouse embryonic stem cells.*”
reserved for those most severely affected to one in which
younger children with early signs of sickle-related morbid- Hemoglobin C Disease and Trait
ity are targeted for treatment.** Umbilical cord blood units
(CBUs) are now considered an acceptable source for Hemoglobin C (HbC) disease is found almost exclusively in
hematopoietic stem cell transplantation (HSCT) when the black population. HbC differs from normal HbA by the
HLA-identical donors are unavailable. CBUs have been single amino acid substitution of lysine for glutamic acid in
used successfully in HLA-matched sibling HSCT for chil- the sixth position from the NH, terminal end of the B chain
dren with sickle cell disease. (Fig. 11-12). This represents the same substitution point as in
It is now possible to diagnose efficiently the presence or HbS but with a positively charged amino acid. The structural
absence of sickle cell disease in embryos obtained with formula for HbC, the presence of which 1s often referred to as
in-vitro fertilization, prior to implantation.*° This powerful HbC disease, is a,B.°°"’*. HbC is seen with great frequency
technique may help carrier couples by allowing them to select in West Africa, particularly northern Ghana, where the inci-
a healthy child without facing the decision of whether or not dence is 17% to 28%.'° In the United States, only 0.02% of
to abort an affected fetus. blacks have HbC disease.'°° The clinical manifestations are
Gene therapy for HbSS disease is also currently being in- mild chronic hemolytic anemia with associated splenomegaly
vestigated. The goal of gene therapy is to replace the B’-globin and abdominal discomfort. The red cell morphology is typi-
gene with a wild-type B-globin gene in order for the affected cally normocytic, and normochromic or hyperchromic, with
patient to produce healthy cells rather than sickle cells. The numerous target cells (50% to 90%) and occasionally micros-
success of gene therapy depends on the ability of researchers pherocytes, fragmented cells, and folded cells (Fig. 11-13).
to isolate, enrich, and insert genes into hematopoietic pluripo- HbC crystals (Fig. 11—14) or “bar of gold” crystals occur more
tential stem cells and generate safe, stable, erythroid-specific often in the red cells of individuals who have undergone
replacement gene expression at a level that is sufficient to have splenectomy than in those whose spleen is intact. Figure 11—15
a clinical effect.45*7 Successful gene therapy has been shown is a scanning electron micrograph (SEM) of hemoglobin C
in a mouse model of immunodeficiency where embryonic crystals. These crystals can be demonstrated in wet prepara-
stem cells from a Rag2 gene deficient mouse were obtained by tions by washing the red cells and then suspending them in a
implanting nuclei of skin cells cultured from the diseased sodium citrate solution.'’ The reticulocyte count is slightly
mouse into donor mouse oocytes, followed by insertion of the increased. Hemoglobin bands, at alkaline pH, reveal approxi-
missing Rag2 gene by homologous recombination, differenti- mately 95% HbC plus A,, less than 7% HbF, and no HbA.
ation of the embryonic stem cells into hematopoietic stem Hemoglobins E, Oa,., C, and A, all migrate to the same
220 ~~ Chapter 11 Hemolytic Anemias: Intracorpuscular Defects: Ill. The Hemoglobinopathies
}
Figure 1 1-12 ® Amino acid substitution in hemoglobin C. Figure 1 1-14 ™ Hemoglobin C disease (peripheral blood). Note
the particular crystals: “bar of gold” and numerous target cells
(postsplenectomy). (From Bell, A: Hematology. In: Listen, Look and
Learn. Health and Education Resources, Inc., Bethesda, MD, with
permission.).
position at alkaline pH; HbC can be separated from these
other hemoglobins at acid pH (see Fig. 11-10).
HbC trait, «.8,B,5"s, is present in 2% to 3% of Hemoglobin E Disease and Trait
American blacks, and these individuals are clinically asymp-
tomatic.°° The only significant finding on the peripheral blood Hemoglobin E (HbE) disease occurs with greatest frequency in
smear is targeting. At alkaline pH, there is approximately Burma, Thailand, Cambodia, Laos, Malaysia, and Indonesia.'*°
60% HbA and 40% HbC plus A. This variant is now prevalent in the United States because of
the influx of refugees from Southeast Asia. The homozygous
state (a,B,°°C"'>s) presents with little or no anemia, target cells,
Hemoglobin D Disease and Trait and microcytic, hypochromic red cell indices. Alkaline elec-
trophoresis reveals approximately 95% to 97% HbE plus A,,
Hemoglobin D (HbD) disease has several variants. The most
and the remainder of the hemoglobin is HbF. HbE migrates
common variant in American blacks is HbDpunjay, Which is
with HbC and HbO,,,,, at alkaline pH, but migrates with HbA
synonymous with HbD,. anscies. [ts frequency is less than
at acid pH. HbE trait (a@,8,8 °C") is asymptomatic clinically.
0.02%.°° Both the homozygous (a,B,'7'S""S"") and the het-
Microcytosis, target cells, and approximately 70% HbA and
erozygous (a,8,B,'7'S""S") states (where glycine is substi-
30% HbE plus A, are noted on routine electrophoresis.'°? HbE
tuted for glutamic acid) are asymptomatic. The peripheral
is slightly unstable, and there is an associated thalassemic com-
blood smear is unremarkable, except for a few target cells.
ponent with this hemoglobin variant. This is responsible for the
HbD migrates electrophoretically to the same position as
microcytosis and the lower than expected quantified value of
HbS and HbG at alkaline pH but migrates with HbA at acid
HbE in HbAE.*!
pH. HbD is a non-sickling soluble hemoglobin.
It has been postulated that HbE may protect against
malaria, because areas such as Thailand that are highly
endemic for malaria also have a high incidence of the HbE
gene. Some authors attribute this effect to the fact that the
Common Uncommon
Se iicnatlonic «values are approximate. There is a tremendousreovantabiiny en diseatesgroups and betweenni individual
patients of the same group, particularly with regard to clinical severity.
SS = sickle cell anemia; SB° Thal = sickle beta zero thalassemia; SB* Thal = sickle beta plus thalassemia; S HPFH =
HbS in association with the hereditary persistence of fetal hemoglobin syndrome; SC = hemoglobin SC disease;
nRBCs = nucleated red blood cells.
Source: Charache, S, et al (eds): Management and Therapy of Sickle Cell Disease. US Department of Health and Human
Services, Public Health Service, National Institutes of Health NIH Pub 91:2117, August 1991.
Unstable Hemoglobins
More than 130 unstable hemoglobins (Fig. 11-20) associated
with hemolytic anemias have been described.*°° Unstable
hemoglobins are hemoglobin variants in which amino acid
Increased O, Affinity Decreased O, Affinity-May substitutions or deletions have weakened the binding forces
and Polycythemia Have Mild Anemia or Cyanosis _ that maintain the structure of the molecule. The instability may
if OB, ilrinos vee “og@™B, cause hemoglobin to denature and precipitate in the red cells
268, Hb 01,%4A50B,, as Heinz bodies. Most unstable hemoglobin variants are inher-
SO oped Hb pisiacite pee ited as autosomal dominant disorders.°° However, absence of a
02877" HD agenogi 2Bo" > positive family history is not always helpful, as new mutations
Dempsey aB5"*™ Abpeth sre are common. Many mutations producing unstable hemoglo-
px Crestiane) ae pana “B04 binopathies are single amino acid substitutions in either the
ara OBC cea 4: a-, B-, y-, or 6-globin chains that affect a few key areas of the
Hb pene 09!" hemoglobin structure. By far, the majority of these substitutions
are in the B-globin chain, followed by an a-chain substitution
224 Chapter 11 Hemolytic Anemias: Intracorpuscular Defects: III. The Hemoglobinopathies
100 - =
80 Hb Rainier
Q } F Hb Seattle
Seay
=
Hb A. a
—_—
5 y x Hb Kansas
® 40 J
A >
O
20 eff
0 +=
0 20 40 60. 80
Op» pressure (mm Hg) Figure 1 1-20 @ Unstable hemoglobin: Hb Zurich (peripheral
blood). (From Bell, A: Hematology. In: Listen, Look and Learn.
Figure | 1-19 @ Oxygen equilibrium curve of whole blood from Health and Education Resources, Inc., Bethesda, MD, with
subjects with Hb Rainier, Hb Seattle, Hb Kansas and normal permission.)
control (HBA).
Methemoglobinemia
Methemoglobinemia is a clinical condition with methemoglo-
bin levels greater than 1% of the total hemoglobin.°° Methe-
moglobin contains the oxidized ferric form of iron (Fe**)
rather than the ferrous form (Fe?*). In this state, the molecule Case Study
is unable to bind oxygen, which results in cyanosis. The blood
is chocolate-brown in color. In general, there are three causes A 13-year-old black girl was admitted to the hospital appear-
of methemoglobinemia: ing acutely ill with fever and abdominal pain. On physical
examination, an enlarged spleen was evident. Laboratory
1. Hemoglobin M variants (dominant inheritance) test results were as follows:
2. NADH-methemoglobin reductase deficiency (recessive
inheritance; see Chap. 10) Hgb 5.0 g/dL
3. Toxic substance (acquired; see Chap. 10) Het 15%
RBC count 1.4 * 10”/L
There are five variants of hemoglobin M (Table 11-19), WBC count PDS AD ING:
which result from a single amino acid substitution in the a- or Reticulocyte count 1%
B-globin chain that stabilizes iron in the ferric form. If the Differential
substitution occurs in the a chain, cyanosis is present at birth. Segmented neutrophils 62%
Cyanosis does not occur with a B-chain substitution until Bands 12%
approximately 6 months of age.” This correlates with the Lymphocytes 19%
Monocytes 4%
switch from y to B chains. The presumptive diagnosis of
Eosinophils 2%
HbM is made from the absorption spectra of hemolysates, and Basophils 1%
hemoglobin electrophoresis on agar gel at pH 7.1.°° Patients RBC indices Normal
have obvious cyanosis but otherwise are generally asympto- Platelet count 400 X 10°/L
Peripheral blood smear (see Fig. 11-16)
matic. No specific treatment is indicated or possible.
ANSWERS
Case Seti yout #
1. Numerous target cells are present on the peripheral
blood smear along with some cells that appear to have
QUESTIONS shadows of precipitating intraerythrocytic crystals.
1. Describe the morphological features of this peripheral 2. The data suggest a diagnosis of HbSC disease.
blood smear. 3. The crystals in HbSC disease appear to be only partially
2. In reviewing the electrophoretic data, what diagnosis is formed, or there may be more than one formation. The
suggested? crystals in the red cells often are described as having a
3. Comment on crystal formation in this condition. glove-shaped appearance with several “fingers” protruding.
4. Discuss the clinical presentation of the patient. Is 1t con- 4. Generally HbSC disease has a milder presentation than
sistent with HbSC disease? HbSS disease; however, this patient appears to be expe-
5. This girl’s parents have no hematologic problems; there- riencing a severe episode of SC crisis. This is indicated
fore, for her to have HbSC disease, what would be their by her acute illness, abdominal pain, and decrease in
most likely genotypes? hemoglobin and hematocrit without an increase in the
6. The inheritance of structurally abnormal hemoglobins reticulocyte count.
follows simple mendelian laws. With parents having the 5. With no hematologic problems, one parent would be
trait form of HbS and HbC, what would be the expected expected to have HbS trait (HbAS), whereas the other
genotypes in any of four children? would most likely have HbC trait (HDAC).
continued 6. The probability for each birth would be 25% for HbAA,
25% for HbAS, 25% for HbAC, and 25% for HbSC.
Jicervom’
1. Which of the following is not a characteristic of hemo- ae 5. Which of the following are crises associated with sickle
globinopathies? cell anemia?
a. Abnormal hemoglobins are synthesized. a. Aplastic crisis with low reticulocyte count, and
b. Result from inherited abnormalities or genetic mutations. infections
Cc)AU are manifested in clinically significant conditions. b. Hemolytic crisis with splenic sequestration, decreased
d. Result in a defect in structural integrity or function of emoglobin and hematocrit, increased reticulocyte
the hemoglobin molecule. count, and jaundice
2. Which of the following are used in the nomenclature c. Vaso-occlusive or painful crises with severe pain,
system for abnormal hemoglobins? tissue damage, and necrosis
a. Capital letters d. All of the above
b: Names of places 6. What are the therapeutic goals in the treatment of sickle
c. Names of chains and substitutions cell anemia?
d\All of the above a. Decrease microvascular entrapment of sickled cells or
3. What is the amino acid substitution found in sickle cell change the volume of RBCs
anemia? b. Modify oxygen affinity or solubility of sickle hemo-
ubstitution of valine for glutamic acid in the sixth globin
position from the NH,-terminal 8 chain c. Increase production of fetal hemoglobin
b. Substitution of lysine for glutamic acid in the sixth aa of the above
position from the NH,-terminal 8 chain 7. What is the amino acid substitution found in HbC
c. Substitution of lysine for glutamic acid in the 26th disease?
position from the NH,-terminal ® chain a. Substitution of valine for glutamic acid in the sixth
d. Substitution of valine for glutamic acid in the 121st osition from the NH,-terminal B chain
position from the NH,-terminal 8 chain (Ob ubstitndes of lysine for glutamic acid in the sixth
4. What factors contribute to the sickling of RBCs? position from the NH,-terminal 6 chain
a. Increase in pH and oxygenation c. Substitution of lysine for glutamic acid in the 26th
(b)Decrease in pH and oxygenation, and dehydration position from the NH,-terminal ® chain
c. Increase in pH and decrease in oxygenation d. Substitution of valine for glutamic acid in the 121st
d. Decrease in dehydration and increase in pH and position from the NH,-terminal B chain
oxygenation
Chapter 11 Hemolytic Anemias: Intracorpuscular Defects: III. The Hemoglobinopathies a i)~—
8. Which of the following is not true for HbC disease? © Hemoglobin electrophoresis at alkaline pH
a. Mild anemia d.{RBC indices
b. Numerous target cells
10. Which of the following is a cause of methemoglobinemia?
c. Crystals in red cells
a. HbM variants :
(A)HbC can be separated from other hemoglobins at an
b. NADH-diaphorase deficiency
alkaline pH
c. Toxic substances
9. Which finding would be most useful in establishing a di- d,\All of the above
agnosis of HbSC disease?
a. Target cells and sickled cells on peripheral blood smear See answers at the back of this book.
_b. Severe anemia: Increased reticulocyte count
= Hemoglobin S (HbS) is produced when valine substitutes mHbC disease presents with a normocytic, and nor-
for glutamic acid in the sixth position of the B chain. mochromic or hyperchromic anemia, and numerous target
cells, microspherocytes, schistocytes, folded cells, and
m= Hemoglobin C (HbC) is produced when lysine replaces “bar of gold” crystals.
glutamic acid at position six of the B chain.
g Electrophoresis in HbC disease shows 95% HbC plus A),
m Sickle cell anemia (HbSS disease) is the most common
and less than 7% HbF.
type of sickle cell disease and represents the homozygous
form in which the individual inherits a double dose of the @ Hemoglobin SC (HbSC) disease occurs when an HbS gene
abnormal gene that codes for HbS. is inherited from one parent and an HbC gene is
inherited from the other; morphology includes target cells,
m=The characteristic morphology in HbSS disease is the folded red cells, and glove-shaped intracellular crystals.
sickled cell, which increases the viscosity of the blood,
leading to hypoxia, painful crises, and infarction of the # Methemoglobinemia occurs when levels exceed 1% of
spleen, kidney, and bone marrow. total hemoglobin and may be the result of hemoglobin M
(HbM) variants, NADH-diaphorase deficiency, or toxic
mw The three types of crises associated with sickle cell dis- substances.
ease are aplastic, hemolytic, and vaso-occlusive (painful).
w Unstable hemoglobins are hemoglobin variants in which
g In sickle cell disease HbS constitutes 80% or more of the amino acid substitutions or deletions have weakened the
hemoglobin content in addition to HbF plus A). binding forces that maintain the structure of the molecule.
mw Sickle cell trait represents the heterozygous form of sickle g High-affinity hemoglobins, associated with a shift to the
cell disease in which individuals inherit both a normal B- left, bind oxygen more readily and release it less easily to
globin gene and a sickle globin gene. the tissues.
w Individuals with sickle cell trait produce both HbA and w Low-affinity hemoglobins, associated with a shift to the
HbS in a ratio of 60:40. right, release oxygen quite readily to the tissues.
228 Chapter 11 Hemolytic Anemias: Intracorpuscular Defects: Ill. The Hemoglobinopathies
REFERENCES and response to inhibition of angiotensin- . Zimmerman, SA, et al: Sustained long-
converting enzyme. N Engl J Med term hematologic efficacy of hydrox-
. Huisman, TH, et al: A syllabus of human SAORI, IG. yurea at maximum tolerated dose in chil-
hemoglobin variants, ed 2. The Sickle . Serjeant, GR, and Serjeant, BE: Sickle dren with sickle cell disease. Blood
Cell Anemia Foundation, Augusta, GA, Cell Disease, ed 3. Oxford University 103:2039, 2004.
1998. Press, New York, 2001. 35). Saunthararajah, Y, et al: Effects of 5-aza-
. Pennsylvania State University: HbVar: A . Lee, MT, et al: Stroke prevention in 2°-deoxycytidine on fetal hemoglobin
database of human haemoglobin variants sickle cell anemia (STOP): Extended levels, red cell adhesion, and hematopoi-
and thalassemias [WWW document], follow-up and final results. Blood etic differentiation in patients with sickle
available at: http://globin.cse.psu.edu, 108:847, 2006. cell disease. Blood 102:3865, 2003.
accessed on March 6, 2005. . Adams, RJ: Stroke prevention and treat- 36. Atweh, GF, et al: Sustained induction of
ie>) . Steinberg, MH: Genetic modulation of fetal hemoglobin by pulse butyrate ther-
ment in sickle cell disease. Arch Neurol
sickle cell anemia. Proc Soc Exp Biol apy in sickle cell disease. Blood
58:565, 2001.
Med 209:1, 1995. . Adams, RJ, et al: Discontinuing prophy- QIEn IR), WOOLY),
Herrick, JB: Peculiar elongated and lactic transfusions used to prevent stroke . Cao, H, et al: Induction of human
sickle-shaped red corpuscles in a case of in sickle cell disease. N Engl J Med gamma globin gene expression by his-
severe anemia. Arch Intern Med 6:517,
353:2769, 2005. tone deacetylase inhibitors. Blood
1910. 2D, Lange, RD, et al: In vitro differences in 103:701, 2004.
Nn . Pauling, L, et al: Sickle cell anemia, a
behavior of sickle cell anemia and trait 38. De Franceschi, L, et al: Oral magnesium
molecular disease. Science 110:543,
erythrocytes produced by variations in pidolate: Effects of long-term adminis-
1949.
O, tension and pH. J Lab Clin Med tration in patients with sickle cell dis-
. Sickle Cell Disease Guideline Panel:
36:848, 1950. ease. Br JHaematol 108:284, 2000.
Sickle cell disease: Screening, diagnosis,
U3), Kark, JA, et al: Sickle-cell trait as a risk 3Y). Brugnara, C: Therapeutic strategies for
management, and counseling in newborns
factor for sudden death in physical train- prevention of sickle cell dehydration.
and infants. Clinical Practice Guideline
ing. N Engl J Med 317:781, 1987. Blood Cells Mol Dis 27:71, 2001.
No 6. AHCPR Pub No 93-0562. Agency
24. Clarke, GM, and Higgins, TN: Labora- AO, Orringer, EP, et al: Purified poloxamer
for Health Care Policy and Research, tory investigation of hemoglobinopathies 188 for treatment of acute vaso-occlusive
Public Health Service, US Department
and thalassemias: Review and update. crisis of sickle cell disease: A random-
of Health and Human Services,
Clin Chem 46:1284, 2000. ized controlled trial. JAMA 286:2099,
Rockville, MD, April 1993.
UD, National Newborn Screening and Genet- 2001.
~—. Steinberg, MH: Management of sickle
ics Resource Center: National Newborn 4 — . Ou, J, et al: L-4F, an apolipoprotein A-1
cell disease. N Engl J Med 340:1021,
Screening Status Report. [WWW Docu- mimetic, dramatically improves vasodi-
IQ).
ment], available at http://genes-r-us. lation in hypercholesterolemia and sickle
ge Eaton, WA, and Hofrichter, J: Sickle cell
uthscsa.edu/, accessed on March 9, 2006. cell disease. Circulation 107:2337, 2003.
hemoglobin polymerization. Adv Protein
26. Henthorn, JS, et al: Neonatal screening 42. Wang, WC, et al: High risk of recurrent
Chem 40:63, 1990.
for sickle cell disorders. Br J Haematol strokes after discontinuance of five to
Stuart, MJ, and Nagel, RL: Sickle-cell
124:259, 2004. twelve years of transfusion therapy in
disease. Lancet 364:1343, 2004.
. Daniel, YA, et al: Rapid and specific patients with sickle cell disease.
. Steinberg, MH: Sickle cell anemia and
detection of clinically significant haemo- J Pediatr 118:377, 1991.
fetal hemoglobin. Am J Med Sci
globinopathies using electrospray mass 43. Vermylen, C, et al: Haematopoietic stem
308(5):259, 1994.
spectrometry-mass spectrometry. Br J cell transplantation for sickle cell ane-
. Hebbel, RP: Beyond hemoglobin
Haematol 130:635, 2005. mia: The first 50 patients transplanted in
polymerization: The red blood cell mem-
28. Gaston, MH, et al: Prophylaxis with oral Belgium. Bone Marrow Transplant 22:1,
brane and sickle disease pathophysiol-
penicillin in children with sickle cell 1998.
ogy. Blood 77:214, 1991.
anemia: A randomized trial. N Engl J 44. Walters, MC, et al: Impact of bone mar-
. National Institutes of Health: The Man-
Med 314:1593, 1986. row transplantation for sickle cell disease:
agement of Sickle Cell Disease, ed 4.
. Charache, S, et al: Effect of hydroxyurea An interim report. Blood 95:1918, 2000.
NIH Publication N. 02-2117. Division of
on the frequency of painful crises in 45. Walters, MC: Stem cell therapy for
Blood Diseases and Resources, National
sickle cell anemia. N Engl J Med sickle cell disease: Transplantation and
Heart, Lung and Blood Institute,
SLIT, IOS. gene therapy. Hematology (Am Soc
National Institutes of Health, Bethesda,
30. Steinberg, MH, et al: Fetal hemoglobin Hematol Educ Program) 66, 2005.
MD, June 2002.
in sickle cell anemia: Determinants of 46. Xu, K, et al: First unaffected pregnancy
. Bunn, HF: Pathogenesis and treatment of
response to hydroxyurea. Multicenter using preimplantation genetic diagnosis for
sickle cell disease. N Engl J Med
Study of Hydroxyurea. Blood 89:1078, sickle cell anemia. JAMA 281:1701, 1999.
SET, ISIS,
INS7/. 47. Larochelle, A, and Dunbar, CE: Genetic
. Castro, O, et al: The acute chest syn-
. Bridges, KR, et al: A multiparameter manipulation of hematopoietic stem
drome in sickle cell disease: Incidence
analysis of sickle erythrocytes in patients cells. Semin Hematol 41:257, 2004.
and risk factors. The Cooperative Study
undergoing hydroxyurea therapy. Blood 48. Rideout, WM IIL, et al: Correction of a
of Sickle Cell Disease. Blood 84:643,
1994.
88:4701, 1996. genetic defect by nuclear transplantation
3. Ferster, A, et al: Hydroxyurea for treat- and combined cell and gene therapy. Cell
. Platt, OS, et al: Mortality in sickle cell
ment of severe sickle cell anemia: A 109:17, 2002.
disease. Life expectancy and risk factors
for early death. N Engl J Med pediatric clinical trial. Blood 88:1960, 49. Chang, JC, et al: Correction of the sickle
1996. cell mutation in embryonic stem cells.
331(15):1022, 1994.
. Maier-Redelsperger, M, et al: Fetal Proc Natl Acad Sci USA 103:1036, 2006.
. Nagel, RL, and Roth, EF, Jr: Malaria and
hemoglobin and F-cell responses to 50. Nagel, RL: Disorders of hemoglobin
red cell genetic defects. Blood 74:1213,
1989. long-term hydroxyurea treatment in function and stability. In Handin, RJ,
young sickle cell patients. The French et al (Eds): Blood: Principles and Prac-
. Falk, RJ, et al: Prevalence and patho-
Study Group on Sickle Cell Disease. tice of Hematology. Lippincott-Raven,
logic features of sickle cell nephropathy
Blood 91:4472, 1998. Philadelphia, pp 1591-1644, 1995.
Chapter 11 Hemolytic Anemias: Intracorpuscular Defects: III. The Hemoglobinopathies bse,
Lhe
51. Lawrence, C, et al: The unique red cell literature. Arch Pathol Lab Med 56. Nagel, RL, et al: HbS-Oman heterozy-
heterogeneity of SC disease: Crystal for- 127:e135—-138, 2003. gote: A new dominant sickle syndrome.
mation, dense reticulocytes, and unusual 54. Vargas-Gonzalez, R, et al: Renal Blood 92:4375, 1998.
morphology. Blood 78:2104, 1991. medullary carcinoma in a six-year-old 57. Wild, BJ, and Bain, BJ: Detection and
52. Huisman, THJ: Combinations of 8 chain boy with sickle cell trait. Patholo Oncol quantitation of normal and variant
abnormal hemoglobins with each other Res 9:193, 2003. haemoglobins: An analytical review.
or with 8-thalassemia determinants with Sp). Noguera-Irizarry, WG, et al: Renal Ann Clin Biochem 41:355, 2004.
known mutations. Influence on pheno- medullary carcinoma: Case report and
type. Clin Chem 43:1850, 1997. review of the literature. Am J Clin Oncol
53. Dimashkieh, H, et al: Renal medullary 26:489, 2003.
carcinoma: A report and review of the
Chapter
Hemolytic Anemias
Intracorpuscular Defects:
IV. Thalassemia
Russell Aaron Higgins, MD
Chantal Ricaud Harrison, MD
Introduction OBJECTIVES
Genetics of Hemoglobin
At the end of this chapter, the learner should be able to:
Synthesis
Pathophysiology of 1. Name the hemoglobin defect of thalassemia.
Thalassemia 2. Describe the different types of mutation involved in alpha («) and beta (8)
Thalassemia Syndromes thalassemia.
Beta Thalassemia
3. Describe the clinical expression of different gene combinations of a and B
Alpha Thalassemia
thalassemia.
Delta—Beta Thalassemia
and Hemoglobin Lepore 4. Describe the condition known as hereditary persistence of fetal hemoglobin (HPFH).
Syndrome
5. List the complications of a regular blood transfusion program for patients with
Hereditary Persistence of
thalassemia major.
Fetal Hemoglobin
Pancellular and Heterocellular 6. Name the most characteristic laboratory findings for the diagnosis of thalassemia.
HPFH
Thalassemia Associated with 7. List red cell indices that help to distinguish thalassemia from iron deficiency.
Hemoglobin Variants 8. Describe the appearance of the peripheral smear in thalassemia.
A Broad Clinical Classification
of Thalassemia Syndromes 9. Name the test used as a population screening test for thalassemia carriers.
Laboratory Diagnosis of 10. List conditions in which quantitation of hemoglobin F is important.
Thalassemia
11. Describe the use of routine chemistries for differentiation of thalassemia from
Routine Hematology
iron-deficiency anemia.
Procedures
Flow Cytometry
Hemoglobin Electrophoresis
Hemoglobin Quantitation
Routine Chemistry
Differential Diagnosis of
Microcytic, Hypochromic
Anemia
Treatment of Thalassemia
Blood Transfusion in
Thalassemia
Curative Treatment of
Thalassemia
Prevention of Thalassemia
Case Study 1
Case Study 2
Case Study 3
230
Chapter 12 Hemolytic Anemias Intracorpuscular Defects: IV. Thalassemia 231
SSS§ « Thalassemia
(57) B Thalassemia
RSS88 o + B Thalassemia
Figure 12-1 ® World distribution of alpha («) and beta (8) thalassemia.
Paha
»
Table 12-1 Composition of Hemos ob
-and Abnormal Hemoglok
Globin Chains Hemoglobin
State
Adult a>
05>
Fetus a*y>
ay,
Embryo O4€> Gower 2
Oe Gower |
Or Portland
a Thalassemia Ba H
4 Bart’s
The ¢- and a-globin genes are found on chromosome 1. Transcriptional mutants affecting one of the promoter
16 and the genes for €-, “y-, “y-, 5-, and B-globins on chromo- sequences
some 11. All globin genes consist of three exons separated by 2. RNA processing mutants, usually affecting introns at the
two introns, with 5° promoter sequences and untranslated splice junction sites or in the consensus sites
regions in 5’ and 3” directions. Each globin gene spans approx- 3. RNA translational mutants due to nonsense or frameshift
imately 1.5 kilobases (Kb). mutations, which produce premature termination and con-
There are two closely linked genes, both active and cod- sistently result in complete lack of production of B chains
ing for identical a globin chains, although at different levels of 4. Miscellaneous mutations affecting initiation codon, 5° cap
activity in the normal adult. The a,-globin gene is expressed at site, or cleaving or polyadenylation sites at the 3° end.’
two to three times the rate of the a,-globin gene.* Detailed
In all cases the final result is a decreased or absent production
mapping of the DNA shows great similarity between the two
of the B-globin chain.
a-globin genes and between the two regions immediately
upstream (5°) of each a-globin gene.*? There are three areas of
homology in DNA blocks called x, y, and z (Fig. 12-2), includ- Pathophysiology of Thalassemia
ing or juxtaposed to the a-globin genes. These homologous
blocks render this area more susceptible to mispairing. Cross- In thalassemia, a decrease in production of one of the globin
ing over in this area of chromosome 16 may result in deletions chains causes a decrease in the amount of normal physiologic
of all or part of one a-globin gene. Occasionally a chromo- hemoglobin produced, resulting in a microcytic, hypochromic
some with three a-globin genes can be produced. This anemia. Because a- and B-globins associate in a stoichiometric
explains why the majority of a thalassemias are the result of a fashion to produce hemoglobin A (a,8;), decreased production
gene deletion. of one globin chain, as occurs in thalassemia, results in a rela-
On the other hand, the majority of 8 thalassemias are the tive excess of the other globin chain. For example, 6 tha-
result of point mutation affecting production of the B-globin lassemia results in an excess of unmatched a globin, which then
chain. A diagram of the B-globin gene fine structure, includ- accumulates in red blood cells. In both a thalassemia and 8 tha-
ing the areas involved in the regulation of expression, is lassemia, this excess globin is responsible for the damage to the
depicted in Figure 12—3. Between 5 and 20 Kb upstream of red blood cell or red blood cell precursors, manifesting as either
the e-globin gene is a B-locus control region (B-LCR), which peripheral hemolysis or ineffective erythropoiesis, respectively.
is involved mainly in the regulation of the switching on and In the case of a thalassemia, the excess y chains and B chains
off of the different B-like genes. Between 30 and 105 bases can form stable tetramers: hemoglobin Bart’s (y,) and hemoglo-
upstream of the B-globin gene lie the three classic promoter bin H (8,), respectively. However, these hemoglobins are phys-
sequences TATA, CAT, and CAC boxes. The point mutations iologically useless and will precipitate in older red cells, causing
resulting in B thalassemia can be classified into the following a shortened red cell life span. These abnormal hemoglobins
categories: may be detected in the peripheral blood as a diagnostic clue for
a By)
———————_:._vWVvO$S
5! a2
Eee] | rr eR Ee
x y Z xX y 7a
Figure | 2-2 m The areas of homology (x, y, and z) can lead to mispairing.
Chapter 12 Hemolytic Anemias Intracorpuscular Defects: IV. Thalassemia 233
cious 6 B 3)
nH
Chromosome 11
Figure 12-5 ™ Diagram of the B-globin gene expression: (top) schematic of the ___location of the B-like genes on chromosome 11 with the
locus control region (LCR) on the 5’ end; (bottom) exploded B-globin gene depicting the three exons and two introns with the three
promoter areas upstream.
a thalassemia. In the case of 8 thalassemia, the excess a chains adult hemoglobin, hemoglobin A (a,8,). The transition, or
form a, precipitates, which cause apoptosis of the red cell pre- switch, from fetal to adult hemoglobin begins in the third
cursors in the bone marrow, resulung in ineffective erythro- trimester and continues until complete at about 6 months;
poiesis.*” In severe forms of B thalassemia there is a selective therefore, infants with B thalassemia syndromes do not pre-
survival of cells producing hemoglobin F, which then becomes sent clinically until after birth. In the heterozygous form, tha-
a diagnostic clue for a homozygous form of 8 thalassemia. lassemia minor, hemoglobin A, is characteristically increased
Although both ineffective erythropoiesis and hemolysis play through an unclear mechanism. In contrast, the homozygous
roles in a and B thalassemia syndromes, hemolysis predomi- forms of B thalassemia, 8 thalassemia major and intermedia,
nates in severe a thalassemia whereas ineffective erythropoiesis characteristically have an increased hemoglobin F as
predominates in severe B thalassemia (Fig. 12-4). described in the preceding text.
The genetic background for 8 thalassemia is very hetero-
geneous. Almost 200 different mutations have been described;
Thalassemia Syndromes however, only some 20 mutations account for more than 80% of
all 8-thalassemia genes worldwide. A specific group of muta-
Beta Thalassemia
tions (four to six) is characteristically found in each geographic
8 Thalassemia does not manifest in utero because the fetus area.° Clinically, these haplotypes may be broadly subdivided
produces fetal hemoglobin, hemoglobin F (a,y,), rather than into B° and B* thalassemia.
Excess o
damages
precursors in
marrow causing
ineffective
erythropoiesis
Figure 12-4 @ Diagram of globin chain imbalance. Excess globin chains precipitate and damage red blood cells and their precursors.
Destruction of red blood cells within the bone marrow (ineffective erythropoiesis) predominates in severe B thalassemia. In contrast, hemoglobin
H precipitates and damages circulating red blood cells (hemolysis) in severe a thalassemia. Notice the changes in hemoglobin constitution of
the peripheral blood with thalassemia: hemoglobin F is increased in severe B thalassemia and hemoglobin H is detectable in severe
a thalassemia.
234 Chapter 12 Hemolytic Anemias Intracorpuscular Defects: IV. Thalassemia
8 thalassemia major, but survive into adulthood without a will be a greater variety in severity of disease, as there may be
large blood transfusion requirement. one, two, three, or four a-globin genes affected in one patient.
Patients with B thalassemia intermedia usually present at a Another characteristic of a thalassemia is the fact that
somewhat older age—generally after the age of 2—and with a the decreased or absent a chain production will result in
slightly higher level of hemoglobin (below 9 to 10 g/dL) than excess y chains during and shortly after fetal life and in
patients with 8 thalassemia major. There is great overlap excess 8 chains later on. This causes formation of stable
between the two conditions at presentation; however, it is very tetramers, such as y, (hemoglobin Bart’s) and 8, (hemoglo-
important to differentiate between them, as the only therapy for bin H), which can be detected by hemoglobin electrophore-
thalassemia major is regular blood transfusion in conjunction sis. These stable, nonfunctional tetramers precipitate in
with iron chelation, whereas the management of thalassemia older red cells, forming inclusion bodies and interfering
intermedia involves mostly supportive therapy with only occa- with membrane function, which results in decreased red
sional blood transfusion under special circumstances. cell survival and may induce a hemolytic crisis during
The serum bilirubin level is significantly more elevated infectious episodes.
in patients with 8 thalassemia intermedia than in those with B
thalassemia major. These patients may develop the physical a’ THALASSEMIA (a THALASSEMIA 1) HAPLOTYPES
and bony characteristics of B thalassemia major including a Thalassemia and « thalassemia | have been used inter-
hepatosplenomegaly. The anemia usually becomes worse changeably in the past; however, «° thalassemia is the preferred
with infections, pregnancy, or folic acid deficiency states. term for the description of the genetic determinant. The «° tha-
These patients are susceptible to frequent, sometimes severe, lassemia haplotype contains deletions of both a genes, a2 and
infections, and gallbladder problems owing to the formation al, on the same chromosome. As a consequence of this deletion
of gallstones. Children usually have an acceptable level of there is a complete absence of production of « chains from the
growth and development (although puberty may be delayed affected chromosome. Because both a-globin genes are deleted,
by a few years), and they reach adulthood if infections are the haplotype can be designated — —, and a patient who is
controlled and if they enjoy good nutrition with particular homozygous for the a° thalassemia mutation would be desig-
emphasis on prevention of folic acid deficiency. They may nated — —/— —. There are at least 21 major a°-thalassemia genes,
become transfusion dependent if severe hypersplenism which differ in the amount of DNA that has been deleted from
occurs. This usually requires splenectomy. Women with the chromosome.°® Each gene appears to be characteristic of a
8 thalassemia intermedia may become pregnant and may certain population in the world. «° Thalassemia genes are found
require blood transfusions as well as folic acid supplementa- frequently in Southeast Asia and less frequently in the Mediter-
tion throughout pregnancy. In spite of the lack of transfusion, ranean area. They also occur sporadically in other parts of the
patients with 6 thalassemia intermedia develop iron overload world. This gene may be recognized in adults through the detec-
as a result of increased gastrointestinal absorption. tion of small amounts of ¢-globin chain.'?
HEMOGLOBIN CONSTANT SPRING from individuals who have non-deletion mutations (— —/a'a or
The a-globin variant Constant Spring, or a, is the result of a a'a /a'a). Clinically, hemoglobin H disease is characterized by
point mutation in the «,-globin gene and may be considered an a variable degree of microcytic, hypochromic anemia.
a* thalassemia haplotype because it affects only one of Hemolytic crises may occur with infections. Because there is a
the two a-globin genes. The point mutation alters the natural switch from fetal (y) to adult (8) globins around the time of
stop codon, allowing translation to continue until the next stop birth, infants and adults have different hemoglobin composi-
codon farther downstream on the mRNA. The end result is an tions. Adults with hemoglobin H disease will have from 5% to
a-globin variant with 31 extra amino acids at its tail end. a 40% hemoglobin H; the remainder is mostly hemoglobin A with
combines with B-globin to produce hemoglobin Constant a small amount of hemoglobin A, and hemoglobin Bart’s.
Spring, Hb CS, which can be detected on hemoglobin Infants who later develop hemoglobin H disease usually have
electrophoresis at alkaline pH as a slow migrating band. between 19% and 27% hemoglobin Bart’s at birth, with the
Heterozygous patients are asymptomatic without hematologic remainder composed of hemoglobin F and hemoglobin A.
abnormalities and produce only small amounts of Hb CS (1%), Hemoglobin H and hemoglobin Bart’s can easily be identified
while homozygotes have a thalassemia minor with microcyto- by hemoglobin electrophoresis, because they migrate anodal to
sis and 5% to 8% Hb CS. Hb CS is found in Southeast hemoglobin A at alkaline pH (Fig. 12-9). These are sometimes
Asia, where it can combine with other prevalent a thalassemia referred to as fast hemoglobins. In addition, hemoglobin H
haplotypes to produce a thalassemia syndromes. shows a characteristic appearance of multiple ragged inclusions
in many red cells after incubation with brilliant cresyl blue, the
CLINICAL EXPRESSION OF THE DIFFERENT GENE so-called golf-ball appearance (Fig. 12-10).
COMBINATIONS (a THALASSEMIAS) a Thalassemia minor, also called a° thalassemia trait or
a Thalassemia can be divided into four clinical categories, a thalassemia | trait, is caused by defects of two of the four
depending on the severity of the disease. The most severe a-globin genes. This is usually the result of heterozygosity for
expression of a thalassemia 1s the hemoglobin Bart’s hydrops the a° thalassemia haplotype (— —/aa) but could also be the
fetalis syndrome, which is caused by homozygosity of the a° result of homozygosity for the a* thalassemia haplotypes
thalassemia gene haplotype (— —/— —). This is a lethal disease, (-a/-a). The condition is characterized by the presence at
and infants with hemoglobin Bart’s hydrops fetalis die either birth of 5% to 15% hemoglobin Bart’s, which disappears with
in utero or soon after birth. They produce no @ chains, and the development and is not replaced by hemoglobin H. Adults,
only hemoglobins found are hemoglobin Bart’s (y,) and therefore, will have a normal hemoglobin electrophoretic
hemoglobin Portland (C,y.). Because hemoglobin Bart’s is pattern. There is a minimal amount of anemia with slight
useless as an oxygen carrier, survival of these fetuses into the hypochromia and microcytosis present. The mean corpuscu-
third trimester or until birth is entirely due to the presence of lar volume (MCV) is usually between 70 and 75 fL. This
hemoglobin Portland. This condition is quite common in South- condition exists in 3% of African Americans and may be
east Asia in the -SEA/-SEA form but is also found sporadically confused with iron deficiency.
in the Mediterranean area in the form of -MED/—MED. The last category of a thalassemia 1s the silent carrier of
The second most severe clinical expression of a a thalassemia, also called a* thalassemia trait or a tha-
thalassemia is hemoglobin H disease. In this entity only one lassemia 2 trait. This is the result of a defect in one of the four
a-globin gene out of four is functioning. This is usually the a-globin genes and is characterized by the presence of a very
result of a compound heterozygosity of the deletional a° tha- small amount (up to 2%) of hemoglobin Bart’s at birth; after
lassemia and a*-thalassemia haplotypes (—/-c) but can also arise the disappearance of hemoglobin Bart’s during development,
+ _—
H x
Figure 12-9 m Diagram of the migration of the different hemoglobins at different pH: (7) normal adult (A, A2); (2) homozygous Hb Lepore
(F, Lepore); (3) HbH/Constant Spring disease a“a/(Constant Spring, A>, A, Bart’s H); (4) compound heterozygous HbE/f-thalassemia (E, F);
(5) Hb Bart’s hydrops fetalis syndrome (Portland, Bart’s); (6) HbS/C disease (S, C).
238 — Chapter 12 Hemolytic Anemias Intracorpuscular Defects: IV. Thalassemia
ee
used to identify hemoglobin Bart’s hydrops fetalis as early as
the first trimester.'*
DAP
Figure 12-10 ® Hemoglobin H inclusions (supravital stain). (From HEMOGLOBIN H DISEASE Hemoglobin H disease is sim-
Bell, A: Hematology. In: Listen, Look and Learn. Health and Education ilar to ® thalassemia intermedia in that it covers a wide
Resources, Inc., Bethesda, MD, with permission.) range of clinical severity from the patients with very mild
anemia to patients potentially requiring regular transfusion.
The age at presentation varies from 0 to 74 years old.’
no recognizable hematologic abnormality is present, except Infants will have near normal hemoglobin levels and no
for a borderline low MCV (78 to 80 fL). This condition is splenomegaly, so the diagnosis must come from clinical
found in up to 30% of African Americans. suspicion and laboratory testing. Most cases are discovered
In the simplest terms, deletional type mutations result in in adulthood and will have episodic periods of anemia, pal-
predictable clinical syndromes such that four deletions produce lor, and weakness during which they may be discovered to
hemoglobin Bart’s hydrops fetalis, three deletions produce have a hypochromic, microcytic anemia, leading to an
hemoglobin H disease, two deletions produce a thalassemia eventual diagnosis. These episodes of anemia may be asso-
minor, and one deletion produces a silent carrier (Fig. 12-11). ciated with concurrent infections, medications, or other ill-
Because non-deletion mutations and hemoglobin variants can nesses. Other clinical features may include scleral icterus,
also combine with deletional a-globin gene mutations, the hepatosplenomagaly, gallstones, and bone marrow expan-
actual molecular basis for a thalassemia syndromes is more het- sion. The bone changes are present in about one third of
erogeneous. The different genetic backgrounds associated with cases but are milder than that seen in severe B thalassemia.
the four different clinical expressions of @ thalassemia are sum- Splenomegaly, if present, may worsen the anemia and
marized in Table 12-2. sequester platelets, so these patients may benefit from a
Mother’s haplotype
Father’s haplotype
6 <<
x —mmen:
Silent carrier a thalassemia minor Hb H disease
<——
ae os aS >t S<
a thalassemia minor Hb H disease Hb Bart’s
Figure 12-11 @ Simplistic look at the interactions of deletional mutations of « thalassemia. The severity of disease is predictable and
depends on the number deleted a-globin genes.
Chapter 12 Hemolytic Anemias Intracorpuscular Defects: IV. Thalassemia 239
Chromosome
Genes J —O asq
aa = normal haplotype; -a = deletion of one a-globin gene; a°Sa = Hb Constant Spring; a'a = nondeletion « thalassemia
gene; — = deletion of both a-globin genes (Note: the clinical phenotype resulting from the combination of these haplotypes
is found at the intersection of the corresponding column and row); N = normal clinical phenotype; « carrier = silent carrier
of a thalassemia, 5%—15% Hb Bart’s at birth, mild anemia; thal = « thalassemia minor, 0%-2% Hb Bart’s at birth, minimal
hematologic changes; H = hemoglobin H disease; Bart’s = hemoglobin Bart’s hydrops fetalis.
Deletion
mutation in mutation in
Sy promoter Ay promoter
region region |
|
Loss of competition Increased Increased
from deleted genes transcription of Sy transcription of Ay
allows increased y
transcription
Increased hemoglobin F
Figure | 2-15 @ Mutations seen in HPFH with hemoglobin F in 20% range. Mutations producing HPFH in this range have common themes.
One common molecular mechanism (left) is the deletion of the 8 and 6-globin genes, (88)°. This eliminates the competition of the locus
control region for the 8 and 8 promoters and allows increased y transcription. Another common theme is a point mutation in the Sy or Ay
promoters which increases the association of the locus control region with the respective y-globin gene and increases its transcription.
Chapter 12 Hemolytic Anemias Intracorpuscular Defects: IV. Thalassemia 241
Deletion Deletion
s /
Increased y globin chain compensates for loss of 6 and 8 globin chains
Figure 12-14 (8p)? Deletions can be found in both HPFH and df thalassemia. A greater amount of hemoglobin F is seen in HPFH than in
5B thalassemia. Two hypotheses for increased production of y globin expression in HPFH are shown above. The 5B deletion along with an
intergenic sequence responsible for y silencing during development is shown at the /eft. A second hypothesis in which a deletion results in
the juxtaposition of a downstream enhancer with the y-globin gene, where it can have a positive effect on y transcription, is shown at the
right. The mechanisms are not exclusive of one another and may coexist. Complete compensation for loss of 8 and B globins with increased
y globin prevents a globin imbalance between a and non-a globin chains in HPFH.
genes. There are two main theoretical models to explain the This technique is discussed later in this chapter with the other
different hemoglobin F output between (68) HPFH and (6B)° laboratory methods useful in the diagnosis of thalassemia.
thalassemia (Fig. 12—14). In the first model, a (68)? HPFH
deletion brings a downstream enhancer into proximity of the PANCELLULAR HPFH
-globin gene. The enhancer then has a positive effect on Various forms of (58)° HPFH have been described, all of which
transcription of the y-globin gene. This has been supported by produce a pancellular distribution of hemoglobin F. One com-
mouse experiments in which HPFH breakpoint sequences mon form is the African (HPFH 1), in which there is a deletion
increase y transcription.'® In the second model, a region of the 6- and B-globin genes and increased synthesis of “y and
between the y and 6-globin genes, which may be responsible “vy chains, which almost completely compensate for the lack of
for y silencing around the time of birth, is deleted in HPFH.
Evidence that this intergenic region may also play a role is
supported by the demonstration of increased ‘y transcription
with its deletion.'”
Homozygotes
Homozygous B° Thalassemia major
Homozygous B* Thalassemia major or intermedia
Homozygous 5B Thalassemia intermedia
Homozygous Hb Lepore Thalassemia major or intermedia
Homozygous HPFH Thalassemia minima
Homozygous a°-thalassemia (—/—) Hemoglobin Bart’s hydrops fetalis
Homozyous a*-thalassemia (—o/—c) Thalassemia minor
Compound Heterozygotes
Compound heterozygous B°/severe B* Thalassemia major
Compound heterozygous B**/severe B* or B° Thalassemia major oi intermedia
Compound heterozygous 58/B° Thalassemia major or intermedia
Compound heterozygous Lepore/8B Thalassemia intermedia
Compound heterozygous Lepore/B° or B* Thalassemia major
Compound heterozygous HPFH/B’ or B* Thalassemia minor
Compound heterozygous E/f° or * Thalassemia major or intermedia
Compound heterozygous C/B° or B* Thalassemia intermedia or minor
Compound heterozygous S/B° or B* Sickle cell disease
Compound heterozygous a-thalassemia (—/—a) Thalassemia intermedia (hemoglobin H disease)
Heterozygotes
Heterozygous 8° or B* Thalassemia minor
Heterozygous B** Thalassemia minima
Heterozygous 68 Thalassemia minor
Heterozygous Lepore Thalassemia minor
Heterozygous HPFH Thalassemia minima
Heterozygous a° thalassemia Thalassemia minor
Heterozygous a* thalassemia Thalassemia minima
strongly suspected on the basis of the results of routine hema- MCH is usually below 22 pg and the MCV below 70 fL,
tology procedures. whereas the hemoglobin level is in the 9 to 11 g/dL range.
In heterozygous thalassemia, there is often a relative
Routine Hematology Procedures erythrocytosis. That is, the RBC count is high relative to the
hemoglobin level. A general rule is that, in non-thalassemic
AUTOMATED BLOOD CELL ANALYZER individuals, the hemoglobin is three times the RBC count.
Automated blood cell analyzers are commonly used in devel- Therefore a patient with a RBC count of4 million will have a
oped countries and provide hemoglobin levels, hematocrit, and hemoglobin level of approximately 12 g/dL. However,
red blood cell indices (see Chap. 31). The red bluod cell indices thalassemic patients have a relative erythrocytosis, so the
are invaluable to clinicians and laboratory personnel, who are hemoglobin level will be much less than three times the RBC
sorting through the differential diagnosis of microcytic count. For example, an individual with heterozygous B tha-
hypochromic anemia. The thalassemias in general are charac- lassemia may have a RBC count of 4 million and a hemoglo-
terized by a decrease in hemoglobin level, hematocrit, mean bin level of 9 g/dL. Demonstrating a relative erythrocytosis in
corpuscular volume (MCV), and mean corpuscular hemoglo- an individual with a low MCV should alert the interpreter of a
bin (MCH) in conjunction with a normal-to-increased red possible thalassemia, and further testing is indicated.
blood cell (RBC) count, a normal to mildly decreased mean
corpuscular hemoglobin concentration (MCHC), and a normal PERIPHERAL SMEAR EXAMINATION
red cell volume distribution width (RDW). The only exception The careful examination of a well prepared peripheral smear
is thalassemia major, in which the degree of anisocytosis is is essential to the diagnosis of thalassemia.
such that the RDW is increased. The decrease in MCV is usu-
ally striking and disproportionate to the decrease in hemoglo- WRIGHT’S STAIN In homozygous B and compound heterozy-
bin and hematocrit. This fact, in conjunction with the relatively gous non-a thalassemia, the peripheral smear demonstrates
high RBC count and the normal RDW, offers a useful discrim- extreme anisocytosis and poikilocytosis with bizarre shapes, tar-
ination index between heterozygous a or B thalassemia and get cells, ovalocytes, and large numbers of nucleated red cells
iron deficiency.'? In iron deficiency, the RDW is increased and (see Fig. 12-5). There is marked hypochromia and microcyto-
the decrease in MCV is less striking and observed only when sis. In heterozygous f thalassemia, the cells are hypochromic
the anemia is more severe. In heterozygous thalassemia, the and microcytic with a mild to moderate degree of anisocytosis
244 Chapter 12 Hemolytic Anemias Intracorpuscular Defects: IV. Thalassemia
and poikilocytosis. Target cells are frequent, and basophilic stip- quality of the technique, as this technique is very sensitive to
pling is often seen (see Fig. 12-6). The peripheral smear of a many variables. Flow cytometry can also be used to assess he-
patient with the sickle cell thalassemia syndrome can be differ- moglobin F distribution (see later) and may suffer less from
entiated from that of a patient with pure sickle cell anemia by the confounding variables of the Kleihauer—Betke test.
the presence of hypochromia, microcytosis, numerous target This stain is very useful in demonstrating the distribution
cells, and only an occasional sickled cell. of hemoglobin F and can be used to differentiate between
In hemoglobin H disease, the peripheral smear demon- pancellular and heterocellular HPFH. It is also useful in dif-
strates hypochromia with microcytosis, target cells, and mild ferentiating heterozygous 58 thalassemia from heterozygous
to moderate anisopoikilocytosis. Patients with heterozygous pancellular HPFH, because the former usually has a hetero-
a’ thalassemia usually demonstrate a mild hypochromia and cellular distribution of hemoglobin F.
microcytosis, whereas those with heterozygous a* tha-
lassemia usually have a perfectly normal peripheral smear. OSMOTIC FRAGILITY
The red cells of patients with homozygous or heterozygous 8
SUPRAVITAL STAINS The reticulocyte count is usually ele- thalassemia, hemoglobin H disease, and «° thalassemia trait
vated up to 10% in hemoglobin H disease and up to 5% in have a decreased osmotic fragility. This fact is not very use-
homozygous B thalassemia but is disproportionately low in ful for diagnostic purposes in a specific patient, but it is the
relation to the degree of anemia in the latter condition. basis of a simple, inexpensive method of screening for the
In hemoglobin H disease, incubation of the red cells with thalassemia carrier state in large populations.
brilliant cresyl blue stain causes in vitro precipitation of hemo-
globin H owing to the redox action of the dye. This results in a
Flow Cytometry
characteristic appearance of the majority of the red cells, which
display multiple discrete inclusions, the appearance of which Flow cytometry of red blood cells using fluorescently labeled
has often been compared to that of a golf ball (see Fig. 12-10). anti-hemoglobin F antibodies can be used to determine if the
Occasionally, and after extensive searching, such cells contain- distribution of hemoglobin F is pancellular or heterocellular.
ing hemoglobin H inclusions can be found in the a° thalassemia Red blood cells are permeabilized and then incubated with
carrier. anti-hemoglobin F before being loaded onto the flow cytome-
In patients with homozygous § thalassemia or hemoglo- ter. As red blood cells pass through the laser in single file, they
bin H disease who have undergone splenectomy, incubation are simultaneously counted and assessed for the presence of
of the blood with methyl violet stain can demonstrate Heinz hemoglobin F. The end result is either one peak, representing
body—like inclusions, which represent in vivo precipitation of a single population of red blood cells containing hemoglobin
the abnormal hemoglobin. F, or two peaks, representing separate populations with and
without hemoglobin F.
ACID ELUTION STAIN The acid elution technique (see Flow cytometry is useful in the same situations where
Chap. 31), originally described by Kleihauer and Betke, is Kleihauer—Betke acid elution is useful. Individuals with
based on the fact that at an acid pH of about 3.3, hemoglobin hemoglobin F in the 20% range may have either heterozygous
A is eluted from an air-dried, alcohol-fixed blood smear, HPFH or heterozygous 58 thalassemia, and the flow cytome-
whereas hemoglobin F is resistant to elution. After such treat- try method can identify the pancellular and heterocellular dis-
ment and subsequent staining with eosin or erythrosin, normal tribution of hemoglobin F, respectively” (Fig. 12-16).
adult red cells appear as very faint ghosts. Red cells contain-
ing hemoglobin F demonstrate a variable amount of stain,
Hemoglobin Electrophoresis
depending on the amount of hemoglobin F present. A con-
trolled preparation containing a mixture of adult and cord cells Hemoglobin electrophoresis plays an important role in the diag-
must also be stained and examined in parallel to check the nosis of thalassemia by allowing the detection of increased
200 200
160 160
120 120
Counts 80
Counts
40
levels of hemoglobin A, and hemoglobin F as well as the pres- Citrate agar gel electrophoresis, which is performed at
ence of abnormal hemoglobins, such as hemoglobin H, hemo- an acid pH between 5.9 and 6.2, is helpful in identifying
globin Bart’s, hemoglobin Lepore, hemoglobin Constant hemoglobin variants that can be inherited along with tha-
Spring, or other structurally abnormal hemoglobins that can be lassemia.
found in association with thalassemia (hemoglobin S, hemoglo-
bin C, hemoglobin E). Table 124 contains a summary of the
different patterns of the hemoglobins present in the non-c tha- Hemoglobin Quantitation
lassemia syndromes. Although an experienced observer can detect an increased
Routine hemoglobin electrophoresis to confirm the diag- level of hemoglobin A, or hemoglobin F on cellulose acetate
nosis of thalassemia is performed at an alkaline pH around or starch gel electrophoresis, actual quantitation is necessary
8.4 on cellulose acetate, starch, or agarose gels. At alkaline pH, to truly establish the diagnosis of thalassemia.
the hemoglobins migrate from the most cathodal to the most
anodal in the following order: first hemoglobin Constant Spring,
then hemoglobins A,, C, and E migrate in the same band; next
hemoglobins S and Lepore, again in the same band; then hemo-
Q
globin F, followed by hemoglobin A, hemoglobin Portland, nN
“Oo
hemoglobin Bart’s, and finally, hemoglobin H. The different
patterns of migration of these hemoglobins are illustrated in
Figure 12-9. An example of alkaline electrophoresis on agarose
gel with some commonly encountered entities is shown in
€
Figure 12-17. Cellulose acetate or starch gel electrophoresis can
be performed at low to neutral pH to detect hemoglobin H and
hemoglobin Bart’s easily, as they migrate anodally (.e., in the
direction opposite to the other hemoglobins) at this pH.
CELLULOSE ACETATE
Cellulose acetate electrophoresis has replaced starch gel elec-
trophoresis in most laboratories, owing to its simple, rapid
method. It uses a smaller sample than starch gel electrophoresis, - +
and minor components such as hemoglobin Constant Spring
and small amounts of hemoglobin A, may be overlooked. Small . A2 SF AM
amounts of hemoglobin A in the presence of mostly hemoglobin CD
F also can be difficult to detect. aie
O
OTHER GELS
Starch gel electrophoresis is a little more cumbersome and Figure 12-17 m Electrophoresis at alkaline pH. Lanes 7 and 7 are
controls with hemoglobins C, S, F, and A. Lanes 3 and 4 are
time consuming. The results of the starch gel electrophoresis normal individuals (A, = 2.5%). Lane 5 is an individual with
are similar to those of the cellulose acetate procedure. Elec- 6 thalassemia minor (A, = 4.7%). Lane 6 is an individual with
trophoresis with starch gel is better at defining the presence of a thalassemia minor; a thalassemia minor has a pattern with a
hemoglobin Constant Spring and should always be used if normal A>. Lane 2 is an individual with B thalassemia minor (A; =
such a variant is suspected. 5%) and “Swiss HPFH” (F, = 7%).
246 Chapter 12 Hemolytic Anemias Intracorpuscular Defects: IV. Thalassemia
Iron deficiency
a Thalassemia
8 Thalassemia
Hemoglobin E disease
Anemia of chronic disease
Sideroblastic anemia
CO iastente
1. What is the hemoglobin defect found in thalassemia syn- c. B Thalassemia minor (8 */B)
dromes? d. Hemoglobin H disease (—a/— —)
a. Abnormal incorporation of iron molecule
(Defective production of the globin portion 4, What is the term for the clinical course of homozygous
(d8)° thalassemia?
c. Excessive production of porphyrins
a. Thalassemia minor
d. Amino acid substitution
b. Thalassemia major
2. What type of globin chains and hemoglobin are charac- c. Thalassemia trait
teristics of severe a thalassemia? d. {Thalassemia intermedia
a. Two a chains and two B chains (HbA)
5. Hereditary persistence of fetal hemoglobin (HPFH) is
b. Two a chains and two 6 chains (HbA,)
characterized by the persistence of fetal hemoglobin into
(¢)Four 8 chains (HbH) or four y chains (Hb Bart’s)
adult life. What are the clinical manifestations of this
d. Two @ chains and two y chains (HbF)
condition?
3. Which type of thalassemia has primarily hemoglobin a. Chronic anemia with skeletal abnormalities caused by
Bart’s and shows the following clinical expressions: excessive erythropoiesis
infants die in utero or soon after birth, severe anemia, b. Asymptomatic except during pregnancy or stressful
marked hepatomegaly and splenomegaly, and situations
ascites? c. Hydrops fetalis syndrome
(a. Homozygous «° thalassemia (— —/— —) CoN significant abnormalities for heterozygous; minor
b. Homozygous B° thalassemia (B°/B") 7 ymptoms for homozygous
250 Chapter 12 Hemolytic Anemias Intracorpuscular Defects: IV. Thalassemia
@ Hereditary persistence of fetal hemoglobin (HPFH) com- (MCV), and mean corpuscular hemoglobin (MCH), in con-
prises a group of conditions characterized by the persis- junction with a normal to increased red cell count.
tence of fetal hemoglobin synthesis into adult life without w Hemoglobin electrophoresis aids in the diagnosis of
producing significant hematologic abnormalities. thalassemia by detecting increased levels of hemoglobin
g Thalassemia has been associated with a number of hemo- A, and hemoglobin F, as well as other abnormal
globin structural variants, including hemoglobin S, hemo- hemoglobins.
globin C, and hemoglobin E.
@ The thalassemias are generally characterized by a decrease
in hemoglobin level, hematocrit, mean corpuscular volume
REFERENCES . Mourad, FH, et al: Comparison between differentiation from iron deficiency. Am
desferrioxamine and coibined therapy J Clin Pathol 80:31, 1982.
Ih. Cooley, TB, and Lee, P: A series of cases
with desferrioxamine and deferiprone in 20. Hoyer, JD, et al: Flow cytometric mea-
with splenomegaly in children with ane- surement of hemoglobin F in RBCs:
iron overloaded thalassemia. Br J
mia and peculiar bone changes. Trans Diagnostic usefulness in the distinction
Haematol 121:187, 2003.
Am Ped Soc 37:29, 1925. 12. Bradai, M, et al: Hydroxyurea can elimi- of hereditary persistence of fetal hemo-
NO. Nagel, RL, and Roth, EF: Malaria and
nate transfusion requirements in children globin (HPFH) and hemoglobin S-HPFH
red cell genetic defects. Blood 74:1213, with severe B-thalassemia. Blood from other conditions with elevated
1989. 102:1529, 2003. hemoglobin F. Am J Clin Pathol
~Liebhaber, SA, et al: Human a-globin
. Tang, WT, et al: Immunocytological test 117:857, 2002.
gene expression. The dominant role of to detect adult carriers of (S*"/) dele- . Suh, DD, et al: Influence of hemoglobin S
the a2-locus in mRNA and protein syn- tional a-thalassemia. Lancet 342:1145, adducts on hemoglobin A2 quantification
thesis. J Biol Chem 261:15327, 1986. by HPLC. Clin Chem 42:113, 1996.
1993.
. Liebhaber, SA, et al: Homology and con- . Leung TN, et al: Thalassaemia screening . Schrier, SL, and Angelucci, E: New
certed evolution at the al and «2 loci of in pregnancy. Curr Opin Obstet Gynecol strategies in the treatment of the tha-
human a-globin. Nature 290:26, 1981. EI 2Y), KOO. lassemias. Annu Rev Med 56:157, 2005.
. Higgs, DR, et al: The molecular pathol- . Gibbons, R, et al: The a thalassemias . Sevilla, J, et al: Hematopoietic transplan-
ogy of the thalassemias. In Weatherall, and their interactions with structural tation for bone marrow failure syn-
DJ, and Clegg, JB (Eds): The Tha- haemoglobin variants. In Weatherall, DJ, dromes and thalassemia. Bone Marrow
lassemia Syndromes, ed 4. Blackwell Sci- and Clegg, JB (Eds): The Thalassemia Transplant 35:S17, 2005.
entific Publications, Oxford, 2001, p 133 Syndromes, ed 4. Blackwell Scientific 24. Rivella, S, et al: A novel murine model of
ON. Huisman, THJ, et al: A syllabus of tha- Cooley anemia and its rescue by lentiviral-
Publications, Oxford, 2001, p 484.
lassemia mutations. The Sickle Cell Ane- . Katsantoni, EZ, et al: Persistent -y-globin mediated human B-globin gene transfer.
mia Foundation. Augusta, GA, 1997. expression in adult transgenic mice is Blood 101:1902, 2003.
. Ho, PJ, and Thein, SL: Gene regulation mediated by HPFH-2, HPFH-3, and DS. Ravindran, MS, et al: 8 thalassemia car-
and deregulation: A B globin perspec- HPFH-6 breakpoint sequences. Blood rier detection by ELISA: A simple
tive. Blood Rev 14:78, 2000. 102:3412, 2003. screening strategy for developing coun-
. Yuan, J, et al: Accelerated programmed . Chakalova L, et al: The Corfu 58 tha- tries. J Clin Lab Anal 19:22, 2005.
cell death (apoptosis) in erythroid precur- lassemia deletion disrupts y-globin gene . Chow, J, et al: Evaluation of single-tube
sors of patients with severe B-thalassemia silencing and reveals post-transcriptional osmotic fragility as a screening test for
(Cooley’s anemia). Blood 82:2, 1993. regulation of HbF expression. Blood thalassemia. Am J Hematol 79:198,
. Poortrakul, P, et al: A correlation of ery- 105:2154, 2005. 2005.
throkinetics, ineffective erythropoiesis, . Emburg, SH, et al: Concurrent sickle-cell
and erythroid precursor apoptosis in Thai anemia and a-thalassemia. Effect on See the Bibliography for this chapter at the
patients with thalassemia. Blood 96:7, severity of anemia. N Engl J Med back of the book.
2000. 306:270, 1982.
. Cao, A: Diagnosis of B-thalassemia . Johnson, CS, et al: Thalassemia minor:
intermedia at presentation. Birth Defects Routine erythrocyte measurements and
22a MOS8:
Chapter
Hemolytic Anemias
Extracorpuscular Defects
Denise M. Harmening, PhD, MT(ASCP), CLS(NCA)
Louann W. Lawrence, DrPH, MT(ASCP)SH, CLSpH(NCA)
Ralph Green, BAppSci(MLS), FAIMLS
Carl R. Schaub, MD
252
Chapter 13 Hemolytic Anemias: Extracorpuscular Defects 253
immunoglobulin (Fig. 13-1). Clq then causes the activation C5 convertase (C4b2a3b) cleaves C5 into the compo-
of Clr, which then activates Cls. (A bar across the top of a nents C5a, which is released into the plasma and acts as an
complement component denotes its active form as shown in anaphylatoxin and a chemotactic agent, and C5b, which binds
Figures 13-1 and 13-2.) C4 is the second complement protein C6 and C7 to the cell membrane. Membrane-bound C5b67
to be activated. This occurs when Cls cleaves C4 into its acti- causes binding of C8, resulting in immediate ion flux into the
vated components, C4a, which remains in the plasma, and cell and the beginning of cell lysis. The C5b678 complex can
C4b, of which a small number of molecules attach to the cell bind up to six C9 molecules, together forming the membrane
membrane, with the rest remaining in the plasma in the inac- attack unit, C5b6789, which causes cell lysis and accelerated
tive form. C2 attaches to C4b in the presence of magnesium movement of ions into the cell. With the binding of C9, the
and is then cleaved by Cls into a and b subunits. C4b2a com- rate of cell lysis is greatly accelerated (see Fig. 13-1).
bines with C4b and forms the enzyme C3 convertase (C4b2a), Complement activity is regulated by certain inhibitors
and C2b is released into the plasma. (Cls inhibitor, C3b inactivator, C4 inactivator) and by the
Amplification of complement activity now occurs with the instability of certain components (C4b2a, and C4b2a3b).'
action of C3 convertase on C3. C3 convertase (C4b2a) cleaves
C3 into its active components, C3a and C3b, and is able to cleave ALTERNATE (PROPERDIN) PATHWAY The alternate, or
hundreds of C3 molecules. C3a is released into the plasma and properdin, pathway of complement activation also results in cell
acts as an anaphylatoxin. C3b binds to the cell membrane and lysis, but by a different mechanism and group of proteins. The
combines with C4b2a to form another enzyme, C5 convertase alternate pathway bypasses the complement components Cl,
(C4b2a3b). Some of the C3b molecules attach to other sites on C2, and C4 and enters at C3. This pathway consists of a distinct
the cell, are inactivated (iC3b), or are cleaved by C3 inactivator group of proteins: complement component C3; factor B, which
to C3c, which 1s released into the plasma, and to C3d, an inactive is enzymatically cleaved into fragments Bb (biologically active)
subunit that remains attached to the cell. The components C4, and Ba; factor D, which cleaves factor B; properdin (P), a serum
C2, and C3 are referred to as the enzyme activation unit. protein that stabilizes the C3bBb complex; and factor H (C3b
inactivator accelerator), which aids in controlling activation of
Cl (Clq, Clr, Cls—recognition unit) the alternate pathway? (see Fig. 13-2).
Clq + IgG or IgM + Caté
mace
y
ace C3
C3 +C3bBb (C3 Convertase)
C3a ,C3 inactivator (factor |, factor H) C8b inactivator ae
YA
¢ Vi ee C3a
N
.
|
C5 G5
ten
C5a
C5b + C6 + C7 C5b + C6 + C7
C5b67 + C8
<—
|<
ante+ C9 oTSi +~ @)
O a| QO foe)
o
=—— OO
<——
C5b6789 (membrane attack unit) oO a oy <4foe)©
|
Cell Lysis
<—o
Cell Lysis
Figure 13-1 @ Classical pathway of complement activation. Figure 15-2 @ Alternate pathway of complement activation.
254 Chapter 13. Hemolytic Anemias: Extracorpuscular Defects
Mechanisms IgM or IgG3, IgG1, IgG2 (two IgG IgM and/or IgG sensitization with/
molecules within close proximity of each without iC3b (inactivated complement)
other) activate complement to completion Cell-mediated phagocytosis
Antibody-dependent cellular
cytotoxicity (ADCC)
Organ Occurs within blood vessels Spleen: IgG alone or IgG +
iC3b-coated cells
Liver: iC3b alone or IgG +
iC3b-coated cells
Laboratory findings Hemoglobinemia Spherocytosis
Hemoglobinuria
Serum haptoglobin: marked decrease Serum haptoglobin: decreased
Indirect bilirubin: elevated Indirect bilirubin: elevated
Lactate dehydrogenase (LD): elevated Lactate dehydrogenase (LD): elevated
Positive direct antiglobulin test Positive direct antiglobulin test
hemolysis, whereas hematuria is not related to hemolysis. process does not always go to completion (C1 through C9). In
Microscopic examination of the urine may be helpful in iden- most cases, complement activation is stopped by an inhibitory
tifying intact red cells to distinguish hematuria from hemo- factor (control mechanism) at the C3b stage. C3b is cleaved to
globinuria. In chronic intravascular hemolysis, hemosiderin form iC3b. If activation is stopped, iC3b is further broken
may appear in the urine (hemosiderinuria). down into C3dg, which then remains attached to the red cell
In hemolysis, free hemoglobin in the blood is bound by membrane.”
a molecule called haptoglobin, which is consumed in the Red cells coated with IgG! or IgG3 are preferentially
process. Within hours of intravascular hemolysis, haptoglobin removed in the spleen® rather than the liver, because blood
is depleted. As little as 5 mL of lysed red cells can bind all of passing through the spleen becomes hemoconcentrated, alter-
the available haptoglobin. However, decreased haptoglobin is ing the ratio between free [gG in the plasma and cell-bound
not specific for intravascular hemolysis, since it is also seen IgG. Free IgG can bind to Fe receptors, blocking their ability
in extravascular hemolysis. In addition, haptoglobin is rapidly to bind the IgG that is attached to red cells. The condition of
synthesized in the liver, and can return to normal levels within hemoconcentration in the spleen shifts the ratio of free IgG to
24 hours. Because haptoglobin is an acute phase reactant pro- red cell—bound IgG in favor of the red cell—bound IgG. The
tein, concentrations may vary considerably, depending on activated form of complement (C3b) or its inactivated form
several factors, such as underlying disease processes. Conse- (iC3b) are not present in the free form in plasma; therefore,
quently, care must be taken when interpreting haptoglobin
levels as an indicator of hemolysis. Ji is advisable to have a
“baseline” haptoglobin level to compare to the haptoglobin INTRAVASCULAR HEMOLYTIC EVENT
level following suspected hemolysis. See Figure 13-3 for the
sequence of laboratory findings. Serum haptoglobin
Hemoglobinuria
EXTRAVASCULAR IMMUNE HEMOLYSIS Extravascular
hemolysis is the phagocytosis of red cells by fixed phagocytes
within the mononuclear phagocyte system (MPS), formerly
called the reticuloendothelial system. The two major organs of Level
——~> —
the MPS are the spleen and the liver. In the red pulp region of
Hemosiderinuria
the spleen, macrophages with Fc receptors (see ADCC, earlier)
line the splenic cords. Antibody-coated red cells interact with
the Fe receptors, resulting in complete or partial phagocytosis.
In the case of partial phagocytosis, part of the red cell membrane
is removed. If the red cell membrane is able to repair itself, the Time (days)
normal biconcave disk is converted to a sphere-shaped red cell
called a spherocyte. Spherocytes lack deformability and become Figure 13-3 @ Indicators of acute intravascular hemolysis. Within
a few hours of an acute hemolytic event, free hemoglobin is cleared
physically trapped in the spleen; those that do escape the spleen
from plasma and the serum haptoglobin falls to undetectable levels:
can be seen in the peripheral blood and their presence 1s indica- hemoglobinuria ceases soon after. If no further hemolysis occurs,
tive of immune-mediated hemolysis (Fig. 13-4). the serum haptoglobin level recovers, and methemalbumin disap-
As previously mentioned, both IgG and IgM antibodies pears within several days. The urinary hemosiderin can provide
are capable of activating complement. However, the activation more lasting evidence of the hemolytic event.
256 Chapter 13 Hemolytic Anemias: Extracorpuscular Defects
Antigen
Number and density
Cellular distribution
Presence of soluble antigen
Figure 13-4 = Autoimmune hemolytic anemia (peripheral blood). Complement
Note (A) spherocytes and (B) polychromasia. Concentration of complement factors
Concentration and activity of regulating factors
the hemoconcentration of blood in the spleen does not con- Mononuclear Phagocytic System
Activity of phagocytic cells (influenced by underlying
tribute significantly to the destruction of complement-coated
disease processes, generation and activity of lymphokines
red cells.!° However, cells coated with both IgG and and interleukins, and any concurrent drug therapy)
C3b/iC3b are phagocytized more efficiently in the spleen than
if they are coated by IgG alone.'®
The liver has the largest concentration of macrophages
with receptors specific for immune complexes; thus, the liver 1. Alloimmune: The patient produces alloantibodies to for-
is the major site of removal for red cells coated with comple- eign red cell antigens introduced through transfusions,
ment or heavily coated with IgG.** There is very little pregnancy, or organ transplantation.
removal of red cells coated with small amounts of IgG in the 2. Autoimmune: The control mechanism preventing autoreac-
liver, because of the high concentration of free IgG located tive antibodies is lost and antibodies directed against the
there. Cells sensitized with both IgG and iC3b/C3d are patient’s own red cells develop.
removed in the liver and spleen. 3. Drug-induced: The patient produces antibodies directed at
a particular drug, its metabolites, or red cells coated with
Laboratory Findings the drug. These antibodies then destroy red cells.
Spherocytes are commonly seen on the peripheral blood smear
when extravascular immune hemolysis has occurred. Serum
ALLOIMMUNE HEMOLYTIC ANEMIA
bilirubin (primarily indirect) and LD are usually elevated, and
Alloimmunization is the process in which the immune system
urobilinogen may be increased in urine and stool specimens.
of an individual is stimulated by a foreign antigen and pro-
The DAT is usually positive. The DAT (also called the antihu-
duces the corresponding antibody. The antibody produced by
man globulin test, AHG, and previously called the direct
this immune response is termed an alloantibody. The anti-
Coombs’ test) detects antibody and/or complement coating
body coats the foreign red cells introduced into the circula-
patient red cells. In the DAT, patient red cells are washed to
tion, resulting in shortened red cell survival. Alloantibody
remove adherent proteins, and then reacted with reagent con-
production may result from:
taining high-titer, polyspecific antibodies against both IgG (all
subclasses) and complement, and examined for resulting red |. Transfusion of blood (antibodies are produced against for-
cell agglutination; this is called the polyspecific DAT. If this test eign donor red cell antigens)
is positive, testing is repeated with monospecific reagents that 2. Pregnancy (antibodies are produced against “foreign” anti-
recognize IgG and complement (C3d or C3b+C3d) separately, gens on fetal cells released into the maternal circulation)
to determine which is involved.'' The laboratory findings in 3. Organ transplantation (antibodies are produced against for-
immune hemolysis are summarized in Table 13-2. The factors eign antigens on the transplanted organ or “passenger
that influence immune hemolysis are listed in Table 13-3. cells” that may be released into the recipient).
ACUTE HEMOLYTIC TRANSFUSION REACTIONS Acute posttransfusion sample as early as 48 hours after the transfusion.
hemolytic transfusion reactions (acute HTRs) are character- This type of reaction is termed delayed, because it takes time for
ized by acute intravascular hemolysis and are associated with the patient to produce sufficient antibody to destroy the trans-
the ABO blood group antibodies. The immunoglobulins asso- fused cells. Characteristically the reaction may occur anywhere
ciated with the ABO blood group are IgM or both IgM and from 2 to 10 days after transfusion.'*
IgG. In individuals who have both types (IgM and IgG), the The symptoms of delayed HTRs are usually mild and
majority of antibody is IgM with a minor amount of IgG." nonspecific; therefore, delayed HTRs may not be recognized
Ordinarily, an individual possesses antibodies directed toward by clinicians.!° Symptoms include mild fever, mild jaundice,
the A and/or B antigens absent from his or her own red cells. and an unexpected fall in hemoglobin (or conversely, lack of
As previously stated, IgM antibodies are efficient activators expected rise in hemoglobin after transfusion). Laboratory
of complement, which results in immediate destruction of the findings are those associated with extravascular hemolysis, but
transfused cells. most cases are subclinical and discovered only serologically
Symptoms of acute HTRs are variable. Typical symp- (through direct antiglobulin test, antibody screening, and com-
toms are fever, shaking, chills, and pain or a burning sensation patibility tests).'°
along the infusion site. Other symptoms include nausea, vom- Treatment is rarely necessary, and investigation focuses
iting, lower back pain, hypotension, and chest pain. See Table on accurately identifying the antibody to ensure that blood for
13-4 for a list of clinical features. An incoherent, unconscious, future transfusions will be negative for the foreign antigen
or anesthetized patient cannot verbalize his or her symptoms that corresponds to the patient’s antibody. The antibodies
and is therefore at greater risk of receiving one or more units most commonly implicated in delayed HTRs are listed in
of incompatible blood undetected. Table 13-5, and the laboratory findings are summarized in
The laboratory findings in acute HTRs are those associ- Table 13-6.
ated with intravascular hemolysis (see Table 13-2). The treat-
ment of acute HTR focuses on prompt termination of the HEMOLYTIC DISEASE OF THE NEWBORN Hemolytic dis-
transfusion and treatment of any signs or symptoms of shock ease of the newborn (HDN) is an immune hemolytic disorder
with supportive measures. Stroma from hemolysed red cells in which fetal or newborn red cells are destroyed by maternal
can clog and damage renal glomeruli; thus, intravenous fluids alloantibodies. In HDN, maternal—fetal blood group incompat-
are administered to maintain renal function. ibility is always present. Maternal IgG antibodies directed
against “foreign” antigens on fetal red cells cross the placenta
DELAYED HEMOLYTIC TRANSFUSION REACTIONS and destroy the red cells in the fetal circulation. Only IgG
Delayed hemolytic transfusion reactions (delayed HTR) are antibodies can cross the placenta; IgM cannot, and does not
associated with antibodies to blood groups other than the ABO cause HDN.
blood group. Antibodies implicated in delayed HTR are usually Pregnant women can become immunized to “foreign”
IgG, which may activate complement, causing sensitization of fetal red cell antigens when fetal blood enters the maternal cir-
the red cells with C3 but seldom leading to intravascular hemol- culation during pregnancy or at delivery (fetal-maternal hemor-
ysis. Delayed HTRs are the most common type of transfusion rhage). The amount of whole blood exchanged is normally only
reactions. They are caused by an anamnestic or secondary | mL. There is some evidence that as little as 0.03 mL of red
immune response to the transfused red cells. This occurs in pre- cells can stimulate an immune response.'*'” Thus, it is possible
viously immunized patients whose alloantibody level (after the to become immunized from miscarriages and abortions. Blood
initial stimulation) has dropped to serologically undetectable transfusions can also stimulate an immune response, as men-
levels. As a result, the initial antibody screening and compatibil- tioned earlier in the discussion of alloimmune hemolytic
ity tests on the patient’s pretransfusion sample are negative.
When the patient is re-exposed to the foreign red cell antigen
(transfused), an anamnestic response is mounted by the recipi- ys Table 13-5 Antibodies Most
ent. The titer rises and the antibody may be detected in the . Commonly Implicated
in Delayed Hemolytic :
_ Transfusion Reactions Cae
Table134 Clinical Features of (DHTRs) :
: - Acute Hemolytic Antibody Blood Group System
-Transfusion Reactions.
Anti-Jk*
Fever Hemoglobinuria Anti-K
Chills Shock Anti-c
Chest pains Generalized bleeding Anti-E
Hypotension Oliguria Anti-Fy*
Nausea Anuria Anti-Jk°
Flushing Back pain Anti-C
Dyspnea Pain at infusion site Anti-e
258 Chapter 13 Hemolytic Anemias: Extracorpuscular Defects
aie ABO Rh
reactivity). In 80% of the cases, the immunoglobulin is IgG is also lackingSo in Rh,,,, null cells.2? On rare occasions, other specifici-
alone or IgG together with IgA, IgM, and/or C3d.*! Comple- ties have also been reported.*'??!
ment proteins act synergistically with immunoglobulins to
cause red cell hemolysis. In fact, the severity of hemolysis is DAT-Negative AIHA
greater in the presence of complement in addition to IgG.*!? Occasionally (in 1% to 3% of patients) the DAT result is repeat-
Although not evaluated as a part of routine diagnostic testing, edly negative in a patient who has clear evidence of hemolysis
the presence of IgA, IgM, or both, may be found in addition with no other apparent cause.**** These patients represent a
to IgG if appropriate antisera are used.*! WAIHA can present small group that is referred to as having DAT-negative AITHA.*°
several difficult problems in serologic testing, which fall into More sensitive techniques for the detection of IgG or C3d, or
two categories: both, on red cells have shown that many of these patients have
increased levels of these immunoproteins on their cells. The
1. The patient’s red cells are strongly coated with autoanti-
routine direct antiglobulin test (DAT) can detect immunoglob-
body, which interferes with phenotyping.
ulin sensitization of as little as 200 molecules of IgG per red
2. Autoantibody present in the serum may mask an underly-
cell.*! More sensitive techniques are capable of detecting as
ing alloantibody.
few as 20 IgG molecules.*° When interpreting DAT results, it is
important to remember that a positive DAT alone is not indica-
Autoantibody Specificity
tive of immune hemolysis, but 1f hemolysis is present or sus-
The autoantibodies produced in WAIHA usually react with all
pected, it could be the result of immune mechanisms. The DAT
cells tested. Serologic studies have suggested that some autoanti-
result can be positive in up to 8% of hospitalized patients who
bodies are directed at Rh blood group antigens because of their
have no signs or symptoms of hemolysis.*°°! In most of these
lack of reactivity with Rh,,,, cells (cells which lack all Rh blood
group antigens).'? However, further analysis of these autoantibod-
ies with apparent Rh specificity has demonstrated that the reactiv-
ity is actually directed at another red cell membrane protein which
(by -
Hemolytic Anemia
Hodgkin’s disease, non-Hodgkin’s lymphoma, multiple
myeloma, and Waldenstrom’s macroglobulinemia
Autoimmune disorders such as systemic lupus erythematosis,
rheumatoid arthritis, scleroderma, and pernicious anemia
Warm AIHA Other neoplastic disorders, including carcinomas of the ovary,
Cold agglutinin syndrome breast, lung, colon, pancreas, thymus, kidney, and uterus
Paroxysmal cold hemoglobinuria Viral infections, including hepatitis B and hepatitis A
Drug-induced Chronic inflammatory disorders including ulcerative colitis
260 Chapter 13. Hemolytic Anemias: Extracorpuscular Defects
patients, the positive result reflects complement sensitization, of these alternative therapies is variable, and they are used
probably secondary to the disease process from which the only in selected cases.“ 25,50,51
patient is suffering.
IgA antibodies have also been reported to cause AIHA COLD AUTOAGGLUTININS
with characteristics of WAIHA or, less commonly, cold agglu- Normal Cold Autoagglutinins
tinin disease.*’*’ These would also result in a negative DAT Cold reacting autoantibodies (autoagglutinins) are present in
result since IgA does not fix complement in vivo.” all normal human sera.*?** The specificity of these cold
autoantibodies includes anti-I, anti-H, and anti-IH. Practically
Laboratory Diagnosis all adults have I and H antigens present on their red cells.
Typically, patients with WAIHA exhibit a moderate to severe Generally, most examples of anti-I, anti-H, and anti-IH have
normocytic anemia with increased reticulocyte count. The no clinical significance, and these autoantibodies are often too
blood smear can display classic signs of extravascular hemoly- weak to be detected via routine serologic testing, owing pri-
sis: polychromasia reflecting reticulocytosis (see Fig. 13-4) marily to their low concentration in the serum and their narrow
and spherocytosis. Occasionally, nucleated red blood cells may thermal range (4°C to 22°C). The characteristics of normal
be seen. On rare occasions, WAIHA is associated with reticulo- cold autoantibodies found in healthy adults with those of
cytopenia (decreased reticulocyte count). Reticulocytopenia pathologic cold autoantibodies are compared in Table 13-11.
associated with intense hemolysis indicates a lack of bone mar- The benign autoagglutinins differ in many ways from the
row response, and is associated with a high mortality rate. pathologic cold autoagglutinins that produce cold agglutinin
Patients also show accumulation of the products of red cell syndrome (or cold AIHA). The fundamental characteristic that
catabolism: hyperbilirubinemia (especially unconjugated), and differentiates benign autoagglutinins from pathologic autoag-
increased LD. glutinins is the thermal amplitude: pathologic cold autoagglu-
tinins may react at or above 30°C.»
Treatment
Therapy in WAIHA is aimed at treating the underlying disease
Pathologic Cold Autoantibodies
if one is present. Measures to support cardiovascular function
Pathologic cold autoantibodies can be divided into:
are important in patients who are severely anemic. Transfusion
is usually avoided if possible, as this may only accelerate the 1. Primary (idiopathic) cold agglutinin disease (primary CAD)
hemolysis instead of ameliorating the anemia. However, trans- 2. Cold agglutinin syndrome secondary to infection (sec-
fusion should be used in life-threatening situations. ondary CAD)
As all donor blood is invariably incompatible, it is general 3. Paroxysmal cold hemoglobinuria (PCH)
practice to use donor blood that is least reactive in the cross-
match and that is antigen negative for any clinically significant Cold Agglutinin Syndrome (CAS)
alloantibodies that may be present in the patient’s serum.*!** Primary CAD Primary cold agglutinin also called cold
Blood is transfused slowly, in small volumes (100 mL), and the hemagglutinin disease or idiopathic cold AIHA, represents
patient observed closely for any adverse reactions.**** Some approximately 16% of the cases of AIHA.*' Primary CAD is
hematologists advocate the use of phenotypically similar blood a chronic condition and occurs predominantly in older indi-
irrespective of its degree of incompatibility in the crossmatch. viduals, with a peak incidence after 50 years of age.*! It is
The rationale for this approach is that patients with autoim- found in all racial groups, affecting both men and women.
mune antibodies may be more likely to produce alloimmune Although the disease is often idiopathic, a careful evaluation
antibodies, which can be masked by the autoantibodies. How- of the patient may reveal the presence of a lymphoprolifera-
ever, one study indicates that the incidence of alloimmuniza- tive disorder,'’ other malignancy, or infection. Because of this
tion or adverse hemolytic transfusion reactions in patients with association, it is prudent to investigate patients for possible
WAIHA is no greater than the incidence found in other multi- malignancy when they present with a pathologic cold autoan-
transfused patient populations.*° tibody and no other obvious cause, such as infection. This is
Corticosteroids are usually the first line of treatment. illustrated by one series of 78 patients with persistent cold
Corticosteroids such as prednisone produce their effect by: agglutinin disease. On investigation, 28 of these patients were
found to have no underlying malignancy, 6 had chronic lym-
1. Reduction of antibody synthesis*’
2. Altered antibody avidity‘ phocytic leukemia, 31 had non-Hodgkin’s lymphoma and
13 had Waldenstrom’s macroglobulinemia.*©
3. Depression of macrophage activity,*® which reduces the
clearance of antibody-coated red cells”! CAD is a hemolytic anemia produced by an autoanti-
body that reacts optimally at 4°C, but has a wide thermal
Splenectomy is usually considered as the next step if corti- amplitude, reacting at temperatures greater than 30°C as
costeroid therapy is ineffective. Splenectomy decreases the well.*!»’°* The antibody is usually an IgM immunoglobulin,
production of antibody and removes the primary site of which quite efficiently activates complement.** Antibody
red cell destruction.*! Immunosuppressive drugs, intravenous specificity in this disorder is almost always anti-I,5*** less
immunoglobulin, antilymphocyte globulin, anti-CD20 commonly anti-i, and rarely anti-Pr.*+
(Rituximab)*” or plasma exchange may be used in patients CAD is rarely severe and is usually seasonal, as the cold
who do not respond to conventional therapy. The success rate winter months often precipitate the signs and symptoms of a
Chapter 13 Hemolytic Anemias: Extracorpuscular Defects 261
Characteristic Pathologic
chronic hemolytic anemia. Acrocyanosis, also called mance of blood counts and preparation of blood smears may
Raynaud’s phenomenon’ (symptoms of cold intolerance, be difficult. The patient’s mean corpuscular volume (MCV)
such as pain and a bluish tinge in the fingertips and toes, from an automated cell counter will be erroneously high and
owing to vasospasm), is frequently the patient’s main com- the red blood cell count erroneously low due to clumping of
plaint, along with a sense of numbness in the extremities red blood cells. This causes unrealistic values in other calcu-
when exposed to the cold. These symptoms occur because the lated parameters, such as hematocrit, mean corpuscular
cold autoantibody agglutinates the individual’s red cells in the hemoglobin (MCH) and mean corpuscular hemoglobin con-
capillaries of the skin, causing local blood stasis.°? During centration (MCHC). The blood sample should be warmed to
cold weather, the temperature of an individual’s skin and 37°C for 30 to 60 minutes and reassayed on the automated cell
exposed extremities can fall to as low as 28°C, activating the counter to obtain accurate values. In severe cases, replace-
cold autoantibody. This activated cold antibody agglutinates ment of plasma in the prewarmed blood specimen with an
red cells and fixes complement as the erythrocytes flow equal volume of warm saline may be necessary to obtain
through the capillaries of the skin. When the erythrocytes accurate CBC results.
return to the body core (where the temperature is 37°C), the The tendency for spontaneous autoagglutination of red
cold agglutinin elutes off the red cells, leaving activated com- cells from these patients dictates that serum samples must be
plement behind. Hemolysis occurs from the completion of the maintained and separated at 37°C to obtain accurate results for
complement cascade or by removal of red cells sensitized the antibody titer and thermal amplitude studies.*? Similarly,
with C3b/iC3b by macrophages in the liver (see the earlier samples for the determination of DAT results must be collected
section on Intravascular Hemolysis for a review). If any red into ethylene diaminetetraacetic acid (EDTA) to inhibit any in
cells coated with C3b/iC3b escape destruction in the liver, vitro attachment of complement to the cells after collection.
their complement proteins are further degraded to C3d, for A simple serum screening procedure can be performed
which there are no receptors on macrophages.’ The patient’s by testing the ability of the patient’s serum to agglutinate nor-
DAT result will be positive with monospecific anti-C3d mal saline-suspended red cells at 20°C and 4°C. If this test
antiglobulin reagents. result is positive, further steps must be taken to determine the
This hemolytic episode is not associated with fever, titer and thermal amplitude of the cold autoantibody; if nega-
chills, or acute renal insufficiency, as would be characteristic tive, the diagnosis of CAD is unlikely.*”
of patients with paroxysmal cold hemoglobinuria (see later The peripheral blood smear in patients with CAD may
discussion). Hemoglobinemia and, less frequently, hemoglo- show agglutination (clumping of red cells) (Fig. 13-5). The
binuria may be detected after exposure to the cold. Patients clinical criteria for diagnosis of CAD are summarized in
also display weakness, pallor, and weight loss, which are Table 13-12.
characteristic symptoms of chronic anemia. CAS usually
remains quite stable, and when it does progress, it intensifies CAD Secondary to Infections Cold agglutinin syndrome can
gradually. Other clinical features of CAS may include jaun- also occur as a transient disorder that is secondary to infec-
dice and splenomegaly. tions. Episodes of cold autoimmune hemolytic anemia often
occur after upper respiratory infections. Approximately 50%
Laboratory Findings Most patients with CAD present with of patients suffering from pneumonia caused by Mycoplasma
reticulocytosis and a positive DAT result (polyspecific and pneumoniae have elevated titers (greater than 1:64) of cold
C3d). In some cases grossly visible agglutination of anticoag- autoagglutinins.*'°”°* Secondary CAS develops in the second
ulated whole blood samples occurs as the blood cools to room or third week of the patient’s illness, and a rapid onset of
temperature. As a result of this autoagglutination, perfor- hemolysis with symptoms of pallor and jaundice is usually
262 Chapter 13. Hemolytic Anemias: Extracorpuscular Defects
Treatment
Treatment of primary CAD, secondary CAD, and the anemia
Figure 15-5 ® Cold hemagglutinin disease (peripheral blood). associated with infectious mononucleosis is similar. In many
Note the autoagglutination of red cells. cases, the disease is self-limited and requires no treatment.
Patients with persistent disease may be instructed to avoid the
cold, keep warm, or move to a milder climate.® It is interesting
found. Resolution of the episode usually occurs in 2 to
3 weeks, as the hemolysis is self-limited.°* The offending cold to note that one ingenious physician contacted the National
Aeronautics and Space Administration (NASA) for an environ-
autoantibody is an IgM immunoglobulin with characteristic
anti-I specificity. Very high titers of the cold autoagglutinin mentally controlled spacesuit so his patient would not have to
are seen almost exclusively in patients with mycoplasma remain indoors during the acute phase of the disease.®* Corticos-
pneumonia. It has been reported that the cold agglutinin pro- teroids have been used, but have limited success.°! Plasma
duced in this infection is an immunologic response to the exchange has been used in acute cases to provide temporary
mycoplasma antigens, and these antibodies cross-react with removal of antibodies.*°°' Patients with severe disease unre-
the red cell I antigen.®*! sponsive to conventional therapy have been successfully treated
The antibodies produced in both primary CAD and CAD with anti-CD20 (Rituximab).” Splenectomy is ineffective
secondary to mycoplasma pneumonia have anti-I specificity. because extravascular hemolysis resulting from complement
They differ in that the autoantibody in primary CAS is invari- sensitization occurs predominantly in the liver.
ably monoclonal (IgM with kappa [k] light chains only), Transfusion is rarely required. If blood is needed, the
whereas the autoantibody produced secondary to infection is blood should be ABO/Rh compatible and lack any antigens
polyclonal (IgM with both k and lambda [A] light chain for which the patient has an alloantibody. Blood should be
types).° The monoclonality of the autoantibody in primary warmed using a blood warmer and transfused slowly, with
CAS suggests a possible underlying lymphoproliferative constant monitoring of the patient for adverse reactions.”
disorder.
Paroxysmal Cold Hemoglobinuria
Infectious Mononucleosis Paroxysmal cold hemoglobinuria (PCH) is the least common
Infectious mononucleosis may also be associated with a type of AIHA, representing only 1% to 7% of patients.?!
hemolytic anemia resulting from a cold autoagglutinin. Many It occurs most commonly in children, associated with viral
studies have reported an association of anti-i production in disorders such as measles, mumps, chickenpox, infectious
mononucleosis, and the poorly defined ‘flu syndrome.””°
Although PCH is transient and self-limited, severe hemolysis
may occur.
Originally, PCH was described in association with The other aliquot is cooled at 4°C for 30 minutes and then
syphilis, in which an autoantibody was formed in response to incubated at 37°C for another 30 minutes. Both samples are
Treponema pallidum organism, the causative agent of the dis- then centrifuged and observed for hemolysis. A positive test
ease.’' However, with the discovery and use of antibiotics, result is present when hemolysis is seen in the sample placed
PCH is no longer commonly associated with syphilis. at 4°C and then at 37°C, and no hemolysis in the control
Red cell destruction in PCH is the result of a cold-reacting sample. The Donath—Landsteiner test is summarized in
IgG autoantibody (always polyclonal) termed an autohe- Table 13-15.
molysin. This autohemolysin binds to the P antigen on the
patient’s red cells at lower temperatures. Hemolysis occurs Treatment Protection from cold exposure is the only useful
when the red cells are rewarmed on return to the normal body therapy for PCH. For acute postinfection forms of PCH, the
temperature. The autohemolysin fixes complement, and the hemolysis usually terminates spontaneously after resolution of
sensitized cells undergo complement-mediated intravascular the infectious process.” If anemia is severe, transfusions may
hemolysis.” The PCH autoagglutinin attaches only to red cells be required. The same transfusion protocol as in CAD applies.
at cooler temperatures and then activates complement in Characteristics of warm and cold autoimmune hemolytic ane-
warmer temperatures. Thus, the antibody is called a biphasic mias are reviewed and compared in Table 13-16.
hemolysin. It is also called the Donath—Landsieiner antibody,
and its specificity is anti-P.*’ MIXED AUTOIMMUNE HEMOLYTIC ANEMIA In the past
As the name of PCH implies, paroxysmal or intermittent two decades, a number of reports have drawn attention to
episodes of hemoglobinuria occur on exposure to the cold. the occurrence of mixed AIHA, in which patients exhibit
These acute attacks may be characterized by a sudden onset
of fever, shaking chills, malaise, abdominal cramps, and back
pains.”? All the signs of intravascular hemolysis are evident,
including hemoglobinemia, hemoglobinuria, and hyperbiliru-
binemia (see Fig. 13-3). This results in a severe and rapidly Whole Blood Whole Blood
Control Test
progressive anemia. Polychromasia, nucleated red blood
cells, and poikilocytosis are demonstrated in the peripheral Procedure
blood smear. The symptoms and signs may resolve in a few 1. 30 min He
hours or persist for days. Splenomegaly and renal insuffi- 2. 30 min BRE
ciency may also develop. PCH and cold agglutinin syndrome 3. Centrifuge
are compared and contrasted in Table 13-14. and observe
The Donath—Landsteiner test is the classic diagnostic Results
test for PCH, developed by the two physicians for whom it is Positive No hemolysis Hemolysis
named. A blood sample drawn from the patient is split into Negative No hemolysis No hemolysis
two aliquots maintained at different temperatures. One Inconclusive Hemolysis Hemolysis
aliquot, used as the control, is kept at 37°C for 60 minutes.
264 Chapter 13 Hemolytic Anemias: Extracorpuscular Defects
Drug +
X fir Complex
a
Complement
RBC
Membrane
Modification
yt .
Non-specific
Drug r% K F protein
adsorption
Antibody \. tyx =*
ys
Proteins oF. \d
@<@
Figure 13-7 ™ Immune complex mechanism. (From Petz, LD, and Figure 13-8 ® Membrane modification mechanism. (From Petz,
Garratty, G [Eds]: Acquired Immune Hemolytic Anemias. Churchill LD, and Garratty, G [Eds]: Acquired Immune Hemolytic Anemias.
Livingstone, New York, 1980, with permission.) Churchill Livingstone, New York, 1980, with permission.)
266 Chapter 13. Hemolytic Anemias: Extracorpuscular Defects
Immunoglobulin Polyclonal IgG; Polyclonal IgM in infection Polyclonal IgG Polyclonal IgG
characteristics IgM, and IgA may Monoclonal k chain
also be present; rarely IgM in cold agglutinin
IgA alone disease
Complement Variable Always Always Depends on
activation mechanism of
drug, antibody,
and RBC
interaction
Thermal 20°C-37°C; 4°C—32°C optimum 4°C; 4°C-20°C; 20°C-37°C;
reactivity optimum 37°C occasionally to 37°C biphasic optimum 37°C
hemolysin
Titer of free Low (< 1:32) High (> 1:1000 at 4°C) Moderate to Depends on
antibody May only be low (< 1:64) mechanism of
detectable using drug, antibody,
enzyme treated cells and RBC
interaction
Reactivity of eluate Usually panreactive Nonreactive Nonreactive Panreactive with
with antibody Aldomet-type
screening cells antibody.
Nonreactive in
all other
circumstances
Most common Anti-Rh precursor =| Anti-P Anti-e-like;
specificity -common Rh -j Aldomet, antidrug
-LW -Pr
-En*/Wr?
-U
Site of RBC Extravascular: Extravascular: predominantly Intravascular Intravascular and
destruction predominantly liver, rarely intravascular spleen
spleen with some
liver involvement
LIFE CYCLE The malarial parasite has a complex life cycle. activation with increased monocyte/macrophage activity,
The insect vector is the Anopheles mosquito, of which numer- which promotes extravascular hemolysis of both infected and
ous species can transmit the parasite. Figure 13-9 illustrates noninfected red cells in the spleen.*’**
the malarial life cycle. Anemia associated with malaria is normocytic nor-
mochromic. Leukopenia is present in many cases, as is
CLINICAL PRESENTATION The most commonly reported thrombocytopenia (particularly in individuals with P. falci-
symptoms are fever, malaise, headache, chills, and sweats. parum infections). Diagnosis of malaria is made by exami-
Some patients show classic periodic episodes of fever nation of a peripheral blood smear. Blood should be taken
and chills, correlating with rupture of infected erythrocytes. just prior to the onset of fever because the parasitemia is
Nausea, vomiting, and diarrhea may be present. Quite often greatest at this time. However, this is possible only if classic
classic periodic episodes of fever and chills are absent, and periodic episodes of fever and chills are present. It is best
these symptoms are mistakenly attributed to a viral infection, to make blood smears from a fingerstick. If anticoagulated
allowing the malaria to go untreated. It is always advisable to blood must be used, smears should be made as soon as
inquire if the patient has been overseas and which countries possible to prevent changes in erythrocyte and parasite
have been visited. morphology. Examination of an unfixed Giemsa- or
Splenomegaly is present in 40% to 50% of patients with Wright-stained blood smear (thick preparation) is per-
acute malaria. It is present in virtually all patients with formed to ascertain the presence of malarial parasites.
chronic malaria, accounting for the high incidence of Staining is usually performed at a pH of 7.2 to enhance the
splenomegaly in the tropics.*® blue staining of the parasites’ cytoplasm. Determination of
the species of parasite may be made on this smear if sch-
LABORATORY DIAGNOSIS Hemolysis in malaria occurs izonts are present (by the number of merozoites within the
intravascularly as a result of direct red cell destruction by schizont). If only trophozoites are present, the examination
the parasite. In addition, malarial infection causes immune of a thin blood smear is necessary. Often more than one
268 Chapter 13 Hemolytic Anemias: Extracorpuscular Defects
Infections
Intracellular Malaria Physical disruption and immune
Babesiosis Physical disruption
Extracellular Bartonella Direct action on RBC membrane
and MPS sequestration
Clostridium Enzymatic action on RBC membrane
Bacterial sepsis: meningococcal, Physical disruption secondary to DIC
pneumococcal
Viral Unknown
Mechanical
Macroangiopathic Cardiac prosthesis Physical disruption because of shear stress
March hemoglobinuria Physical disruption
Microangiopathic Hemolytic uremic Physical disruption
syndrome (HUS)
Thrombotic thrombocytopenic Physical disruption
purpura (TTP)
Chemical and physical agents
Oxidative agent Dapsone at high dosage Direct oxidation of RBC membrane components
Nonoxidative agents Lead Alteration of RBC membrane components
Venoms Possible direct effect on RBC membrane by enzymes
Osmotic effect Water (drowning or water irrigation Osmotic lysis
during surgery)
Burns Localized dehydration
Acquired membrane disorders Vitamin E deficiency; abetalipo- RBC membrane oxidation; lack of membrane
proteinemia deformability
Liver disease Lipid abnormalities of RBC membrane lead to
decrease in deformability
Renal disease Retained metabolic products cause membrane
changes, leading to a decrease in deformability
Hypersplenism Sequestration of normal cells
parasite is present in a single cell (Fig. 13-10). Plasmodium tickborne in humans, but has also been transmitted by blood
falciparum gametocytes have a characteristic banana or transfusion.?! In the United States, cases are most common in
crescent shape, assisting in the identification. Occasionally New England and the upper Midwest, owing to the presence of
it is possible to see irregular purple inclusions called infected ticks and their hosts in those areas.”* Infection tends to
Maurer’s dots in the red cell cytoplasm, which are probably be self-limited, although in elderly, immune-compromised, or
breakdown products of hemoglobin. Plasmodium vivax asplenic patients it can follow an acute and fatal course.??
gametocytes are round, ameboid forms that expand and Patients usually present with a history of malaise, headache, and
distort the red cell. Bluish purple inclusions called fever, sometimes associated with vomiting and diarrhea. In
Schiiffner’s dots are often seen in red cells infected with P. splenectomized patients this condition can progress to rigors,
vivax and P. ovale (Fig. 13-11). The features of the differ- acute intravascular hemolysis with associated hemoglobinemia,
ent malarial parasites infecting humans are summarized in hemoglobinuria, jaundice, and renal failure.
Table 13-21.
Immunoassays help screen large numbers of patients for LABORATORY DIAGNOSIS Diagnosis of the disease is made
the presence of infection. Flow cytometry has also been used via examination of the peripheral blood, where parasites very
to show the presence of malarial parasites in red cells. similar to P. falciparum are seen in the red cells (Fig. 13-12).
Features that distinguish babesiosis from malaria are the forma-
BABESIOSIS tion of tetrads of merozoites (Maltese cross), absence of pig-
Infection by the organism Babesia represents a zoonotic ment granules in infected erythrocytes, and the presence of
infection, as humans are not natural hosts for the parasite. The extracellular merozoites.”” A history of possible exposure to
disease is carried by ticks (Lxodes scapularis) and normally ticks and a lack of recent travel to areas where malaria is
infects cattle, deer, and rodents.” The disease is usually endemic help in making the correct diagnosis. Serologic tests
Chapter 13 Hemolytic Anemias: Extracorpuscular Defects 269
Rupture of schizont
to release merozoites
into bloodstream
Sporozoites platon GS — =
asexual mutipcaton, Ss —> E=t—
-@-
forming schizonts
a Merozoites enter red blood
Exoerythrocytic cell, forming ring forms
cycle called trophozoites
ra Siam
HUMAN
MOSQUITO Some merozoites
form 9 & O' gametocytes
NG Mature is after several erythrocytic
gametocytes cycles
~——
Sporozoites stored Gametocytes
in salivary glands ingested by
‘ awaiting new Gametocyte union Anopheles
human host forms ookinetes mosquito
which produce (sexual cycle)
thousands of
sporozoites
Figure 15-9 @ Malarial life cycle in humans and mosquitoes. Beginning of cycle is indicated by an asterisk.
Figure 1S: 10 m Ringed forms of Plasmodium falciparum in red Figure 15-11 @ Late stages of Plasmodium vivax malaria.
blood cells (RBCs). Note that the same RBCs may be infected with Schuffner’s dots. Note and contrast the platelet on the RBC (center)
more than one ring. with the ring form of malaria toward the periphery (arrow).
270 Chapter 13. Hemolytic Anemias: Extracorpuscular Defects
for antibodies to Babesia by immunofluorescent assay or testing does not appear to be any intermediate host. The organisms
by polymerase chain reaction (PCR) have been described.”** adhere to the red cell surface and appear as gram-negative rods
in the acute phase of the disease. In the recovery phase, they
assume a coccoid appearance.
Extracellular Infections The disease has two clinical phases. The first is the
BARTONELLOSIS (OROYA FEVER)
hemolytic phase (Oroya fever), which may not occur in all
This disease is restricted to northern areas of South America, patients. When it does occur, there is a rapid onset with marked
including Peru, Ecuador, and Columbia. The name Oroya fever intravascular hemolysis. Red cells are also sequestered in the
derives from the city of Oroya in the Peruvian Andes, where spleen and liver.*° The anemia can be quite severe, and blood
many railroad construction workers were affected by the disease smears show many nucleated red cells and a reticulocytosis.”°
in the late 1800s. It is also referred to as Carrion’s disease,” Antibiotic therapy, including penicillin, streptomycin, and tetra-
named after the medical student who died as a result of a self- cyclines, is effective in treating patients in this stage of the
experiment designed to determine the nature of the infection. infection.*° The second stage of the disease (verruca peruviana)
Bartonellosis has a high fatality rate in nonimmune is nonhematologic and involves the development of verrucous
patients and is caused by the organism Bartonella bacilliformis. nodes (warty tumors) over the patient’s face and extremities.
Infection is transmitted by the sand fly (Phlebotomus), and there
CLOSTRIDIUM PERFRINGENS (WELCHID
This organism is a gram-positive, spore-forming bacillus that
is responsible for the development of gas gangrene. Infections
Ss with this organism are generally located in deep tissues where
anaerobic conditions required for the organism’s survival
exist. The organism is normally present in the environment
and may infect tissues exposed by trauma and surgical proce-
dures. There is a high incidence of the infection in septic
abortions.”’ The organism is responsible for extensive tissue
damage resulting from the release of enzymes and toxins.
Septicemia caused by C. perfringens may produce an acute
intravascular hemolytic process resulting from the release of
an alpha (a) toxin or lecithinase. This process, combined with
phospholipases and possibly proteinases also produced by the
organism, acts on the red cell membrane to cause its destruc-
tion and subsequent lysis of the cell.°* Hemolysis is often
severe, with marked hemoglobinemia and hemoglobinuria.
Figure
Acute renal failure may develop quite rapidly, and the prog-
13-12 ® Comparison of babesiosis (left) and malaria (right).
nosis is generally poor.””'®? Microspherocytes, hemolysed
Chapter 13 Hemolytic Anemias: Extracorpuscular Defects 27]
Mechanical Etiologies
The passage of red cells through the vascular system subjects
the cell to a wide range of environmental conditions. As red
cells travel around the body, shear forces are highly variable
Figure 13-15 @ Peripheral blood showing red cell fragmentation
and are influenced by:
with thrombocytopenia. (A) Polychromasia and (B) nucleated RBCs
1. The surface conditions of the blood vessel from a patient with thrombotic thrombocytopenia purpura (TTP).
2. The size of the vessel lumen
3. The rate at which the cell is moving (Fig. 13-14). The reticulocyte count and serum LD level are
4. The number of other cells present at the same time. Other usually elevated.** Leukocytes are usually normal, and platelets
environmental conditions the cel! is exposed to include are often reduced because of their interaction with the abnor-
changes in pH, electrolytes, and protein concentration. mal surface.'”* Decreased haptoglobin, mild hemoglobinemia,
mild hemosiderinuria,'" and occasionally hemoglobinuria are
When these factors cause mechanical rupture of the cell mem-
present (depending on the amount of red cell destruction).
brane, intravascular hemolysis results, accompanied by the
Treatment of cardiac hemolysis can range from supportive
presence of red cell fragments or schistocytes on peripheral
therapy, which may include iron supplementation and transfu-
blood smear (Fig. [os
sion, to surgically correcting the faulty valve or vessel.!°
CARDIAC PROSTHESIS
MARCH HEMOGLOBINURIA
Historically, hemolytic anemia associated with prosthetic
This form of hemolytic anemia was first described in the late
heart valves was a frequent complication ofcardiac corrective
1800s in a young German soldier who demonstrated frank
surgery. Innovative changes in design and composition of
hemoglobinuria following a field marching exercise.'!°° The
valves has reduced mechanical hemolysis to a rare and minor
anemia has been described in individuals involved in strenu-
complication.'°!
ous and sustained physical activity.'"’ Similar traumatic red
The primary cause of hemolysis is mechanical trauma to
cell destruction has been reported in a practitioner of karate!
red blood cells, resulting from turbulence of flow through the
and a conga drum player.'°”’ The cause of the anemia is com-
prosthesis.'°* The severity of the anemia is highly variable in
plex, involving:
patients with heart valve prostheses. Mild, compensated
hemolysis is common; overt anemia is unusual, and rarely is |. Direct physical disruption of red cells as they flow through
the anemia severe enough to require transfusion.'°! the capillaries of the feet or hands
The peripheral blood smear shows many fragmented 2. [ron loss in sweat
cells (schistocytes), helmet cells, and occasional spherocytes 3. Adaptation to a right-shifted oxygen dissociation curve.!!°
Viruses Protozoa
Hiei
ie
ie
Figure 13-16 m Peripheral blood from a patient with extensive Figure 15-18 @ Acanthocytosis from a patient with abetalipro-
burns. Note the typical (A) microspherocytes and (8) membranous teinemia. (From Hyun, BH, et al: Practical Hematology. A Laboratory
fragments. (From Bell, A: Hematology. In Listen, Look and Learn. Guide with Accompanying Filmstrip. WB Saunders, Philadelphia,
.) 1975, with permission.)
Health and Education Resources, Inc., Bethesda, MD, with permission
274 Chapter 13 Hemolytic Anemias: Extracorpuscular Defects
Case Study 2 |
A 38-year-old man was diagnosed with pneumonia 3 weeks
previously. He returned to the hospital because he was expe-
riencing severe weakness and shortness of breath on slight
exertion. The patient was pale and jaundiced. Laboratory
findings revealed severe normocytic anemia. The peripheral
blood smear demonstrated marked agglutination of erythro-
cytes. The crossmatch and antibody screen tests were both
strongly positive at the room temperature phase (20°C).
On further testing, the patient’s DAT was also strongly pos-
itive, with complement proteins. The Donath—Landsteiner
test was negative. Thermal amplitude studies revealed
immunoglobulin reactivity up to 34°C.
Figure 13-19 @ Renal disease (peripheral blood). Note the presence
of (A) burr cells, (B) thorn cell, (©) blister cell, and (D) schistocyte. QUESTIONS
(From Bell, A: Hematology. In: Listen, Look and Learn. Health and
Education Resources, Inc., Bethesda, MD, with permission.) 1. What is the most likely diagnosis?
2. What other laboratory test values may be affected by the
agglutination of RBCs? What is the remedy?
echinocytes (Fig. 13-19), which have numerous small spines 3. What is the treatment for this patient?
over their entire surface. Other conditions in which
echinocytes are seen include pyruvate kinase deficiency and ANSWERS
bleeding associated with peptic ulcer disease.
1. Cold agglutinin syndrome, secondary to pneumonia
(probably Mycoplasma).
2. Certain complete blood count (CBC) values from an
automated instrument will be erroneous because of the
Case Study 1 agglutination of RBCs at room temperature. The blood
should be warmed to 37°C and rerun through the
A 26-year-old white woman diagnosed with rheumatoid instrument.
arthritis came to the emergency department with complaints 3. This cold agglutinin syndrome would be expected to be
of weakness and shortness of breath. Laboratory findings self limited; meanwhile the patient should avoid the cold.
revealed a severe normocytic anemia. The patient had
received many red cell transfusions over the past 8 years,
but had not received any transfusions in over a year. The
peripheral blood smear demonstrated mild anisocytosis,
spherocytosis, and moderate polychromasia. The cross-
match and antibody screen tests were weakly positive at the
Case Study 3
antiglobulin phase. On further testing, the patient’s direct
A 50-year-old African American man had recently returned
antiglobulin test (DAT) was also weakly positive, in poly-
from a trip to Africa to visit relatives. He complained of
specific and IgG phases. The effort to obtain compatible
headaches, fatigue, and general malaise over the past 2 weeks.
blood for transfusion was complicated because the patient’s
The patient then developed a high fever followed by severe
serum reacted weakly with all cells tested.
chills, which persisted for approximately a day. The fever and
chills subsided, and the patient thought he was getting better
QUESTIONS until last night, when he experienced another bout of fever and
1. What is the most likely diagnosis? chills. Laboratory findings revealed the patient was anemic and
2. Is this hemolysis likely to be intravascular or extravas- slightly leukopenic. The laboratory noted that odd, ring-like
cular? Why? inclusions were seen in approximately 10% of his red cells,
3. What other laboratory tests may be requested to aid in along with rare blue, crescent-shaped forms that appeared to be
the diagnosis and what are the expected results? extracellular. On further testing the DAT was negative. The
crossmatch and antibody screening tests were also negative.
ANSWERS
QUESTIONS
1. Warm autoimmune hemolytic anemia, secondary to
rheumatoid arthritis. 1. What is the most likely diagnosis?
2. Extravascular hemolysis. Immune hemolysis with IgG, 2. What other laboratory tests should be requested to aid in
complement negative, occurs in the spleen. the diagnosis?
3. Indirect bilirubin (increased), LDH (increased), reticulo- 3. What is the vector for this disease? What are the reasons
cyte count (increased). for a recent increased incidence of this disease?
continued
Chapter 13 Hemolytic Anemias: Extracorpuscular Defects 275
ANSWERS
1. Malaria, most likely Plasmodium falciparum.
2. Thick smear preparation, stained with Giemsa or
Wright’s stain; it is best to use fingerstick blood rather
than anticoagulated blood.
3. The vector is the Anopheles mosquito. The incidence of
malaria is increasing because the parasite has developed
resistance to antimalarial drugs, and the mosquito vector
has developed resistance to some insecticides.
Quess ons
1What are the mechanisms of immune hemolysis? sg Ss Which is not a characteristic of warm autoimmune
a. IgG or IgM antibodies that activate the classical com- hemolytic anemia? ;
plement pathway a. Variable anemia
b. Antibody-dependent cellular cytotoxicity (ADCC) b. Reticulocytosis and spherocytosis
mediated by NK cells, monocytes/macrophages, and c. Positive result for Donath—Landsteiner test
granulocytes d. DAT result usually positive for both IgG and C3d
c. Complete or partial phagocytosis of antibody-coated . What are features of cold agglutinin syndrome?
erythrocytes a. Usually an IgM antibody
d. All of the above b. Reticulocytosis and positive DAT
to . Which would best distinguish hemolytic anemia caused c. Tendency for spontaneous autoagglutination of RBC
by immune mechanisms from other hemolytic anemias? samples
a. Presence of spherocytes on peripheral blood film d. All of the above
b. Increased reticulocyte count . What is the principle of the Donath—Landsteiner test?
c. Enlarged spleen a. Antibody binds red cells at 37°C and causes lysis
d. Positive DAT at 4°C.
. Which is true concerning autoimmune hemolytic anemia? b. Antibody binds red cells at 4°C and causes lysis
a. Majority of cases are of the “cold” type at 37°C.
b. Seen in transfusion reactions c. Antibody binds red cells at 4°C or 37°C and causes
c. Is demonstrated in hemolytic disease of the newborn immediate lysis.
d. Antibodies are produced against one’s own erythro- d. Antibody binds red cells at 4°C or 37°C but causes
cyte antigens lysis only at 4°C.
What is the process in which the immune system pro- . What are causes for nonimmune hemolytic anemia?
duces antibodies to foreign red cell antigens introduced a. Infections
into their circulation through transfusion, pregnancy, or b. Mechanical, chemical, and physical agents
organ transplantation? c. Acquired membrane disorders
a. Alloimmune hemolytic anemia d. All of the above
b. Autoimmune hemolytic anemia 10. Which of the following organisms is (are) associated
c. Drug-induced immune hemolytic anemia with hemolytic anemia?
d. None of the above a. Mycoplasma pneumoniae
. What causes hemolytic disease of the newborn (HDN)? b. Clostridium perfringens
a. Maternal IgG antibodies, formed as a result of a previ- c. Babesia microti
ous blood exposure or pregnancy, cross the placenta d. All of the above
and attach to fetal cells. . Which of the following, as measured via an automated
b. Fetal IgG antibodies cross the placenta and attach to hematology instrument, would most likely be affected by
maternal red cells. a cold agglutinin?
c. Maternal IgM antibodies, formed as a result of a pre- a. Hemoglobin
vious blood exposure or pregnancy, cross the placenta b. Hematocrit
and attach to fetal cells. c. Platelet count
d. Fetal IgM antibodies attach to fetal red cells and cross d. Leukocyte count
the placenta to enter the mother’s circulation.
276 Chapter 13 Hemolytic Anemias: Extracorpuscular Defects
Il. Roitt, I, et al: Roitt’s Essential Im- hemolytic disease and its prevention. autoimmune hemolytic anemia 1s an
munology, ed. 11. Blackwell, Boston, In: Garratty, G (Ed): Hemolytic IgM-IgG immune complex disease.
2006. Disease of the Newborn. AABB, Autoimmunity 25:272, 2005.
i). Petz, LD: Evaluation of patients with
Arlington, VA, 1984. 313): Mauro, FR, et al: Autoimmune
. Jakobowicz, R, et al: Immunization of hemolytic anemia in chronic lympho-
immune hemolysis. Immunohematol
Rh negative volunteers by repeated cytic leukemia: Clinical, therapeutic,
20(3):167, 2004.
injections of very small amounts of Rh and prognostic features. Blood
. Jones, DRE: Detection of red cell sen-
positive blood. Vox Sang 23:376, 95:2786, 2000.
sitization by antibody and comple-
LOW . Sachs, UJ, et al: Does a negative direct
ment: Current practice and future per-
. Keith, LG, and Berger, GS: The risk of antiglobulin test exclude warm
spectives. Comp Haematol Int 3(4),
ISOs Rh immunization associated with autoimmune haemolytic anemia?
abortion, spontaneous and induced. A prospective study of 504 cases. Br J
. Garratty, G: Autoimmune hemolytic
In Frigoletto, FD, et al (Eds): Rh Haematol 132:655, 2006.
anemia. In Garratty, G (Ed): Immuno-
Hemolytic Disease: New Strategy for . Garratty, G: Immune hemolytic anemia
biology of Transfusion Medicine. CRC
Eradication. GK Hall, Boston, 1982. associated with negative routine serol-
Press, Boca Raton, FL, 1994.
. Siegel, DL, and Silberstein, LE: Red ogy. Semin Hematol 42:156, 2005.
. Devine, DV: Complement. In Anderson,
blood cell autoantibodies. In Ander- . Mauro, FR, et al: Autoimmune
KC, and Ness, PM (Eds): Scientific
son, KC, and Ness, PM (Eds): Scien- hemolytic anemia in chronic leukemia:
Basis of Transfusion Medicine: Implica-
tific Basis of Transfusion Medicine: Clinical, therapeutic, and prognostic
tions for Clinical Practice, ed. 2. WB
Implications for Clinical Practice, ed. features. Blood 95:2786, 2000.
Saunders, Philadelphia, 2000.
2. WB Saunders, Philadelphia, 2000. . Sokol, RJ, et al: IgA red cell autoanti-
. Pineda, AA, et al: Trends in the inci-
. Dacie, JV, and Worlledge, SM: Auto- bodies and autoimmune hemolysis.
dence of delayed hemolytic and
immune hemolytic anemias. Prog Transfusion 37:175, 1997.
delayed serologic transfusion medi-
Hematol 6:82, 1969. . Reusser, P, et al: Autoimmune
cine. Transfusion 39:1097, 1999,
. Petz, LD, and Garratty, G: Acquired hemolytic anemia associated with
~—. Zupanska, B: Cellular immunoassays
Immune Hemolytic Anemias, ed 2. IgA—Diagnostic and theraputic
and their use for predicting the clinical
Churchill Livingstone, New York, aspects in a case with long term fol-
significance of antibodies. In Garratty,
2004. lowup. Acta Hematol 77:53, 1987.
G (Ed): Immunobiology of Transfu-
. Hoffman, PC: Immune hemolytic . Pereira, A, et al: Anti-Sa Cold agglu-
sion Medicine. CRC Press, Boca
anemia—selected topics. Am Soc tinin of IgA class requiring plasma
Raton, FL, 1994.
Hematol 2006. exchange therapy as early manifesta-
oo. Anderson, DR, and Kelton, JG: Mecha-
23. Gehrs, BC, and Friedburg, RC: Au- tion of multiple myeloma. Ann Hema-
nisms of intravascular and extravascu-
toimmune hemolytic anemia. Am J (WON! (CLOPSNS), WINS).
lar cell destruction. In Nance, SJ (Ed):
Hematol 69:258, 2002. 40. Russell, MW, and Mansa, B:
Immune Destruction of Red Blood
24. Rosse, WF, Hillmen, P and Schreiber, Complement-fixing properties of human
Cells. AABB, Arlington, VA, 1989.
AD: Immune-Mediated Hemolytic IgA antibodies: Alternative pathway
\O. Rozsnyay, X, et al: Distinctive role
Anemia. American Society of Hema- complement activation by plastic bound,
of IgGI and [gG3 isotypes in FcR- tology, 2004. but not specific antigen bound, IgA.
mediated function. Immunology . Domen, RE: An overview of immune Scand J Immunol 30:175, 1989.
66:491, 1989. hemolytic anemias. Cleve Clin J Med 4]. Wikman, A, et al: Characterization of
. Horsewood, P, and Kelton, JG:
65:89, 1998. red cell autoantibodies in consecutive
Macrophage-mediated cell destruction. . Sallah, S, et al: Future development of DAT-positive patients with relation to
In Garratty, G (Ed): Immunobiology of lymphoproliferative disorders in in vivo haemolysis. Ann Hematol
Transfusion Medicine. CRC Press, patients with autoimmune hemolytic 84:150, 2005.
Boca Raton, FL, 1994. anemia. Clin Cancer Res 7:791, . King, KE, and Ness, PM: Treatment of
i — . Beadling WV, and Cooling L: 2001. autoimmune hemolytic anemia. Semin
Immunohematology. In McPherson, . Naithani, R, et al: Autoimmune Hematol 42:137, 2005.
RA, and Pincus, MP (Eds): Henry’s hemolytic anemia in children. 43 . Petz, LD: A physician’s guide to trans-
Clinical Diagnosis and Management Pediatr Hematol Oncol 24:309, 2007. fusion in autoimmune haemolytic
by Laboratory Methods, ed 21. . Young, PP, et al: Autoantibody forma- anaemia. Br J Haematol 124:712,
Saunders Elsevier, Philadelphia, tion after alloimmunization: Are blood 2004.
2006. transfusions a risk factor for alloim- 44, Ness, PM: How do I encourage clini-
. Freedman, J, and Semple, JW: Com- mune hemolytic anemia. Transfusion cians to transfuse mismatched blood to
plement in transfusion medicine. In 44:67, 2004. patients with autoimmune hemolytic
Garratty, G (Ed): Immunobiology of 2S), Sokol, RJ, et al: Post-transplant anemia in urgent situations? Transfu-
Transfusion Medicine. CRC Press, immune mediated hemolysis. Transfu- sion 46:1859, 2006.
Boca Raton, FL, 1994. sion 42:198, 2002. 45. Phillipe, P: Autoimmune hemolytic
13}. Brecher ME (Ed): Technical Manual, 30. Hendrick, AM: Autoimmune anemia: diagnosis and management.
ed 15. AABB, Bethesda, MD, 2005. hemolytic anemia—A high risk disor- Presse Med May 2, 2007.
14. Klein, H, and Anstee, D (Eds): der for thromboembolism. Hematol- 46. Buetens, OW, and Ness, PM: Red
Mollison’s Blood Transfusions in ogy 8:53, 2003. blood cell transfusion in autoimmune
Clinical Medicine, ed 11. Blackwell, al Lee, E, Redman, M, and Win, N: Do hemolytic anemia. Curr Opin Hematol
Oxford, 2006. patients with autoantibodies or clini- 10:429, 2003.
. Talano, JA, et al: Delayed hemolytic cally insignificant alloantibodies AT Petz, LD: A physician’s guide to trans-
transfusion reaction/hyperhemolysis require an indirect antiglobulin test fusion in autoimmune haemolytic
syndrome in children with sickle cell crossmatch? Transfusion 47:1290, anaemia. Br J Haematol 124:712,
disease. Pediatrics 111(6 Pt 1):e661-5, 2007. 2004.
June, 2003.
27 8 Chapter 13. Hemolytic Anemias: Extracorpuscular Defects
48 . Biodi, CS, et al: Use of the ery- 65. Grotto, D, et al: Clinical and laboratory 83. Snow, RW, et al: The global distribution of
throphagocytosis assay for predicting the presentation of EBV positive infectious clinical episodes of Plasmodium
clinical consequences of immune blood mononucleosis in young adults. Epi- falciparum malaria. Nature 434:214, 2005.
cell destruction. Clin Lab 50:265, 2004. demiol Infect 131:683, 2003. 84. Krogstad, DJ: Malaria as a reemerging
49, D’ Arena, G, et al: Rituximab therapy for 66. Rosenfield, RE, et al: Anti-i, a frequent disease. Epidemiol Rev 18:77, 1996.
chronic lymphocytic leukemia- cold agglutinin in infectious mononucle- 85. Kain, KC, and Keystone, JS: Malaria in
associated autoimmune hemolytic ane- osis. Vox Sang 10:631, 1965. travelers. Infect Dis Clin North Am
mia. Am J Hematol 81:598, 2006. oF Horwitz, CA, et al: Cold agglutinins in 12:267, 1998.
. Berentsen, S, et al: Rituximab for primary infectious mononucleosis and heterophile- 86. Buetler, E: Hemolytic anemia due to
chronic cold agglutinin disease: A antibody-negative mononucleosis-like infections with microorganisms. In
prospective study of 37 courses of ther- syndromes. Blood 50:195, 1977. Beutler, E, et al (Eds): Williams Hematol-
apy in 27 patients. Blood 103:2925, 2004. 68. Bell, WR, et al: Cold agglutinin ogy, ed 7. McGraw-Hill, New York, 2005.
. D’ Arena, G, et al: Rituximab for warm- hemolytic anemia: Management with an 87. Lamikanra, AA, et al: Malarial anemia:
type idiopathic autoimmune hemolytic environmental suit. Ann Intern Med Of mice and men. Blood 110:18, 2007.
anemia: A retrospective study of 11 adult 106:243, 1987. 88. Casals-Pascual, C, et al: Suppression of
patients. CEur JHaematol 79:53, 2007. 69. Schéllkopf, C, et al: Rituximab in erythropoiesis in malarial anemia with
oy Beck, ML: The I blood group collection. chronic cold agglutinin disease: A hemozoin in vitro and in vivo. Blood
In Moulds, JM, and Woods, LI (Eds): prospective study of 20 patients. Leuk 108:2569, 2006.
Blood Groups: P, I, Sda and Pr. AABB, Lymphoma 47:253, 2006. 89. Hawkes, M, and Kain, KC: Advances
Arlington, VA, 1992. 70. Anstall, HB, and Blaylock, RC: The in malaria diagnosis. Expert Rev Anti
53: Issitt, PD: Applied Blood Group Serol- P blood group system: Biochemistry, Infect Ther 5(3):485, 2007.
ogy, ed 3. Montgomery Scientific, genetics and clinical significance. In 90. Rodgers, SE, and Mather, TN: Human
Miami, FL, 1985. Moulds, JM, and Woods, LL (Eds): Babesia microti incidence and Ixodes
54. Issitt, PD: Cold reacting autoantibodies Blood Groups: P, I, Sd* and Pr. AABB, scapularis distribution, Rhode Island,
outside the I and P blood groups. In Arlington, VA, 1992. 1998-2004. Emertg Infect Dis 13:633,
Moulds, JM, and Woods, LL (Eds): ae Gertz, MA: Cold hemolytic syndrome. 2007.
Blood Groups: P, I, Sda and Pr. AABB, Am Soc Hematol:19, 2006. 9 pane. Jacoby, GA, et al: Treatment of transfu-
Arlington, VA. 1992. Te: Kumar, ND, et al: Paroxysmal cold sion-transmitted babesiosis by
55) Gertz, MA: Cold hemolytic syndrome. hemoglobinuria in syphilis patients. exchange transfusion. N Engl J Med
Am Soc Hematol 2006. Genitourin Med 69:76, 1993. 303:1098, 1980.
56. Reardon, JE, and Marques, MB: Labora- 2 Gupta, R, et al: A prospective compari- 2 Della-Giustina D, et al: Transfusion-
tory evaluation and transfusion support son of four techniques for diagnosis of acquired babesiosis in a nonendemic
of patients with autoimmune hemolytic paroxysmal nocturnal hemoglobinuria. area. Mil Med 170:295, 2005S.
anemia. Am J Clin Pathol 125(Supp)): Int J Lab Hematol 29:119, 2007. OB: Hutchnigs, CL, et al: New insights into
S71, 2006. 74. Mauro, FR, et al: Autoimmune the altered adhesive and mechanical
ilk Mougey, R: Cold autoimmune hemolytic hemolytic anemia in chronic lympho- properties of red blood cells parasitized
anemia: A review of clinical and labora- cytic leukemia: Clinical, therapeutic, and by Babesia bovis. Mol Microbiol
tory considerations. Immunohematology prognostic features. Blood 95:2786, 65:1092, 2007.
1:1, 1984. 2000. 94. Chisholm, ES, et al: Indirect immuno-
5 oo. Johnson, ST, et al: One center’s experi- S: Young, PP, et al: Autoantibody formation fluorescence test for human Babesia
ence: The serology and drugs associated after alloimmunization: Are blood trans- microti infection: Antigenic specificity
with drug-induced immune hemolytic fusions a risk factor for autoimmune Am J Trop Med Hyg 35:921, 1985.
anemia—a new program. Transfusion hemolytic anemia? Transfusion 44:67, 5). Higgins, R: Emerging or re-emerging
47:238, 2007. 2004. bacterial zoonotic diseases: Bartonel-
oy) Sokol, RJ, et al: Cold haemagglutinin 70. Shulman, JA, et al: Autoimmune hemolytic losis, leptospirosis, Lyme borreliosis,
disease: clinical significance of serum anemia with both cold and warm autoanti- plague. Rev Sci Tech Off Int Epiz
haemolysins. Clin Lab Haematol 22:337, bodies. JAMA 253:1746, 1985. 23:569, 2004.
2000. Vie Wautier, JL, and Rouger, P: Drug- 96. Manfredi, R, et al: Bartonellosis: light
60. Wilson, ML, et al: Mycoplasma pneumo- induced hemolytic anemia. Transfus and shadows in diagnostic and thera-
niae associated with hemolytic anemia, Clin Biol 8:377, 2001. peutic issues. Clin Microbiol Infect
cold agglutinins, and recurrent thrombo- 78. Garratty, G, and Petz, LD: Drug-induced 11:167, 2005.
sis. South Med J 100:215, 2007. immune hemolytic anemia. Am J Med 7. Nadisauskiene, RJ, et al: Puerperal
6 ome. Havouis, S, et al: Transgenic B lympho- Stosskhey, IQ 7Sy. Clostridium perfringens sepsis in a
cytes expressing a human cold agglutinin WS: Garratty, G, et al: The effect of methyl- patient with granulocytopenia. Gynecol
escape tolerance following experimental dopa and procainamide on supressor cell Obstet Invest 65:32, 2007.
infection of mice by Mycoplasma pulmo- activity in relation to red cell autoanti- 98. Simpkins, H, et al: Structural and
nis. Eur JImmunol 32:1147, 2002. body production. Br J Haematol 84:310, compositional changes in the red cell
62. Roeicke, D, et al: Coexisting anti-I/i plus 1OOBy membrane during Clostridium welchii
anti Pr cold agglutinins in individual 80. Ackroyd, JF: The immunological basis infection. Br J Haematol 21:173,
sera. Infus Ther Transfus Med 27:149, of purpura due to drug hypersensitivity. NOTAG
2000. Proc R Soc Med 55:30, 1962. WD). Loran, MJ, et al: Massive hemolysis
3. Leo, A, et al: Increase of post-infection 81. Jamin, D, et al: An explanation for non- associated with Clostridium perfrin-
cold agglutinins after infection with immunologic adsorption of proteins onto gens sepsis. Am J Emerg Med 24:881,
Mycoplasma pneumoniae, varicella- red blood cells. Blood 67:993, 1986. 2006.
zoster and rubella virus. Beitr Infus Ther . Mueller-Eckhardt, C, and Salama, A: 100. Bianco, M, and Ferreirés, C: Massive
Transfus Med 32:197, 1994. Drug-induced immune cytopenias: A haemolysis by Clostridium perfringens:
64. Jenkins, WJ, et al: Infectious mononucle- unifying pathogenic concept with special Quick diagnosis in a patient with severe
osis: An unsuspected source of anti-i. emphasis on the role of drug metabo- sepsis. Ann Biol Clin (Paris) 65:181,
Br JHaematol 11:480, 1965. lites. Trans Med Rev 4:69, 1990. 2007.
Chapter 13 Hemolytic Anemias: Extracorpuscular Defects 279
101. Erslev, AJ: Traumatic cardiac hemolytic itil, Bahloul, M, et al: Thrombotic microan- 120. Fonte, R, et al: Anaemia and abdominal
anemia. In Beutler, E, et al (Eds): giopathies. Incidence, pathogenesis, pain due to occupational lead poison-
Williams Hematology, ed 7. McGraw- diagnosis, treatment and prognosis. ing. Haematologica 92(3 Suppl):
Hill, New York, 2005. J Mal Vasc 32:75, 2007. ECRO7, 2007.
102 . Strémer, H, et al: Management of 2. Davis, A, and Barnes, C: Thrombotic 12 — . Gunay, N, et al: A series of patients in
patients with prosthetic heart valves. thrombocytopenic purpura in childhood: the emergency department diagnosed
Herz 31:455, 2006. An uncommon but life-threatening with copper poisoning: Recognition
103. Harker, LA, and Slichter, SJ: Studies of cause of thrombocytopenia. Paediatr equals treatment. Tohoku J Exp Med
platelet and fibrinogen kinetics in Child Health 43:640, 2007. 209(3):243, 2006.
patients with prosthetic heart valves. INAL3s. George, JN: Evaluation and manage- (22: Arnon, R, et al: Wilson disease in chil-
N Engl J Med 283:1302, 1970. ment of patients with thrombotic throm- dren: Serum aminotransferases and
104. Vesey, JM, and Otto, CM: Complica- bocytopenic purpura. J Intens Care Med urinary copper on triethylene tetramine
tions of prosthetic heart valves. Curr 22:82, 2007. dihydrochloride (trientine) treatment.
Cardiol Rep 6:106, 2004. 114. Martinez, J: Microangiopathic J Pediatr Gastroenterol Nutr 44:596,
105. Mecozzi, G, et al: Intravascular hemol- hemolytic anemia. In Beutler, E, et al 2007.
ysis in patients with new-generation (Eds): Williams Hematology, ed 7. 123. Boulard, M, et al: The effect of copper
prosthetic heart valves: A prospective McGraw-Hill, New York, 2005. on red cell enzyme activities. J Clin
study. J Thorac Cardiovase Surg 115. Hebbel, RP, and Eaton, JW: Pathobiol- Invest 51:459, 1972.
123:550, 2002. ogy of heme interaction with the ery- 124. Anderson, PC: Spider bites in the
106. Fleischer, R: Uber eine neute form von throcyte membrane. Semin Hematol United States. Dermatol Clin 15:307,
hamoglobinurie beim menschen. Berlin 26:136, 1989. DOT,
Klin Wochenschr 18:691, 1881. 116. Mullick, P, et al: Aniline-induced . Gibley, RL, et al: Intravascular hemoly-
107. Gilligan, DR, et al: Psychologic methaemoglobinaemia in a glucose- sis associated with North American
intravascular hemolysis of exercise: 6-phosphate dehydrogenase enzyme crotalid envenomation. J Toxicol Clin
Hemoglobinemia and hemoglobinuria deficient patient. Anaesth Intens Care Toxicol 36:337, 1998.
following cross-country runs. J Clin 35:286, 2007. 126. Abubakar, MS, et al: Purification and
Invest 22:859, 1943. Ih. Lee, JJ, et al: Hemolytic anemia as a activity of two phospholipase enzymes
108. Streeton, JA: Traumatic hemoglobinuria sequela of arsenic intoxication follow- from Naja nigricolis nigricolis
caused by karate exercises. Lancet ing long-term ingestion of traditional Reinhardt venom. J Biochem Mol
POLO. Chinese medicine. Korean Med Sci Toxicol 17:53, 2003.
109. Furie, B, and Penn, AS. Pigmenturia 19:127, 2004. . Byard, RW, et al: Drowning, haemodi-
from conga drumming: Hemoglobinuria 118. Beutler, E: Hemolytic anemia due to lution, haemolysis and staining of the
and myoglobinuria. Ann Intern Med chemical and physical agents. {n Beutler, intima of the aortic root-preliminary
80:727, 1974. E, et al (Eds): Williams Hematology, observations. J Clin Forensic Med
110. Erslev, AJ: March hemoglobinuria and ed 7. McGraw-Hill, New York, 2005. 13:121, 2006.
sports anemia. In Beutler, E, et al (Eds): IIS), Kales, SN, et al: Hematopoietic toxicity
Williams Hematology, ed 7. McGraw- from lead-containing Ayurvedic med-
Hill, New York, 2005. ications. Med Sci Monit 13:295, 2007.
Chapter
Hypoproliferative
Anemia
Anemia Associated with Systemic
Diseases
Carmen J. Julius, MD
Carl R. Schaub, MD
Introduction OBJECTIVES
Anemia of Chronic Disease At the end of this chapter, the learner should be able to:
The Inflammatory Response
and Body Defense l. Describe anemia of chronic disease.
Mechanisms Zz. List the three major types of diseases that cause anemia of chronic disease.
Etiology and
Pathophysiology 3 . Identify laboratory findings characteristic of anemia of chronic disease.
Characteristics 4 . Define the pathophysiology of anemia of chronic disease.
Treatment
>: Describe the treatment of anemia of chronic disease.
Anemia Associated
with Renal Disease 6 . Describe the pathophysiology of anemia associated with renal insufficiency.
and Renal Failure
in. Describe the treatment of anemia of renal insufficiency
Etiology and Pathophysiology
Characteristics 8 . Describe the characteristics of the red blood cells in anemia associated with liver
Treatment disease.
Anemia Associated with . Describe the etiology of anemia associated with alcoholism.
Liver Disease
Etiology and . List the three categories of causes of the anemia associated with malignancy and
some of the mechanisms under each category.
Pathophysiology
Characteristics . Describe the pathophysiology and characteristics of anemia of human immunodefi-
Treatment ciency virus infection and the acquired immunodeficiency syndrome.
Anemia Associated with . List the mechanisms of anemia of infancy and anemia associated with prematurity.
Alcoholism/Alcohol Abuse
Etiology and . List the causes of “anemia” of improperly drawn laboratory samples.
Pathophysiology . Describe the way(s) in which “anemia” of improperly drawn laboratory samples may
Characteristics be detected.
Treatment
Anemia Associated with
Endocrine
Disease/Disorders
Adrenal Insufficiency
Thyroid Disease
Hyperparathyroidism
Hypogonadism
Pituitary Dysfunction
280
Chapter 14 Hypoproliferative Anemia: Anemia Associated with Systemic Diseases 281
immune activation has suggested a variety of other, perhaps modern era, when many infections can be rapidly and effec-
more descriptive names ranging from “anemia of inflamma- tively treated with antibiotics, it is the chronic, the unsus-
tion” to “cytokine-mediated anemia,” but ACD seems to have pected, or the antibiotic-resistant infection that will more
persisted as the accepted terminology, and will be used here. commonly cause ACD. All of the chronic inflammatory disor-
ACD is defined by a modest normocytic or sometimes ders (also called autoimmune, collagen vascular, connective
microcytic anemia that presents with a decreased serum iron tissue or rheumatologic diseases) have the ability to produce
concentration despite ample reticuloendothelial iron stores ACD because they are, by nature, inflammatory diseases.
(measured by serum ferritin or bone marrow iron stains). Anemia is the most common hematologic abnormality seen in
Thus, the diagnosis of ACD should be made only when these patients with rheumatoid arthritis, systemic lupus erythemato-
findings are present, and the use of ACD as a wastebasket sus, mixed connective tissue disease, scleroderma, dermato-
term should be resisted. Anemias associated with other some- myositis, and Sjégren’s syndrome.® Chronic rejection after
times chronic diseases such as renal failure, liver disease, or solid organ transplantation is also commonly accompanied by
endocrine diseases are not included in ACD, and are dis- ACD.>° Recently, an anemia in acutely ill patients in the inten-
cussed separately below. sive care setting has been described which otherwise has all
the characteristics of ACD®; surprisingly, not all patients with
ACD need have a truly chronic disease (Table 14-4).
The Inflammatory Response and Body In some patients the diagnosis of ACD may provide an
Defense Mechanisms initial clue leading to the detection of a previously undiscov-
ered infection, chronic inflammatory disease, or occult malig-
THE GENERAL INFLAMMATORY RESPONSE (FIRST
nancy. This justifies the use of the CBC in health screening
LINE OF DEFENSE)
and in long-term serial evaluation of patients.
Inflammation is one of the body’s responses to tissue injury
ACD is due primarily to a decrease in red blood cell
from physical agents, foreign organisms, and immune reactions
(RBC) production by the bone marrow, with secondary con-
in the host. Inflammatory and hemostatic responses occur
tribution by decreased RBC survival. The pathophysiology of
simultaneously to control any damage at the injured area (see
ACD is multifactorial and complex, but is ultimately driven
Chaps. 26 and 27). The coagulation cascade and the comple-
by the immune response. Cytokines produced by inflamma-
ment, fibrinolytic, and kinin systems interact to modulate
tory cells produce:
inflammation (see Chaps. 24, 26, and 27).
During the inflammatory process, complement can be 1. Dysregulation of iron homeostasis
activated by the antibody-induced classical pathway, or 2. Impaired proliferation of erythroid precursors
directly by microorganisms via the alternative pathway. 3. Decreased erythropoietin response to anemia
The presence of C3a, C5a, and other chemotaxins attracts 4. Shortened RBC lifespan’
phagocytes to the site of injury, where they recognize and
Each of these mechanisms contributes to ACD (Table 14-5).
phagocytose foreign substances or organisms. Neutrophils,
monocytes, and macrophages possess receptors for comple-
DYSREGULATION OF IRON HOMEOSTASIS
ment that can induce both exocytosis of granules (containing
Dysregulation of iron homeostasis, also called reticuloen-
proteolytic enzymes, free ion radicals, and other inflamma-
dothelial iron block, refers to the inability to use iron properly.
tory metabolites) and endocytosis of complement-coated
It is primary in the pathogenesis of ACD.*° Pro-inflammatory
foreign substances.
cytokines such as interleukin-1 (IL-1) and tissue necrosis
Inflammation will continue as long as injury and damage
factor-alpha (TNF-a) divert iron from the circulation into stor-
continue. When the source of inflammation is persistent and
age sites in the reticuloendothelial system.'° This effect of
the condition is chronic, mediators from the humoral and cell-
reducing the availability of iron for hematopoiesis, despite
mediated immune responses contribute to the onset of anemia
otherwise ample iron stores in reticuloendothelial storage
(Fig. 14-1). The functions of the various types of T-cell lym-
sites, produces a functional iron-deficiency state. A decrease in
phocytes in cell-mediated immunity are listed in Table 14-1.
intestinal iron absorption and impaired iron reutilization by the
The cytokines that contribute to inflammation are listed in
hepatocytes’! also occurs in ACD patients.
Table 14—2 and the functions of macrophages are summarized
The dysregulation of iron homeostasis in ACD is prob-
in Table 14-3.
ably mediated by hepcidin, a recently described iron regulat-
ing protein and acute phase reactant.'? It acts to decrease iron
Etiology and Pathophysiology absorption from the small intestine and block iron release
from macrophages.'*'* In animal models, inflammation,
Anemia of chronic disease occurs in patients with chronic (or probably acting through production of interleukin-6 (IL-6),
occasionally acute) immune activation. This includes chronic increases the release of hepcidin, which causes decreased
infections (bacterial, fungal, parasitic, and mycobacterial) serum iron.'* Because iron is important for the growth of
such as chronic osteomyelitis or tuberculosis; chronic inflam- many bacteria, this probably acts as a defense against infec-
matory diseases such as rheumatoid arthritis, systemic lupus tion. Hepcidin plays a central role in the development of
erythematosus, vasculitis, sarcoidosis, or inflammatory bowel ACD by decreasing the availability of iron for hemoglobin
disease; and malignant tumors, particularly carcinomas. In the synthesis.
Chapter 14 Hypoproliferative Anemia: Anemia Associated with Systemic Diseases WN 83
Ag presentation
to T cell :
MHC
IL-2 ~~
yas,
S
! |
B Humoral Immunity | C Cellular Immunity |
|
| |
| Lymphokines T-suppressor/cytotoxic |
| (CD8) |
| |
| CSF T cell. >|
| { MAF == oy |
| ieogtc De) | AgClass| Altered |
| 3 | BFU-E Nite
AD
self-cell ||
| | |Suppression
| | and Md ; |
, GM-CFU : :
production ee
| increase |
| Activated macrophage |
| | y * phagocytosis |
| | Leukocyte ¢ monokine |
| | production production |
| y |
|
| | IL-1 |
! |
Lysozyme and |
| lactoferrin |
| | release |
ais oe ae Le AL ene ER Ea, a a
Figure 14-1 @ Mechanism of humoral and cellular immunity. A. The antigen phagocytized by the APC is digested, and small antigenic frag-
ments or epitopes are associated with the class Il MHC and presented to aT cell with a receptor specific for the antigen. Formation of the
antigen-receptor complex between the two cells and IL-1 secreted by the APC provide the signals for the T cell to be activated and secrete
IL-2 for its autostimulation and proliferation to effector T-helper cells. 8B. Humoral immunity. The T-helper effector cell (CD4) and some of its
lymphokines provide the necessary signals for the B cell with the same antigen specificity to be activated and proliferate to B memory and
antibody-producing plasma cells. C. Cellular immunity. Some of the lymphokines from the activated CD4 celis and the complex formed
between antigens associated with class | MHC on altered self-cell and T-cytotoxic cell (CD8) receptors cause the activation of the CD8 cells,
which mediate the cytotoxic killing of the altered self-cells. Some of the other lymphokines produced also play significant roles in
hematopoiesis and activation of phagocytic cells. (Ag = antigen; APC = antigen-presenting cell; MHC = major histocompatibility complex;
IL-1 = interleukin-1; IL-2 = interleukin-2; CSF = colony-stimulating factor; BFU-E = burst-forming unit-erythroid; GM-CFU = granulocyte/
macrophage-colony-forming units; PMN = polymorphonuclear neutrophils; MAF = macrophage-activating factor.)
Macrophages in the bone marrow demonstrate SUPPRESSION OF ERYTHROPOIESIS AND THE ROLE
increased iron stores despite the low serum iron. The iron is OF CYTOKINES
trapped within the reticuloendothelial system and is unable The bone marrow is morphologically normal in ACD (except
to be fully utilized in erythropoiesis. Serum ferritin is for increased iron stores), but is unable to increase erythro-
increased reflecting the increased reticuloendothe lial iron poiesis in response to the anemia.*”'’'* Increased cytokines
stores (see Chap. 6). This results in the paradoxical ferroki- including IL-1, TNF-a and interferon gamma (IFN-y) act to
netics typical of ACD: ample iron stores with a functionally suppress erythropoiesis by the bone marrow by increasing
iron-deficient or sideropenic state. This also increases the apoptosis (cell death) in erythroid precursors.’ Sera from
amount of free erythrocyte protoporphyrin (FEP), which is patients with chronic inflammatory disorders (e.g., rheuma-
a porphyrin ring without iron,'® reflecting decreased heme toid arthritis, systemic lupus erythematosus, and juvenile
production. rheumatoid arthritis) inhibit erythropoiesis in vitro.'°?° The
284 Chapter 14 Hypoproliferative Anemia: Anemia Associated with Systemic Diseases
: oo
jHl y.
ae Ge oe
Lipopolysaccharide
emergence of malignant Q
cells, or autoimmune %
dysregulation
|
Immune effector
Hepcidin
mechanisms ©
Stomach
Duodenum
Transferrin-
bound iron
Decreased Fe?+
Increased absorption
ferritin o Transferrin
O32
receptor
Sincreased eS
) © Fe2+02.0
@ ©
/ @
Degradation and Macrophage
phagocytosis of Divalent metal
senescent ef transporter 1
& 6
erythrocytes @ ©0
~ Ferroportin 1
&
S) pk kee
oD :
Decreased
Fe?+
© Inhibit
erythrop
Key
Activates
SSS Secretes/produces
Stimulates
Inhibits
Increases uptake
Decreases
Figure 14-2 @ Pathophysiological mechanisms underlying anemia of chronic disease. In A, the invasion of microorganisms, the emergence of
malignant cells or autoimmune dysregulation leads to activation of T cells (CD3+) and monocytes. These cells induce immune effector mecha-
nisms, thereby producing cytokines such as interferon-y (from T cells) and tumor necrosis factor-a (TNF-«), interleukin-1, interleukin-6 and
interleukin-10 (from monocytes or macrophages). In B, interleukin-6 and lipopolysaccharide stimulate the hepatic expression of the acute-phase
protein hepcidin, which inhibits duodenal absorption of iron. In C, interferon-y, lipopolysaccharide, or both increase the expression of divalent
metal transporter 1 on macrophages and stimulate the uptake of ferrous iron (Fe?*). The anti-inflammatory cytokine interleukin-10 upregulates
transferrin receptor expression and increases transferrin-receptor-mediated uptake of transferrin-bound iron into monocytes. In addition, activated
macrophages phagocytose and degrade senescent erythrocytes for the recycling of iron, a process that is further induced by TNF-a through
damaging of erythrocyte membranes and stimulation of phagocytosis. Interferon-y and lipopolysaccharide down-regulate the expression of the
macrophage iron transporter ferroportin 1, thus inhibiting iron export from macrophages, a process that is also affected by hepcidin. At the same
time, TNF-a, interleukin-1, interleukin-6 and interleukin-10 induce ferritin expression and stimulate the storage and retention of iron within
macrophages. In summary, these mechanisms lead to a decreased iron concentration in the circulation and thus to a limited availability of iron for
erythroid cells. In D, TNF-a and interferon-y inhibit the production of erythropoietin in the kidney. In £&, TNF-a, interferon-y and interleukin-1
directly inhibit the differentiation and proliferation of erythroid progenitor cells. In addition, the limited availability of iron and the decreased bio-
logic activity of erythropoietin lead to inhibition of erythropoiesis and the development of anemia. Plus signs represent stimulation and minus signs
inhibition. (From Weiss G, and Goodnough LT: Anemia of chronic disease. N Engl } Med 352:1013, 2005, with permission. Copyright © 2005
Massachusetts Medical Society. All rights reserved.)
Chapter 14 Hypoproliferative Anemia: Anemia Associated with Systemic Diseases 287
The serum ferritin concentration is usually increased, of choice is correction of the underlying infection, inflamma-
reflecting the block in iron utilization with accumulation of tory disorder, or malignancy. If this is unsuccessful, or if
storage iron. Because serum ferritin is also an acute-phase a more rapid correction of hemoglobin levels is required,
reactant, it may be a poor indicator of iron stores in chronic recombinant human erythropoietin (rHuEpo) therapy or red
inflammatory diseases. Other acute phase reactants will also cell transfusion may also be considered.
be elevated in these patients, including C-reactive protein Use of rHuEpo is sometimes effective in ACD and may
and fibrinogen. Nonspecific measures of inflammation decrease the need for transfusion. rHuEpo seems to partially
including increased “sed rate” (erythrocyte sedimentation counteract the antiproliferative effects of cytokines.*?*°
rate)*®*' and lymphopenia (decrease in the absolute lympho- Patients with decreased serum erythropoietin levels are most
cyte count below 1500 cells/uL) are also commonly seen. likely to benefit from rHuEpo administration.***
Serum erythropoietin level is generally normal in ACD, but Despite the low serum iron levels, there is not a true iron
if decreased, may predict a better response to treatment with deficiency in ACD. Instead, there is a lack of iron availability.
recombinant human erythropoietin (rHuEpo).*?** Treatment of this anemia with iron in the absence of a true
In ACD patients with more severe anemia and microcy- superimposed iron deficiency is ineffective, and in some cases
tosis, making the distinction from iron-deficiency anemia may can actually be harmful. An excess of iron could increase the
become important. This can usually be accomplished by mea- virulence of an infecting organism and exacerbate an under-
suring transferrin or TIBC (low normal to decreased in ACD, lying infection.
increased in iron deficiency) and serum ferritin (increased in Transfusion offers another treatment possibility in
ACD, decreased in iron deficiency) (Table 14-6). Some patients with more severe anemia and no sign of remission in
authors have proposed using soluble transferrin receptor to their disease,'' but only after a proper evaluation of body iron
make this distinction (increased in iron deficiency),*** but stores, possibly including a bone marrow evaluation, has
one study found soluble transferrin receptor no more useful taken place.
than TIBC in distinguishing ACD from iron deficiency.*° Free
erythrocyte protoporphyrin (FEP) is elevated because limited
iron is available to the red cell precursors for incorporation Anemia Associated with Renal Disease
into the porphyrin ring. Thus the RBC cannot complete pro- and Renal Failure
duction of the heme moiety for incorporation into the hemo-
Renai disease is associated with a wide variety of hematologic
globin molecule.
abnormalities. These include anemia, abnormal platelet func-
Difficult cases may require bone marrow examination. In
tion, abnormal white blood cell function,’ and coagulopathy.
ACD, iron stain on the bone marrow aspirate will show normal
The latter usually results from the direct effect of uremia on
to increased iron in marrow macrophages but decreased iron in
platelet and coagulation factor function. Anemia almost invari-
erythroid precursors (sideroblasts).*”** This again reflects the
ably accompanies significant chronic renal insufficiency or renal
reticuloendothelial iron block with decreased availability of
failure.
iron for erythropoiesis.
Indirect Effects
Dilutional anemia
Hypersplenism, erythrocyte sequestration Patients with chronic liver disease or liver failure and
Hemolytic anemia
Spur-cell anemia (acanthocytosis)
end-stage cirrhosis have decreased capacity for the production
RE macrophage activity of coagulation factors by the liver, especially the vitamin
Malnutrition K-dependent factors (see Chaps. 24 and 26). This may be
Protein, folate, iron, and vitamin deficiencies a cause of chronic blood loss or severe bleeding associated
Anemia of chronic disease with hemostatic challenges (such as surgery). Microcytic,
hypochromic anemia may develop as a result of acute or, more
often, occult chronic blood loss similar to the mechanisms
severity of liver disease. Target cells and acanthocytes are seen of iron-deficiency anemia as outlined elsewhere (see Chap. 6).
on the peripheral blood smear in cases of chronic liver disease Patients with chronic liver disease may also have hyper-
and are a consequence of the altered lipid production seen with splenism, a condition found in 15% to 20% of patients with
liver dysfunction (Fig. 14-4 and Chap. 3). Macrocytosis with cirrhosis and advanced liver disease.°’ This condition is a
the formation of target cells is seen on the peripheral blood cause of platelet sequestration leading ultimately to thrombo-
smear in cases of chronic liver disease with a level of obstruc- cytopenia and associated chronic bleeding tendencies,
tive jaundice or hepatocellular cholestasis affecting lipid especially gastrointestinal and mucosal surface bleeding.
metabolism. Target cells are due to increased membrane lipid Hypersplenism can also lead to decreased red cell survival as
(cholesterol and phospholipids) content while acanthocytes— RBCs are trapped in this major reticuloendothelial system
usually seen in cases of severe, chronic, liver disesase®™— organ.
are due to excess membrane cholesterol (Table 14-9).
Hemolytic anemia may occur because of the acanthocytes
Characteristics
(“spur cells”) due to the RBC membrane’s rigidity or lack of
permeability®° decreasing the deformability of the cell as it Overall, anemia associated with chronic liver disease alone
passes through the spleen. (without complications) is mild to moderate and normo-
cytic, normochromic. It is hypoproliferative, as demon-
strated by absent reticulocytosis, and may show variable
RBC morphology including moderate numbers of round
macrocytes. Increasing macrocytes may even cause the
MCV to be modestly elevated.
The anemia may be complicated by hemolysis or
decreased red blood cell survival due to destruction of acan-
thocytes,® in which case reticulocytosis may be present, or by
the simultaneous effects of iron deficiency (from blood loss)
or folate deficiency (predominantly from poor dietary habits).
Serum ferritin is increased in chronic liver disease reflecting
a chronic inflammatory response. Decreased plasma iron may
be due to the same mechanism seen in ACD or may be due to
decreased transferrin synthesis related to liver cell synthetic
x dysfunction.
Macrocytosis can be seen in patients with cirrhosis,
obstructive jaundice, and other types of liver disease, often
without accompanying anemia.® These macrocytes are
Figure 14-4 m Peripheral blood smear from an individual with
round, slightly flattened RBCs with a slight increase in size
severe liver disease. Target cells (arrows) are caused by altered lipid
metabolism with subsequent effects on red blood cell membrane. and volume. The MCV in liver disease is usually between
290 Chapter 14. Hypoproliferative Anemia: Anemia Associated with Systemic Diseases
100 and 110 fL. These macrocytes are due to abnormal mem- precursors in the bone marrow is observed in alcoholic
brane cholesterol and phospholipid content. They are not, patients.°7' >> This toxic change is often associated with
however, the macro-ovalocytes seen in megaloblastic anemia abnormal hemoglobinization of the red blood cell cytoplasm.
(see Fig. 7-4).°° Bone marrow examination shows macronor- The mitochondria in red blood cell precursors—altered by
moblastic RBC precursors but no megaloblastic changes in alcohol’s effects—develop abnormal heme synthesis with sec-
early red blood cell precursors or the myeloid series cell ondary accumulation of iron. Surrounding the nucleus, these
changes such as giant “band” forms, or hypersegmented neu- abnormal iron-encrusted mitochondria form ringed sideroblasts
trophils that are typical of a true megaloblastic anemia.*®* (demonstrated upon iron staining of bone marrow aspirates or
The survival of these macrocytic red cells is not decreased. biopsies) in as many as one-fourth of hospitalized alcoholic
This type of macrocytosis does not respond to vitamin B,, or patients.” This phenomenon, also seen in chloramphenicol
folate therapy but usually resolves only after improvement, if (drug) toxicity, results in red blood cell membrane damage.”
any, in liver disease and function. Chloramphenicol (an older, discontinued antibiotic), which has
Oval megaloblastic macrocytes caused by folate defi- severe bone marrow effects associated with RBC precursor
ciency are another cause of macrocytosis, may also be seen in maturation arrest,’””* shares similar features with heavy and
some patients with liver disease, and are caused by poor nutri- prolonged alcohol abuse which too may result in overall,
tion, usually as a result of chronic alcohol intake.°* Alcoholism decreased bone marrow cellularity.’*’? Alcohol and its metabo-
is acommon cause of end-stage liver disease (see below). lites then are suppressants of hematopoietic-progenitor cell
Reticulocytes are also a cause of macrocytes in liver dis- production”’’”*”**° but fall short of causing a complete matura-
ease. These macrocytic cells are younger red blood cells that tion arrest or bone marrow aplasia. Thus, alcohol use leads to a
demonstrate polychromasia as seen on the peripheral blood hypoproliferative anemia.
smear. They are associated with blood loss or complications Macrocytosis is very common in chronic alcoholics and
seen in liver disease (see above). Therefore, macrocytes in may not show megaloblastic features.°**°*' Even in alcoholics
liver disease have different morphologic features depending who are well nourished or who have only mild liver disease,
on their cause. macrocytosis may persist unaccompanied by any megaloblastic
The anemia of chronic liver failure is often complicated changes.**® The mechanism of macrocytosis due to alcoholism
by portal hypertension, hypersplenism, and increased plasma alone is unknown.
volume as is seen with splenomegaly. The RBC mass is Megaloblastic anemia, when seen in alcoholics, may be
“pooled” in the venous system, sequestered in the spleen, or associated with direct effects of alcohol on folate metabolism
diluted in an increased plasma volume. These factors can (e.g., antifolate effect of ethanol, ethanol-induced increased
even yield a falsely decreased hematocrit, which may cause urinary folate excretion, ethanol-induced interruption of nor-
difficulty in evaluating the degree of anemia. mal enterohepatic circulation—usually seen in cirrhosis, or
ethanol-induced intestinal folate malabsorption).°+”? How-
ever, it is usually related to dietary folate deficiency due to
Treatment
malnutrition in these patients.**Anemia of chronic disease
Correction of any dietary deficiencies (lack of iron or folate) is also common in alcoholics as in other patients with chronic
can help the anemia of chronic liver disease. However, reso- liver disease. The element of malnutrition may give rise to
lution of the liver disease itself corrects the macrocytosis, decreased hemoglobin production or synthetic capacity by the
acanthocytosis, and anemia. In most instances, advanced liver liver affected by alcohol use/abuse.
disease and hypersplenism, when present, are irreversible. In With increasing chronic liver damage and disease, the
these cases, dietary supplementation (iron, vitamin B,,, and alcoholic patient is prone to hypersplenism and all of the
folate) can be efficacious in preventing any hematologic same effects of cirrhosis and end-stage liver disease outlined
sequelae; however, bizarre red cell morphology and low- in the preceding text.
grade anemia may persist due to the liver disease alone.
Characteristics
Anemia Associated with Alcoholism/ The anemia associated with alcoholism is usually macrocytic
Alcohol Abuse and hypoproliferative with similar morphologic changes as
described above with other liver disease. The change in cell
One of the most common causes of liver disease is excessive size persists until 1 to 4 months after the patient stops con-
alcohol ingestion (see Table 14-8). A wide array of hemato- suming ethanol.*
logic abnormalities occur as complications of acute and Iron deficiency due to chronic blood loss (e.g., bleeding
chronic alcoholism.°*° from esophageal varices formed as a consequence of cirrho-
sis) and folate deficiency related to poor nutrition may occur
Etiology and Pathophysiology together in severe, chronic liver disease due to alcohol use. In
this case, the MCV may even be normal, but it will be as a
Ethanol has direct toxic effects on red blood cell precursors, result of the average of two disparate populations of RBCs.
bone marrow cellularity, and peripheral red cell morphol- The red blood cell distribution width (RDW) will be greatly
ogy. Vacuolization of erythroblasts and red blood cell increased.
Chapter 14 Hypoproliferative Anemia: Anemia Associated with Systemic Diseases 291
anemia is mild to moderate in degree, normocytic, nor- It also secretes gonadotropins that control the production of
mochromic, and is associated with a decreased or (inappropri- androgens and estrogens as well as adrenocorticotrophins that
ately) normal reticulocyte count; it is not associated with control the production of adrenal cortical hormones. Thus,
neutropenia or thrombocytopenia. Anemic patients tend to pituitary function has far-reaching consequences. To correct
have more severe hyperparathyroidism with higher levels of this anemia, patients usually require the exogenous replace-
parathyroid hormone, serum calcium, and alkaline phos- ment of thyroid hormone, androgens, and/or estrogens, as
phatase. They are also more likely to show bone cysts, subpe- well as the administration of corticosteroids. All glands
riosteal bone resorption, and marrow fibrosis.” affected by the pituitary dysfunction are bypassed by this
The pathogenesis of anemia of hyperparathyroidism is exogenous hormone replacement necessitated because the
unclear. Parathyroid hormone may decrease proliferation of “master gland” is not functional.
erythroid precursors in the bone marrow.°? Bone marrow A summary of endocrine disorders including the
examination in these patients also tends to show more exten- type/morphology of the anemia, the pathogenesis, and the
sive marrow fibrosis in anemic patients, but myelophthisic treatment is outlined in Table 14-10.
changes in the peripheral blood are not seen. ”'
The treatment of this anemia involves the treatment of
the underlying hyperparathyroidism. Patients undergoing Anemia Associated with Malignancy
parathyroidectomy generally show improvement of their ane-
Anemia is very common in patients with both hematologic and
mia, usually to normal.”
nonhematologic malignancies (Fig. 14-5). The type of abnor-
Patients with renal failure frequently develop secondary
mality depends on a multitude of factors, including the type of
hyperparathyroidism. In these patients, it is difficult to sort
cancer, the site or sites in the body involved by the malignancy,
out the contribution of the anemia of hyperparathyroidism
the patient’s treatment with chemo- or radiation therapy, and the
from that of anemia of renal insufficiency (see preceding
extent of involvement of the hematopoietic tissues (i.e., bone
text). However, it has been noted that effective medical treat-
marrow) by the primary malignancy (Table 14-11). Anemia
ment of these patients’ hyperparathyroidism sometimes
may be the presenting symptom of an otherwise occult (uniden-
improves anemia and reduces the requirement for rHuEpo.”*
tified) malignancy. Anemia may be caused by decreased RBC
production, increased red blood cell destruction, or the toxic
Hypogonadism effects of the treatment of the malignancy. The focus in this
chapter and section is on decreased RBC production due to
Androgens are other hormones that have important roles as direct effects of malignancy on the hematopoietic bone marrow.
stimulants of erythroid activity, both physiologically and ther-
apeutically. For instance, the higher normal range for HGB
concentrations in men as compared to women reflects the Etiology and Pathophysiology
effect of androgens. Healthy men have HGB levels up to
DIRECT EFFECTS
1.0 g/dL higher than those of healthy women.
When the bone marrow is infiltrated by a malignant tumor
Androgens seem to promote erythropoiesis in at least
(also called myelophthisis), it can lead to anemia (i.e.,
two ways: (1) They increase the production of erythropoietin
myelophthisic anemia). Leukoerythroblastosis, which refers
by the kidney and (2) they stimulate the marrow in conjunc-
to the presence of both immature white blood cells (WBCs)
tion with erythropoietin. They are occasionally used pharma-
and immature—including nucleated—RBCs in the peripheral
cologically to treat refractory anemias. An example of this is
blood, is frequently seen on the peripheral blood smear along
Oxymetholone.
with teardrop RBCs (Fig. 14-6). The term can refer to bone
Men with hypogonadism as well as boys and elderly men
marrow infiltration by malignant cells or to marrow replace-
have hemoglobin levels similar to those seen in adult females.
ment by other processes (e.g., fibrosis, military tuberculosis,
In these instances, serum androgen (i.e., testosterone) levels are
osteopetrosis, etc.). Although leukoerythroblastosis can also
decreased while follicle-stimulating hormone and luteinizing
be seen in reactive and congenital conditions, it is the result
hormone levels (i.e., pituitary hormones) are increased in an
of myelophthisic processes in approximately two-thirds of
attempt by the pituitary to stimulate the nonfunctioning gonads.
cases.” Indeed, most individuals would reserve the term
In a mature man with gonadal hypofunction, the decreased
leukoerythroblastosis for the myelophthisic variant of this
androgen levels result in a mild normocytic, normochromic ane-
disorder only. In most cases of malignancy involving the bone
mia accompanied by a reticulocytopenia that may be reversed
marrow, some degree of reactive, space-occupying fibrosis
with the administration of exogenous androgens.
accompanies bone marrow infiltration by malignant cells.
Leukoerythroblastosis with “teardrop” RBCs is often a
Pituitary Dysfunction clue to marrow infiltration by the tumor with associated bone
marrow fibrosis (Fig. 14~7) as opposed to a reactive process
A mild, normocytic, normochromic anemia is seen in patients that usually demonstrates leukoerythroblastosis and normal
with hypopituitarism. Reticulocytosis is absent in this RBC morphology (Fig. 14-8). Marrow fibrosis for any
chronic, hypoproliferative anemia. The pituitary gland is reason, including desmoplastic (i.e., characterized by or caus-
responsible for the secretion of thyroid-stimulating hormone. ing the growth of fibrous tissue) reaction associated with
Chapter 14 Hypoproliferative Anemia: Anemia Associated with Systemic Diseases 293
metastatic tumor to the bone marrow, can lead to “‘teardrop”- Blood loss can be another direct effect of malignancies'©°
shaped RBCs and leukoerythroblastosis. Marrow invasion as can chronic or acute disseminated intravascular coagula-
can occur with solid tumors, particularly metastatic small cell tion (DIC) (see Chap. 27). The latter is the result of activation
carcinoma of the lung; breast carcinoma; prostate cancer; gas- of the coagulation system by metastatic carcinoma with high
trointestinal tumors; and, in some instances, lymphoma.’*”” cell “turnover” or death and significant release of tissue
The anemia is due to a “crowding out” of normal bone mar-
row elements by the malignancy. Essentially, the bone mar-
row space loses both its hematopoietic cellularity and its
proper microenvironment, resulting in a hypoproliferative
anemia. Extensive marrow invasion/involvement can lead to
pancytopenia.
Direct Effects
Replacement of marrow by malignant cells
Primary hematologic malignancy
Ineffective erythroid production
Qualitative reduction in erythropoiesis
Metastatic marrow infiltration
Quantitative reduction in erythropoiesis
Replacement of marrow by fibrosis
Acute and chronic blood loss
Indirect Effects
Anemia of malignant disease/anemia of chronic disease
Anemia of associated organ failure (e.g., renal, hepatic)
Malnutrition and vitamin deficiency
Microangiopathic hemolytic anemia
Immune hemolytic anemia
Treatment-Associated Anemia
Immediate
Chemotherapy
Radiation therapy
Late
Secondary myelodysplasia or leukemia
Idiopathic
Depleted marrow reserve
Figure 14-5 m Peripheral blood from a patient with disseminated
Microangiopathic hemolytic anemia
carcinoma. Note presence of (A) schistocytes and (B) helmet cells.
and (for example postmitomycin chemotherapy)
(From Bell, A: Hematology, In: Listen, Look, and Learn, Health
n.)
Education Resources, Inc., Bethesda, MD, with permissio
294 Chapter 14. Hypoproliferative Anemia: Anemia Associated with Systemic Diseases
SO
“Sar
,
INDIRECT EFFECTS
Malignant tumors elicit a chronic inflammatory response that Radiation therapy is also toxic to bone marrow stem
commonly causes anemia of chronic disease. cells when bone marrow is included within the field of irradi-
ation. Although radiation therapy may be focused directly on
TREATMENT/THERAPY-ASSOCIATED EFFECTS a metastatic lesion within bone, hematopoietic bone marrow
Transient pancytopenia is expected in any patient undergoing may be secondarily involved during radiation therapy for
aggressive treatment for hematologic or nonhematologic lesions within other organs.
malignancies. Most combination chemotherapy protocols use The sequence of recovery in bone marrow is quite vari-
one or more of the alkylating agents, which are directly toxic able. Both erythroid and granulocytic regeneration usually
to rapidly dividing cells, including both rapidly proliferating precede megakaryocyte regeneration'®'; however, the half-life
tumor cells and normally dividing hematopoietic cells. When of RBCs is considerably longer than the half-lives of the other
given frequently and in intervals, alkylating agents cause a two cell lines. Therefore, RBC recovery in the peripheral
dramatic decrease in the peripheral blood cell counts. blood often lags behind white blood cell and platelet recovery.
The anemia in this instance is hypoproliferative due to the
immediate toxic damage to hematopoietic precursors by
chemotherapeutic reagents or radiation therapy.
Morphology Pathophysiology
The introduction of highly active antiretroviral therapy same time, a gradual switch to renal production takes place.'**
(HAART) has greatly reduced the anemia of HIV infec- This switch is not complete until about | to 2 months after
tion.'?7° Potential mechanisms include a reduction in oppor- birth.'**
tunistic infections, a lessening of the effects of ACD, and an Therefore, the more responsive kidney cannot and does
overall improvement in nutritional status related to a lessen- not play a part in responding to anemia for some time after
ing of the overall HIV infection.'*? While some have found birth. A contributing factor is the increased rate of clearance
that HAART lessened the effects of AZT on the anemia of of erythropoietin from neonatal plasma as well as_ its
HIV,!* others hold that AZT may still exacerbate the anemia increased volume of distribution in most infants on birth and
of HIV despite its use in HAART.'*° Persistent anemia and early life!*?'*!4? in addition to rapid neonatal growth in the
transfusion dependence despite HAART is now considered a first few weeks of life requiring a concomitant expansion of
poor prognostic indicator.'°°'?! RBC mass'** in the face of a shorter lifespan of the neonatal
Therefore, in HIV infection, decreased production of RBC as compared to the adult RBC.'** This leads to a relative
RBCs can be attributed to several possible mechanisms— decrease in erythropoietin and a poor response of erythropoi-
including therapeutic interventions. These are outlined in etin to anemia. Varying levels of anemia are due to the many
Table 14-13. physiologic factors that are present during the first | to
2 months of life (Table 14-14).
The anemia is usually well tolerated in term infants.
Anemia of Infancy Indeed, it is commonly referred to as the “physiologic” ane-
mia of infancy because it occurs in the absence of any recog-
Etiology and Pathophysiology
nized nutritional deficiency and is characteristic of even the
Infants are born with HGB values of 17.0 g/dL or higher. Dur- healthiest of infants.'°*'*!
ing the first month of life, all infants experience a decrease in
their circulating RBC mass. The concentration of HGB
declines rapidly from this highest (birth) level to the lowest Characteristics
value of any time during the first few years of life.'°* The The decrease in RBC mass leads to a mild, normocytic, nor-
nadir of the HGB level approaches 9.0 g/dL within the first 10 mochromic anemia with reticulocytopenia which corrects
to 12 weeks of life.'°? specifically as the production of erythropoietin increases.
This decline in red blood cell mass is due to many factors. Other cell lines such as platelets and WBCs are not affected.
The lack of permeability of the placenta to maternal system
erythropoietin is one contributing cause,'**'°° yet the most
prominent is the predominance of erythropoietin production Treatment
by the neonatal liver.'*’'*? The neonatal liver is less sensitive
to anemia and tissue hypoxia than is the kidney.'*”'*° At term Vitamin and hematinic supplementation do not alter the
birth, 70% to 75% of the erythropoietin produced in response decrease in bone marrow erythropoiesis.'*! If anemia causes
to anemia continues to be produced by the liver.'** At the symptoms (e.g., clinical evidence of hypoxia), packed RBC
units are the product of choice for transfusion. However, in the
absence of symptoms, no treatment is required for this physio-
logic anemia of infancy as it usually corrects on its own.
Anemia Associated with Prematurity Vitamin and hematinic supplementation is not as impor-
tant during the first 1 to 2 months of life as it is after this time.
Etiology and Pathophysiology Therefore, if a premature neonate is likely to be hospitalized
The anemia associated with prematurity is a subset of the for a long period of time, it is advisable to supplement his diet
anemia of infancy. Premature infants experience the same with iron and certain vitamins, including vitamin E.'°*'*’ This
alterations in the production, plasma distribution, and clear- can prevent an anemia of prematurity from becoming worse
ance of erythropoietin as do term neonates. While some or from persisting as a nutrient deficiency anemia (e.g., iron
have related the premature infant’s blunted erythropoietin deficiency or vitamin B,,/folate deficiency).
response to the erythrocyte’s amount of fetal HGB and the rHuEpo has emerged as a possible treatment for the ane-
Shape of the infant’s oxygen—hemoglobin dissociation mia of prematurity.'**'°°>-!>4 Progenitor cells committed to
curve,'** the decreased response of erythropoietin to oxy- erythroid differentiation are present in infants with anemia of
prematurity, and these maintain normal responsiveness to ery-
gen tension is still the key factor to this anemia.!*+!45
In fact, preterm infants demonstrate earlier and more severe
thropoietin.'° Although some early evidence suggested that
the use of rHuEpo led to a reduction in the requirement for
declines in HGB values as compared to full-term
blood transfusion in the premature infant,'°*'°°'°’ much of
infants.'*°'*” As such, HGB levels may approach nadirs
the current evidence fails to support this finding in the anemia
of 8.0 g/dL in >1.0 kg and 7.0 g/dL in preterm infants
of prematurity.'*?!°*'°° The exact subgroup of premature
weighing less than 1.0 kg.!*”
neonates that is best helped by this treatment is still subject to
Although adequate oxygenation may be maintained in
investigation.'**'*’ Reviews show that rHuEpo can stimulate
symptom-free infants with anemia of prematurity through a
erythropoiesis.'°'°’ The failure to mount an appropriate,
host of adaptive physiologic mechanisms,'** an added factor
endogenous erythropoietin response may explain the HGB
(or complicating factor) in this clinical scenario is blood
levels seen in premature infants in the months following
loss.'** Many premature infants experience respiratory dis-
birth.'* Iron supplementation with rHuEpo use,'*”!°* as is
ease or problems necessitating ventilatory assistance and long
employed in the treatment of the anemia of renal disease/
hospital admissions while others require a long hospital stay
renal failure, is indicated here, as these neonates too may
because of the need for nutrient supplementation by the use of
become iron-deficient after stimulation with pharmacologic
special feeding tubes.or intravenous catheters. Whatever the
doses of erythropoietin.'*?'®'7° Since the spectrum of use of
reason, the more ill the infant, the longer the hospital-based
*Hupo is not clear and has not succeeded in the elimination
care and the more frequent the blood sampling from the pre-
or marked reduction of RBC transfusions in premature
mature infant. Therefore, in addition to all of the factors out-
neonates, the focus has shifted to making transfusion of these
lined for the anemia of infancy, frequent blood drawing for
patients as safe as possible by a host of mechanisms. '4?!7))!7?
laboratory specimens is added as a contributing factor to the
anemia of prematurity (see Table 14-14). It stresses the oth-
erwise physiologic anemia present in these infants and makes
the anemia of prematurity much more severe than the benign “Anemia” Associated with Improperly
“physiologic” event that is the anemia of infancy'*? because Collected Laboratory Samples:
the small or critically ill, premature infant requires special, Preanalytical Anemia
and sometimes intensive, care.
Etiology and Pathogenesis
Characteristics A final “anemia” deserves mention. That is, the anemia associ-
ated with improperly collected laboratory samples. It is not an
The anemia is normocytic and normochromic with reticulocy- anemia at all, but rather a specimen adequacy problem. It is a
topenia. The severity of anemia varies with the degree of laboratory problem—a preanalytical problem but has the poten-
blood loss from laboratory sampling in concert with the level tial to become a true clinical patient management problem.
of contribution of the other causes of anemia in the neonatal Most laboratory specimens are drawn from the venous
patient (see Table 14-14). Other blood constituents such as system of a patient. They are usually drawn from direct
platelets and WBCs are not greatly affected. venipuncture. Certain patients (e.g., hematology/oncology
patients or intensive care unit patients) have central venous
catheters or peripheral intravenous (IV) lines in place because
Treatment
of the nature of their primary disease or their hospital care.
If signs of hypoxemia are present, RBC transfusion is indi- Due to many circumstances, it is often easier to collect a spec-
cated.'*3 Indeed, replacement of blood drawn for laboratory imen from the indwelling catheters in these patients than to
studies is the predominant indication for RBC transfusions access a new vein by venipuncture.
given to neonates.'*° These are small volume RBC transfu- Care must be taken to avoid a dilutional effect on the
sions'33"'5! because the key is the maximum amount of RBCs specimen due to catheter “flush” fluid or concurrent IV fluid
for the minimum transfused volume necessary. RBC transfu- therapy. Blood removed directly from a catheter is not true,
sions prevent any untoward effects of “high-output” conges- whole blood but a mixture of catheter solution (usually an
tive heart failure as a result of the infant’s anemia.'** anticoagulant) and the patient’s whole blood. This “anemia”
298 Chapter 14 Hypoproliferative Anemia: Anemia Associated with Systemic Diseases
Characteristics CBC
Urinalysis
In these cases, a normocytic, normochromic “anemia” will be Urine reagent strip: WBC: 11.8 X 10°/L
seen when the laboratory specimen is analyzed. Usually, other 4+ urine protein RBC: 29 < 1077/5
peripheral blood cells (platelets and WBCs) will also be 1+ hemoglobin Heb: 8.3 g/dL
decreased because the entire blood sample has been diluted. A Urine microscopic: Het: 25%
comparison with previous complete blood count (CBC) values a Ae:| ae oe
(the laboratory delta check) is most helpful here, as a sharp or Moderate leukocytes MCHC: 30%
significant change in the patient’s hemoglobin or hematocrit is Casts: few hyaline RDW: 14.7%
noted most of the time. This reflects the comparison of a previ- and few granular Platelets 315 x
ous, properly drawn sample—or one drawn from the patient : 10°/L :
prior to the placement of an IV line or a central venous Blood Chemistry eae es
ee BUN 113 mg/dL 1+ poikilocytosis
catheter—to one that is improperly drawn and therefore diluted. Cre ee mg/dL. i+ oeois Re
Het: 25% She was given 3 to 4 L of 0.9% saline, which stabilized her
blood pressure enough to allow her to be transferred back to
MCV: 90 fL
surgery. Laboratory tests then revealed:
RDW: 12.5% (normal 11%-14%)
WiBEz6 OP a6 7/15
Erythrocyte sedimentation rate (ESR): 100 mm/h (normal
0—10 mm/h) Hgb: 7.5 g/dL
Rheumatoid factor: Positive Het: 20%
Platelets: 100 < 10°/L
QUESTIONS The evening shift medical technologist noted a significant
1. What is the patient’s diagnosis? change in the patient’s hemoglobin level over the course of
2. What could cause the increase in ESR? 1 to 2 hours in the recovery room by the use of a delta check
3. Why was the patient anemic? (1.e., a significant change in laboratory values). The technol-
ogist called the recovery room to report the now lower value
and to ask about proper specimen collection. The call was
transferred to the operating room, where the patient was
now undergoing an emergency laparotomy. The technolo-
gist reported the value to the physician (surgeon), who
Case Study 3 explained that the patient’s abdomen was now full of blood
(approximately 2 L) because of a damaged artery that was
A 29-year-old woman with cholecystitis (inflammation of not “tied off’ during surgery. The surgeon had just sutured
the gallbladder) was taken to elective surgery, where she the artery and was closing the abdominal wound after evac-
underwent a cholecystectomy. Her preoperative laboratory uating and suctioning the blood from the cavity.
values revealed:
Os estioms
1. Which of the following lists of laboratory findings would c Suppression of erythropoiesis by cytokines
be most characteristic of the anemia of chronic disease? d. All of the above
a. Normocytic, hypochromic anemia; high serum iron . What are the main factors responsible for reducing ery-
level: increased TIBC; decreased ferritin levels. thropoiesis in anemia of chronic disease?
b. Normocytic, normochromic anemia; low serum iron a. Cytokines from macrophages and lymphocytes
level: decreased TIBC; increased ferritin levels. b. Antibodies from B lymphocytes
c. Microcytic, hypochromic anemia; normal serum iron c. Erythropoietin from the kidney
level: decreased TIBC; normal ferritin levels. d. Polyamines associated with renal failure
d. Macrocytic, normochromic anemia; low serum iron
4. What is the treatment for anemia of chronic disease?
level; decreased TIBC; decreased ferritin levels.
a. Blood transfusion
i) Which of the following are causes for the anemia of b. Iron therapy
chronic disease? c. Treatment of the underlying inflammatory process
a. Shortened RBC lifespan d. Human recombinant IL-1
b. Impaired iron metabolism
300 Chapter 14 Hypoproliferative Anemia: Anemia Associated with Systemic Diseases
b. Difect effects of alcohol ibe), Delta check of hemoglobin values for a patient helps with:
QO . Iron deficiency
a. anemia of infancy
d. Vitamin B,, and folate deficiency b. anemia of endocrine disorders
. What are the typical hematologic findings associated c. anemia due to improperly collected laboratory samples
with anemia from endocrine dysfunction? d. anemia due to liver disease/alcoholism
a. Mild normocytic, normochromic anemia
b. Anemia, abnormal platelets, and coagulopathy See answers at the back of this book.
@ The major causes for the anemia of renal insufficiency are dysfunction, adrenal insufficiency, Addison disease, thyroid
(1) decreased production of erythropoietin by damaged disease, hyperparathyroidism, and hypogonadism.
kidneys, (2) suppression of erythropoiesis by the uremic
m Anemia seen in malignancies is dependent on the type of
toxins that accumulate in renal failure, and (3) slightly to
cancer, the site of infiltration, the patient’s therapy, and
moderately decreased RBC lifespan. Other causes of ane-
the extent of bone marrow involvement.
mia may complicate the anemia of renal insufficiency.
# Myelophthisis refers to infiltration of the bone marrow by
m The anemia of renal insufficiency is hypoproliferative
a malignant tumor; the most characteristic peripheral
(normal to decreased reticulocyte count); it is normocytic
blood morphology associated with myelophthisis is
and normochromic, with the presence of burr cells.
leukoerythroblastosis (the presence of immature myeloid
m Chronic liver disease may show RBC morphology that precursors and nucleated RBCs) with teardrop-shaped red
includes macrocytosis, target cells, acanthocytes, and cells.
thrombocytopenia if hypersplenism is present; a micro-
m= Chemotherapy agents result in a transient pancytopenia
cytic, hypochromic anemia is evident in the presence of
resulting from alkylating agents that are directly toxic to
chronic blood loss with iron deficiency.
rapidly dividing cells.
m The bone marrow in chronic liver disease shows nor- m Anemia associated with HIV infection is characterized as
moblastic or macronormoblastic maturation in RBC a normocytic, normochromic, hypoproliferative anemia
precursors. RBCs have decreased survival because of with a decreased reticulocyte count and dysplastic bone
increased membrane lipids and lack of deformability marrow features; 85% of patients with acquired immun-
(e.g., acanthocytes or “spur cell” anemia).
odeficiency syndrome (AIDS) are anemic.
mwAnemia caused by alcoholism is the result of ethanol m@Anemia of infancy is physiologic and due to the switch
toxicity to precursor cells in the bone marrow. The bone from liver to renal erythropoietin production.
marrow shows macrocytosis, vacuolization of erythrob-
lasts, and abnormal hemoglobinization of erythroid series m Anemia associated with prematurity is a subset of the ane-
precursors. The peripheral blood shows macrocytosis and mia of infancy and even though influenced by erythropoi-
may show spur cells. etin production shift from liver to kidney, is also affected
by blood loss anemia due to or associated with the inten-
w A dimorphic blood picture may be seen in anemia of alco- sive care rendered to many of these premature infants.
holism because of iron deficiency caused by chronic
blood loss; the MCV is normal and the RDW increased.
mw Anemia of improperly collected laboratory samples is
often first suspected by a failed “delta” check on a given
m Anemia associated with endocrine disease is usually nor- patient/patient sample. It may be related to sample dilu-
mocytic and normochromic and may be caused by pituitary tion with intravenous fluids.
REFERENCES 8. Means, RT: Advances in the anemia of regulated by anemia, hypoxia and inflam-
chronic disease. Int J Hematol 70:7, 1999. mation. J Clin Invest 110:1037, 2002.
I Cash, JM, and Sears, DA: The anemia of 9). Ludwiczek, S, et al: Cytokine-mediated 16. Cartwright, GE, and Lee, GR: The ane-
chronic disease: Spectrum of associated regulation of iron transport in human mia of chronic disorders. Br JHaematol
diseases in a series of unselected hospi- monocytic cells. Blood 101:4148, 2003. Petal, USA
talized patients. Am J Med 87:638, 1989. 1, Alverez-Hernandez, X, et al: Induction 17. Papadaki, HA, et al: Bone marrow prog-
tw. Schilling, RF: Anemia of chronic dis- of hypoferremia and modulation of enitor cell reserve and function and stro-
ease: A misnomer (editorial). Ann Intern macrophage iron metabolism by tumor mal cell function are defective in
Med 115:572, 1991. necrosis factor. Lab Invest 61:319, 1989. rheumatoid arthritis: evidence for a
bo. Weiss, G: Pathogenesis and treatment of rt — . Damon, LE: Anemias of chronic disease tumor necrosis alpha-mediated effect.
anaemia of chronic disease. Blood Rev in the aged: Diagnosis and treatment. Blood 99:1610, 2002.
16:87, 2002. Geriatrics 47:47, 1992. 18. Papadaki, HA, et al: Anemia of chronic
. Means, RT: Recent developments in the 12. Nemeth, E, et al: Hepcidin, a putative disease in rheumatoid arthritis is associ-
anemia of chronic disease. Curr Hematol mediator of anemia of inflammation, is a ated with increased apoptosis of bone
Rep 2:116, 2003. / type II acute phase protein. Blood marrow erythroid cells: improvement fol-
. Weiss, G, and Goodnough, LT: Anemia 101:2461. 2003.
lowing anti-tumor necrosis factor-alpha
of chronic disease. N Engl J Med 13. Nemeth, E, et al: IL-6 mediates hypofer- antibody therapy. Blood 100:474, 2002.
352: 101 1, 2005. i remia of inflammation by inducing the 19. Liu, H, et al: Suppression of haematopote-
- Corwin, HL, and Krantz, SB: esacre of synthesis of the iron regulatory hormone sis by IgG auto-antibodies from patients
the critically ill: “Acute” anemia of hepcidin. J Clin Invest 113:1271, 2004. with systemic lupus erythematosus (SLE).
chronic disease. Crit Care Med 29:S199, 14. Laftah, AH, et al: Effect of hepcidin on Clin Exp Immunol 100:480, 1995.
2001. intestinal iron absorption in mice. Blood 20. Allen, DA, et al: Inhibition of CFU-E
_ Means, RT, and Krantz, SB: Progress in 103:3940, 2004. colony formation in uremic patients with
understanding the pathogenesis of the 15 . Nicholas, G, et al: The gene encoding the inflammatory disease: Role of IFN-gamma
anemia of chronic disease. Blood iron regulatory protein hepcidin is and TNF-alpha. J Invest Med 47:204, 1999.
80:1639, 1992.
302 Chapter 14. Hypoproliferative Anemia: Anemia Associated with Systemic Diseases
. Wang, CQ, et al: Interferon gamma receptor values in the anemia of rheuma- . Hsu, C: Epidemiology of anemia associ-
exerts its negative regulatory effect pri- toid arthritis. Arthritis Rheum 47:166, ated with chronic renal insufficiency.
marily on the earliest stages of murine 2002. Curr Opinion Nephrol Hypertension
erythroid progenitor cell development. 36. Wians, FH, et al: Discriminating 133792002;
J Cell Physiol 162:134, 1995. between iron deficiency anemia and ane- 54. Kazmi, WH, et al: Anemia: An early
. Denza, H, et al: Association between the mia of chronic disease using traditional complication of chronic renal insuffi-
activation of macrophages, changes of indices of iron status verses transferring ciency. Am J Kidney Dis 38:803, 2001.
iron metabolism and the degree of ane- receptor concentration. Am J Clin Pathol 23): Chandra, M, et al: Serum immunoreac-
mia in patients with malignant disorders. 115:112, 2001. tive erythropoietin levels in patients with
Eur JHaematol 48:244, 1992. Be Ellis, LD, et al: Marrow iron: An evalua- polycystic disease as compared with
ii)o>). Bertero, MT, and Caligaris-Cappio, F: tion of depleted stores in a series of other hemodialysis patients. Nephron
Anemia of chronic disorders in systemic 1.332 needle biopsies. Ann Intern Med 2E EG), XS.
autoimmune diseases. Haematologica 61:44, 1964. 56. Eckardt, KU, et al: Erythropoietin in
se ID), OM 38. Gardner, DL, and Roy, LM: Tissue iron polycystic kidneys. J Clin Invest
24. Maciejewski, JP, et al: Nitric oxide sup- and the reticuloendothelial system in 84:1160, 1989.
pression of human hematopoiesis in rheumatoid arthritis. Ann Rheum Dis ah Kausz, AT, et al: Anemia management in
vivo: Contribution to inhibitory action of KY Asy, WXSII. patients with chronic renal insufficiency.
interferon-gamma and tissue necrosis . Means, RT, and Krantz, SB: Inhibition of Am J Kidney Dis 36(Suppl):S39, 2000.
factor-alpha. J Clin Invest 96:1085, human erythroid colony-forming units 58. Besarab, A, and Levin, A: Defining a
1995. by gamma interferon can be corrected by renal anemia management period. Am J
PIS. Means, RT: Erythropoietin in the treat- recombinant human erythropoietin. Kidney Dis 36(Suppl):S13, 2000.
ment of anemia in chronic infectious, Blood 78:2564, 1991. 3Y) Kausz, AT, et al: Intraperitoneal erythro-
inflammatory, and malignant disease. AQ. Spivak, JL: Iron and the anemia of poietin in children on peritoneal dialysis:
Curr Opin Hematol 2:210, 1995. chronic disease. Oncology (Huntingt) A study of pharmacokinetics and effi-
. Jelkmann, W: Proinflammatory cytokines 16(Suppl 10):25, 2002. cacy. Am J Kid Dis 34:651, 1999.
lowering erythropoietin production. J 4 —. Zachee, P, et al: Hematologic aspects of 60. Santoro, A: Anemia in renal insufficiency.
Interferon Cytokine Res 18:555, 1998. end-stage renal failure. Ann Hematol Rev Clin Exp Hematol (Suppl) 1:12, 2002.
fe Chambers, JK: Epoetin alfa: Focus on 69:33, 1994. 6 font. Hayashi, T, et al: Cardiovascular effect
inflammation and infection. Case study S wo . Macdougall, IC: Role of uremic toxins in of normalizing the hematocrit level dur-
of the anemia patient. ANNA J 25:353, exacerbating anemia in renal failure. ing erythropoietin therapy in predialysis
1998. Kidney Int Suppl 78:S67, 2001. patients with chronic renal failure. Am J
28. Greendyke, RM, et al: Serum levels of . McGonigle, RJ, et al: Erythropoietin Kidney Dis 35:250, 2000.
erythropoietin and selected other deficiency and inhibition of erythro- 62. Muirhead, N: Erythropoietin and renal
cytokines in patients with anemia of poiesis in renal insufficiency. Kidney Int transplantation. Kidney Int (Suppl)
chronic disease. Am J Clin Pathol 25:437, 1984. 69:S86, 1999.
101:338, 1994, 44. Means, RT: Anemias secondary to chronic . Silverberg, DS, et al: Intravenous iron
29. Yucel-Lindberg, T, et al: Signal transduc- disease and systemic disorders. In Greer, for the treatment of predialysis anemia.
tion pathways involved in the synergistic JP, et al (Eds): Wintrobe’s Clinical Hema- Kidney Int (Suppl) 69:S79, 1999,
stimulation of prostaglandin production tology, ed 11. Lippincott, Williams & 64. Girard, DE: Hematologic effects of acute
by interleukin-1 beta and tumor necrosis Wilkins, Philadelphia, 2004, p. 1451. and chronic alcohol abuse. In Bagby, GC
factor alpha in human gingival fibrob- 4S. Zappacosta, AR, et al: Normalization of (Ed): Hematologic Aspects of Systemic
lasts. J Dent Res 78:61, 1999. hematocrit in patients with end-stage renal Disease. Hematol Oncol Clin North Am
. Vreugdenhil, G, et al: Anaemia of disease on continuous ambulatory peri- IES ZIPS LO Sie
chronic disease in rheumatoid arthritis. toneal dialysis. Am JMed 72:53, 1982. 65. Allen, DW, and Manning, N:
Raised serum interleukin-6 (IL-6) levels 46. Wallner, SF, et al: The effect of serum Cholesterol-loading of membranes of
and effects of IL-6 and anti-IL-6 on in from patients with chronic renal failure normal erythrocytes inhibits phospholipid
vivo erythropoiesis. Rheumatol Int on erythroid colony growth in vitro. repair and arachidonoyl-CoA:
10:127, 1990. J Lab Clin Med 92:370, 1978. 1-palmitoyl-sn-glycero-3-phosphocholine
Ww . Maccio, A, et al: Hemoglobin levels 47. Ohno, Y, et al: Inhibitors of erythroid acyl transferase. A model of spur cell
correlate with interleukin-6 levels in colony forming cells (CFU-E and anemia. Blood 87:3489, 1996.
patients with advanced untreated epithe- BFU-E) in sera of azotemic patients with 66. Cooper, RA: Hemolytic syndromes and
lial ovarian cancer: role of inflammation anemia of renal disease. J Lab Clin Med red cell membrane abnormalities in liver
in cancer-related anemia. Blood 106:362, 92:916; 1978: disease. Semin Hematol 17:103, 1980.
2005. . Desforges, JF: Anemia in uremia. Arch 67. O’Reilly, RA: Splenomegaly at a United
Wwi). Ludwig, H, et al: Prediction of response Intern Med 126:808, 1970. States County Hospital: Diagnostic eval-
to erythropoietin treatment in chronic . Loge, JP, et al: Characterization of the uation of 170 patients. Am J Med Sci
anemia of cancer. Blood 84:1056, 1994. anemia associated with chronic renal 312:160, 1996.
. Henry, D, et al: Prediction of response to insufficiency. Am J Med 24:4, 1958. 68. Seppa, K, et al: Blood count and hema-
recombinant human erythropoietin therapy . Kaye, M: The anemia associated with tologic morphology in nonanemia
in cancer patients. Blood 85:1676, 1995. renal disease. J Lab Clin Med 52:83, macrocytosis: Differences between alco-
34. Suominen, P, et al: Single values of 1958. hol abuse and pernicious anemia. Alco-
serum transferring receptor and transfer- Nn —. Eckardt, KU: Pathophysiology of renal hol 10:343, 1993.
ring receptor ferritin index can be used anemia. Clin Nephrol 53(Suppl):S2, 2000. 69. Zima, T: Ethanol metabolism and patho-
to detect true and functional iron defi- 2. Hsu, C, et al: Epidemiology of anemia biochemistry of organ damage—1992. IV.
ciency in rheumatoid arthritis patients associated with chronic renal insuffi- Ethanol in relation to the cardiovascular
with anemia. Arthritis Rheum 43:1016, ciency among adults in the United system. Hematologic, immunologic,
2000. States: results from the third national endocrine disorders and muscle and bone
35 . Fitzsimons, EJ, et al: Erythroblast iron health and nutrition examination survey. damage caused by ethanol. Fetal alcohol
metabolism and serum soluble transferring J Am Soc Nephrol 13:504, 2002. syndrome. Sb Lek 94:303, 1993.
Chapter 14 Hypoproliferative Anemia: Anemia Associated with Systemic Diseases 303
70. Meagher, RC, et al: Suppression of 90. Mallette, LE, et al: Primary hyper- 108. Treacy, M, et al: Peripheral blood and
hematopoietic-progenitor-cell prolifera- parathyroidism: Clinical and biochemi- bone marrow abnormalities in patients
tion by ethanol and acetaldehyde. cal features. Medicine 53:127, 1974. with HIV-related disease. Br JHaema-
N Engl J Med 307:845, 1982. 9 — . Mallette, LE: Anemia in hypercalcemic tol 65:289, 1987.
TAL McCurdy, PR, and Rath, CE: Vacuolated hyperparathyroidism: Renewed interest 109. Scadden, DT, et al: Pathophysiology and
nucleated bone marrow cells in alco- in an old observation. Arch Intern Med management of HIV-associated hemato-
holism. Semin Hematol 17:100, 1980. WIEST, WT logic disorders. Blood 74:1455, 1989.
WX: Chanarin, I: Haemopoiesis and alcohol. Wee Boxer, M, et al: Anemia in primary . Bodey, GP: Community respiratory
Br Med Bull 38:81, 1982. hyperparathyroidism. Arch Intern Med viral infections in the immunocompro-
Wx McCurdy, PR, et al: Abnormal bone- spose, IDI. mised host: Past, present, and future
marrow morphology in acute alcoholism. WB. Potasman, I, et al: The role of sec- directions. Am J Med 102:77, 1997.
N Engl J Med 266:505, 1962. ondary hyperparathyroidism in the ane- . Folks, TM, et al: Infection and replica-
74. Sullivan, LW, and Hebert, V: Suppres- mia of chronic renal failure. Nephron tion of HIV-1 in purified progenitor
sion of hematopoiesis by ethanol. J Clin SIS), NS Fs53). cells of normal human bone marrow.
Invest 43:2048, 1964. 94. Argiles, A, et al: Medical treatment of Science 242:919, 1988.
WS Jarrold, T, et al: Bone marrow-erythroid severe hyperparathyroidism and its 2. Neal, TF, et al: CD34+ progenitor cells
morphology in alcoholic patients. Am J influence on anaemia in end-stage renal from asymptomatic patients are not a
Clin Nutr 20:716, 1967. failure. Nephrol Dial Transplant 9:1809, major reservoir for human immunodefi-
76. Savage, D, et al: Anemia in alcoholics. 1994. ciency virus-1. Blood 86:1749, 1995.
Medicine (Baltimore) 65:322, 1986. 9 Nn. Weick, JK, et al: Leukoerythroblastosis: . Molina, JM, et al: Lack of evidence for
Vike Rosenbach, LM, et al: Chloramphenicol Diagnostic and prognostic significance. infection of or effect on growth of
toxicity: Reversible vacuolization of ery- Mayo Clin Proc 49:110, 1974. hematopoietic progenitor cells after in
throid cells. N Engl J Med 263:724, 1960. 96. Shamdas, GJ, et al: Leukoerythroblastic Vivo or in vitro exposure to human
78. McCurdy, PR: Chloramphenicol bone anemia in metastatic prostate cancer. immunodeficiency virus. Blood
marrow toxicity. JAMA 176:106, 1961. Clinical and prognostic significance in 76:2476, 1990.
12: Lieber, CS: Metabolism and metaboiic patients with hormone-refractory dis- 114. Shen, H, et al: Intrinsic human immun-
effects of alcohol. Semin Hematol 17:85, ease. Cancer 71:3594, 1993. odeficiency virus type | resistance of
1980. OF. Wong, KF, et al: Solid tumor with ini- hematopoietic stem cells despite core-
80. Seppa, K, et al: Evaluation of macrocy- tial presentation in the bone marrow— ceptor expression. J Virol 73:728, 1999.
tosis by general practitioners. J Stud A clinicopathologic study of 25 adult 5. Chelucci, C, et al: In vitro human
Alcohol 57:97, 1996. cases. Hematol Oncol 11:35, 1993. immunodeficiency virus-1 infection of
8 — . Seppa, K, et al: Macrocytosis as a conse- 98. Lebtahi, N, et al: Evaluating bone mar- purified hematopoietic progenitors in
quence of alcohol abuse among patients in row metastasis of neuroblastoma with single-cell culture. Blood 85:1181, 1995.
general practice. Alcoholism 15:871, 1991. iodine-123-MIBG scintigraphy and NGS Sato, T, et al: HIV infection of
82. Wu, A, et al: Folate deficiency in the MRI. J Nucl Med 38:1389, 1997. megakaryocytic cell lines. Leuk Lym-
alconholic—its relationship to clinical and 99. Broll, R, et al: Tumor cell dissemina- phoma 36:397, 2000.
haematological abnormalities, liver dis- tion in bone marrow and peritoneal cay- WEY, Sutor, GC, et al: Thrombotic microan-
ease and folate stores. Br J Haematol ity. An immunocytochemical study of giopathies and HIV infection: Report of
29:469, 1975. patients with stomach or colorectal two typical cases, features of HUS and
83. Hines, JD: Reversible megaloblastic and carcinoina. Langenbecks Arch Chir TTP, and review of the literature. Infec-
sideroblastic marrow abnormalities in alco- 381:51, 1996. tron 27a O99)
holic patients. Br JHaematol 16:87, 1969. 100. Ou, JH: Transitional cell carcinoma in 118. Harriman, GR, et al: Vitamin B,, mal-
84. Eichner, ER, and Hillman, RS: The evo- dialysis patients. Eur Urol 37:90, 2000. absorption in patients with acquired
lution of anemia in alcoholic patients. 101. Foucar, K: Effects of therapy, transplan- immunodeficiency syndrome. Arch
Am J Med 50:218, 1971. tation and detection of minimal residual Intern Med 149:2039, 1989.
85. Lindenbaum, J: Hematologic complica- disease. In Foucar, K (Ed): Bone Marrow iI), Burkes, RL, et al: Low serum cobal-
tions of alcohol abuse. Semin Liver Dis Pathology. American Society of Clinical amin levels occur frequently in the
7:169, 1987. Pathologists Press, Chicago, 1995. acquired immune deficiency syndrome
86. Maitra, A, and Abbas, AK: The 102. Harris, CE, et al: Peripheral blood and and related disorders. Eur JHaematol
endocrine system. In Kumar, V, bone marrow findings in patients with 38:141, 1987.
Abbas, AK and Fausto, N (Eds): acquired immune deficiency syndrome. 120. Revell, P, et al: Folic acid absorption in
Robbins Pathologic Basis of Disease, Pathology 22:206, 1990. patients infected with human immunod-
ed 7. Elsevier-Saunders, Philadelphia, 103. Cosby, C, et al: Hematological compli- eficiency virus. J Intern Med 230:227,
2005, pp. 1155-1226. cations and quality of life in hospital- 1991.
87. Orwoll, ES, and Orwoll, RL: Hemato- ized AIDS patients. AIDS Patient Care . Richman, DD, et al: The toxicity of azi-
logic abnormalities in patients with STDS 14:269, 2000. dothymidine (AZT) in the treatment of
endocrine and metabolic disorders. In 104. Spivak, JL, et al: Hematologic abnormal- patients with AIDS and AIDS-related
Bagby, GC (Ed): Hematologic Aspects ities in the acquired immune deficiency complex. N Engl J Med 317:192, 1987.
of Systemic Disease. Hematol Oncol syndrome. Am J Med 77:224, 1984. . Krown, SE, et al: Phase II, randomized,
Clin North Am 1:261, 1987. 105. Spivak, JL, et al: Acquired immune open-label, community-based trial to
88. Colon-Otero, G, et al: A practical deficiency syndrome and pancytopenia. compare the safety and activity of com-
approach to the differential diagnosis JAMA 250:3084, 1983. bination therapy with recombinant
and evaluation of the adult patient with 106. Zon, LI, et al: Haematologic manifesta- interferon-alpha2b and zidovudine
macrocytic anemia. Med Clin North Am tions of the human immune deficiency versus zidovudine alone in patients
HOSS, WE virus (HIV). Br J Haematol 66:251, 1987. with asymptomatic to mildly sympto-
89. Nomura, S, et al: Case report: Hypothy- 107. Zon, LI, and Groopman, JE: Haemato- matic HIV infection. HIV Protocol
roidism as a possible cause of an logic manifestations of the human C91-253 Study Team. J Acquir Immune
acquired reversible hemolytic anemia. immune deficiency virus (HIV). Semin Defic Syndr Hum Retrovirol 20:245,
Am J Med Sci 302:23, 1991. Hematol 25:208, 1988. (YES).
304 Chapter 14. Hypoproliferative Anemia: Anemia Associated with Systemic Diseases
. Snower, DP, and Weil, SC: Changing 141. Ruth, V, et al: Postnatal changes in 158. Ohls, RK, et al: Effects of early ery-
etiology of macrocytosis. Zidovudine as serum immuno-reactive erythropoietin thropoietin therapy on the transfusion
a frequent causative factor. Am J Clin in relation to hypoxia before and after requirements of preterm infants below
Pathol 99:57, 1993. birth. J Pediatr 116:950, 1990. 1250 grams birth weight: A multicenter,
124. Eyer-Silva WA, et al: Macrocytosis in 142. George, JW, et al: Response of rhesus randomized controlled trial. Pediatrics
patients on Stavudine. Scan J Infect Dis monkeys to recombinant human ery- 108:934, 2001.
33:239, 2001. thropoietin: Comparison of adults and 159. Yeo, CL, et al: Effect of recombinant
. Glaspy, JA, and Chap, L: The clinical infants (abstract). Pediatr Res 25:269A, human erythropoietin on transfusion
application of recombinant erythropoi- 1989. needs in preterm infants. J Paediatr
etin in the HIV-infected patient. In Spi- 3. Stockman, JA, et al: The anemia of pre- Child Health 37:352, 2001.
vak, JL (Ed): Hematol Oncol Clin maturity: Factors governing the erythro- 160. Meyer, MP, et al: Recombinant erythro-
North Am 8:945, 1994. poietin response. N Engl J Med poietin and blood transfusion in
126. Phair, JP, et al: Recombinant human 296:647, 1977. selected preterm infants. Arch Dis
erythropoietin treatment: Investigational 144. Brown, MS, et al: Decreased response Child Fetal Neonatal Ed 88:F41, 2003.
new drug protocol for the anemia of the of plasma immunoreactive erythropoi- 161. Avent, M, et al: A comparison of high
acquired immunodeficiency syndrome. etin to “available oxygen” in anemia of versus low dose recombinant human
Overall results. Arch Intern Med prematurity. J Pediatr 105:793, 1984. erythropoietin versus blood transfusion
153326695 1993: 145. Stockman, JA, et al: Anemia of prema- in the management of anemia of prema-
Wie Semba, RD, et al: Highly active anti- turity: Determinants of the erythropoi- turity in a developing country. J Trop
retroviral therapy associated with etin response. J Pediatr 105:786, 1984. Pediatr 48:227, 2002.
improved anemia among HIV-infected 146. Stockman, JA: Anemia of prematurity. 162. Franz, AR, and Pohlandt, F: Red blood
women. AIDS Patient Care STDS Current concepts in the issue of when cell transfusions in very and extremely
15:473, 2001. to transfuse. Pediatr Clin North Am low birthweight infants under restrictive
123: Moore, RD, and Forney, D: Brief Soe MEOS6: transfusions guidelines: Is exogenous
Report: Anemia in HIV-infected 147. Stockman, JA: Anemia of prematurity. erythropoietin necessary? Arch Dis
patients receiving highly active anti- Clin Perinatol 4:239, 1977. Child Fetal Neonatal Ed 84:F96, 2001.
retroviral therapy. J Acquir Immune 148. Lachance, C, et al: Myocardial, erythro- 3. Amin, AA, and Alzahrani, DM: Effi-
Defic Syndr 29:54, 2002. poietic, and metabolic adaptations to cacy of erythropoietin in premature
129. Semba, RD, et al: Improvement of ane- anemia of prematurity. J Pediatr infants. Saudi Med J 23:287, 2002.
mia among HIV-infected injection drug 125:278, 1994. 164. Vamvakas, EC, and Strauss, RG: Meta-
users receiving highly active antiretro- 149. Strauss, RG: Controversies in the man- analysis of controlled clinical trials
viral therapy. J Acquir Immune Defic agement of the anemia of prematurity studying the efficacy of rHuEPO in
Syndr 26:315, 2001. using single-donor red blood cell trans- reducing blood transfusions in the ane-
. Buskin, SE, and Sullivan, PS: Anemia fusions and/or recombinant human ery- mia of prematurity. Transfusion 41:406,
and its treatment and outcomes in per- thropoietin. Trans Med Rev 20:34, 2006. 2001.
sons infected with human immunodefi- 150. Lenes, BA, and Sacher, RA: Blood 165. Strauss, RG: Managing the anemia of
ciency virus. Transfusion 44:826, 2004. component therapy in neonatal medi- prematurity: Red blood cell transfusions
13 — . Sullivan, PS, et al: Epidemiology of cine. Clin Lab Med 1:285, 1981. versus recombinant erythropoietin.
anemia in human immunodeficiency . Strauss, RG, et al: Commentary on Trans Med Rev 15:213, 2001.
virus (HIV)-infected persons: Results small-volume red cell transfusions for 166. Doyle, JJ: The role of erythropoietin in
from the multistate adult and adolescent neonatal patients. Transfusion 30:565, the anemia of prematurity. Semin Peri-
spectrum of HIV disease surveillance 1990. natol 21:20, 1997.
project. Blood 91:301, 1998. Ik, Meyer, MP, et al: Recombinant human 167. Widness, JA, and Strauss, RG: Recom-
. Dallman, PR: Anemia of prematurity. erythropoietin in the treatment of the binant erythropoietin in treatment of the
Annu Rev Med 32:143, 1981. anemia of prematurity: Results of a premature newborn. Semin Neonatol
3. Strauss, RG: Current issues in neonatal double-blind, placebo-controlled study. 3:163, 1998.
transfusions. Vox Sang 51:1, 1986. Pediatrics 93:918, 1994. 168. Brown, MS, et al: Postnatal changes in
. Widness, JA, et al: Erythropoietin Il). Frenck, RW, and Shannon, KM: erythropoietin levels in untransfused pre-
transplacental passage: Review of ani- Hematopoietic growth factors in pedi- mature infants. J Pediatr 103:612, 1983.
mal studies. J Perinat Med 23:61, 1995. atrics. Curr Opin Pediatrics 5:94, 1993. 169. Schaefer, RM, and Schaefer, L: The
135 . Schneider, H, and Malek, A: Lack of per- 154. Ohls, RK, and Christensen, RD: hypochromic red cell: A new parameter
meability of the human placenta for ery- Recombinant erythropoietin compared for monitoring of iron supplementation
thropoietin. J Perinat Med 23:71, 1995. with erythrocyte transfusion in the during rhEPO therapy. J Perinatol Med
136. Malek, A, et al: Lack of transport of treatment of anemia of prematurity. 232835 1995.
erythropoietin across the human pla- J Pediatr 119:781, 1991. 170. Kumar, P, et al: Recombinant human
centa as studied by an in vitro perfusion IIS). Shannon, KM, et al: Circulating ery- erythropoietin therapy for treatment of
system. Pflugers Arch 427:157, 1994. throid progenitors in the anemia of pre- anemia of prematurity in very low birth
. Zanjani, ED, et al: Studies in the liver maturity. N Engl J Med 317:728, 1987. weight infants: A randomized, double-
to kidney switch of erythropoietin pro- 156. Shannon, KM, et al: Recombinant blind, placebo-controlled trial. J Perina-
duction. J Clin Invest 67:1183, 1981. human erythropoietin stimulates ery- tol 18:173, 1998.
. Strauss, RG: Erythropoietin in the thropoiesis and reduces erythrocyte 17 ion. van Straaten HLM, et al: Evaluation of
pathogenesis and treatment of neonatal transfusions in very low birth weight a strategy to limit blood donor exposure
anemia. Transfusion 35:68, 1995. preterm infants. Pediatrics 95:1, 1995. in high risk premature newborns based
. Strauss, RG: Erythropoietin and neona- . Ohls, RK, et al: The effect of erythro- on clinical estimation of transfusion
tal anemia. N Engl J Med 330:1227, poietin on the transfusion requirements need. J Perinatol Med 28:122, 2000.
1994. of preterm infants weighing 750 grams 172. Strauss, RG: How I transfuse red blood
140 . Bauer, C: Erythropoietin: From gene or less: A randomized, double-blind, cells and platelets to infants with the
structure to therapeutic applications. J placebo-controlled study. J Pediatr anemia and thrombocytopenia of pre-
Perinat Med 23:77, 1995. 131:661, 1997. maturity. Transfusion 48:209, 2008.
PART 3
WHITE BLOOD CELL DISORDERS
Chapter
Neutrophils OBJECTIVES
Neutrophil Function
At the end of this chapter, the learner should be able to:
Disorders of Neutrophils
Eosinophils tie Characterize the changes in neutrophil count and morphology that develop in
response to bacterial infection.
Basophils
. Define the three modes of neutrophilic migration.
Monocytes
Absolute Monocytosis: Oo . Describe the changes in migration pattern and appearance that occur in a neutrophil
Reactive versus Malignant as a result of chemoattractant stimulation.
Causes
. Characterize the sequence of events that occur during phagocytosis.
Lymphocytes
Definition of Lymphocytosis . Define neutropenia.
Lymphocyte Morphology . Identify three causes of acquired neutropenia.
Causes of Reactive
Lymphocytosis OO}
=
a . Describe the etiology and classic clinical features of Chédiak—Higashi syndrome,
Laboratory Examination May-—Hegglin anomaly, Alder—Reilly anomaly, and Pelger—Huét anomaly including
Lymphocytopenia the associated changes in neutrophil morphology.
Case Study 1 . Describe the inheritance of and molecular basis for chronic granulomatous disease.
Case Study 2 . Compare and contrast three white cell anomalies in regard to morphology.
. Define lymphocytosis.
305
306 Chapter 15 Cell Biology, Disorders of Neutrophils, Infectious Mononucleosis, and Reactive Lymphocytosis
16. Describe how the Epstein-Barr virus affects the B- and T-lymphocyte populations.
17. Utilize laboratory results for distinguishing infectious mononucleosis from other
lymphocytoses.
, Blood vessel
with PMN
Activation
Diapedesis
_ Neutrophil Migration
Chemotaxis
= —> pyioge ofMigration _ Characteristics
{Ee ) Endothelial
naMN fe \ ceils Locomotion Cot Nondirectional
Chemokinesis Nondirectional acceleration
> d
322 NS
2 i
&
| of migration speed
ee i PMN Opsonization Chemotaxis Directional
Bacteria
(mS 7 »)
fira |’ Pa aia Att achment
ANNA, =
Chemontiractans LER
QUANTITATIVE DISORDERS
NEUTROPHILIA Neutrophils play a central role in removing
infectious and inflammatory agents that challenge host immu-
nity. Within minutes, neutrophils are stimulated to migrate
from circulation through junctions between endothelial cells
to the site of infection or inflammation. The classic response
to infectious and inflammatory processes is an increase in the
relative number of neutrophils, termed neutrophilia. The
accelerated release of neutrophils from the bone marrow
reserve is accompanied by a “shift to the left” that is defined
as an increased number of metamyelocytes and band forms in
Figure 15-3 m Electron microscopy of phagolysosome formation.
(A) Staphylococci lie within phagocytic vesicles limited by sacs formed the circulating pool.? An increase in circulating neutrophils
from inverted pieces of the neutrophil membrane. Cytoplasmic gran- and immaturity is similarly observed in the early stages of
ules are approaching the phagocytic vesicles. (B) Higher magnifica- neoplastic conditions, such as chronic myeloid leukemia
tion shows degranulation with the discharge of granule contents (CML), and other chronic myeloproliferative disorders (see
into the vicinity of the staphylococcus.
Chaps. 17 and 18). The major distinction is that the myeloid
precursors released during the infectious response are more
limited to the metamyelocyte and band stages; whereas, in
the lytic activity of granular enzymes within the phagolyso- neoplastic processes, earlier precursor cells such as myelo-
some, which leads to eventual killing and digestion. cytes, promyelocytes, and blasts are also present. Table 15—4
Microorganism destruction (killing) is accomplished by lists the causes of secondary reactive neutrophilia. Leukocyte
oxygen-dependent and non-oxygen-dependent mechanisms.
alkaline phosphatase (LAP), by cytochemical staining, is use-
The non-oxygen-dependent mode of killing the internalized ful for differentiating neutrophilic response to infection
particle is represented by an alteration in pH and the release
(leukemoid reaction) from chronic myeloid leukemia, where
of the lysosomal and proteolytic enzymes into the phagolyso-
the LAP is increased in leukemoid reaction and decreased in
some.' The lytic enzymes possess bacteriocidal activity that
cleaves segments of the bacterial cell wall. Alternatively,
oxygen-dependent killing involves the release of nictinomide
adenine dinucleotide phosphatase (NADPH) oxidase, which
mediates the production of the active oxygen metabolites
superoxide and hydrogen peroxide during a process known as
the respiratory burst.° These active oxygen metabolites are
capable of microorganism injury. To enhance the antibacterial
activity further, superoxide and hydrogen peroxide react with
other products of phagocytosis such as myeloperoxidase
(in primary granules of neutrophils and monocytes) to pro- ¢ Infections
duce highly toxic agents such as hypochlorous acid (i.e., 1. Bacterial (most common)
household bleach).’* 2. Toxoplasma, mycobacterium, leptospiral, and viral (less
common)
¢ Inflammatory Disorders
¢ Tissue Necrosis
Disorders of Neutrophils
1. Burns
2. Trauma
Disorders of the neutrophilic cell line may predispose an
3. Infarct
individual to recurrent bacterial infections that are resistant to 4. Acute gout
treatment. Neutrophilic disorders are classified as quantitative ° Metabolic
and qualitative, reflecting changes in neutrophil number and 1. Uremia
function, respectively. Quantitative disorders may present a 2. Ketoacidosis
3. Eclampsia
significant decrease (or increase) in the absolute neutrophil
* Other Causes Unrelated to Infection or Inflammatory
number and are referred to as neutropenia and neutrophilia, Process
respectively. Neutropenia limits the potential for effective 1. Stress
mobilization and killing at the site of microbial invasion.? 2. Rigorous exercise
Alternately, qualitative disorders are marked by neutrophil 3. Smoking
dysfunction as a result of impaired migration or altered 4. Pregnancy
5. Trauma
bacteriocidal activity. As noted, phagocytosis occurs in a com-
6. Acute hemorrhage or hemolysis
plex sequence of events ranging from activation, migration, 7. Postsplenectomy
and diapedesis to the recognition, ingestion, and killing of an
Chapter 15 Cell Biology, Disorders of Neutrophils, Infectious Mononucleosis, and Reactive Lymphocytosis 309
Figure 15-4 m Toxic granulation (peripheral blood). Note the Figure 15-6 ®@ Vacuolated neutrophils suggesting the presence of
prominent dark-staining granules. infection or a severe inflammation.
310 Chapter 15 Cell Biology, Disorders of Neutrophils, Infectious Mononucleosis, and Reactive Lymphocytosis
‘Table,15-6 Classification of
_ Neutropenia — Drug Class Drug Prototype
GVHD = graft-versus-host disease; BM = bone marrow; CMPD = chronic myeloproliferative disorders; AML
= acute
myelocytic leukemia; PNH = paroxysmal nocturnal hemoglobinuria.
fects Motility/Chemotaxis Defects
1. Chronic granulomatous disease 1. Lipid storage diseases (1.e., Gaucher’s)
2. Immunodeficiency disorders with 2. Complement disorders
decreased immunoglobulins 3. Chediak—Higashi syndrome
3. Complement disorders 4. Specific granule deficiency
4. Myeloperoxidase 5. Actin and microtubular defects
5. Specific granule deficiency 6. Hyperimmunoglobulin E syndrome
Chronic Granulomatous Disease present with deficiencies in one of the cytosol subunits (see
Chronic granulomatous disease (CGD) is a familial, heteroge- Table 15-15). Regardless of the subunit defect, superoxide
neous disorder of the neutrophil that may be chronic or inter- cannot be produced, and, in turn, bacterial killing is ineffec-
mittent. The mode of inheritance of most cases is X-linked tive.2* Ingestion of bacteria, degranulation, and phagolyso-
recessive; however, autosomal recessive cases have been some formation are normal.
described. CGD is the best-understood disorder of neutrophils. Diagnosis is established by demonstrating a bacterio-
It is seen in | out of every 500,000 individuals who exhibit cidal defect resulting from the absence of the oxidative
recurrent bacterial and fungal infections, generally during the burst on the nitroblue tetrazolium test (NBT), a luminol-
first year of life.!? The congenital abnormality is attributable to enhanced assay, or by measuring respiratory burst activity
a failure in the activation of the respiratory burst that results in with flow cytometry.*! The X-linked-recessive inheritance
little or no superoxide production and is the result of mutations may be confirmed by studying the family history and
in the genes encoding the subunits of the enzyme, NADPH performing molecular gene analysis.” Indicators of CGD
oxidase.”’ The characteristic clinical picture of these patients is include the presence of disease in male members of
of lymphadenitis, deep tissue infections such as osteomyelitis, the maternal family and intermediate to low neutrophil
and visceral and hepatic abscesses, recurrent pulmonary infec- activity in the mothers and sisters of affected boys. In most
tions, organomegaly, and infected eczematoid rash.*'** CGD cases, the female relatives are clinically well, but occasion-
patients exhibit neutrophilia rather than neutropenia. The hall- ally they may present with an increased susceptibility to
mark of CGD is the formation of granulomas during chronic infections or a syndrome resembling systemic lupus
inflammatory reactions that keep the organisms localized. erythematosus.
Life-threatening infections are often caused by Aspergillus, S. Progress has been made in modalities of treatment and
aureus, and enteric organisms.” the prognosis of CGD is improving. Aggressive prophylac-
The first cases of CGD were observed in boys presenting tic antibiotic therapy should be initiated as soon as a diag-
with severe bacterial infections by the first year of life, and were nosis is made. Gamma (y) interferon has been effective in
believed to be inherited as a sex-linked recessive disorder. Most limiting the frequency of infections, and granulocyte trans-
of these patients died of septicemia or chronic pulmonary disease fusions are useful in patients who respond poorly.*? Bone
in early childhood. Additional cases of CGD were identified in marrow and cord blood stem cell transplantation has been
girls who exhibited separate but genetically similar defects. It is proven to be successful when performed in children, even
now recognized that CGD may be inherited in a sex-linked- during ongoing infection; however, the rate of success is
recessive or autosomal-recessive pattern, depending on the sub- dependent on the HLA-match and supportive therapy with
unit of NADPH oxidase affected* (Table 15—15). both G-CSF and granulocyte infusions.*° In vitro studies of
Chronic granulomatous disease results from one of four gene correction for CGD have been carried out using
single gene mutations in NADPH oxidase, which is composed peripheral blood progenitor cells, making the prospect of a
of oxidase components known as phagocytic oxidase subunits, cure most encouraging.”!
or phox.*® The interaction of these subunits results in the for-
mation of superoxide during the respiratory burst. Each phox DISORDERS OF ABNORMAL NEUTROPHIL MORPHOLOGY
subunit is identified as a glycoprotein (gp) or a protein (p). Granulocytes with abnormal morphology can be acquired or
Two of the subunits, gp91-phox and p22-phox, are located in inherited. The acquired disorders of neutrophil morphology
the plasma membrane and together form the enzyme are listed in Table 15-16. Although rare, several hereditary
cytochrome b. The other subunits, p47-phox, p67-phox, and abnormalities in neutrophil morphology may be observed in
p40-phox, reside in the cytosol. Stimulation of NADPH oxi- patients without an association with infection, neutrophilic
dase leads to the migration of the cytosol subunits to the dysfunction, or altered neutrophil number. Such abnormali-
plasma membrane.”’ With the help of secondary mediators, the ties in neutrophilic morphology are collectively referred to as
phox subunits assemble into the active oxidant, superoxide.?’ “white blood cell anomalies”. The inherited disorders of
The classic X-linked form demonstrates a total absence of neutrophil morphology are listed and their characteristics
cytochrome b; whereas, most autosomal-recessive cases summarized in Table 15-17.
Hypersegmentation and hyposegmentation of the
nucleus are WBC anomalies that reflect the number of
segmented lobes demonstrated in the mature neutrophil.
Table 15-15 Molecular Basis of Hypersegmentation describes larger-than-normal neutrophils
~ Chronic Granulomatous with six or more nuclear lobes present (see Chaps. 5 and 7).
Disease A similar hypersegmentation of eosinophils has been
reported, with five or more nuclear lobes present. Hyperseg-
Cytochrome b Mode of Neutrophil Structure mentation in the granulocytes may be an indicator of an
Subunit Affected Inheritance Inveae acquired anemia such as megaloblastic anemia, or of a
p47-phox
benign autosomal-dominant condition known as hereditary
Autosomal Cytosol
p67-phox Autosomal Cytosol
hypersegmentation of neutrophils.*?
gp9 1-phox X-linked Plasma membrane Hyposegmentation of the nucleus is characteristic of
p22-phox Autosomal Plasma membrane Pelger—Huét anomaly, a condition in which the nucleus is
found to be bilobed or to have no lobulation whatsoever."!
Chapter 15 Cell Biology, Disorders of Neutrophils, Infectious Mononucleosis, and Reactive Lymphocytosis SS
¢ Hypersegmentation
1. Megaloblastic anemia
2. Myeloid malignancies (i.e., AML, myelodysplastic
syndromes)
¢ Hypogranularity
1. Myeloid malignancies (i.e., AML, myelodysplastic
syndromes)
¢ Pseudo-Pelger—Huét
1. Myeloid malignancies (i.e., AML, myelodysplastic
syndromes)
2. Drugs (sulfonamides, colchicines, etc.)
¢ Cytoplasmic Inclusions Figure 15-9 @ Pelger-Huét anomaly (peripheral blood). (From
1. Cryoglobulinemia Hyun, BH, et al: Practical Hematology. A Laboratory Guide with
* Giant Neutrophils Accompanying Filmstrip. WB Saunders, Philadelphia, 1975, with
1. AIDS infection permission.)
2. G-CSF therapy
Severity-
Mild 700-1400/mm*
(0.7-1.4 X 10°/L)
Moderate 1500-4900/mm?
(1.5-4.9 x 10°/L)
Severe >5000/mm*
(>5.0 X 10%/L)
=a,
RNA." This anomaly is also associated with thrombocytope-
nia and giant platelets, and variable neutropenia. Many of
these patients may be clinically asymptomatic (see Chap. 25).
Chediak—Higashi, previously discussed under neu-
trophils with abnormal function, is also classified as a neu- ae
ti pat
sth
Adenovirus
Chickenpox
Cytomegalovirus
EBV (infectious mononucleosis)
Hepatitis
Herpes simplex
Herpes zoster
Human immunodeficiency virus (HIV)
Influenza
Paramyxovirus (mumps)
Rubella (measles)
Bacterial
Figure 15-15 @A small plasmacytoid lymphocyte with coarse Brucellosis
chromatin and an eccentric nucleus. Paratyphoid fever
Pertussis (whooping cough)
Tuberculosis
Morphological criteria alone usually enable one to dis- Typhoid fever
tinguish reactive lymphocytoses from malignant disorders.
Drug Reactions
When there is ambiguity, clinical history, cell typing, and During recovery from acute infections (especially in children)
serologic findings may help in distinguishing these entities. If
necessary, a bone marrow or lymph node biopsy may lead to Miscellaneous
the correct diagnosis. The most common causes of a reactive Acute infectious lymphocytosis
Allergic reactions
lymphocytosis are shown in Table 15-26. Malignant condi- Autoimmune diseases
tions that may be confused with a reactive lymphocytosis are Hyperthyroidism
listed in Table 15-27. Malnutrition
Rickets
Syphilis
Causes of Reactive Lymphocytosis Toxoplasmosis
INFECTIOUS MONONUCLEOSIS
HISTORY In 1907, Turk described clinical symptoms in sev-
eral patients who probably suffered from infectious mononu- terminology of morphology is seldom used today. Infectious
cleosis (IM) caused by EBV infection.*° Reactive lymphocyte mononucleosis is a historical term. We now know that the
morphology and clinical symptoms were correlated in 1920 large mononuclear cells originally described in IM are lym-
by Sprunt, who named the syndrome “infectious mononucle- phocytes and not monocytes.
osis.”*? Downey, in 1923, described in further detail the Later, in 1932, Paul noticed that serum from patients with
unusual lymphocyte morphology associated with IM.*° IM contained antibodies against sheep erythrocytes.*' This dis-
Although morphology is still important, Downey’s descriptive covery was the basis for the Monospot test (Ortho Pharmaceu-
ticals). This contemporary test is the most commonly used and
the simplest method available for measuring IgM heterophile
antibodies. Heterophile antibodies are antibodies that also react
with cells of other species. In particular, the IgM antibodies
react with sheep and horse erythrocytes. The antibodies are not
absorbed by guinea pig kidney in the Monospot test and, thus,
if present, agglutinate horse erythrocytes.
The DNA virus responsible for IM was first observed in
lymphoblasts cultured from patients with Burkitt’s lym-
phoma**** (see Chap. 21). This virus is now known as the
Epstein-Barr virus (EBV). In addition to Burkitt’s lymphoma,
EBV has been associated with other malignancies, including
B-cell lymphoproliferative syndromes, nasopharyngeal carci-
noma, Hodgkin lymphoma, and gastric carcinoma.*! The
malignancies appear to occur after the virus has been dormant
in the host for years.
Figure 15-16 @A large reactive lymphocyte with prominent In regard to acute viral illness, EBV was first linked as
nucleolus (immunoblast). the causative agent for IM by Henle in 1967.4 He was able to
Chapter 15 Cell Biology, Disorders of Neutrophils, Infectious Mononucleosis, and Reactive lymphocytosis 321
limited success has been seen using y-interferon to treat compli- be seen in 4% of patients.°* Morphologically, these cells
appear similar to those seen in CLL/small lymphocytic lym-
cations such as pneumonia or encephalitis.°°
phoma (SLL). In pertussis, immunologic marker studies
Antibiotics are not useful unless there are complications
reveal that the lymphocytes are predominantly helper T
such as streptococcal pharyngitis. The fatality rate of IM is
cells.°° Prominent lymphadenopathy is notably unusual in
approximately | in 3000 cases.*° In acute cases, complete
children with pertussis or infectious lymphocytosis.
recovery usually occurs within 2 months, and recurrences are
extremely rare. It should be noted that not all children who
MALIGNANT CONDITIONS
have acquired immunity had a prior history or clinical symp-
Malignant disorders that may be confused with reactive lym-
toms of IM. EBV infections in immunocompromised patients
phocytoses are listed in Table 15—27. In general, the malig-
may be life threatening. In view of the increasing incidence of
nant cells in leukemia and lymphomas are monotonous in
patients with immunodeficencies, the morbidity rate is likely
appearance and, therefore, are morphologically different from
to increase.
the reactive lymphocytes seen in benign conditions. Malig-
nant lymphoid cells in leukemias and lymphomas are mono-
CYTOMEGALOVIRUS INFECTION
Cytomegalovirus (CMV) belongs to the herpes virus family clonal when immunophenotypically analyzed.
and is endemic worldwide. The virus may be transmitted by The morphology of lymphoblasts seen in acute lym-
oral, respiratory, and sexual means or by blood transfusion phoblastic leukemia (ALL) is different from that of reactive
and organ transplantation. Patients with CMV infection may lymphocytes. ALL is morphologically classified as L1, L2, or
have recurrent infection or reactivation of a latent infection, L3, depending on the morphology of the cells (see Chap. 16). In
as seen in herpes simplex infection, or may be reinfected with contrast to reactive lymphocytes, blasts of L2 leukemia have
fine chromatin and prominent nucleoli (Fig. 15-17). The blasts
a different CMV strain. CMV infection is the most common
cause of heterophile-negative IM. The diagnosis is made by of L1 leukemia are monotonous in appearance and do not have
demonstrating the presence of IgM antibodies to CMV or by as much cytoplasm as do reactive lymphocytes (Fig. 15-18).
shell vial tissue cultures. The N:C ratio is much higher in LI blasts than in reactive
lymphocytes. In L3 blasts (Fig. 15-19), there are abundant
CLINICAL MANIFESTATIONS In the majority of immuno- cytoplasmic vacuoles that are lacking in reactive lymphocytes.
competent individuals, CMV infection is usually asympto- The lymphoid cells present in prolymphocytic leukemia (PLL)
matic. The clinical manifestations reflect the involved may also be confused with reactive lymphocytes. The promi-
organ(s). When CMV infection occurs in previously healthy nent, usually single, nucleoli that are present in PLL are shown
individuals, the symptoms mimic EBV infection. Symptoms in Figure 15-20. Reactive lymphocytes have more abundant
include fever, sore throat, splenomegaly, lymphadenopathy, cytoplasm and less prominent nucleoli than prolymphocytes.
and myalgia. Morphological changes in lymphocytes are Severe cytopenias that occur in acute leukemias are
indistinguishable from those seen in EBV infection. Because infrequent in benign lymphocytosis. With anemia or severe
of liver involvement, mild to moderate elevations of liver thrombocytopenia, the diagnosis of leukemia must be
function tests are common. considered (see Chap. 16).
In immunocompromised individuals, CMV infection may Patients with lymphoma may have a leukemic phase
be life threatening. Because CMV infection is endemic, par- with circulating malignant cells. Except for the large-cell
enterally acquired infection from transfusions is a concern, lymphomas, circulating lymphoma cells may be easily
especially with the increase in the numbers of individuals who distinguished from reactive lymphocytes. The nuclear
are immunocompromised from chemotherapy, immunosuppres-
sive drugs, or human immunodeficiency virus (HIV) infection.
BACTERIAL INFECTIONS
Lymphocytosis in bacterial infections occurs more commonly
in chronic infections and during the recovery period follow-
ing acute infections. The organisms most often responsible
include Brucella, Mycobacterium tuberculosis, and spiro-
chetes. Marked lymphocytosis with mature-appearing lym- Figure 15-17 mL2 leukemia. The L2 lymphoblasts have fine chro-
phocytes is seen in children with pertussis (whooping cough), matin with a moderate amount of cytoplasm and nucleoli. Note the
and absolute lymphocyte counts exceeding 50 X 10°/L may prominent nucleolus in one of the lymphoblasts (arrow).
Chapter 15 Cell Biology, Disorders of Neutrophils, Infectious Mononucleosis, and Reactive Lymphocytosis eS)i)Ww
iL
Figure 15-18 @L1 leukemia. These L1 lymphoblasts have a high Figure 15-20 @ Prolymphocytes with moderate amounts of cyto-
N:C ratio (scant cytoplasm), fine chromatin, and indistinct nucleoli. plasm and prominent nucleoli.
Except for the fine chromatin, note the similarity to mature
lymphocytes. These cells should not be confused with reactive
lymphocytes. Note the large “smudge cells” (arrows). observer. A reactive lymphocyte with abundant cytoplasm (low
N:C ratio) and indented cytoplasmic borders (large arrow) is
shown in Figure 15-13. The less experienced observer may
clefting seen in the leukemic phase of follicular small
mistake these unusual-appearing reactive lymphocytes for
cleaved-cell lymphoma is shown in Figure 15-21. Fortu-
monocytes or blasts. However, as seen in Figure 15—22, mono-
nately, in large-cell lymphoma, malignant cells in the periph-
cytes typically have linear condensation of chromatin, whereas
eral blood are rare. Prominent generalized lymphadenopathy
blasts (see Fig. 15-17) have chromatin strands that are finer and
is unusual in IM, but may occur in non-Hodgkin’s or evenly dispersed.
Hodgkin lymphomas (see Chap. 21).
The lymphoblasts in L2 leukemia or the monoblasts in
MS5a ieukemia (a category of acute myeloid leukemia) may also
Laboratory Examination be mistaken for reactive lymphocytes. An MSa, monoblastic
leukemia 1s illustrated in Figure 15-23. Monoblasts have a low
A variety of laboratory procedures may help in the correct N:C ratio but fine chromatin is present as well as prominent
diagnosis of patients with an absolute lymphocytosis. CBC, nucleoh. The fine chromatin and prominent nucleoli (arrow)
serology, and microbiologic culture are often performed. Of present in an L2 lymphoblast is shown in Figure 15—17. With
these procedures, a CBC with differential and serologic stud- reactive lymphocytes, a careful review of the cell morphology
ies are the most useful. will reveal that overall, the chromatin is not fine enough for the
Proper evaluation of the peripheral blood smear is crucial cells to be classified as blasts and that the overall morphology
for the correct differential diagnosis in patients with an absolute is variable and not monotonous as seen in leukemias.
lymphocytosis. Especially in IM, reactive lymphocytes are Serologic tests play a critical role in establishing the
prominent and should easily be identified by the experienced diagnosis in patients with an absolute lymphocytosis. In the
1 Ss a
Se
4
Beane %
Figure 15-19 m L3 leukemia. Note the abundant cytoplasmic vac- Figure 15-21 ™ Lymphocytes from a patient with follicular small
uoles and clumped chromatin. cleaved-cell lymphoma. Note the prominent nuclear clefting.
324 Chapter 15 Cell Biology, Disorders of Neutrophils, Infectious Mononucleosis, and Reactive Lymphocytosis
Figure 15-22 ™ This monocyte has very fine granular cytoplasm, Figure 15-25 @ MSa leukemia. These monoblasts have abundant
cerebriform nucleus, linear condensation of chromatin, and no cytoplasm (low N:C ratio), fine chromatin with some chromatin
nucleolus. clumping, and prominent nucleoli.
proper clinical setting, a positive Monospot (heterophile anti- The Monospot test is most frequently used for diagnos-
body) test with reactive lymphocytes in the peripheral blood ing IM, because the rise in titer of heterophile antibodies
(see Fig. 15-13) is diagnostic of IM. A variety of antibodies parallels the rise in titer of the more specific EBV antibod-
formed by humoral responses may be measured when it is ies. The Monospot test is much quicker, easier to use, and
necessary to elucidate further the etiology of lymphocytosis less expensive than measuring viral-specific antibodies. If
in difficult cases or in cases in which the Monospot test is the Monospot test is initially negative, it should be repeated
negative” (Table 15—28). in | week, if IM is suspected. Serum may be analyzed for
In the first week of viral infections, IgM antibodies are antibodies to specific EBV antigens; antibodies to CMV and
formed against viral capsid antigens. During the second hepatitis may be measured, and viral cultures may be per-
week, as the immunologic response matures, IgG antibod- formed. Some of the possible testing strategies are dia-
ies are formed. The pattern of the immunologic response grammed in Figure 15—25.
and lymphocytosis that is expected in patients with IM In cases where malignancy is being considered, lympho-
is illustrated in Figure 15-24. A rise in titer should be cytes may be further characterized by flow cytometric
demonstrated by comparing serum obtained during acute immunophenotypic analysis. With leukemias and lym-
and convalescent phases. Also, by using enzyme-linked phomas, flow cytometric immunophenotyping usually reveals
immunosorbent assay (ELISA) techniques, antibodies to a monoclonal cell population.°’ As previously described in
CMV and hepatitis may be measured. Finally, viral cultures IM, B cells are infected by EBV with most of the reactive
may be performed. lymphocytes representing polyclonal T cells.
©
rsZ 50
oO = Se ee Be ee OOF
Op |
0 1 5 3 4 5 6 > g antibodies
Heterophile Negative Y
TIME IN WEEKS FROM ONSET OF ILLNESS (0) Infectious Mononucleosis Sa
Tel
‘Table 15-29 Causes of Secondary Reactive Lymphocytopenia (Absolute Counts: srt <
OX 10°/L in Adults; < 2.0 x 10°9/L in Children)
1, The enz matic contents of primary (azurophilic) gran- i, (Defective bacterial killing
ules include: 4 . d. Abnormal phagolysosome formation
a. NADPH oxidase and hydrogen peroxide 9. Identify the disease characteristic(s) associated with
b. Cytochrome b and collagenase
Chediak—Higashi syndrome.
ee ea lysozyme artial albinism, recurrent infections, and mild bleed-
. Alkaline phosphatase
and gelatinase
ing tendencies
2. Directional migration toward a gradient stimulated by a b. Granulomas, osteomyelitis, and hepatic abscesses
emoattractant is referred to as: c. Hypopigmentation of skin and chronic, swollen lymph
ee nodes :
b. Random mobility d. Periodic pneumonia that may result in lesions called
c. Opsonization pneumatoceles
d. Chemokinesis
10. Pelger—Huét anomaly may be described by:
3. The marking of an invading microbe with IgG and com- a. Large neutrophils and hypersegmentation of the
plement to facilitate recognition is referred to as: nucleus with greater than six lobes
Chemokinesis b. Dark-staining, coarse granules in cytoplasm of neu-
Soin ona trophils, eosinophils, basophils, and monocytes
c. Phagolysosome fusion c. Pale blue inclusions of the cytoplasm of neutrophils
d. Signal transduction and giant platelets
4. Which sequence reflects the correct order for phago- Hyposegmentation of the nucleus with the majority of
cytosis? neutrophils being bilobed or monolobed ,
a. Release of cytoplasmic granules; binding of particle; 11. Reactive lymphocytes may best be distinguished from
ingestion; fusion of phagolysosome blasts by the presence of which of the following mor-
b. Ingestion; binding of particles; fusion of phagolyso- phological features?
some; release of cytoplasmic granules a. Prominent nucleoli
Binding of particle; ingestion; fusion of phagolyso- Fine chromatin
some; release of cytoplasmic granules Heterogeneous cell population
d. Fusion of phagolysosome; binding of particle; release . High N:C ratio
of cytoplasmic granules; ingestion 12. Which of the following antigens is detectable first by
5. In oxygen-dependent killing, the enzyme responsible for ELISA?
meditating the production of active oxygen metabolites a. EBNA
during the respiratory burst is: BV-VCA (IgM)
a. Myeloperoxidase c. EBV-VCA (IgG)
b. Lysozyme d. Heterophile
c. Lactoferrin 13. The Epstein-Barr virus infects which of the following cells?
@ NADPH oxidase a. Helper T lymphocytes
6. The two most important biochemical products of the res- b.)\Cytotoxic T lymphocytes
piratory burst that are involved with particle digestion c. B lymphocytes
during active phagocytosis are: d. NK cells
a. Lactoferrin and gelatinase 14. What is the most frequent cause of a heterophile
b. Superoxide dismutase and catalase (Monospot) negative mononucleosis-like syndrome?
. Glutathione peroxidase and copper-zinc enzymes _ HIV
Superoxide anion and hydrogen peroxide (som
7. The morphological characteristic(s) associated with c. Hepatitis C
hediak—Higashi syndrome is (are): d. Toxoplasma gondii
a lysosomal granules 15. In which of the following conditions are reactive lym-
b. Hypersegmented agranular neutrophils with vacuolization phocytes found?
c. Prominent dark-staining granules and pyknotic nuclei a. Infectious mononucleosis
d. Pale blue inclusions in cytoplasm of neutrophils and b. CMV infection
giant platelets c. Rubella
8. The defect in chronic granulomatous disease is attributed to: (4 All of the above
a. Delayed degranulation
b. Inefficient ingestion of microbes
328 Chapter 15 Cell Biology, Disorders of Neutrophils, Infectious Mononucleosis, and Reactive Lymphocytosis
REFERENCES NT. Starkebaum, G: Chronic neutropenia 3)9) Sims, KL: Peripheral eosinophilia and
ile Gabrilovich, DI: The Neutrophils: New associated with autoimmune disease. diagnosis of hypereosinophilic syn-
Semin Hematol 39(2):121, 2002. drome. Lab Med 37:440, 2006.
Outlook for Old Cells. Imperial College
. Haddad, E, et al: Treatment of 34. Tefferi, A: Blood eosinophilia: A new
Press, London, 2005.
Chediak—Higashi syndrome by allogenic paradigm in disease classification, diag-
. Nathan, DG, and Oski, FA: Hematology
bone marrow transplantation: Report on nosis, and treatment. Mayo Clinic Proc
of Infancy and Childhood, ed 6. WB
10 cases. Blood 85:3328, 1995. 80:75, 2005.
Saunders, Philadelphia, 2003,
. Winkelstein, JA, et al: Chronic granulo- WWN . Gotlib, J: Molecular classification and
pp 882-951.
matous disease. Report on national pathogenesis of eosinophilic disorders:
. Viswanathan, K, and Dhabhar, FS:
registry of 368 patients. Medicine 2005 update. Acta Haematol 114:7,
Stress-induced enhancement of leuko-
(Baltimore) 79(3):155, 2000. 2005.
cyte trafficking into sites of surgery or
. Lambeth, JD: Regulation of the phago- . Vardiman, JW, et al: The World Health
immune activation. Proc Natl Acad Sci
cyte respiratory burst oxidase by protein Organization (WHO) classification of the
USA 102(16):5808, 2005. Epub 2005
interactions. J Biochem Mol Biol myeloid neoplasms. Blood 100(7):2292,
Apr 7.
33(6):427, 2000. 2002.
. Fox, SE, et al: The effects and compara-
i) . Carnide, EG, et al: Clinical and labora- Sie Jaffe, E, et al: The World Health Organi-
tive differences of neutrophil specific
tory aspects of chronic granulomatous zation Classification of Tumors, Pathol-
chemokines on neutrophil chemotaxis of
disease in description of eighteen ogy and Genetics, Tumors of
the neonate. Cytokine 29(3):135, 2005.
patients. Pediatr Allergy Immunol Haematopoietic and Lymphoid Tissues.
Epub 2004 Dec 8.
16(1):5, 2005. IARC Press, Lyon, 2001, pp 91-117.
Nn . Walker, A, et al: Regulation of neutrophil
. Segal, BH, et al: Genetic, biochemical, . Peterson, L: Reactive disorders of
apoptosis and removal of apoptotic cells.
and clinical features of chronic granulo- lymphocytes. In: Kjeldsberg, CR (Ed),
Curr Drug Targets Inflamm Allergy
matous disease. Medicine (Baltimore) Practical Diagnosis of Hematologic
4(4):447, 2005.
79(3):155, 2000. Disorders | Benign Disorders. American
. O'Flaherty, JT, and Cordes, JF: Huinan
. Johnston, RB: Clinical aspects of chronic Society for Clinical Pathology, Chicago,
neutrophil degranulation responses to
granulomatous disease. Curr Opin 2006, pp 259-276.
nucleotides. Lab Invest 70(6):816, 2004.
Hematol 8(1):17, 2001. 3). Sprunt, TP, and Evans, FA: Mononuclear
. Kannan, S: Neutrophil degranulation:
. Heyworth, PG, et al: Chronic granulo- leukocytosis in reaction to acute infec-
Therapeutic targets in [NTP]O mediated
matous disease. Review. Curr Opin tions (“infectious mononucleosis’).
neutrophil degranulation. Med Hypothe-
Immunol 15(5):578, 2003. Bull Johns Hopkins Hosp 31:410,
ses 63(2):325, 2004.
. Ezekowitz, RA: Update on chronic gran- 1920.
Stark, MA, et al: Phagocytosis of apop-
ulomatous disease: The concept of the AO. Downey, H, and McKinlay, CA: Acute
totic neutrophils regulates granulopoiesis
near-normal host. Review. Curr Clin Top lymphadenosis compared with acute
via IL-23 and IL-17. Immunity
Infect Dis 20:325, 2000. lymphatic leukemia. Arch Intern Med
DAG rea, ANOS. . Kamani, NR, and Infante, AJ: Chronic B27S2 1923)
. Wintrobe, MM: Greer, JP (Ed),
granulomatous disease and other disor- 41. Paul, JR, and Bunnell, WW: The pres-
Wintrobe’s Clinical Hematology, ders of neutrophil function. Review. ence of heterophile antibodies in infec-
ed 11. Philadelphia, Lippincott Williams Clin Rev Allergy Immunol 19(2):141, tious mononucleosis. Am J Med Sci
& Wilkins, 2004. 2000. 183:90, 1932.
. Kobayashi, SD, et al: Neutrophils in the . Geiszt, M, et al: Chronic granulomatous 42. Epstein, MA, et al: Virus particles in
innate immune response. Arch Immunol disease: More than the lack of cultured lymphoblasts from Burkitt’s
Ther Exp (Warsz) 53(6):505, 2005. superoxide? Review. J Leuko Biol lymphoma. Lancet 1:702, 1964.
i— ee. Foucar, K: Reactive disorders of granu-
69(2):191, 2001. 43. Pagano, JS: Epstein-Barr virus: The first
locytes and monocytes. In: Kjeldsberg, . Brown, JR, et al: Diminished production human tumor virus and its role in cancer.
CR (Ed), Practical Diagnosis of Hemato- of anti-inflammatory mediators during Proc Assoc Am Phys 111:6, 1999.
logic Disorders | Benign Disorders. neutrophil apoptosis and macrophage 44. Henle, G, et al: Relation of Burkitt’s
American Society for Clinical Pathology, phagocytosis in chronic granulomatous tumor-associated herpes-type virus to
Chicago, 2006, pp 191-276. disease (CGD). J Leuko Biol 73(5):591, infectious mononucleosis. Proc Natl
. Wagner, C, et al: Cellular inflammatory 2003. Acad Sci USA 59:94, 1968.
response to persistent localized WS), Stasia, MJ, et al: Characterization of six 45. Kobbervig, C, et al: Infectious mononu-
Staphylococcus aureus infection: Pheno- novel mutations in the CYBB gene lead- cleosis progressing to fatal malignant
typical and functional characterization of ing to different sub-types of X-linked lymphoma: A case report and review of
polymorphonuclear neutrophils (PMN). chronic granulomatous disease. Hum the literature. Leuk Lymphoma
Clin Exp Immunol 143(1):70, 2006. Genet 116(1-2):72, 2005. Epub 2004 44(7):1215, 2003.
138 Chen, Y, et al: Neutrophil activation by Nov 6. 46. Infectious mononucleosis fact sheet.
murine retroviral infection during 30. Bhattacharya, A, et al: Successful N S W Public Health Bull 16(9-10):168,
chronic ethanol consumption. Alcohol umbilical cord blood stem cell trans- 2005.
38(2):109, 2003. plantation for chronic granulomatous 47. Ebell, MH: Epstein-Barr virus infectious
. Capsoni, F, et al: Primary and sec- disease. Bone Marrow Transplant mononucleosis. Am Fam Physician
ondary autoimmune neutropenia. Arthri- 31(5):403, 2003. 70(7):1279, 2004.
tis Res Ther 7(5):208, 2005. Epub 2005 Sil Mamishi, S, et al: A case of invasive 48. Hiroshima, K, et al: Fatal infectious
Aug 31. aspergillosis in CGD patient successfully mononucleosis with evidence suggestive
. Bauduer, F: G-CSF: A very efficient treated with Amphotericin B and INF- of the development of B cell lymphoma.
therapy for chronic autoimmune neu- gamma. Ann Clin Microbiol Antimicrob Pathol Int 53(9):642, 2003.
tropenia. A brief review of the literature. 4:4, 2005. 49. Cyran, EM, et al: Intravenous gamma-
Hematol Cell Ther 40:189, 1998. . Kjeldsberg, CR (Ed): Practical Diagnosis globulin treatment for immune thrombo-
16. Christensen, RD, and Calhoun, DA: of Hematologic Disorders | Benign Dis- cytopenia associated with infectious
Congenital neutropenia. Clin Perinatol orders. American Society for Clinical mononucleosis. Am J Hematol 38:124,
31(1):29, 2004. Pathology, Chicago, 2006. 1991.
330 Chapter 15 Cell Biology, Disorders of Neutrophils, Infectious Mononucleosis, and Reactive Lymphocytosis
50. Andersson, J: EBV, everybody’s virus: from human herpesvirus-6 (HHV-6) in 56. Ventura, KC, and Hudnall, SD: Hemato-
The management of Epstein-Barr virus patients with mononucleosis-like ill- logic differences in heterophile-positive
infections. Herpes 3:55, 1996. nesses not resulting from Epstein-Barr and heterophile-negative infectious
. Rosenberg, ES, et al: Acute HIV infec- virus or cytomegalovirus. Am J Clin mononucleosis. Am J Hematol
tion among patients tested for mononu- Pathol 93:776, 1990. 76(4):315, 2004.
cleosis [letter], N Engl J Med 340:12, 54. Hodge, G, et al: A marked decrease in . Dunphy, CH: Applications of flow
1999. L-selectin expression by leucocytes in cytometry and immunohistochemistry to
. Steeper, TA, et al: Heterophile-negative
nNiw) infants with Bordetella pertussis infec- diagnostic hematopathology. Arch Pathol
mononucleosis-like illnesses with reac- tion: Leucocytosis explained? Respirol- Lab Med 128:1004, 2004.
tive lymphocytosis in patients undergo- ogy 8(2):157, 2003. kek Peterson, L: Lymphocytopenia. In:
ing seroconversions to the human OD: Chen, X, et al: Pertussis toxin as an Kjeldsberg, CR (Ed), Practical Diagnosis
immunodeficiency virus. Am J Clin adjuvant suppresses the number and of Hematologic Disorders | Benign
Pathol 90:169, 1988. function of CD4+CD25+ T regulatory Disorders. American Society for Clinical
. Steeper, TA, et al: The spectrum of clini- cells. Eur J Immunol 36(3):671, Pathology, Chicago, 2006, pp 271.
cal and laboratory findings resulting 2006.
Chapter
Introduction to Leukemia
and the Acute LeuKemias
Ken Gatter, MD, JD
Frank Cruz Ill, MD
Rita Braziel, MD
Introduction OBJECTIVES
Definition and Overview
Comparison of Acute At the end of this chapter, the learner should be able to:
and Chronic Leukemia L. Define leukemia.
Historical Perspectives
Etiology and Risk Factors 2 . Compare and contrast acute versus chronic leukemia, and leukemia versus lymphoma.
Classifications 3 . Compare and contrast acute myeloid and acute lymphoblastic leukemia.
Introduction to Acute 4. Describe the FAB classification and list the characteristics of each type.
Leukemia
Incidence 5 . List and describe each of the WHO subtypes of acute lymphoblastic and acute
Clinical Features myeloid leukemia, paying particular attention to immunophenotypic and cytogenetic
Laboratory Evaluation of features.
Acute Leukemia . Describe characteristic morphology and cytochemical staining patterns for each of
Acute Myeloid Leukemia the subtypes of acute myeloid leukemia.
FAB Classification of AML
. List the WHO categories of acute myeloid leukemias with recurrent cytogenetic
WHO Classification of AML
abnormalities.
Acute Lymphoblastic
. List the criteria for immunologic classification of acute lymphoblastic leukemia,
Leukemia
including early precursor B- and T-cell acute lymphoblastic leukemias.
Classification of ALL and
Introduction . Describe the role of genetic analysis of newly diagnosed leukemia and know some of
Review of Lymphocyte the prognostic implications of the more common genetic alterations seen in acute
Ontogeny leukemias.
FAB Classification of ALL
. Describe the morphologic and cytogenetic abnormalities associated with acute
WHO Classification of ALL
promyelocytic leukemia (FAB M3) and understand some of the clinical implications.
Burkitt’s Leukemia/
Lymphoma (Mature Lt Correctly identify a blast on a peripheral blood smear or bone marrow aspirate, be
B-Cell ALL) able to distinguish between the typical morphologic features of a lymphoblast and a
Childhood versus Adult ALL myeloblast and list some of the flow cytometry antibodies useful for distinguishing
Treatment of Acute between myeloid and lymphoid.
Leukemia
Treatment of ALL
Treatment of AML
Measurement of Response
to Therapy for Acute
Leukemia
Case Study 1
Case Study 2
Case Study 3
oS)Lo —
332. Chapter 16 Introduction to Leukemia and the Acute Leukemias
Introduction
Definition and Overview
Leukemia is a malignant disease of hematopoietic tissue,
characterized by replacement of normal bone marrow ele-
ments with abnormal (neoplastic) blood cells. These Type of Leukemia % of Cases
leukemic cells are frequently (but not always) present in the
peripheral blood and commonly invade reticuloendothelial Acute myelogenous (AML)
Acute lymphocytic (ALL)
tissue, including the spleen, liver, and lymph nodes. They
Chronic myelogenous (CML)
may also invade other tissues, infiltrating any organ of the Chronic lymphocytic (CLL)
body. If left untreated, leukemia eventually causes death.
Leukemia can be a rapidly progressive disease character- Source: Cancer Facts and Figures 2006. American
Cancer Society.
ized by an abnormal expansion of immature cells or blasts
(acute leukemia), or a slowly progressive disorder charac-
terized by an abnormal expansion of mature cells (chronic
leukemia). Acute and chronic leukemias are compared in 60 years of age or older, with the most prevalent incidence in
Table 16-1. the 70- to 90-year-old range! (see Chap. 17).
Leukemia can be divided into two major cell types: Leukemia can strike anyone of either sex. The frequency
myelogenous and lymphocytic. Each has an acute and chronic of all types of leukemia is slightly higher among males
form, which leads to four major types of leukemias: than females, with approximately 57% of new cases of
leukemia diagnosed in males in 2006.'In terms of race and
¢ Acute myeloid leukemia ethnicity, leukemia rates are higher among Americans of
¢ Chronic myeloid leukemia European descent than among African Americans, and the
e Acute lymphoblastic leukemia lowest among American Indians and Alaskan natives.' In the
¢ Chronic lymphocytic leukemia
United States, an estimated 208,080 individuals are living
The overall incidence of leukemia in the United States is 8 to with leukemia. !
10 new cases per 100,000 individuals per year. In 2006, an
estimated 35,070 new cases of leukemia were diagnosed in
the United States.' The percentages of new cases in 2006 for
Comparison of Acute and Chronic
each type of leukemia are estimated in Table 16—2.' Leukemia
Most cases of leukemia occur in older adults, with acute
The clinical and laboratory features of acute and chronic
myeloid leukemia (AML) and chronic lymphocytic leukemia
leukemia differ in a number of respects, as summarized in
(CLL) being the most common types. As the elderly popula-
Table 16—1. Patients with acute leukemia usually present with
tion continues to increase, leukemia will strike many more
a sudden onset of symptoms, and if left untreated, the disease
adults than children. Currently, the ratio of adult cases to child-
runs a rapidly fatal course of 6 months or less. In contrast,
hood cases is 10:1.' More than 50% of all cases of leukemia
patients with chronic leukemia tend to have an insidious
occur after the age of 64.' In children, the most common form
onset and a more indolent clinical course, usually lasting 2 to
of leukemia is acute lymphoblastic leukemia (ALL). Chronic
6 years. Mean survivals in chronic leukemia also increase as
leukemia, whether of lymphoid or myeloid lineage, is gener-
diagnostic sensitivities increase and patients are diagnosed at
ally considered to be a disease of adults. CLL is extremely rare
an earlier stage in their disease.
in children and is unusual before the age of 40 (see Chap. 20).
Patients with acute and chronic leukemia also show dif-
CML increases dramatically among individuals who are
ferences in their hematologic parameters. In general, bone
marrow failure and its sequelae are much more prominent in
the initial presentation of acute leukemia than in that of the
chronic form. In acute leukemia, there is a loss of normal
bone marrow function. Anemia is consistently observed in the
acute leukemic patient, although its severity is variable.
Thrombocytopenia is common. The white blood cell count
Chronic varies; it may be markedly elevated with numerous blasts, or
Adults it may be normal, or in some cases, even decreased. Typically
Clinical onset Insidious the various normal circulating white blood cells are dimin-
Course (untreated) 2-6 yr ished in number, resulting in a neutropenia. In chronic
Leukemic cells Immature Mature leukemic patients, anemia is often mild at presentation. The
Anemia Mild to severe Mild
Thrombocytopenia Mild to severe platelet count is usually normal, but it may actually be
Mild
White blood cell count Variable Increased increased in CML. Chronic leukemias almost always present
Organomegaly Mild Prominent with elevated white blood cell counts, which are often very
high (> 50,000/uL).!
Chapter 16 Introduction to Leukemia and the Acute Leukemias 333
Both acute and chronic leukemia patients can display Bone marrow and stem cell transplantation have improved the
enlargement of the spleen, liver, or lymph nodes, but this is survival of many patients with leukemia, especially those
more consistently seen in patients with chronic leukemia, in with acute forms. The effectiveness of treatment is most
whom organomegaly tends to be more prominent and can notable for children with acute lymphoblastic leukemia
occasionally be massive. (ALL). Before the 1960s, childhood ALL was universally
fatal, but with modern treatment regimens the survival rates
Historical Perspectives have dramatically improved over the past 30 years. The
majority of children with ALL younger than 15 years of age
The initial description of leukemia as a clinical entity was are now cured.' The survival rates, however, are not as
made by John Bennett in Scotland and Rudolf Virchow in impressive for adults with ALL and AML. For example, ALL
Germany, who independently published their findings in patients older than 60 years of age have five year survival
1845. They described a series of autopsy studies of victims of rates below 20%.° Similarly, AML patients older than
a progressive chronic disorder of unknown origin, in which 60 show dismal survival rates in most instances.? In addition,
enlarged spleens and purulent-appearing blood were found. survival rates among adults with leukemia show differences
The blood, when examined microscopically, revealed an according to race. Whites have slightly higher rates than
astounding increase in “coloriess” corpuscles. Bennett ini- many minority groups.'
tially suggested that the marked increase in white blood cells
was the result of an inflammatory process. Virchow chose the
term Weisses Blut (white blood), which was later translated Etiology and Risk Factors
into Greek as leukemia. He proposed that leukemia was
The origin of leukemia at the genetic level appears, in most
caused by a neoplastic proliferation of white blood cells. The
cases, to be related to mutation and altered expression of onco-
ensuing debate between Bennett and Virchow continued for
genes and tumor suppressor genes. Most oncogenes regulate
several years, but eventually even Bennett rejected inflamma-
cell proliferation and differentiation. Abnormal oncogene or
tion as the etiology of leukemia.
tumor suppressor gene expression induced by translocation
Virchow continued his study of leukemia, and defined
and genetic fusion or mutation often results in unregulated cel-
two groups characterized predominantly by either splenic or
lular proliferation. Although the events that lead to this are not
nodal involvement. Today these are recognized as chronic
entirely understood, a number of host and environmental fac-
myelogenous leukemia and chronic lymphocytic leukemia,
tors have been identified that are associated with increased risk
respectively (see Chaps. 17 and 20).
of leukemia transformation. The epidemiologic aspects of
In 1857 Friedreich gave a classic account of a rapidly
acute leukemia have been reviewed* and are summarized in
progressive form of leukemia, to which Epstein applied the
Table 16-3.
term acute in 1889. Further classification was made possible
in 1877 by Paul Ehrlich’s discovery of a tri-acid stain that per-
mitted the morphological characterization of blood cells. Classifications
Examination of the morphology enabled investigators to
show that acute leukemia was associated with immature cells Leukemia is classified according to cell type—with regard to
whereas chronic leukemia was associated with mature, well both cell maturity and cell lineage. Cell maturity is used to dis-
differentiated cells. At the turn of the century, Naegeli tinguish between acute and chronic forms of leukemia. When
described the myeloblast and divided the acute leukemias into the malignant cells are immature (stem cells, blasts, or other
myeloblastic and lymphoblastic forms. A decade later, immature precursors), the leukemia is classified as acute; when
Shilling described a monoblastic (blasts with the morphologic the cells are predominantly mature, it is described as chronic. In
features of immature monocytes) variant of the myeloblastic general these two groups correspond to a rapid (acute) or slow
form. Thus, the main morphological variants of acute and (chronic) natural history. Leukemias are further defined accord-
chronic leukemia were well established by 1930. Since that ing to cell lineage as lymphoid or myeloid. The term myeloid
time classification of leukemia has been refined, and clini- (from myelo, Greek for marrow, and eidos, form) encompasses
cally distinct subgroups have been characterized. granulocytic, monocytic, megakaryocytic, and erythrocytic
Today the clinical laboratory plays an important role in the leukemias. Today, there are two major classifications: the
diagnosis and classification of leukemia. The morphological French-American-British (FAB) and the World Health Organi-
analysis of leukemia is now augmented by cytochemical, cyto- zation (WHO), which are presented later in this chapter.
genetic, immunologic, and molecular techniques. Together The FAB and WHO classification schemes broadly divide
these methods are used to delineate specific categories of acute leukemia into acute lymphoblastic leukemia (ALL) and
leukemia for which distinct treatment protocols are used. acute myeloid leukemia (AML). Alternate names for AML
Certainly the most significant application of the biologic include “acute nonlymphoblastic leukemia (ANLL),” “acute
understanding of leukemia has been the improvement in out- myelogenous leukemia,” “‘acute myeloblastic leukemia,” and
come as a result of continued advances in treatment. Complex “acute granulocytic leukemia.”
therapeutic protocols with multiple cytotoxic drugs and radi- The types of acute leukemias, their common abbrevia-
ation, as well as more “targeted,” less toxic approaches such tions, the FAB classification, and their alternate names are
as tyrosine kinase inhibitors and protease inhibitors are used. listed in Table 16-4.
334 Chapter 16 Introduction to Leukemia and the Acute Leukemias
7 thine ae
taple 16-3 Host and Environmental Factors Associated with an Increased Risk
of Leukemia
eae ae = acuteSlimehe
AML= acute Ferelsbiasticle leukemia; TdT = ne Rope teens transferase;
CNS= central nervous system.
Source: From Kjeldsberg, CR (Ed): Practical Diagnosis of Hematologic Disorders. ASCP Press, Chicago, 1989, p 349,
with permission.
336 Chapter 16 Introduction to Leukemia and the Acute Leukemias
Feature
nucleoli are often present. The typical lymphoblast (Fig. 16—2) important aid in the classification of acute leukemia because
is a smaller cell with scant cytoplasm. The nuclear chromatin they identify cellular components that are associated with
often appears denser than in the myeloblast, and nucleoli are specific cell lines. For example, a positive myeloperoxidase
usually indistinct when present. For a review of morphologi- or Sudan black B stain indicates myeloid differentiation, and
cal descriptions of the blast stage, see Chapter 1. a positive nonspecific esterase stain indicates monocytic dif-
Granulocytic differentiation is suggested by the presence ferentiation. When any of these stains are positive even in a
of azurophilic granules. A very helpful morphological feature is relatively small percentage of blasts, lymphoid origin is ruled
the Auer rod, the presence of which excludes ALL. Auer rods out, with rare exceptions. Thus, the cytochemical stains help
are cytoplasmic inclusions that result from an abnormal fusion distinguish between ALL and AML. They are also used to
of primary granules and are pathognomonic for a myeloprolif- subclassify AML.
erative process, particularly AML. On Romanowsky-stained The cytochemical reactions are performed by applying
smears, Auer rods appear as pink- or purple-staining rods or staining techniques to peripheral blood smears, bone mar-
splinter-shaped inclusions (see Fig. 16-1). They are present in row smears, or touch preparations. Fresh preparations are
up to 60% of patients with AML,” but it may take a long, care- preferred, especially for enzyme reactions. Control smears
ful review of the blood or marrow smear to find them; given can be fixed in the appropriate fixative, allowed to air-dry,
their diagnostic importance, this search is well worth the effort. and stored at —20°C for future staining. Caution should be
In acute promyelocytic leukemia, Auer rods are easy io find, taken when interpreting cytochemical stains. It is the
some cells having “bundles” of cigar-shaped rods and are leukemic cell population whose identity (cell lineage) is in
termed “faggot cells.” question; therefore, a positive reaction is determined by
finding positive staining in the leukemic blasts rather than in
CYTOCHEMISTRY mature cells. For a description of all the cytochemical stains
Special stains are used to identify chemical components of cells including principles of reactions, methods and interpreta-
such as enzymes or lipids. These cytochemical stains are an tions, refer to Chapter 36. The cytochemical reactions that
Figure 16-1 m™ Myeloblast (note the Auer rod). Figure 16-2 ™Lymphoblasts (peripheral blood).
338 Chapter 16 Introduction to Leukemia and the Acute Leukemias
Myeloperoxidase Mainly primary Late myeloblasts, granulocytes; Valuable in that the primary
granules; Auer rods monocytes less intensely granules are not always
visible; separates AML
(+ blasts) from ALL (— blasts)
Sudan black B Phospholipids: Late myeloblasts, granulocytes; Parallels peroxidase, but
sterols, neutral fats monocytes less intensely smears do not need to be
fresh
Specific esterase (Naphthol Cytoplasm Neutrophilic granulocytes; Parallels peroxidase, but
AS-D chloroacetate) mast cells less sensitive; useful on
paraffin-embedded tissues
Nonspecific esterase Cytoplasm Monocytes; focal staining in Useful for determining
Alpha-naphthy! acetate T cells; ANAE also + degree of monocytic
(ANAE) and butyrate in megakaryocytes differentiation; separates
mono (+) from myelo
(—) blasts
Periodic acid—Schiff Glycogen and related Lymphocytes, granulocytes, Helpful in supporting
substances megakaryocytes diagnosis of
erythroleukemia
are useful in the classification of acute leukemia are summa- granulocytic cells and, to a lesser extent, in monocytic lyso-
rized in Table 16-8. somal granules.
The SBB is the most sensitive stain for granulocytic pre-
MYELOPEROXIDASE (MPO) Peroxidase is present in the cursors, with a staining pattern that generally parallels the
primary granules of myeloid cells (Fig. 16-3). These granules myeloperoxidase stain. As with the peroxidase stain, the SBB
first appear in the early promyelocyte (late blast) and persist is used to differentiate AML from ALL. Positivity seldom
through subsequent stages of cell maturation. Monocytes occurs in lymphoid cells; SBB is less specific than MPO. The
have variable staining with peroxidase and are most often SBB stain, whose reactivity does not diminish with time, is
only weakly positive. This enzyme is not present in lympho- particularly useful for specimens that are not fresh.
cytes or their precursors and is, therefore, useful in differenti-
ating AML from ALL. It is more specific for granulocytic SPECIFIC ESTERASE (NAPHTHOL AS-D CHLOROACETATE)
differentiation than the Sudan black B stain. The specific esterase stain (Fig. 16-5), commonly referred to
as chloroacetate esterase (CAE), roughly parallels the perox-
SUDAN BLACK B (SBB) Phospholipids, neutral fats, and idase and SBB stains, although it is not as sensitive. It is
sterols are stained by Sudan black B (SBB) (Fig. 16-4). Phos- negative in eosinophils and monocytes, but positive in neu-
pholipids occur both in primary and secondary granules of trophils, basophils, mast cells and their precursors. Its most
Figure 16-3 ™ Myeloperoxidase positivity in acute promyelocytic Figure 16-4 @ Sudan black B positivity in acute myeloblastic
leukemia. leukemia (AML), M2.
Chapter 16 Introduction to Leukemia and the Acute Leukemias 339
Be ea
Figure 16-6 m™ Nonspecific esterase (alpha-naphthy! butyrate) pos- Figure 16-7 ® Periodic acid-Schiff positivity in acute lymphoblastic
itivity in acute monocytic leukemia (M5). leukemia (ALL). Note the “block” staining pattern.
340 Chapter 16 Introduction to Leukemia and the Acute Leukemias
paragraphs briefly introduce the immunologic cell marker The availability of myeloid-specific monoclonal anti-
procedures. Their utility in the evaluation of acute leukemia is bodies has permitted surface marker analysis of AML. The
discussed in the individual sections on AML and ALL. rationale for this is based on the notion that AML is a clonal
disorder derived from a myeloid stem line, which displays the
CELL SURFACE MARKERS Cell surface markers are pro- surface membrane antigens expressed in normal myeloid dif-
teins on the cell membrane that can be detected using flow ferentiation pathways. A schematic representation of surface
cytometry and immunohistochemistry. Different stages of antigen expression in normal myeloid differentiation is shown
maturation express different proteins. Some proteins are pre- in Figure 16-8.
sent early in development whereas others do not appear until Surface marker analysis has begun to replace conven-
much later. Still other proteins may appear, disappear, and tional cytochemical methods for lineage determination in
then reappear at a later stage of development. This unique acute leukemia. Surface marker studies are used to distin-
expression of proteins enables the diagnostician to use these guish AML from ALL, particularly when the leukemic cells
proteins as markers of both cell lineage and maturation stage. are poorly differentiated or negative and indistinct with cyto-
Although some surface markers can be detected with chemical stains, such as in the case of AML, minimally
polyclonal antisera, many can be detected only with mono- differentiated.
clonal antibodies. Some antibodies that are commonly used to When surface marker analysis is performed to distinguish
evaluate leukemias and other lymphoproliferative and myelo- AML from ALL, it is important to choose a panel of markers
proliferative disorders are listed in Table 16-9. that includes antibodies to several myeloid-associated antigens
od
table 16-9 Monoclonal Antibodies Used for Study of Leukemia _
and Lymphoma 4
A
(CD33) Myo r ae r
(CD13) My7 |
HLA-
DR
Figure 16-8 ® Distribution of myeloid and monocytic surface antigens with bars depicting the strength of antigen reactivity. The wider the
bar, the stronger the reaction, the narrowing indicating a weaker reaction. PMN = polymorphonuclear neutrophil; CFU-GM = colony-forming
unit granulocyte macrophage/monocyte.
(e.g., CD33, CD13, and CD14), as well as B- and T-lymphoid Cytoplasmic markers are very useful in assessing cell
antigens.'' Multiple myeloid lineage-specific markers are nec- lineage in ALL. Cytoplasmic CD3 1s present early in T-cell
essary because no single marker defines all forms of AML. development, and strong expression is seen in most cases of
This topic will be addressed later in the chapter in the section precursor T-cell ALL. Likewise, the presence of cytoplasmic
on “acute leukemias of ambiguous lineage.” CD22 and CD79a aids in defining B-cell lineage in ALL.
The surface phenotypes expressed in cases of AML do not The presence of cytoplasmic IgM heavy chain (\) is a
correlate absolutely with the WHO classification or with spe- defining characteristic of pre-B cells; the earlier precursor
cific genetic alterations, although certain trends exist.° For B cells are cytoplasmic pt negative. Pre-B-cell ALL has tradi-
example, cases of AML with monocytic differentiation may tionally been associated with a worse prognosis than early-
express monocyte-related surface markers (CD14), and cases of pre-B-cell ALL. However, the poor prognosis is now known
acute promyelocytic leukemia tend to express a promyelocytic to be more closely linked to the frequently associated translo-
phenotype (CD13 and CD33 positive, HLA-DR and CD34 neg- cation, t(1;19) in pre-B-cell ALL, rather than to the pre-B-cell
ative). Other AML subgroups exhibit widely variable pheno- phenotype.'* Accordingly, cytoplasmic pu staining is no longer
types. Occasionally, mixed lineage phenotypes are encountered, routinely performed in many laboratories; instead, FISH is
with coexpression of myeloid and lymphoid markers. These now used to identify t(1;19) and other prognostically impor-
biphenotypic or bilineage leukemias may reflect origination of tant cytogenetic abnormalities.
the malignant cell line from an early progenitor capable of both Another helpful cytoplasmic marker used in immunophe-
myeloid and lymphoid differentiation. notyping acute leukemias is cytoplasmic myeloperoxidase.
Flow cytometric analysis requires a cell suspension, typi- This myeloid marker is often used as part of a flow cytometric
cally from bone marrow aspirates or peripheral blood. It is panel, in addition to the traditional myeloid markers, CD13
important to have fresh specimens with viable cclls; nonviable and CD33. Immunohistochemical stains for cytoplasmic
cells lead to nonspecific staining, which may make interpreta-
tion impossible. An immunofluorescent method (direct or
indirect) is used to stain the cells, and a flow cytometer is used
to analyze them. (See Chap. 34 for a review of flow cytometry.)
The surface-staining pattern, as observed with a fluorescent
microscope, that is typical of a strongly positive reaction is
shown in Figure 16-9.
myeloperoxidase can also been performed on formalin-fixed, Philadelphia chromosome t(9;22) associated with CML and
paraffin-embedded sections. However, immunohistochemical the translocation t(15;17) consistently observed in acute
staining with currently available antibodies does not seem to promyelocytic leukemia. The WHO classification for AML
be as specific for myeloid lineage as either the traditional includes six entities with recurrent cytogenetic abnormalities,
cytochemical method or flow cytometry. each with clinical significance. Cytogenetic abnormalities
also provide important prognostic information in ALLs. In
TERMINAL DEOXYNUCLEOTIDYL TRANSFERASE Anti- addition to cytogenetic evaluation, flow cytometric analysis
bodies can also be directed at nuclear antigens using the for DNA ploidy status provides useful prognostic information
same cell membrane permeabilization procedure. Terminal mA
deoxynucleotidyl! transferase (TdT) is a unique nuclear enzyme Cytogenetic analysis may be carried out in different
(DNA polymerase) present in stem cells and precursor B- and ways, including karyotyping, FISH, PCR, or sequence analy-
T-lymphoid cells.'* High levels are found in the majority (90%) sis of particular base pairs. Karyotyping evaluates stained
of the lymphoblastic leukemias, including both B- and T-lineage chromosome metaphase preparations to detect the numeric
ALL. TdT, although not lineage specific, provides information and structural karyotype (from karyon, Greek for nucleus,
useful in the distinction of ALL from AML. TdT is present in plus typos, meaning mark) abnormalities. Normal human
most cases of lymphoblastic lymphoma and in approximately cells have 46 chromosomes and are diploid (diplous, double,
one-third of cases of chronic myeloid leukemia in ALL blast plus eidos, form), having two alleles of each gene. That is,
crisis (CML-BC).'* Detectable levels of this enzyme are also in they have two haploid sets of 23 chromosomes. An aneuploid
5% to 10% of the cases of AML,'* although the level is usually population can be either hypodiploid or hyperdiploid, and can
significantly lower than in lymphoblasts.'* be identified as such by DNA flow cytometry as well as by
TdT is detected using a number of immunologic meth- cytogenetic examination.
ods, including flow cytometry and immunohistochemical Karyotyping is capable of detecting structural rearrange-
methods, that laboratories routinely use. It is important to note ments, including translocations, inversions, deletions, duplica-
that only nuclear staining 1s considered a_ positive tions, and isochromosomes. Current standard karyotyping,
reaction on immunohistochemical stains. TdT can also be however, will fail to detect chromosomal abnormalities in
detected by a variety of other immunologic techniques 40 to 50% of AMLs.'° Other methods, such as FISH, can
(ammunocytochemical or immunofluorescent), using fresh detect very small but significant genetic abnormalities that will
smears, touch preparations, or cytopreparations (Fig. 16—10). not be apparent by standard karyotyping. Some mutations
When heparinized specimens are studied, removing the (such as NPM1 and FLT3-ITD mutations), will look normal by
heparin by washing the sample is essential to reducing false- current standard cytogenetic techniques, including FISH.
negative results. Of particular importance to the study of acute leukemia are
the translocations that result from the movement of a DNA
CYTOGENETICS segment from one chromosome to another and that are often
Cytogenetic analysis of leukemic cells is an essential compo- associated with oncogene rearrangement, genetic fusion, and
nent in the evaluation of the newly diagnosed leukemic abnormal gene expression. Translocations result from a recipro-
patient. It plays a major role in diagnosis, subclassification, cal interchange of portions of two nonhomologous chromo-
prognosis, selection of appropriate therapy, and monitoring somes and are the most common structural chromosomal
the effects of'therapy. In the WHO classification a number of abnormalities. A number of these are associated with distinct
chromosomal abnormalities have been associated with dis- subgroups of AML or ALL and have prognostic significance.
tinct forms of leukemia. Classic examples of this are the For example, patients with acute myelomonocytic leukemia
who exhibit an inversion or deletion of the long arm of chromo-
some 16 (inv[16q] or del[16q]) have a longer median survival
than patients with other types of AML.
Chromosomal abnormalities (both numeric and struc-
tural) are found by routine clinical methods in the majority
of patients with acute leukemia. Importantly, just because
current techniques do not identify a genetic abnormality in
a patient’s acute leukemia does not mean that there is none.
As laboratory techniques improve, the percentages of
genetic abnormalities identified in both AML and ALL
will increase. The recent development of fluorescently
labeled chromosome painting probes and automated differ-
ential color analysis (automated spectral karyotyping)
should greatly enhance the sensitivity and accuracy of
karyotypic analysis. This technique holds great promise
for the identification of additional chromosomal abnormal-
Figure 16-10 ® TdT positivity in ALL using immunofluorescence ities in leukemic patients, who frequently have complex
method. karyotypes.'°
Chapter 16 Introduction to Leukemia and the Acute Leukemias 343
Table 16-10 lists common cytogenetic abnormalities assays, but are valuable at diagnosis and relapse. FISH has
associated with acute leukemia. several advantages, including the ability to use cells in either
metaphase or interphase, the ability to use paraffin-embedded
MOLECULAR GENETICS tissue, and the ability to detect some abnormalities not seen
The molecular genetic basis for many of the acute leukemias by conventional karyotyping.'* FISH and PCR are used
has been elucidated in recent years. Extensive studies of the primarily for confirmation of the presence of a suspected
immunoglobulin genes, T-cell antigen receptor genes, and chromosomal abnormality not detected by conventional cyto-
other genes involved in the many recurrent cytogenetic abnor- genetics, for the analysis of specimens that are not suitable for
malities described in the preceding section have been cloned conventional cytogenetics, for confirmation of relapse, and
and characterized.'* DNA FISH probes and polymerase chain for the monitoring of minimal residual disease following
reaction (PCR)-based primers are available for rapid and pre- therapy.'*
cise detection of many specific cytogenetic abnormalities at These methods permit detection of residual leukemic dis-
the molecular level.'° FISH assays are not as sensitive as PCR ease at extremely low levels, and clinical applications of these
techniques are still being developed. The sensitivity of molecu- M3: Promyelocytic; M3m: promyelocytic microgranu-
lar techniques enables detection of leukemic subpopulations lar variant
that are not identifiable by conventional morphology or other M4: Myelomonocytic; M4Eo: myelomonocytic with
methods; therefore, patients who are in clinical remission may bone marrow eosinophilia
be observed to possess minimal residual disease when PCR- M5: Monocytic (a) poorly differentiated and (b) well
based techniques are employed.'*” The significance and clinical differentiated
applications of minimal residual disease need to be explored
Mo: Erythroid
further, because standard treatment regimens are designed to
address those levels of residual disease traditionally detected by M7: Megakaryoblastic
morphologic means. These groups are defined according to the predominant cell type
observed on Romanowsky and cytochemically stained blood
and bone marrow aspirate smears (Table 16-11). Additional
Acute Myeloid Leukemia specialized studies are required to confirm the diagnosis of MO
FAB Classification of AML (AML without cytologic maturation) and M7 (acute megakary-
ocytic leukemia). A summary of the cytochemical reactions in
In 1976 a group of French, American, and British hematol- each type of AML is found in Table 16-12.
ogists, prompted by the need for uniform nomenclature and
classification of acute leukemia, proposed a morphological AML FAB M0
classification scheme for leukemia.'* This scheme, the MO is AML without morphological or cytochemical matura-
French—American—British Classification, or better known tion (Fig. 16-11). It comprises approximately 5% of the adult
as the FAB Classification, proved to be useful in standard- cases of AML.!? AML MO have primitive leukemic blasts that
izing the morphological classification of both acute show no distinctive myeloid morphological features and lack
myeloid.and lymphoid leukemias. In the years since it was reactivity with the conventional battery of cytochemical
first introduced, the FAB cooperative group has made sev- stains (myeloperoxidase, Sudan black B, NSE). Accordingly,
eral modifications, striving to make the classification as flow cytometry or another form of immunophenotyping is
objective and unambiguous as possible.° Although there are required for the diagnosis. Typically, these undifferentiated
still some areas of ambiguity, the FAB system has gained AMLs exhibit immunologic reactivity for at least one
wide acceptance, and continues to be relevant even after the myeloid lineage-specific antigen (CD33, CD13, CD117) in
adoption of the most recent WHO Classification scheme. the absence of lymphoid antigens (particularly CD3, CD79a,
Importantly, the new WHO includes parts of the FAB Clas- CD22). Generally, undifferentiated AML is negative for
sification system. expression of antigens of myelomonocytic differentiation,
The FAB classification of AMLs are divided into the such as CD14 and CD15.'! The prognosis of MO appears to be
following groups: poor compared to other types of AML.
MO: Myeloid without cytologic maturation
AML FAB M1
M1: Myeloid with minimal maturation M1 is AML with minimal morphologic and cytochemical
M2: Myeloid with maturation maturation (Fig. 16-12). It comprises 15% of the adult cases
Myeloid without cytologic maturation: Morphologically undifferentiated leukemic blasts with myeloid immunophenotype
Myeloid without maturation: Marrow leukemia cells are primarily myeloblasts with no azurophilic granules
Myeloid with maturation: Leukemia cells show prominent maturation beyond myeloblast stage
Promyelocytic: Abnormal, hypergranular promyelocytes dominate; Auer rods easily found; increased incidence of DIC; strong
MPO staining
Microgranular variant of M3: Indistinct granules; rare to occasional Auer rods, nucleus often reniform or bilobed; increased
incidence of DIC; strong MPO staining
Myelomonocytic; Both monocytic (monocytes and promonocytes) and myeloid differentiation (maturation beyond myeloblast
Stage)
M4 with bone marrow eosinophilia: Similar to M4 with marrow eosinophilia (abnormal and immature); associated with
abnormal 16q karyotype
Monocytic, poorly differentiated: Monoblasts predominate, typically with abundant cytoplasm and single distinct nucleoli
Monocytic, well differentiated: Predominantly promonocytes in marrow and more pronounced maturation in blood
Erythroleukemia: Dysplastic erythroblasts with multinucleation, cytoplasmic budding, vacuolation, and megaloblastoid
changes
Megakaryoblastic: Wide range of morphology; cytoplasmic projections sometimes present; electron microscopy or
immunocytochemical stains necessary for diagnosis
Chapter 16 Introduction to Leukemia and the Acute Leukemias 345
AKoL
MPO
SBB
CAE
NSE
AcP
PAS hf /-
CrP “=
“Negative reaction is for erythroblasts only; myeloblasts present will stain positive with MPO and SBB.
‘Positive reaction is with a naphthyl acetate and negative reaction is with alpha naphthyl butyrate nonspecific esterases.
+ =blasts stain positive; — = blasts stain negative; +/— = blasts stain negative or weakly positive (< 3%).
MPO = myeloperoxidase; SBB = Sudan black B; CAE = chloroacetate esterase, NSE = nonspecific esterase; AcP = acid
phosphatase; PAS = periodic acid-Schiff; CrP = cyanide-resistant peroxidase; AML = acute myeloblastic leukemia:
APL = acute promyelocytic leukemia; AMML = acute myelomonocytic leukemia; AMoL = acute monocytic
leukemia; EL = erythroleukemia; MegL = megakaryoblastic leukemia; AEoL = acute eosinophilic leukemia.
«
of AML.’ M1 is characterized by poorly differentiated adult cases of AML and is, therefore, the most common
myeloblasts (type | blasts) representing greater than 90% of type.'’ M2 is characterized by myeloblasts (type | and type 2)
nonerythroid cells, with no evidence of maturation to more and greater than 10% of the immature cells are promyelocytes
mature myeloid cells. The nucleus of these type | myeloblasts or later stages of myeloid maturation. M2 is also character-
typically has loose open lacy chromatin, distinct nucleoli and ized by a specific translocation involving chromosome 8 and
immature cytoplasm without any granules. Fewer than 10% 21; t(8;21) in approximately 15% of the cases of M2 AML.
of the myeloblasts are type 2 blasts which have the same mor- Myeloblasts in M2 will stain positive for MPO, SBB, and
phology as type | but also have <15 azurophilic granules in NCAE cytochemical stains (Fig. 16-15). AML with matura-
the cytoplasm. Auer rods may be present in M1 myeloblasts. tion immunophenotypically resembles AML without matura-
M1 myeloblasts will demonstrate a positive reaction with tion, as evidenced by expression of CD13, CD33, and CD117.
MPO, SBB, and NCAE cytochemical stains, demonstrating In addition to these markers, the blasts may also express
myeloid lineage. Immunophenotypically, at least two myeloid CD19, CD34, and CD56."
antigens are expressed (CD13, CD33, CD117, and cytoplas-
mic MPO), but lymphoid antigens are generally absent.'' The AML FAB M3 AND M3m
course of this leukemia is generally described as aggressive. M3 is acute promyelocytic leukemia (APL; Fig. 16-16). It
comprises approximately 10% of the adult cases of AML." It
AML FAB M2 is characterized by abnormal promyelocytes (>30% of non-
M2 is AML with some maturation to or beyond the promye- erythroid cells) with heavy granulation, sometimes obscuring
locyte stage (Figs. 16-13 and 16-14). It comprises 25% of the the nucleus, and, often, abundant cytoplasm. Auer rods are
Figure 16-11 @ Acute myeloblastic leukemia (AML) without mat- Figure 16-12 AML, M1, peripheral blood.
uration, MO, bone marrow.
346 = Chapter 16 Introduction to Leukemia and the Acute Leukemias
Ss.
Figure 16-15 ® AML with maturation, M2, bone marrow. Figure 16-16 @ Acute promyelocytic leukemia (APL), M3, bone
marrow.
Figure 16-18 ® Acute “microgranular” promyelocytic leukemia, Figure 16-19 ™ Acute myelomonocytic leukemia (AMML), M4,
M3m, peripheral blood. bone marrow.
it for therapeutic considerations. Patients with the microgran- vacuolated is also a feature that is often seen in promono-
ular variant tend to have a higher white blood cell count than cytes. Monocytes predominate in the peripheral blood with an
patients with typical APL and may have a shorter survival.'? absolute count greater than 5 * 10°/L (Fig. 16—20).'? Serum
Morphologically, microgranular APL can be mistaken for and urinary muramidase, a lysozyme, is increased in M4. The
acute myelomonocytic or monocytic leukemia. The leukemic myeloblasts must positively stain for myeloperoxidase or
cells appear monocytoid with prominent nuclear folding and SBB. Generally, monoblasts, promonocytes, and monocytes
abundant cytoplasm. The nucleus of most cells in the periph- will stain positive with NSE.
eral blood is reniform or bilobed. Granulation of these cells is AMML expresses myeloid antigens, as would be
scant or absent, although occasional cells with heavy granula- expected. In addition to these, antigens of monocytic differen-
tion are almost always present. The bone marrow aspirate tiation may also be expressed, including CDI1b, CDl1Ic,
may reveal a morphological pattern that more closely resem- CD14, CD64, and CD4.'!7° It should be noted that CD14,
bles typical APL.'? CD11b, CD13, CD15, CD33, and CD64 may be absent or at
The diagnosis of APL, microgranular variant, can be least partially diminished in M4 and M5." Therefore it is
confirmed with cytochemical studies, including myeloperoxi- important to correlate cytochemistry, morphology, and cyto-
dase or Sudan black stain, which are strongly positive. The genetics with flow cytometric immunophenotyping. Exclud-
NSE reaction is usually negative.'* Cytogenetic studies of ing the cases of inv(16) AMML, the cytogenetic features of
microgranular APL reveal the same abnormal karyotype AMML are diverse and not specific for this diagnosis. AMML
t(15;17) that is found in the hypergranular form. may respond to aggressive therapy, but this is varied. Again,
Immunophenotypic analysis of APL typically shows a AMML has a prognosis similar to that of other AMLs.
characteristic phenotype, featuring CD13 and CD33 antigen
expression with the lack of CD34 and HLA-DR expression. AML FAB M4Eo MA4Eo is AMML associated with bone mar-
Monocytic markers (CD14 and CD15) are generally not row eosinophilia in which abnormal and immature
expressed. Reverse transcriptase (RT) -PCR and FISH meth-
ods are also available for detection and rapid characterization
of this disease.'® The prognosis of this disease is comparable to
that of AML M2.
AML FAB M4
M4 is acute myelomonocytic leukemia (AMML,; Fig. 16-19).
M4 is one of the more commonly diagnosed forms of AML,
representing approximately 20% of the adult cases.'” The
leukemic cells of AMML are characterized by both granulo-
cytic and monocytic differentiation. Morphologically,
monoblasts have round nuclei, prominent nucleoli, and deli-
cate chromatin, surrounded by a basophilic cytoplasm that
may contain fine azurophilic granules. The distinction be-
tween monoblasts and promonocytes is often difficult, but
promonocytes generally have a more convoluted nucleus
and a lower nuclear:cytoplasmic ratio than do monoblasts. A Figure 16-20 ®AMML, M4, peripheral blood.
more granular cytoplasm that is less basophilic and more
348 Chapter 16 Introduction to Leukemia and the Acute Leukemias
ee
Figure 16-21 ™ Acute monocytic leukemia (AMoL), poorly differ- Figure 16-25 @ Gum hypertrophy: A clinical manifestation of
entiated, MSa, peripheral blood. acute leukemia (M5).
eosinophils are present. M4Eo has a longer median survival cells in monoblastic leukemia). In contrast, in acute mono-
than M4 and is associated with an inversion of chromosome cytic leukemia (FAB M5b) the majority of monocytic cells
16. Similar to M4, this type of AML shows myeloid and are promonocytes and monocytes; however, the predominant
monocytic differentiation and stains positive with MPO, cell in the peripheral blood is the promonocyte.
SBB, and NSE. MS5a and M5b leukemia both have distinctive clinical
manifestations associated with the monocyte’s propensity to
AML FAB MS5a AND MS5b migrate to extramedullary sites. Skin and gum involvement are
MSa is acute monocytic leukemia (AmoL), poorly differenti- particularly characteristic (Fig. 16-23). Lymphadenopathy fre-
ated and M5b is AmoL, well differentiated (Figs. 16—21 and quently occurs, and sometimes hepatosplenomegaly is promi-
16-22). Acute monoblastic and acute monocytic are some- nent. CNS involvement also has an increased incidence in these
times used as synonyms for MSa and MS5b leukemias, respec- patients. Serum and urinary muramidase is increased in M5a
tively. M5 leukemias comprise approximately 10% of the and M5b. Typically, monoblasts and promocytes are strongly
adult cases of AML.'? These leukemias are characterized by positive for NSE. Monoblasts are usually myeloperoxidase
an excess of cells of the monocytic cell line. In these cases, and SBB negative. In general, MSa and M5b leukemia follows
cells of monocytic lineage (monoblasts, promonocytes, and an aggressive course. MS usually expresses CD64, CD56,
monocytes) must comprise greater than 80% of the leukemic HLA-DR, CD4, CD13, and CD33.'' The monocytic antigen
cells (not overall marrow cells). In the FAB criteria, CD14 is often absent or frequently diminished in MS5."!
leukemias of the monocytic lineage are subdivided into a and
b based upon the relative proportion of monoblasts and AML FAB M6
promonocytes. In cases of acute monoblastic leukemia (FAB M6 is acute erythroleukemia and is characterized by an
MS5a), monoblasts comprise the majority of monocytic cells abnormal proliferation of erythroid and myeloid precursors
(typically monoblasts are greater than 80% of the monocytic (Figs. 16-24 and 16-25). M6 is uncommon, comprising
Figure 16-22 ™ AMoL, well-differentiated, M5b, peripheral blood. Pigure 16-24 m Erythroleukemia, M6, bone marrow.
Chapter 16 Introduction to Leukemia and the Acute Leukemias 349
BM aspiration ——> Hypocellular BM on films the disease. Molecular techniques such as RT-PCR and FISH
Y are used to determine recurring genetic abnormalities in the
CELLULAR BM BM biopsy leukemic cells.
The WHO classification incorporates five major cate-
gories in its classification of acute myeloid leukemias:
ERYTHROBLASTS % OF ALL NUCLEATED BM CELLS (ANC)
1. Acute myeloid leukemias with recurrent genetic abnor-
malities
<50% Erythroblasts 250% Erythroblasts
2. Acute myeloid leukemia with multilineage dysplasia
3. Acute myeloid leukemia, and myelodysplastic syndromes
(MDS), therapy related (t-AML and t-MDS)
NSE stain, but cases with positive NSE activity have been reaction is usually negative, but can be positive.'* Cytogenetic
reported. In addition to characteristic morphology, acute studies of microgranular APL reveal the same abnormal kary-
promyelocytic leukemia (APL) contains a translocation that otype t(15;17) that is found in the hypergranular form, and
results in the fusion of a transcription factor called PML on rapid FISH analysis for the translocation is often valuable for
chromosome 15 with the alpha («)-retinoic acid receptor gene confirming the diagnosis.'’
(RARa) on chromosome 17, giving rise to one of the most Due to the hemorrhagic complications of DIC associated
striking instances of genotype-phenotype correlation in with APL, and the opportunity for a directed therapeutic inter-
pathology: the hybrid gene, PML-RARa.”> Because wild-type vention, namely ATRA, the recognition and characterization of
RARa acts as a transcriptional activator, when translocated, it this disease, when confronted with a new diagnosis of acute
acts as a repressor of transcription leading to a maturation leukemia, is of paramount importance. The wise diagnostician
arrest at the promyelocyte stage.’ In the normal state, RARa will consider APL whenever monoblastic or myelomonoblastic
interacts with transcriptional co-repressors and, the levels of leukemia enters the differential. Immunophenotypic analysis
retinoic acid constitutively present are sufficient for overcom- typically shows a characteristic phenotype, featuring variable
ing this repression. However, the fusion of RARa to PML CD13 and strong CD33 antigen expression with the lack
leads to the formation of a co-repressor complex that is of CD34 and HLA-DR expression. Lymphoid markers such as
markedly enhanced.** The formation of this enhanced CD2 and CD9 may be expressed as well. Monocytic markers
complex is involved in the oncogenesis of APL through and markers of maturity (CD15) are generally not expressed."!
mechanisms that are not entirely understood, but high doses The prognosis of this disease is comparable to that of
of all-trans-retinoic acid (ATRA) are effective as a differenti- AML t(8;21) or AML inv(16). Deaths in APL are typically
ating agent in APL.** ATRA in combination with an anthracy- early and due to hemorrhagic complications. Variant translo-
cline forms the basis for therapy. cations, including t(11;17) and t(5;17), may occur in cases
APL often presents with the signs and symptoms of that are morphologically and immunologically similar to APL
DIC. The majority (80%) of patients with APL present with with the classic t(15;17). Although these translocations also
hemorrhagic manifestations, including petechiae, small involve the RARa gene, t(11;17) is resistant to ATRA.'”
ecchymoses, hematuria, and bleeding from venipuncture and
bone marrow sites.'* The abnormal promyelocytes are rich in AML WITH 11q23 (MLL) ABNORMALITIES The mixed lin-
thromboplastic substances that, if released, trigger DIC. The eage leukemia or myeloid lymphoid leukemia (MLL) gene is
most consistent coagulation abnormalities include a pro- involved in at least 80 different translocations, many of which
longed prothrombin time and thrombin time, elevated fibrin are seen in AML, ALL, and MDS.* In AML, MLL gene
degradation products, and decreased amounts of plasma fib- translocations are associated with either AML with monoblas-
rinogen. Thrombocytopenia, which tends to be more severe tic differentiation or AML associated with previous topoiso-
than in other types of AML, is almost universally present. merase II inhibitor therapy.** Functionally, MLL gene appears
Schistocytes are sometimes evident on the peripheral blood to act to modulate the expression of HOX genes, but the pre-
smear. cise mechanism is not clear. MLL gene translocations typi-
A second form of APL is the microgranular variant cally occur in children or younger adults.* Clinically, patients
(known as M3m in the FAB classification).'* The leukemic may present with extramedullary monocytic sarcoma or
cells of the microgranular APL have primary granules that are DIC.* A global approach to the detection of MLL gene abnor-
not readily visible on Romanowsky-stained smears. These malities is prudent, given the heterogeneity of abnormalities
granules can, however, be demonstrated with strongly posi- that are associated with this gene. Southern blot, RT-PCR,
tive myeloperoxidase staining (in contrast to the negative conventional cytogenetic studies, and FISH analysis all have
staining typical for monoblasts and promonocytes), as well as a role in the characterization of AMLs with such abnormali-
by Sudan black staining.'? The disease also has a high inci- ties. Typically, MLL gene abnormalities denote a poor
dence of DIC, and it is, therefore, important to recognize it for outcome.**
therapeutic considerations. Patients with the microgranular It should be noted that the WHO maintains that a diag-
variant tend to have a higher white blood cell count than nosis of AML can be made in patients with a blast count of
patients with typical APL and may have a shorter survival.'* less than 20% and a clonal recurring genetic abnormalities of
Morphologically, microgranular APL can be mistaken t(8;22), inv(16), t(16;16), and t(15;17).° Notwithstanding, the
for acute myelomonocytic or monocytic leukemia. The WHO also emphasizes that the diagnosis of acute leukemia is
leukemic cells appear monocytoid with prominent nuclear not synonymous with a decision to treat as an acute
folding and abundant cytoplasm. The nucleus of most cells in leukemia.° Many clinical factors should be considered before
the peripheral blood is reniform or bilobed. Granulation of treating a patient with chemotherapy.
these cells is scant or absent, although occasional cells with
heavy granulation are almost always present. The bone mar- AML WITH MULTILINEAGE DYSPLASIA
row aspirate may reveal a morphological pattern that more AML with multilineage dysplasia is an acute leukemia with
closely resembles typical APL."° greater than 20% blasts and dysplasia in two or more cell
The diagnosis of microgranular APL can be confirmed lines.° This dysplasia must be significant, meaning that it must
with cytochemical studies, including myeloperoxidase or be present in greater than 50% of the cells in at least two lines,
and it must be in a pretreatment specimen.° Typically, this is a
Sudan black stain, which are strongly positive. The NSE
352 Chapter 16 Introduction to Leukemia and the Acute Leukemias
disease that occurs in elderly patients in a setting of MDS, battery of cytochemical stains (myeloperoxidase, Sudan black
although it may also occur de novo. Most often patients pre- B, NSE). Accordingly, flow cytometry or another form of
sent clinically with severe pancytopenia. This is a morpholog- immunophenotyping is required for the diagnosis. Typically,
ically heterogeneous disease, and these features are reflected these minimally differentiated AMLs exhibit immunologic
in the immunophenotype, which is highly variable, often reactivity for at least one myeloid lineage-specific antigen
showing aberrant and promiscuous antigen expression. Cyto- (CD33, CD13, CD117) in the absence of lymphoid antigens,
genetic alterations are often complex, but may involve the particularly cCD3, cCD79a, cCD22.'' Generally, minimally
loss of segments of chromosomes 5q and 7q. The prognosis is differentiated AML is negative for expression of antigens of
not favorable."’ myelomonocytic differentiation, such as CD14 and CD15."
Rarely, blasts may also express a lymphoid antigen, but the
AMLs AND MYELODYSPLASTIC SYNDROMES, expression is dimmer than that seen in lymphoid malignan-
THERAPY RELATED cies and there should be more myeloid antigens expressed. In
AML and myelodysplastic syndromes may arise as a result of the differential diagnosis for minimally differentiated AML is
chemotherapy or radiation. Presentation as a myelodysplastic ALL, acute megakaryoblastic leukemia, and acute leukemia
syndrome, presenting as a cytopenia or pancytopenia at some of ambiguous lineage. The prognosis appears to be poor com-
time after treatment, is more common than presenting with pared to other types of AML.°
outright AML. In some cases, the myelodysplastic syndrome
develops into an acute leukemia, but in other cases mortality ACUTE MYELOBLASTIC LEUKEMIA WITH MINIMAL
is due to the long term effects of MDS.” MATURATION Known as AMLM1 under the FAB classifica-
The WHO recognizes two major subtypes of therapy- tion, acute myeloblastic leukemia with minimal maturation is
related AMLs: (1) alkylating agent/radiation related, and composed of predominantly poorly differentiated myeloblasts,
(2) topoisomerase II inhibitor related.© The development of typically representing greater than 90% of nonerythroid cells.**
leukemiais more rapid after topoisomerase II inhibitor therapy, Importantly, myeloblasts predominate and there should be no
with a reported mean of about 33 to 34 months after beginning significant evidence of maturation to more mature myeloid
chemotherapy, compared with about 5 to 6 years after alkylating cells such as neutrophils. The nucleus of the myeloblast typi-
agent/radiation.® As is the case with AML with multilineage cally has a fine, lacy chromatin pattern and distinct nucleoli.'?
dysplasia, immunophenotypic analysis of the increased blast The quantity of cytoplasm is usually moderate, and azurophilic
population in therapy-related AML and MDS shows no specific granules or Auer rods may be seen, but there are fewer than
pattern, rather, the immunophenotype is heterogeneous.° In 10% of the blasts with azurophilic granules. The myeloperoxi-
addition to pan-myeloid antigen expression, the blasts may dase or Sudan black reactions must demonstrate at least 3%
aberrantly express CD56 and CD7."’ positivity in the blast population to document myeloid differen-
The rationale behind the further subdivision of therapy tiation.** Immunophenotypically, at least two myelomonocytic
related AML and MDS is based on distinct cytogenetic and antigens are expressed (CD13, CD33, CD117, and MPO), but
prognostic differences between the two main groups. Therapy- lymphoid antigens are generally absent. The course of this
related AML and MDS related to alkylating agents/radiation leukemia is generally described as aggressive.!!
therapy typically surface 5 to 6 years after therapy and show
clonal evolution from MDS, frequently harboring deletions or ACUTE MYELOBLASTIC LEUKEMIA WITH MATURATION
translocations involving 5q and 7q, in addition to other chro- Morphologically synonymous with the FAB M2, cases of AML
mosomal abnormalities.'’ These leukemias are generally asso- with maturation show a marrow infiltrate with greater than
ciated with a very poor outcome and short survival time. 20% blasts and evidence of maturation to or beyond the
In contrast, AML arising from topoisomerase inhibitor promyelocyte stage.** There should be more than 10% of cells
treatment have a shorter time to onset after treatment, typically at different stages of granulocyte maturation, and the percent-
about 3 years. Second, these therapy related AMLs have differ- age of maturing myeloid cells is typically higher than 10%.”* At
ent cytogenetic abnormalities, the most common of which is a least 3% of the leukemic cells are myeloperoxidase or Sudan
translocation involving 11q23 (the MLL gene).*° Other reported black positive, and this percentage is usually also much
genetic abnormalities in AML after topoisomerase treatment higher.** Monocytes comprise less than 20% of the bone mar-
include t(8;21), inv(16) and even APLs with t(15;17).° Lastly, row; thus NSE activity must not exceed 20% of marrow cells.°
it may be that these patients have a better outcome than those AML with maturation immunophenotypically resembles
with alkylating agent/radiation associated MDS or AML. AML without maturation, as evidenced by expression of CD13,
CD33, and CD117."' In addition to these markers, the blasts
ACUTE MYELOID LEUKEMIA NOT OTHERWISE may also express CD15, CD34, and HLA-DR."'! As described
CATEGORIZED in the preceding text, many cases of AML with these features
ACUTE MYELOBLASTIC LEUKEMIA WITHOUT harbor the t(8;21) and are best classified under AML with
MATURATION Also known as FAB MO, these cases of acute recurrent genetic abnormalities.'’ In the remaining cases, other
myeloid leukemia show no definitive myeloid differentiation translocations including t(6;9) and t(8;16) have been described.
by conventional morphological and cytochemical analysis. but have yet earned specific classifications as recurrent genetic
These AMLs have primitive leukemic blasts that show no dis- abnormalities under the 2001 WHO classification scheme.
tinctive myeloid morphological features and lack reactivity Survival rates are variable, and evidence shows that AML with
(less than 3% of blasts staining positive) with the conventional maturation responds to aggressive therapy.
Chapter 16 Introduction to Leukemia and the Acute Leukemias 353
ACUTE MYELOMONOCYTIC LEUKEMIA Acute myelo- promonocytes are weakly to mildly positive, although not
monocytic leukemia (AMML), M4 in the FAB system, is one uniformly.
of the more commonly diagnosed forms of AML. The leukemic The immunophenotype of these leukemias expresses
cells of AMML are characterized by both neutrophilic and myeloid antigens (CD13, CD33, and CD117) and also expresses
monocytic differentiation. By WHO criteria, not only must markers of monocytic differentiation (CD14, CD64, CD4, and
blasts be greater than or exceed 20%, but neutrophils and pre- lysozyme).** Often, CD34 is not expressed on these cells. While
cursors, as well as monocytes and precursors, must each some differential expression of myeloperoxidase may be seen
comprise equal to or greater than 20% of the bone marrow cel- by flow cytometry (i.e., myeloperoxidase antigen may be seen
lularity.° This criteria of at least 20% monocytes and monocytic in acute monocytic leukemia rather than acute monoblastic
precursors is to separate out cases of AML with or without mat- leukemia), these findings alone are not able to discern specific
uration that have occasional, but not significant, numbers of differences between these two subtypes of leukemia.
monocytes.** Morphologically, monoblasts have round nuclei, Aside from cases of acute monocytic leukemia that fall
prominent nucleoli, and delicate chromatin, surrounded by a under the AML with recurrent genetic abnormalities section,
basophilic cytoplasm that may contain fine azurophilic gran- there are few specific cytogenetic findings that would be
ules. The distinction between monoblasts and promonocytes is suggestive of acute monoblastic or monocytic leukemia. One
often difficult, but promonocytes generally have a more convo- of these is associated with erythrophagocytosis and coagu-
luted nucleus and a lower nuclear:cytoplasmic ratio than do lopathy and is characterized by a t(8;16)(p11;p13) abnormal-
monoblasts; a more granular cytoplasm that is less basophilic ity. In general, acute monoblastic and monocytic leukemia
and more vacuolated than monoblasts. At least 3% of the follows an aggressive course.
myeloid blasts must positively stain for myeloperoxidase. Gen-
erally, monoblasts, promonocytes, and monocytes will stain ACUTE ERYTHROID LEUKEMIA Acute erythroid leukemia,
positively with NSE. which is equivalent to FAB M6, is characterized by an abnor-
AMM expresses myeloid antigens, as would be expected. mal proliferation of erythroid precursors and, generally, an
In addition to these, antigens of monocytic differentiation may aggressive course. Two types are generally recognized. One
also be expressed, including CD11b, CD11c, CD14, CD64 and type of erythroid leukemia is characterized by the neoplastic
CD4.* Excluding the cases of inv(16) AMML that are classified population purely composed of erythroid precursors (pure ery-
as AML with recurrent genetic abnormalities, the cytogenetic throid leukemia, FAB M6b) and immature erythroid precursors
features of AMML are diverse and not specific for this diagno- should be greater than 80% of the cellularity with no significant
sis.°° AMML may respond to aggressive therapy, but this myeloblastic component.® Pure erythroid leukemia has previ-
is varied. Again, AMML has a prognosis similar to that of ously been referred to as DiGuglielmo disease or true
other AMLs. erythroleukemia. The other type of erythroid leukemia has ery-
throid precursors and myeloblasts (erythroleukemia M6a),
ACUTE MONOBLASTIC LEUKEMIA AND ACUTE although with a predominance (greater than or equal to 50%) of
MONOCYTIC LEUKEMIA Synonymous with FAB M5a erythroid precursors. ° In the erythroleukemia type, myeloblasts
and M5b, acute monoblastic and acute monocytic leukemias must be greater than 20% of the nonerythroid population.
are leukemias characterized by a preponderance of cells of Patients with either type of erythroid leukemia have hypercel-
monocytic lineage. In these cases, cells of monocytic lin- lular bone marrows with marked erythroid hyperplasia associ-
eage (monoblasts, promonocytes, and monocytes) must ated with abnormal erythroid forms.° Megaloblastoid changes
comprise greater than 80% of the leukemic cells (not overall in the erythroblasts along with other dysplastic features, includ-
marrow cells). 7* Like the FAB criteria, the WHO further ing bizarre multinucleation, markedly vacuolated cytoplasm of
subdivides leukemias of monocytic lineage based on the rel- erythroblasts, and cytoplasmic budding, are common."* Cyto-
ative proportion of monoblasts and promonocytes. In cases plasmic PAS-positive staining in the neoplastic erythroblasts is
of acute monoblastic leukemia (FAB M35a), monoblasts consistent with the diagnosis of erythroleukemia but is not spe-
comprise the majority of monocytic cells (typically cific; and negative staining does not rule it out.° Myeloblasts in
monoblasts are greater than 80% of the monocytic cells in erythroleukemia show myeloid differentiation and they may
monoblastic leukemia).*? In contrast, in acute monocytic have Auer rods. Abnormal megakaryocytes may also be present.
leukemia (FAB MSb) the majority of monocytic cells are In the differential diagnosis of erythroleukemia is a
promonocytes. myelodysplastic syndrome with excess blasts, as both dis-
Acute monoblastic and monocytic leukemia both have dis- eases have dysplastic erythroid precursors and increased
tinctive clinical manifestations associated with the monocyte’s numbers of blasts (see Chap. 19). One may appreciate the
propensity to migrate to extramedullary sites. Skin and gum overlap between these two diagnoses when considering that
involvement are particularly characteristic. Lymphadenopathy the diagnosis of erythroleukemia can be made according to
frequently occurs, and sometimes the spleen and liver are the criteria of erythroid precursors which are greater than
markedly enlarged. CNS involvement also has an increased 50% of all nucleated cells in the BM and myeloid blasts
incidence in these patients.’ which are more than 20% of the nonerythroid marrow cells.°
The morphologic features of the constituent cells have A diagnosis of MDS may be considered with a blast percent-
been described earlier in the section on AMML. Typically, age significantly less than 20% of NEC.°
monoblasts and promocytes are strongly positive for NSE." Also in the differential with erythroleukemia are
Monoblasts are usually not myeloperoxidase positive, whereas vitamin B,, and folate deficiency. Pure erythroid leukemia
354 Chapter 16 Introduction to Leukemia and the Acute Leukemias
should not correct following vitamin B,,/folate therapy. Also, spontaneously regress without chemotherapy, only to recur in
pure erythroid leukemia will exhibit dysplastic changes in the some cases. Care should, therefore, be taken when assessing
erythroid line that far exceeds those typically seen in vitamin increased blasts in Down neonates. Importantly, not every infant
B,,/folate deficiency, although the latter may at times be strik- with increased megakaryoblasts will undergo spontaneous
ing as well. In children, caution must be taken when the diag- remission, and some neonates, as well as infants and children, are
nosis of pure erythroid leukemia is considered owing to the diagnosed with megakaryoblastic leukemia. In this younger pop-
possibility of congenital dyserythropoietic anemia. The diag- ulation, the translocation, t(1;22)(p13;q13) found in acute
nosis of erythroleukemia should be made only with consulta- megakaryoblastic leukemia is associated with a poor prognosis.’
tion of the clinical history and setting, including a history of
MDS and folate and B,, levels.° ACUTE BASOPHILIC LEUKEMIA In this very rare
The erythroblasts of pure erythroleukemia generally lack leukemia, the differentiation is primarily toward the
myeloperoxidase and do not express myeloid antigens. basophilic lineage. Care should be taken to ensure the
Depending on how immature the erythroid blasts are, they absence of the Philadelphia chromosome, which is the hall-
may or may not express glycophorin A.** Other antigens such mark of chronic myelogenous leukemia (CML), because
as CD41 and CD61, which are typically megakaryocytic anti- CML in blast phase may be indistinguishable. The blasts
gens, may be variably expressed.'' Indeed, distinguishing ery- will generally have a moderately basophilic cytoplasm and
throblasts from megakaryoblasts can be difficult. coarse basophilic cytoplasmic granules similar to those
seen in basophils. Numerous immature basophils may also
ACUTE MEGAKARYOBLASTIC LEUKEMIA (M7) Acute be present, as in CML. Mature basophils are often few in
megakaryoblastic leukemia, which is equivalent to FAB M7, number. Cytochemically, acute basophilic leukemia shows
is a relatively uncommon form of leukemia characterized by metachromatic staining with toluidine blue, but are
neoplastic proliferation of megakaryoblasts and atypical myeloperoxidase negative. The blasts express myeloid
megakaryocytes. Recognition of this entity was aided by the markers and also express CD9.** No specific cytogenetic
use of platelet peroxidase (PPO) ultrastructural studies. PPO, abnormality has been found. This is a very rare leukemia;
which is distinct from myeloperoxidase, is specific for the what information exists about the outcome of patients sug-
megakaryocytic cell line.?! Acute megakaryoblastic leukemia gests a poor prognosis.
is defined as an acute leukemia in which greater than or equal
to 50% of the blasts are of megakaryocytic lineage.° ACUTE PANMYELOSIS WITH MYELOFIBROSIS This, too,
The blasts observed in acute megakaryoblastic leukemia is a rare form of acute leukemia representing 1% to 2% of all
display a wide range of morphology, from small cells with cases.° It is characterized by an acute panmyeloid proliferation
scant cytoplasm and dense chromatin, to large cells with a in the setting of bone marrow fibrosis. This may be seen de novo
moderate amount of cytoplasm and a fine reticulated chromatin or following treatment with alkylating agents.** Clinically,
pattern.“ Cytoplasmic projections or blebs are sometimes pre- severe pancytopenia is present without splenomegaly, corre-
sent, and in some cases azurophilic granules resembling early sponding to the relatively acute onset. The evolution of this dis-
granular megakaryocytes can be seen. The presence of ease process is typically rapid. The peripheral blood shows an
megakaryocytic fragments in the peripheral blood is also sug- erythroleukoblastic picture with dysplastic features and a promi-
gestive of acute megakaryoblastic leukemia. nent megakaryoblastic/megakaryocytic population.° The bone
Conventional cytochemistry can suggest the diagnosis of marrow biopsy is hypercellular, with all lineages showing
acute megakaryoblastic leukemia, but is not definitive.'* The varying amounts of hypercellularity. Bone marrow fibrosis is
myeloperoxidase and Sudan black reactions are negative, prominent.°
whereas acid phosphatase and PAS are usually positive.'* Non- AML with multilineage dysplasia with fibrosis and acute
specific esterase shows dot-like positivity with a-naphthy] megakaryoblastic leukemia are in the differential diagnosis, and
acetate.” distinguishing between these may be difficult. Blast percentages
Electron microscopy for platelet peroxidase may be per- and immunophenotype may help, and in acute panmyelosis with
formed, but the diagnosis of acute megakaryoblastic leukemia myelofibrosis, all three cell lines are hyperplastic. The WHO
is usually made by using an immunophenotypic demonstra- committee recommends that if the acute leukemia is predomi-
tion of platelet-specific antigens. Megakaryoblasts will gener- nantly megakaryoblastic with myelofibrosis, it be classified as
ally express at least one of the platelet associated antigens “acute megakaryoblastic leukemia with myelofibrosis”; and if
CD41, CD42b, and CD61. CD36 and factor VIII antigen the blasts represent a panmyelopathy with myelofibrosis present
expression are typical. In addition to this, the megakary- in the BM, the leukemia should be classified as ‘acute pan-
oblasts may or may not express the myeloid markers CD13 myelosis with myelofibrosis.’° The prognosis for all three enti-
and CD33, or CD34 and CD45.” It is important to rule out ties in the differential is poor.**
lymphoid differentiation.
Acute megakaryoblastic leukemia may arise in the context MYELOID SARCOMA Myeloid sarcoma is an extramedullary
of a myelodysplastic syndrome (MDS), myeloproliferative dis- (outside the bone marrow) tumor mass of myeloid blasts or
ease (MPD), or de novo. Increased megakaryoblasts are also immature myeloid precursors. Myeloid sarcoma may occur in
associated with a transient disorder in neonates with Down many sites in the body, including lymph nodes, skin, sinuses,
syndrome. The increased blasts in Down neonates will often brain, and below the periosteum of bones. Myeloid sarcoma is
Chapter 16 Introduction to Leukemia and the Acute Leukemias = 355
also known as extramedullary myeloid tumor, granulocytic sar- unclassified.''! Typical undifferentiated leukemias show an
coma, chloroma, or if in the skin, leukemia cutis.° This myeloid immunophenotype positive for HLA-DR and CD34, but with
tumor may precede or occur concomitantly with presentation or no lineage-specific markers.** Truly biphenotypic leukemias
signal relapse of AML, MDS, chronic myelogenous leukemia, may also show changes in antigen expression over time and
or other chronic myeloproliferative disease (MPD). If it appear to switch lineages. In adults, most of these leukemias
occurs in the setting of MDS or MPD, it may signal transfor- are treated as AML if the morphology and cytochemistry sug-
mation to blast phase. gest undifferentiated leukemia.”
Three types of myeloid sarcoma are generally recognized
and are based on the degree of maturation: blastic, immature,
and differentiated.’ As their names suggest, blastic myeloid Acute Lymphoblastic Leukemia
sarcoma consists of myeloblasts, while immature myeloid Classification of ALL and Introduction
sarcoma is composed of myeloblasts and promyelocytes, and
differentiated myeloid sarcoma shows maturation to neu- ALL is mostly a disease of children, and treatment has
trophils. Also common is a monocytic form, often seen in skin improved remarkably. About 75% of ALL occur in children
and mucosal surfaces. The genetic abnormalities seen in and most ALL are precursor B cell ALL.*’ Historically, ALL
myeloid sarcoma will typically mirror those of the underlying was universally fatal, but today about 80% of newly diag-
AML or MDS. nosed children with ALL may be cured.** Fewer cases of ALL
Importantly, there is a difference between extramedullary are seen in adults and treatment is significantly less effective.
hematopoiesis, which is typically seen in the liver, spleen, and Today, the classification of ALL relies on immunopheno-
other areas of fetal hematopoiesis, and myeloid sarcoma. The for- typic, molecular, and cytogenetic characteristics of the clonal
mer 1s largely a compensatory mechanism and does not evidence blast population in addition to the morphologic features of
transformation to the blast phase. In contrast, a myeloid sarcoma the blasts. The WHO classification scheme emphasizes
represents uncontrolled spread of a neoplastic proliferation.® immunophenotype and genetics, and does not directly rely on
the FAB morphologic classifications. The focus of the currently
ACUTE LEUKEMIAS OF AMBIGUOUS LINEAGE accepted WHO is on whether the lymphoblastic leukemia is a
The advent of immunophenotyping, molecular, and genetic precursor B or precursor T cell lineage, and whether cytoge-
techniques has resulted in the understanding that some acute netic findings indicate a favorable or unfavorable prognosis.
leukemias lack clear lineage, but show characteristics of both These characteristics, not the morphologic classification of the
myeloid and lymphoid, or both B and T lymphoid lineages.° For lymphoblasts, guide therapeutic decisions. Nonetheless, an
example, lymphoid and myeloid antigens may be coexpressed appreciation of the features of FAB morphologic classification
by a single population of leukemic cells (biphenotypic); there is worthwhile, in part to allow one to help differentiate between
may be biphasic populations with distinct lineage-specific phe- myeloblasts, lymphoblasts and reactive lymphocytes.
notypes (bilineal); or there may be the lack of such markers spe- To fully appreciate the classification of ALL and other
cific for a given lineage (undifferentiated). More importantly, lymphoproliferative disorders (e.g., CLL, lymphoma, and
ambiguous lineage leukemias should be distinguished from multiple myeloma), it is important to understand lymphocyte
AML with aberrant lymphoid antigen expression, or ALL with ontogeny. Lymphoid leukemias and lymphomas are a clonal
one or two myeloid antigens. The hallmark of these rare proliferation of lymphoid cells that have been “frozen” at a
leukemias of ambiguous lineage is that the overall findings are given stage of maturation, typically retaining some features of
insufficient to classify as myeloid or lymphoid (undifferenti- their normally differentiated counterparts. In ALL, the malig-
ated) or the antigen expression is truly ambiguous and shows nant clone has an immunophenotype with features of an early
significant coexpression across lineages.° lymphocyte or “lymphoblast,” typically expressing TdT and
Acute leukemias of ambiguous lineage generally either B or T lymphocyte antigens.
have a poor prognosis, especially in adults. The most
important lineage-specific antigens are cytoplasmic CD79a Review of Lymphocyte Ontogeny
and cytoplasmic CD22 for B-lymphoid lineages, membrane
or cytoplasmic CD3 for T-lymphoid lineages, and myeloper- Lymphocytes originate from pluripotent stem cells that are
oxidase (MPO) for myeloid lineages.** Also important present in the yolk sac, fetal liver, spleen, and bone marrow.
but less specific are CD19 and CD20 for B lymphoid, CD2 At birth and into adulthood, the stem cells are normally found
and CD5 for T lymphoid, CD10 for lymphoid generally, only in the bone marrow, where they respond to specific
and CD13 and CD33 for myeloid lineage.*’ A scoring growth factors (hormone-like substances) that trigger their
system for acute leukemias of ambiguous lineage has been commitment toward B- or T-lymphocyte differentiation. The
proposed by the European Group for the Immunologic microenvironment of these developing cells plays a critical
Classification of Leukemia (EGIL) and is reproduced in role in their maturation: B cells develop in the bone marrow
the WHO classification.2? This scheme assigns different (bursa-equivalent tissue), whereas T cells develop in the thy-
degrees of specificity for various antigens expressed by a mus (from committed stem cells that have migrated there
given leukemia. from the marrow). Lymphocyte maturation in these organs is
With the routine use of flow cytometric immunopheno- antigen independent. After the lymphocytes have matured
typing of acute leukemia, fewer than 4% of cases remain they migrate to the peripheral lymphoid organs, including the
356 Chapter 16 Introduction to Leukemia and the Acute Leukemias
lymph nodes, spleen, and other lymphoid tissues. In these composed of four minigene families including the variable
organs the lymphocytes remain in a resting state until they are (VH), diversity (DH), joining (JH), and constant (CH)
stimulated to undergo antigen-dependent development. regions. The CH region has separate DNA sequences that
encode for the different Ig isotypes including mu (yw), delta
B-LYMPHOCYTE DEVELOPMENT (8), gamma (y), alpha (a), and epsilon (€). The V, D, and
Early B-cell maturation (antigen independent) is divided into J regions are the first to undergo rearrangement, forming a
three stages: early pre-B cell, pre-B cell, and mature B cell VDJ complex. In this process, intervening sequences
(Fig. 16-28). These stages are identified by their expression of (introns) are excised and the V, D, and J regions are spliced
TdT, surface markers (HLA-DR, CD10 [CALLA], CD19, together. Messenger RNA is transcribed from this VDJ
CD20), and immunoglobulin (cytoplasmic or surface Ig).*” The complex along with DNA sequences downstream from it,
early pre-B cell is TdT positive and expresses HLA-DR, CD19, including an intron and the constant u (Cu) region. The
and usually CALLA (CD10). HLA-DR, a histocompatibility- mRNA itself is then spliced to bring the VDJ complex
related antigen, is expressed first, followed by CD19 and then adjacent to the Cu region, creating a template for cytoplasmic
CD10. CD19 is the most sensitive and specific surface marker u-heavy chain synthesis. This process is closely followed by
for early B cells.'' During this stage the immunoglobulin genes a similar rearrangement of the k gene (on chromosome 2),
begin to undergo structural re-arrangement followed by the pro- which, if unsuccessful, is in turn followed by rearrangement
duction of cytoplasmic mu (Cu)-heavy chain.” The presence of of the \ gene (on chromosome 22). As B-cell development
cytoplasmic ul distinguishes the pre-B cell from its predecessor, continues, the heavy chain may undergo additional rearrange-
which otherwise has a similar phenotype. As the cell continues ments in the CH region, initiating an isotype switch from i to
to mature, immunoglobulin light chains are produced, and [gM d. Both IgM and IgD are expressed on the surface of the
is assembled and inserted into the plasma membrane. This sur- majority of mature B cells (Fig. 16-29).
face Ig (slg) is the hallmark of the mature B cell, which no After maturation in the bone marrow, B cells circulate
longer expresses TdT. Each B cell expresses only one type of Ig through the blood to the peripheral lymphoid organs, where
light chain (kappa [k] or lambda [A]). This feature is extremely they remain in a resting state until stimulated by specific anti-
helpful in identifying monoclonal proliferations of mature gens to undergo further development. Activated B cells
B cells, because a normal population of B cells will consist of a undergo clonal expansion, producing daughter cells that
mix of k and A light chain expressing B cells.'* If all the B cells retain the same antibody idiotype (antigen-binding region).
in a population express, for example, k, the population is likely Some daughter cells become memory cells and regain the
clonal, because polyclonal B cells would be a mix of some small mature B-cell morphology and phenotype, whereas oth-
B cells expressing \ along with others expressing k. ers continue development toward a short-lived antibody-
Immunoglobulin genes are rearranged in a unique secreting cell—the plasma cell. During this final development,
process that is normally limited to cells committed to B-cell the Ig heavy chain may undergo another isotype switch, to
differentiation. The Ig genes are composed of discontinu- IgG, IgA, or IgE. The plasma cell produces large quantities of
ous segments of minigene families that, when productively Ig and is characterized by a high concentration of cytoplasmic
rearranged, encode for the heavy chain and the k or A light Ig. It does not express SIg, CD19, nor CD20, although other
chains. The heavy chain gene (on chromosome 14) is antigens are sometimes expressed (e.g., PCA-1, PC-1).!!
~ A
THeL° y
Ny f ay
Antigen & : gy x i
Stem cell DR helper fact
perfactors \ (aa yo
DR pig DR cpig ty
nla
CD19
\CD20
Plasma cell
rN
—_—
Figure 16-28 m B-cell development. Heavy chain (H) and light chain (L) are designated as H®, L° if in embryonic form, and H*, L° if
rearranged. > = cytoplasmic mu; Y = immunoglobulin.
Chapter 16 Introduction to Leukemia and the Acute Leukemias ee)Nn~—
V D J Cu
Germline DNA
(non-rearranged)
| DNA rearrangement
i a] i
|Transcription
RNA splicing
Y DPD st Cy
Messenger RNA
|Translation
Peripheral
Bone marrow Thymus lymphoid
Cortex Medulla organs
Suppressor cell
Thymocytes)
Helper cells)
Suppressor cells)
Mature T cells)
Figure 16-51 i Normal T-cell maturation. T-cell receptor (TCR) 8 and « genes are designated as B° and a° if in the embryonic form, and B*
and a’ if rearranged. The colored blue bars indicate where the antigens are present in the stages of maturation. No color indicates the antigens
are not present. Please note that the CD1 antigen disappears on mature thymocytes. The orange color indicates that as the cells mature they
lose the expression of either CD4 or CD8, with only one of these antigens remaining on each mature thymocyte or T-cell, and not both.
358 Chapter 16 Introduction to Leukemia and the Acute Leukemias
Cytologic Features
rete
aaa
Immunologie
Phenotype
|e oo I OT NIE
factor and is involved in other translocations. Many acute Burkitt’s Leukemia/Lymphoma (Mature
leukemias with an MLL gene abnormality are CD10 negative
B-Cell ALL)
by flow cytometry.*° The t(4;11) may be detected by RT-PCR
method. There is a FISH probe for 11q23, which allows iden- Recall that the FAB classification scheme had three categories
tification of a translocation involving the MLL gene and some of ALL based on blast morphology. L3 lymphoblasts were
other chromosome. The 11q23 abnormality is also seen in Burkitt-like and morphologically had the features of lym-
secondary AML in adults, which occurs when adults get AML phoblasts, so were logically classified as ALL. The WHO
after they have been treated with certain chemotherapy. In Classification, with its emphasis on immunophenotype and
both AML and pediatric ALL, 11q23 abnormalities are asso- cytogenetics, does not include B cell ALL, or the FAB L3
ciated with a poor prognosis.*> subtype of ALL, in its precursor B cell ALL classification,
because Burkitt-like lymphoblasts lack the immunopheno-
PRECURSOR T-CELL ALL typic characteristics of an early B cell, namely the TdT is
About 15% to 25% of all patients with ALL have precursor negative. The leukemic B cells in Burkitt’s leukemia, previ-
T-cell ALL.*°*’ These T-ALL cases can be further categorized ously called B-cell ALL, are derived from a transformed or
into one of the three stages of intrathymocyte development stimulated B cell; they are more mature B cells than the
based on immunophenotype, but the prognostic significance lymphoblasts in precursor B ALL.
of where the lymphoblasts are in these stages of development Burkitt leukemia/lymphoma is rare and most typically
remains uncertain.*°*° Cytoplasmic CD3, CD2, and CD7 are represents the leukemic phase of Burkitt lymphoma in
expressed early in development. Surface CD3 is expressed patients with bulky disease (see Chap. 21). Only rarely will
after cytoplasmic CD3. CD7 is a very reliable marker of patients present with blood and marrow disease clinically
T-cell ALL, but CD2 and CD5 are also expressed in many resembling pre-B ALL and not have bulky lymphoma. Burkitt
cases.'' Approximately 10% of T-cell ALL cases express or L3 morphology can reliably predict the immunophenotype,
CD10 or CALLA, which is also seen on many pre-B ALLs."! which is that of a mature B-cell. There is typically a high con-
Interestingly, CD4 and CD8 are occasionally coexpressed in centration of surface immunoglobulin (usually slgM) and
T lymphoblasts, corresponding to a later stage of thymocyte monoclonal light-chain restriction. CD19, CD20, and HLA-
development, but before selection of either CD4 or CD8 on DR are positive; TdT is negative; and most cases do not
mature T cells. Like the lymphoblasts of pre-B ALL, the lym- express CD10. Virtually all cases have a characteristic
phoblasts of T-cell ALL are TdT positive and are associated translocation involving a rearrangement of the c-myc onco-
with both L1 and L2 morphology (see Table 16—14).°’ gene on chromosome 8 with the immunoglobulin heavy-chain
Patients with T-cell ALL often present with a mediastinal gene on chromosome 14, or the light-chain genes on chromo-
mass, a high white blood cell count (greater than 100 x 10°/L somes 2 or 22 (t[8;14], t[2;8], or t[8;22]).*!
in 50% of cases), hepatosplenomegaly, and early meningeal Patients with Burkitt’s leukemia are treated differently
involvement. Males are affected more often than females, and from patients with pre-B ALL, which has a relatively good
the disease occurs more often in older children. These patients prognosis. The prognosis for Burkitt’s leukemia (B-cell ALL)
generally have a poorer prognosis than those with common has traditionally been worse than for most precursor B-cell
pre-B ALL. Features that most consistently correlate with a ALL, but has improved with newer targeted therapies.
better response to therapy include a low white blood cell level,
younger age (less than 15 years), and L1 morphology.** There
is also some evidence that certain surface markers may be Childhood versus Adult ALL
associated with a better (CD5) or worse (CD3) prognosis.*”**
When patients present with a mediastinal mass, it may be The incidence and prognosis of ALL differs markedly with
difficult to distinguish T-cell ALL from lymphoblastic lym- age. Recall that one of the most important clinical predictors
phoma. The cytologic features of this type of lymphoma are of outcome in ALL 1s age, with patients 10 years old or older,
similar to those of ALL. Although the lymphoma is generally and infants younger than | year having a worse prognosis.
associated with a more mature immunophenotype, both the Overall, about 80% of pediatric patients are alive and disease
lymphoma and the leukemia are composed of TdT positive free at 5 years, compared with significantly lower rates in
lymphoblasts with evidence of T cell lineage.*°® After a vari- adults with ALL.*’ For this reason ALL may be considered in
able time, patients with lymphoblastic lymphoma almost two subcategories: childhood ALL and adult ALL.
always develop bone marrow involvement, which renders There are cytogenetic and morphologic differences
their condition indistinguishable from T-cell ALL. The dis- between adult and pediatric ALL. Ll morphology occurs
tinction between T cell or B cell lymphoblastic leukemia and most frequently in the pediatric age group, whereas the L2
lymphoma is generally based on clinical criteria. Patients who type tends to be seen in adults. The t(9;22), a poor prognosis
first present with prominent marrow and peripheral blood translocation also known as the Philadelphia chromosome,
involvement are usually diagnosed with T-cell ALL, even if has a significantly greater frequency in adult precursor B-cell
they have concomitant mediastinal involvement. In contrast, ALL.*° MLL or mixed lineage leukemia gene abnormalities
a patient who presents with mediastinal involvement but no are typically seen in infant precursor B ALL patients and
marrow or peripheral blood involvement is diagnosed with largely account for the disappointing prognosis for ALL in
lymphoma. patients less than | year old.*? The most common cytogenetic
362 — Chapter 16 Introduction to Leukemia and the Acute Leukemias
abnormality in pediatric ALL is t(12;21), seen in about 25% cytopenias, including severe infections and hemorrhages.
of pediatric cases and associated with a favorable prognosis. Improved supportive care has reduced mortality and morbidity
Many patients diagnosed with ALL will achieve a com- from AML therapy; hematopoietic growth factors given during
plete remission, and many are potentially cured.*? Unfortu- induction accelerate recovery from myelosuppression and are
nately, some patients relapse, and some eventually die from pivotal in timely drug administration during dose-intensive
their disease. The therapy used to achieve a cure has toxic treatment regimens. Results of AL therapy have improved over
effects that are a special concern in growing and developing the past 20 years, but therapies producing cures consistently
children. To select the patients who will have an optimal have yet to be identified for most subtypes of AL. Intensive
response to less aggressive therapy, investigators have identi- investigation in this area is still ongoing, with the goal of devel-
fied prognostic factors and defined clinical risk categories. oping therapies that are more patient- and disease-specific, and
These tools allow clinicians to better select the type and inten- also less toxic. Many new “molecular” markers with potential
sity of therapy to most optimally match each patient’s risk of value in prediction of prognosis and therapeutic decision-
relapse against long-term sequelae of treatment. To date, indi- making have been identified by recent microarray gene expres-
cators of a poor prognosis include older age, a high white blood sion profiling and comparative genomic hybridization. The
cell count (more than 20 < 10°/L), and particular chromosomal integration of these new markers into treatment schema is dif-
abnormalities like t(9;22) BCR-ABL, t(4;11) and other MLL ficult and must be done very carefully; however, although their
abnormalities, t(1;19) PBX—E2A and hypodiploidy.* T-cell or clinical utility is currently limited, these new observations are
mature B-cell phenotypes and L2- and L3-type morphology providing pivotal information about disease mechanisms and
have also been associated with less favorable prognosis.** Chil- potential new drug targets.
dren, with hyperdiploid lymphoblasts (more than 50 chromo-
somes), a low white blood cell count, and t(12;21) TEL-AMLI
have a more favorable prognosis.**
Treatment of ALL
The effective treatment of pediatric ALL is one of the great
successes of clinical oncology, with long-term survival in
Treatment of Acute Leukemia
over 80% of children.'?**°? However, over the same time
Therapy for acute leukemia (AL) is among the most complex of period, adults with ALL have had a much lower cure rate of
any anticancer programs. The goals of therapy for AL are to erad- 20% to 40%.?7837° Although well-recognized age-related dif-
icate the leukemic clone, reconstitute normal hematopoiesis, and ferences in the biology of ALL exist, recent studies suggest
prevent any emergence of resistant leukemic subclones. Anti- that the survival difference between adults and children with
leukemic therapy generally consists of three distinct phases: ALL is due more to differences in treatment than disease
induction, consolidation, and maintenance.*'*? Induction biology or patient characteristics. Most children with ALL are
therapy is designed to attain complete remission as quickly as treated on clinical trials, in academic centers with experi-
possible; its success is the best predictor of long-term disease- enced support teams; children also receive higher doses of
free survival. Remission occurs when the patient’s blood counts some important drugs, and adherence to prescribed drug
return to normal, and bone marrow samples show no sign of dis- doses and schedules is very high in pediatric clinical trials. In
ease. Induction therapy achieves a remission in more than contrast, most adults with ALL are not treated on clinical tri-
95% of children and in about 75% to 89% of adults.*° Induction als or in academic medical centers, and dose reductions are
therapy is usually very intense and lasts about | month.** After much more common. Newer studies find that cure rates of
induction chemotherapy, the next step is usually consolidation adult ALL patients treated on pediatric treatment regimens is
chemotherapy, depending on the patient and type of leukemia. similar to that seen in children, suggesting that ALL therapy
The goal of consolidation therapy is to reduce the number of based on disease biology, not age, should be the standard for
disease cells left in the body. The drugs and doses used during all ALL patients in the future.*”~?
consolidation therapy depend on the patient’s risk factors. Con- Central nervous system (CNS) prophylactic treatment is
solidation therapy is also intense, lasting approximately 4 to standard in ALL therapy during induction and consolida-
8 months.” Maintenance therapy usually starts after a patient tion.’° Although unusual at diagnosis (<10%), CNS disease
stays in remission after induction and consolidation therapy. The can be as high as 50% to 75% at | year in the absence of in-
goal is to destroy any leukemic cells that may remain in order to trathecal (IT) chemotherapy. Craniospinal irradiation is asso-
maintain a disease-free state. Maintenance therapy is less ciated with many neurologic adverse events, including
intense than the other two phases and may last 2 to 3 years.” seizures, dementia, and intellectual dysfunction, so IT and
However, the specific drugs used and their timing and high-dose systemic chemotherapy are typically used today for
doses differs remarkably among different AL subtypes.*°34 CNS prophylaxis.****
Their modes of action differ but, in general, they poison and Intensification of induction therapy by using additional
kill dividing cells, usually by blocking DNA or RNA synthesis. drugs has improved survival in some ALL subtypes. Intensive
The toxicity of chemotherapy for AL must always be balanced consolidation is standard in post-remission ALL therapy, but
with the efficacy of treatment. The consequences of AL therapy specific drug cycles are variable. Regardless of the regimen
can be very serious; treatment-related early death is not uncom- used, strict adherence to protocols, without significant dose
mon and there is often significant morbidity from prolonged delay, appears to be very important for long-term survival of
Chapter 16 Introduction to Leukemia and the Acute Leukemias 363
ALL patients. Post-remission maintenance therapy in ALL is of >10,000, and others have an intermediate risk. APL therapy
also standard, and is sometimes given over 2 to 3 years.*° The is curative in most patients; anticipated cure rates are close to
only exception is mature B-ALL, in which relapses beyond 100% in low-risk, 90% in intermediate-risk, and 70% in high-
the first year are rare.*° There are still many questions about risk patients. The addition of all-trans-retinoic acid (ATRA) to
the optimal drugs, doses, and schedules used for maintenance AML chemotherapy increases the CR rate and dramatically
therapy in the different subtypes of ALL. reduces the relapse rate in APL patients.'*!7°°
The utility of allogeneic stem cell transplant (SCT) as
front-line therapy in ALL is not yet well validated. In some AML WITH INV(16) OR t(8;21)
clinical trials, allogeneic SCT has been done in first complete Appropriate treatment is curative in about 50% of cases in
remission (CR1) and it appears to be of some benefit in high- this group, using standard AML regimens for induction, con-
risk ALL.**°° However, the indications, scheduling, and pro- solidation, and maintenance therapy that include high dose
cedures for SCT in CRI are not yet standardized and it is not cytarabine.©°*? Transplant should not be considered in this
clear that SCT-based therapy in CR1 is associated with an im- AML subtype unless relapse occurs.
proved outcome over chemotherapy alone in most subtypes of
ALL. Larger cooperative group studies are required to evalu- AML IN THE ELDERLY
ate the optimal integration of SCT in front-line ALL therapy. Standard AML chemotherapy in the elderly (>60 years old) is
Targeted therapies have been developed for some ALL associated with a high rate of treatment-related mortality and
subtypes. One example of a targeted therapy is the use of ima- a poor outcome in patients who do survive initial therapy.”” CR
tinib, a tyrosine kinase inhibitor, in addition to chemotherapy rates in these patients are <50%, and the probability of sur-
for Philadelphia chromosome-positive ALL. This has resulted vival at 2 years is about 10%. Patients <80 years of age have
in a considerable improvement in outcome in this formerly a mortality rate of <50%, so transfusion support or palliative
very unfavorable subgroup of ALL patients, without signifi- care may be reasonable options for this age group. Investiga-
cantly increasing toxicity.°’ Another example of a targeted tional drugs and/or targeted therapies at initial diagnosis may
therapy is the addition of an anti-CD20 monoclonal antibody be appropriate for these poor prognosis patients. Less inten-
to therapy for mature B-ALL; this has significantly improved sive induction approaches may also be used in the elderly,
outcome for these patients.°? Recent trends in treatment of such as reduced intensity allogeneic stem cell transplant.’”"™
ALL are focusing on’ immunostimulatory oligonucleotides
and other interventions at the genetic level.**?
Measurement of Response to Therapy
for Acute Leukemia
Treatment of AML
Response to therapy for AL is measured by a number of para-
AML is a very heterogenous disease biologically, and the cyto- meters. Traditionally, a complete remission (CR) has been
genetic and molecular abnormalities detected at the time of defined as normalization of peripheral blood counts and a
diagnosis provide the best information for predicting clinical return to normal bone marrow hematopoiesis. The blast percent-
outcome and stratifying therapy.*?AML is typically divided age should be normal, and there should be no persistence of
into the following subgroups for therapy decisions: (1) acute disease morphologically or with flow cytometric analysis.
promyelocytic leukemia (APL), (2) AML with inv(16) or Cytogenetic analysis is typically performed before and after
(8:21), (3) AML in younger patients with no adverse risk fac- therapy, and the absence of a previously identified cytogenetic
tors, (4) AML in younger patients with adverse risk factors, and abnormality is another important component of CR. However,
(5) AML in the elderly. Within each of these groups, there these traditional measures of treatment response are relatively
are further risk factors that can affect initial and subsequent insensitive. New techniques for detection of a low level of resid-
therapy. These include the presence of high white blood cell ual disease, below the level detected by morphologic review,
counts of >50,000 and leukemic lung infiltration, ®’ which are immunophenotyping, or standard karyotyping, suggest that the
indications for immediate therapy without waiting for results of achievement of a molecular complete remission may be the best
karyotyping. In all AML patients, regardless of therapy, achieve- measure of treatment efficacy in AL. Persistence of detectable
ment and maintenance of a CR is the goal of therapy. Once CR disease results in significantly worse survival.’*”°
is maintained for 3 years, the risk of relapse declines markedly Molecular remission is defined as a level of minimal resid-
to only 5% to 10%.™ Some specific treatment considerations for ual disease (MRD) that is below the detection limit of
different AML treatment groups are discussed below. PCR analysis, which is generally | blast in 10,000 (10+) or
100,000 (10°) normal bone marrow cells. The presence of
APL MRD is felt to be an independent prognostic factor that can
Early recognition of APL is crucial, for a number of reasons. identify primary drug resistance and other factors playing a role
APL patients are at risk for fatal hemorrhage due to dissemi- in therapy response. MRD testing is being used in some AL clin-
nated intravascular coagulation, so the appropriate treatment for ical trials to aid in decision-making about the need for mainte-
APL is different from that employed for other types of AML." nance or other more aggressive therapy following induction.
Low-risk APL patients present with WBC counts <10,000 and However, the predictive value of MRD analysis in ALL depends
platelet counts >40,000; high risk patients have a WBC count on the technical quality of the testing, which is time-consuming,
364 Chapter 16 Introduction to Leukemia and the Acute Leukemias
Table 16-16 Some Targeted Drugs count, 79.2 X 10°/L. A peripheral blood smear was manu-
| sGurrently. ines se oe ally reviewed in the hematology laboratory and the differen-
tial diagnosis included 80% blasts.
_ Development | forAMILaie toe A bone marrow aspirate and biopsy were obtained, which
both showed virtually total replacement of normal elements
Type
with sheets of poorly differentiated cells (Fig. 16-36). The
Epigenetic Therapy aspirate smears showed sheets of blasts. The blasts were rel-
DNA methyltransferase Reverse gene silencing by atively large (15 to 20 um) with a moderate amount of cyto-
inhibitors inhibiting DNA methylation plasm. The nuclei varied in shape from round to oval, and
Histone deacetylase Reverse gene silencing by some were indented or folded; most had several distinct
(HDAC) inhibitors inhibiting histone small nucleoli. An occasional blast had azurophilic granules,
deacetylation but this was the exception. No Auer rods were seen. The
FLT3 Inhibitors AML patients (50%) biopsy specimen was hypercellular, approaching 100% cel-
overexpress FLT3
lularity in some areas. The morphology of the aspirated cells
was similar to those seen in the peripheral blood except that
fewer of the cells had folded nuclei, and, in general, the
expensive, and requires highly-trained staff. In most clinical tri- nuclear-to-cytoplasmic (N:C) ratio was higher. Cytochemi-
cal studies of the aspirate smears were positive: the
als, MRD testing must be done by designated central reference
myeloperoxidase stain was positive in approximately 30%
laboratories.” Detection of MRD at any time point after the start
of the blasts and the NSE (a-naphthy] butyrate) was positive
of consolidation therapy is associated with a high relapse risk in in occasional cells (less than 5%).
most AL subtypes, particularly ALL.” Patients with MRD after
consolidation therapy are candidates for stem cell transplanta-
tion (SCT) or other targeted therapies.
The rapid discovery of novel molecular aberrations and
global gene expression profiles have led to the development
of targeted drugs for AML.” These genomic markers confirm
the heterogeneity of this disease and support the need for
individualized patient treatment. Several of these genomic
markers serve not only as prognostic factors but also as ther-
apeutic targets for smali molecules and other novel com-
pounds. Many new targeted agents are currently in clinical
testing. Some of the classes of targeted drugs currently in
development are listed in Table 16-16.” These targeted
approaches are being investigated for relapsed and refractory
AML of initial complete remission duration of less than
1 year. However, the role of these new targets in leukemoge-
nesis remains to be elucidated.
Figure 16-36 @ Case study—bone marrow (AML).
and leukopenia. She required platelets and packed red blood A specimen was also sent for cytogenetic studies. Inter-
cell transfusions. She also required broad-spectrum antibi- phase FISH studies showed 95% of cells positive for the
otics for fever resulting from neutropenia, although no PML-RARa fusion, indicative of the t(15;17) translocation.
specific pathogen could be identified. Three weeks after her Diagnosis: The WHO diagnosis is acute promyelocytic
induction chemotherapy, a repeat bone marrow biopsy was leukemia with t(15;17). The FAB classification would cate-
performed. There was no evidence of residual leukemia by gorize this as AML M3. Importantly, there is a microgranu-
morphologic examination of the aspirate smears and biopsy; lar variant of APL, in which the abnormal promyelocytes
or flow cytometric analysis of the aspirate; or by the FISH have a paucity of visible granules and the nuclei are typi-
probe looking for MLL abnormalities. She remained in com- cally bilobed. These atypical cells may be confused with
plete remission for 11 months, but then relapsed with monocytic blasts, but an MPO is strongly positive.
20% morphologically atypical blasts in her bone marrow Follow-up: Standard treatment with all-trans retinoic acid
confirmed by abnormal immunophenotype identified by (ATRA) was initiated, followed by cytotoxic therapy.
flow cytometry. Attempts to induce a second remission were Through the course of hospitalization, the patient showed
unsuccessful. She developed progressive hepatomegaly, laboratory evidence of DIC, and required blood support
jaundice, and persistent neutropenia. She also developed from the beginning of therapy to maintain appropriate
multiple infections and died from her disease several hemoglobin, platelet, and fibrinogen levels. However, the
months later. patient’s CBC and clinical status began to improve. A month
This case illustrates a typical course of acute myeloid later, no PML—RARa fusion transcripts were detected by
leukemia. Although the patient was not cured, she did RT-PCR on a bone marrow specimen. He then received
achieve and maintain a complete remission for nearly a full maintenance therapy with ATRA in a standard dose for the
year. The 5-year survival rate for adult AML patients with next 2 years.
MLL abnormalities is about 45%, but it is important to note The case underscores the importance of recognition of
that there are many different kinds of AML with MLL abnor- APL. Proper morphologic diagnosis is imperative, so that
malities. Currently, more than 50 different genes have been the appropriate molecular studies can be requested for con-
identified that translocate with 11q23 and the characteristics firmation and so the patient is treated appropriately as soon
of each of these have yet to be fully described. Therefore, as possible. A delay in treatment can lead to complications
MLL abnormalities have an intermediate to poor prognosis. due to APL-associated coagulopathy, including cerebral
«
hemorrhage. The response to induction therapy with ATRA
and standard chemotherapy has been shown to be in the
range of 90% to 95% in genetically confirmed APL either by
conventional cytogenetics, FISH for t(15;17) or by RT-PCR
for PML-RARa.
Case Study 2
A 32-year-old man presented to the ER complaining of
fever, lethargy, and mucosal bleeding. He said that these
symptoms began 5 to 6 days ago, but that before he had been
in a general state of good health. The patient’s past medical
history was unremarkable. On physical exam, the patient
appeared ill, pale, and in moderate acute distress. Numerous
purpura were found on his upper and lower extremities, Case Study 3
which the patient said erupted over the past several days. No
hepatosplenomegaly or enlarged lymph nodes were noted. T. J. was 4 years old when he first presented to his family
CBC and examination of the peripheral blood smear doctor with a 3-week history of fatigue, weakness, and a
showed mild anemia with numerous schistocytes (red blood persistent sore throat. On physical examination he had a pal-
cell fragments). Severe thrombocytopenia and many circu- pable spleen but no evidence of lymphadenopathy. He
lating atypical mononuclear cells were also seen. Additional appeared pale and had multiple bruises over his lower
laboratory studies showed a markedly prolonged prothrom- extremities. A CBC, platelet count, and differential were
bin time and decreased fibrinogen, indicating a profound carried out, with the following results:
coagulopathy.
A posterior iliac crest bone marrow aspirate and core CBC
biopsy were performed. The bone marrow biopsy and aspi-
WBC DAO 107/15 MCV 87.0 fL
rate smear revealed a markedly hypercellular bone marrow
RBC Alex 102 /E MCH 29.0 pg
with 80% promyelocytes, many with prominent azurophilic Het 21.0% MCHC 33.3 g/dL
granulation and abnormal kidney-bean shaped nuclei. Some Hgb 7.0 g/dL Platelets 6.7 X 10°/L
of the promyelocytes contained multiple Auer rods (“faggot
cells”). Cytochemical staining with myeloperoxidase Differential
(MPO) and Sudan black-B showed strong staining in these
promyelocytes. Flow cytometric analysis of bone marrow PMN 3%
Blasts 97%
showed a myeloid population expressing CD13, CD33,
CD45(dim), CD117, while lacking HLA-DR. continued
continued
366 — Chapter 16 Introduction to Leukemia and the Acute Leukemias
continued
Qu estions
Ye e
6. A 49-year-old woman was admitted to the hospital for 10. What cytochemical stain is best for differentiating AML
easy bruising and menorrhagia. She had evidence of dis- from ALL?
seminated intravascular coagulation. Her white blood cell a. Alpha-naphthyl acetate
count is 3 X 10°/L with 95% large, atypical mononuclear b. Nonspecific esterase
cells and some atypical bilobed cells. Many of these cells c. Myeloperoxidase
are packed with larger, purple-staining granules; some d. Periodic acid—Schiff
have multiple Auer rods; and all are strongly peroxidase-
. How many blast cells in a bone marrow aspirate smear
positive. What is the diagnosis?
are necessary for a diagnosis of acute leukemia using
a. AML, FAB M2
FAB criteria?
b. AML, FAB M3
a. 20%
c. AML, FAB M4
b. 15%
d. AML, FAB M5
C2570
. What is the likely genetic abnormality referring to ques- d. 30%
tion 6?
. How many blast cells in a bone marrow aspirate smear
a. ((8;21) AML/I-ETO
are necessary for a diagnosis of acute leukemia using
b. (4511) AF4-MLL
WHO criteria?
c. t(9;22) BCR-ABL
a. 20%
d. t(15;17) PML—-RARa b. 15%
. A 21-year-old patient’s bone marrow and peripheral Cnzove
blood has medium sized blasts with basophilic cyto- d. 30%
plasm and numerous lipid vacuoles. Cytogenetic studies . Which chromosome abnormality occurs in the FAB M2
show t(8;14) involving MYC on chromosome 8 and the
type of AML?
Ig heavy chain region on chromosome 14. Flow cytome- a. t(8;21)
try is most likely to show which of the following? b. t(9;22)
a. CD37 CD77, CD10, CD45, and TdT Cant Clsyailii/))
b. CD10, CD19, CID20, CD45, and surface « light chain d. t(1;19)
c. CD10, CD19, CD45, TdT, and surface « light chain
d. CD10, CD19, CD34, and no surface light chain or TdT 14. Which cytochemical stain is useful in separating
myeloblasts from monoblasts?
. Which of the following is true about the prognostic a. Myeloperoxidase
implications of cytogenetic findings seen in precursor b. Sudan black B
B-cell ALL? c. Specific esterase
a. Hypodiploidy as determined by flow cytometry DI is d. Nonspecific esterase
associated with a favorable prognosis
b. t(1:19) PBX—E2A is associated with an unfavorable . Which chromosome abnormality occurs in the FAB M3
prognosis type of AML?
. (9:22) BCR-ABL is associated with a favorable
a. t(8;21)
(o)
prognosis bet:22)
Cat@lo: iy)
d. Hyperdiploidy of greater than 50 chromosomes as
determined by flow cytometry DI is associated with a ericlelien)
poor prognosis.
See answers at the back of this book.
36 8 Chapter 16 Introduction to Leukemia and the Acute Leukemias
REFERENCES bone marrow biopsy specimens. Mol 18. Bennett, JM, et al: Proposals for the clas-
Pathol 53(2):83, 2000. sification of acute leukemia. Br J Hema-
— . Cancer
freiFacts and Figures
Concer Society 2008.
2008 The
Cwallable 9. Shafer, JA: Artifactual alterations in tol 33:451, 1976.
Bee Hie, Bae ony ‘: phagocytes in the blood smear. Am J 19. Detailed Guide: Leukemia-Acute
en ie i 6x [ieeen Med Technol 48:507, 1982. Myeloid (AML): How is acute myeloid
Ce ee ce ae MS. et al: Low blast conn leukemia (AML) classified? [On-Line]
i)
Eee ae ie sais ee iat HeLa myeloid disorders with Auer Rods: A Cancer Reference Information, The
ae ‘eae cae a ae : clinicopathologic analysis of 9 cases. American Cancer Society 2005. Avail-
eae ie “atte a Am J Clin Pathol 124(2):191, 2005. able: http://www.cancer.org.
tate eee aoe 8noble lil Dunphy, G: Applications of flow cytome- 20. Miwa, H, et al: Biphasic expression of
(ene oy Pease o7sncbl try and immunohistochemistry to diag- CD4 in acute myelocytic leukemia
1):45 1997 a Ener ae Pathol (AML) cells: AML of monocyte origin
Co ieke : ; ab Med 128: , 2004. and hematopoietic precursor cell origin.
is)
AEGN cle techRSE 12. Wintrobe, MM: Greer, JP (Ed), Wintrobe’s Leukemia 1:44, 1998. ;
Clinical Hematology, ed 11. Lippincott 21. Breton-Gorius, J, et al: Megakaryoblastic
5005, ASH Fetes Book 131
drugs. ay Williams & Wilkins, Philadelphia, 2004. acute leukemia: Identification by the
isondlienDPvand Ross As Epidemiol: 13. Kjedsberg, C, et al: Practical Diagnosis of ultrastructural demonstration of
fia
Beers aia childs ant Hematologic Disorders: 2 Malignant Dis- platelet peroxidase. Blood 51:45,
Rate Semin Oncol 24:3, 1997 orders. ASCP Press, American Society for 1978.
; cee RS. and ences IM: Romie Clinical Pathology, Chicago, 2006. 22. Bennett, JM, et al: Propsed revised crite-
Nn
Roa enn milvelodysplastc 14. Hoffbrand, AV, et al: Essential Hematol- ria for the classification of acute myeloid
ESR OnE ogy. Blackwell, Oxford, 2006. leukemia. Ann Intern Med 103:626,
14(2):98 5007 il), Mrozek, K, et al: Cytogenetics in acute 1985.
wae Ww. a al: The World Health leukemia. Blood Rey 18: 15, 2004. 23. Jaffe, E, et al: The World Health Organi-
OO
Or TION cheat suite 16. Kern, W, et al: Determination of relapse zation Classification of Tumors, Pathol-
weed neoplasms. Blood 100:2292 risk based on assessment of minimal ogy and Genetics, Tumors of
3002. Va Se Ee eS residual disease during complete remis- Hematopoietic and Lymphoid Tissues.
Nera SENIUL CEE Roleto fk BIGGS sion by multiparameter flow cytometry in Lyon, France, IARC Press, 2001, p 91.
~—
SF eo AaNES RIE ae unselected patients with acute myeloid 24. Schoch, C, et al: AML with 11q23/MLL
“a Innch RR CaN Indian ico, leukemia. Blood Rev 104(10), 2004. abnormalities as defined by the
ol siggy 17. Mrozek, K, et al: Clinical relevance of WHO classification: incidence, partner
Microbiol 49(4):524, 2006.
mutations and gene-expression changes chromosomes, FAB subtype, age distrib-
oo . Krober, SM, et al: Acute lymphoblastic
in adult acute myeloid leukemia with ution, and prognostic impact in an unse-
leukaemia: correlation between
normal cytogenetics: Are we ready for a lected series of 1897 cytogenetically
morphological/immunhistochemical
prognostically prioritized molecular clas- analyzed AML cases. Blood 102(7):
and molecular biological findings in
sification? Blood J 109(2):421, 2007. 2395, 2003.
Chapter 16 Introduction to Leukemia and the Acute Leukemias 369
PES. Orazi, A: Histopathology in the diagno- MLL gene are highly curable with use of 54. Clarke, M, et al: CNS-directed therapy
sis and classification of acute myeloid chemotherapy alone: Results from the for childhood acute lymphoblastic
leukemia, myelodysplastic syndromes, Japan Infant Leukemia Study Group. leukemia: Childhood ALL Collaborative
and myelodysplastic/myeloproliferative Blood 107:4663, 2006. Group overview of 43 randomized trials.
diseases. Pathobiology 74(2):97, 4}. Kolitz, JE: Current therapeutic strategies J Clin Oncol 21:1798, 2003.
2007. for acute myeloid leukaemia. Br J 5p Thomas, DA, et al: Chemoimmunother-
26. Haferlach, T, et al: Global approach to Haematol 134:555, 2006. apy with hyper-CVAD plus rituximab for
the diagnosis of leukemia using gene . Ribera, J, et al: Comparison of intensive the treatment of adult Burkitt and
expression profiling. Blood 106(4):1189, chemotherapy, allogeneic, or autologous Burkitt-type lymphoma or acute lym-
2005. stem-cell transplantation as postremis- phoblastic leukemia. Cancer 106:1569,
2p Carroll, WL, et al: Pediatric acute lym- sion treatment for children with very 2006.
phoblastic leukemia. Hematol Am Soc high risk acute lymphoblastic leukemia: 56. Thiebault, A, et al: Adult acute lympho-
Hematol Educ Prog 102, 2003. PETHEMA ALL-93 Trial. J Clin Oncol cytic leukemia study testing chemother-
28. Surveillance Research Program: National 25(1):16, 2007. apy and autologous and allogeneic
Cancer Institute SEER*Stat Software. 43. Relling, MY, et al: Thiopurine methyl- transplantation. A follow-up report of the
(www.seer.cancer.gov/seerstat), version transferase in acute lymphoblastic French protocol LALA 87. Hematol
6.2.3; 2006. leukemia. Blood 107:843, 2006. Oncol Clin North Am 14:1353, 2000.
29. Rolink, AG, et al: Fidelity and infidelity 44, Kosaka, Y, et al: Infant acute lym- Sie Towatari, M, et al: Combination of inten-
in commitment to B-lymphocyte lineage phoblastic leukemia with MLL gene sive chemotherapy and imatinib can
development. Immunol Rev 175:104, rearrangements: outcome following rapidly induce high-quality complete
2000. intensive chemotherapy and hematopoi- remission for a majority of patients with
30. Basso, G, et al: Diagnosis and genetic etic stem cell transplantation. Blood newly diagnosed BCR-ABL-positive
subtypes of leukemia combining gene 104(12):3527, 2004. acute lymphoblastic leukemia. Blood
expression and flow cytometry. Blood 45. Pui C-H, Sandlund JT, Pei D, et al. 104:3507, 2004.
Cells Mol Dis 39(2):164, 2007. Improved outcome for children with 58. Fujii, H, et al: In vivo control of acute
. Ross, M, et al: Classification of pediatric acute lymphoblastic leukemia: Results of lymphoblastic leukemia by immunostim-
acute lymphoblastic leukemia by Total Therapy Study XIIIB at St. Jude ulatory CpG oligonucleotides. Blood
gene expression profiling. Blood Children’s Research Hospital. Blood 109:2008, 2007.
102(8):2951, 2003. 104:2690, 2004. a): Moorman, AY, et al: Prognosis of chil-
32. Holleman, A, et al: The expression of 46, Annino, L, et al: Treatment of adult acute dren with acute lymphoblastic leukemia
70 apoptosis genes in relation to lineage, lymphoblastic leukemia (ALL): Long-term (ALL) and intrachromosomal amplifica-
genetic subtype, cellular drug resistance, follow-up of the GIMEMA ALL 0288 ran- tion of chromosome 21 ((AMP21).
and outcome in childhood acute lym- domized study. Blood 99:863, 2002. Blood 109:2327, 2007.
phoblastic leukemia. Blood 107(3):769, 47. Boissel, N, et al: Should adolescents with 60. Grimwade, D, et al. The importance of
2006. acute lymphoblastic leukemia be treated as diagnostic cytogenetics on outcome in
33) Kebriaei, P, et al: Acute lymphoblastic old children or young adults? Comparison AML: Analysis of 1612 patients entered
leukaemia: Diagnosis and classification. of the French FRALLE-93 and LALA-94 into the MRC AML 10 trial. Blood
Best Pract Res Clin Hamatol 15(4):597, trials. J Clin Oncol 21:774, 2003. 922322, 1998;
2002. 48, Thomas, X, et al: Outcome of treatment 61. Mrozek, K, et al. Cytogenetics in acute
34. Gianluigi, Z, et al: Acute lymphoblastic in adults with acute lymphoblastic leukemia. Blood Rev 18:115, 2004.
leukemia with TEL-AML/ fusion has leukemia: Analysis of the LALA-94 62. Kolitz, JE, et al. Dose escalation studies
lower expression of genes involved in trial. J Clin Oncol 22:4075, 2004. of cytarabine, daunorubicin, and etopo-
purine metabolism and lower de novo 49. Kantarjian, H, et al: Long-term follow- side with and without multidrug resis-
purine synthesis. Blood 104:1435, 2004. up results of hyperfractionated tance modulation with PSC-833 in aun-
SB) Yoshiyuki, K, et al: Infant acute lym- cyclophosphamide, vincristine, doxoru- trated adults with acute myeloid
phoblastic leukemia with MLL gene bicin, and dexamethasone (Hyper- leukemia younger than 60 years: Final
rearrangements: outcome following in- DVAD), a dose-intensive regimen, in induction results of Cancer and
tensive chemotherapy and hematopoietic adult acute lymphocytic leukemia. Leukemia Group B study 9621. J Clin
stem cell transplantation. Blood Cancer 101:2788, 2004. Oncol 22:4290, 2004.
104(12):3527, 2004. . deBont, JM, et al: Significant difference G3: Giles, FJ, et al: Leukapheresis reduces
36. Morton, L, et al: Proposed classification in outcome for adolescents with acute early mortality in patients with acute
of lymphoid neoplasms for epidemio- lymphoblastic leukemia treated on pedi- myeloid leukemia with high white cell
logic research from the Pathology Work- atric vs adult protocols in the Nether- counts but does not improve long term
ing Group of the International Lym- lands. Leukemia 18:2032, 2004. survival. Leuk Lymph 42:67, 2001.
phoma Epidemiology Consortium . Schiffer, CA: Differences in outcome in 64. DeLima, M, et al: Implications of
(InterLymph), Blood 110(2):695, 2007. adolescents with acute lymphoblastic potential cure in acute myelogenous
Bile Pui, C, et al: Childhood acute lym- leukemia: A consequence of better regi- leukemia: Development of subsequent
phoblastic leukemia. Rev Clin Exp mens? Better doctors? Both? J Clin cancer and return to work. Blood
Hematol 6:161, 2002. Oncol 21:7600, 2003. 90:4719, 1997.
38. Larson, RA: Management of acute lym- . Bleyer, A: The adolescent and young 65. Tallman, MS, et al: All-trans retinoic
phoblastic leukemia in older patients: adult gap in cancer care and outcome. acid APL: long-term outcome and prog-
Semin Hematol 43:126, 2006. Curr Probl Pediatr Adolsec Health Care nostic factor analysis from the North
Shy) Bertrand, FE, et al: Pro-B-cell to pre- 35:182, 2005. American Intergroup protocol. Blood
B-cell development in B-lineage acute . Lazarus, HM, et al: Central nervous sys- 100:4298, 2002.
lymphoblastic leukemia expression the tem involvement in adult acute lym- 66. Bloomfield, CD, et al: Frequency of pro-
MLL/AF4 fusion protein. Blood phoblastic leukemia at diagnosis: Results longed remission after high-dose cytara-
98(12):3398, 2001. from the international ALL trial MRC bine intensification in AML varies by
40. Mizutani, S, et al: Infants with acute UKALL-XII/ECOG E2993. Blood cytogenetic subtype. Cancer Res
lymphoblastic leukemia and a germline 108:465, 2006. 58:4173, 1998.
370. Chapter 16 Introduction to Leukemia and the Acute Leukemias
67. Nguyen, S, et al: A white blood cell acute myeloid leukemia: Results from and Reporting Standards for Therapeutic
index as the main prognostic factor in Cancer and Leukemia Group B 8461. Trials in Acute Myeloid Leukemia. J
t(8;21) acute myeloid leukemia (AML): Blood 108:63, 2006. Clin Oncol 21:4642, 2003.
A survey of 161 cases from the French . Goldstone, AH, et al: Attempts to . Marcucci, G, et al: Abnormal cytogenet-
AML Intergroup. Blood 99:3517, 2002. improve treatment outcomes in acute ics at date of morphologic complete
oO . Delaunay, J, et al: Prognosis of myeloid leukemia (AML) in older remission predicts short overall and
inv(16)/t(16;16) acute myeloid leukemia patients: The results of the United disease-free survival, and higher relapse
(AML): A survey of 110 cases from the Kingdom Medical Research Council rate in adult acute myeloid leukemia:
French AML Intergroup. Blood 102:462, AML 11 trial. Blood 98:1302, 2001. Results from Cancer and Leukemia
2003. . Buchner, T, et al: Treatment of older Group B study 8461. J Clin Oncol
. Mrozek, K, et al: Comparison of cytoge- patients with AML. Crit Rev Oncol 22:2410, 2004.
netic and molecular genetic detection of Hematol 56:247, 2005. . Bruggemann, M, et al: Clinical signifi-
t(8;21) and inv(16) in a prospective 74. Hegenbarf, U, et al: Treatment of acute cance of minimal residual disease
series of adults with de novo AML: myelogenous leukemia by low-dose, quantification in adult patients with stan-
A Cancer and Leukemia Group B study. total-body, irradiation-based condition- dard-risk acute lymphoblastic leukemia.
J Clin Oncol 19:2482, 2001. ing and hematopoietic cell transplanta- Blood 107:1116, 2006.
. Estey, E: How I treat older patients with tion from related and unrelated donors. . Kolitz, JE: Current therapeutic strategies
AML. Blood 96:1670, 2000. J Clin Oncol 24:444, 2006. for acute myeloid leukaemia. Br J
— . Farag, SS, et al: Pretreatment cytogenet- 5. Cheson, BD, et al: Revised recommen- Haematol 134:555, 2006.
ics add to other prognostic factors pre- dations of the International Working . Improving Outcomes in Acute Myeloid
dicting remission and long-term outcome Group for Diagnosis, Standardization of Leukemia. EpiCept corporation. Design-
in patients 60 years of age or older with Response Criteria, Treatment Outcomes, Write, 2007.
Chapter
Chronic
Myeloproliferative
Disorders I
Chronic Myelogenous Leukemia
Carl R. Schaub, MD
37]
Shy) Chapter 17 Chronic Myeloproliferative Disorders |: Chronic Myelogenous Leukemia
leukemogens known to increase the risk of development of cluster region) gene on chromosome 22 (Fig. 17-3). The
CML include exposure to ionizing radiation’? (such as was resulting chimeric gene, located on the Ph chromosome and
seen in radiologists prior to the use of safety shielding tech- referred to as the BCR-ABL gene, is transcribed, producing a
niques, patients treated with radiation therapy, and survivors 210-kDa protein with tyrosine kinase activity (called
of nuclear explosions), to cytotoxic drugs, especially alkylat- p2108°*48/). This new BCR—ABL fusion protein causes aber-
ing agents, and to biologically active chemicals such as rant activation of several cellular signaling pathways and is
benzene.'* Detailed studies of atomic bomb survivors in Japan considered essential in the pathogenesis of CML.” The
after World War II found that in Hiroshima, 34 of the BCR-ABL protein causes leukemogenic cell growth in
78 leukemia cases were CML, while only three of the hematopoietic cell lines, and can generate a syndrome in mice
20 leukemias in the survivors in Nagasaki were CML." that is similar to CML.'*** The 5% to 10% of CML patients
Development of CML follows the initial leakemogenic event without an identifiable Ph chromosome will nevertheless have
by several years. However, the vast majority of CML patients a BCR-ABL gene when studied via molecular techniques'®;
report none of the above risk factors or exposures; the cause these patients have the same clinical course and outcome as
in more than 95% of CML cases is unknown. CML is not an Ph chromosome positive patients. Those who do not have an
inherited disease; rather it appears to be acquired, as sug- identifiable BCR-ABL gene are considered atypical CML, a
gested by the rarity of familial aggregations of CML"? and separate disease with a different prognosis and treatment (see
the failure of the second member of pairs of identical twins to Differential Diagnosis below).
develop the chronic leukemia.*? The Ph chromosome is not Depending on the precise break point in the BCR gene,
present in patients’ non-hematopoietic tissues, nor is it found the BCR—ABL fusion protein can vary in size from 185 kDa to
in the parents or offspring of patients. 230 kDa. As stated in the preceding text, nearly all patients
with typical CML in chronic phase have the 210-kDa protein.
However, a subset of CML patients with a 230-kDa protein
Pathogenesis present with a lower WBC count, slower progression to blast
phase, and a better prognosis.”° Patients with Ph chromosome-
CML is a clonal stem cell disorder?! in which the abnormal
positive acute lymphoblastic leukemia may present with either
clone is characterized by the presence of the Ph chromosome.
the 210-kDa protein or a 190-kDa protein. This suggests that
The chromosomal translocation that creates the Ph chromo-
not all Ph chromosomes or BCR-ABL genes are identical, and
some is t(9;22), a reciprocal translocation between the long
also that fusion proteins of different sizes can be correlated
(q) arms of chromosome 9 and chromosome 22. A smaller
with different clinical outcomes.*’
piece of the long arm of chromosome 9 is broken off at band
The Ph chromosome has been found in neutrophil,
q34.1 and translocated to the long arm of chromosome 22; at
monocyte, B lymphocyte, erythrocyte, megakaryocyte, and
the same time, a larger piece of the long arm of chromosome
basophil precursors from CML patients’ blood and bone mar-
22 is broken at band g11.21 and moved to chromosome 9.
row. This suggests that the original cell in which the muta-
This results in a small, abnormal chromosome 22 (the
tion arose was a pleuripotential stem cell, before separation of
Ph chromosome; Fig. 17—2). The notation describing this
the lymphoid and the myeloid lineages. It is the presence
translocation, t(9;22)(q34.1;q11.1), defines both the translo-
of the Ph chromosome carrying the BCR-ABL gene that gives
cation and the specific break points in chromosomes 9 and 22.
the abnormal CML clone its growth advantage over normal
In the translocation, the ABL-/ gene, the cellular homologue
cells and allows them to replace the normal bone marrow
of the Abelson murine leukemia virus oncogene from chro-
elements.”
mosome 9, is brought into contact with the BCR (break
All leukemias appear to be maintained by a pool of self-
renewing malignant cells. In CML, immature granulocytes fill
the bone marrow and are released prematurely into the
peripheral blood in various stages of maturation.*? At the mol-
ecular level, it has been established that it is the chimeric
BCR-ABL fusion protein that plays the central role in
myeloid proliferation and transformation in CML. There is
direct evidence from in vitro studies that the BCR-ABL pro-
tein (p210°***") causes uncontrolled cell growth in
hematopoietic cell lines.”’ Introduction of the BCR-ABL gene
into the bone marrow of mice leads to the development of a
myeloproliferative syndrome that closely resembles human
CML, proving that the BCR-ABL gene alone is sufficient to
establish the leukemic phenotype in vivo. Effects of the
BCR-ABL fusion protein in the CML cells include increased
Figure 17-2 @ A metaphase spread from a patient with CML proliferative capacity, a slight delay in maturation,?! and a lack
showing t(9;22). The abnormal chromosomes 9 and 22 are marked of responsiveness to the normal regulators of growth (e.g.,
with arrows. The Philadelphia chromosome is the abnormal chro- cytokines or the bone marrow microenvironment).*?*° The
mosome 22 on the right; the normal chromosome 22 is on the left. abnormal protein also seems to prevent apoptosis (normal
Chapter 17 Chronic Myeloproliferative Disorders |: Chronic Myelogenous Leukemia aD
9 Hybrid
gene
ber-abl
123 4
Hybrid
mRNA
(spliced)
ber-abl
Normal
c-abl
mRNA
Inactive
Selective Tyr-phosphorylation protein kinase
advantage of protein(s) substrates AcIve
CML @ of Ph- regulating growth Sort
positive and/or differentiation kinase ber-abl c-abl
clone of hematopoietic 210 kD 145 kD
progenitor cells
A
Figure 17-5 ® Molecular basis of the Philadelphia (Ph) chromosome. (A) Sequence of molecular and biochemical events involved in gener-
ating the Ph chromosome and its phenotypic consequences. (B) Southern blot analysis of DNA from CML cells analyzed with a bcr probe to
show clonal rearrangements in the ber region. Lane 7: Ph-positive CML DNA showing one rearranged band; lane 2: Ph-positive CML as in
lane 1 but with a different break point in the bcr region; lane 3: Ph-negative leukemic cell DNA showing no rearranged bcr; and lane 4:
molecular weight markers. (From Greaves, MF: Cellular identification and markers. In Zucker-Franklin, D, et al [eds]: Atlas of Blood Cells.
Lea & Febiger, Philadelphia/EE edi-ermes, Milano, Italy, 1988, p 43, with permission.)
programmed cell death) in the CML clone.'*** The BCR-ABL sweats and weight loss; bone tenderness and aching related to
fusion gene also leads to defective adhesion of the CML clone marrow expansion; and complaints related to an accompanying
to bone marrow stromal components, resulting in the early anemia." On physical examination, patients usually have mini-
release of immature forms into the bloodstream.***° mal to moderate splenomegaly, and occasionally hepatomegaly
The genetics of CML can occasionally be even more as well. Lymphadenopathy, which is rarely seen except late in
complex. Approximately 5% of CML patients have the deleted the course of the disease, is associated with a poor prognosis.
portion of chromosome 22 translocated to other chromosomes, Some patients will have bleeding complications related to qual-
such as t(4;22), t(12;22), and t(19;22).*’ These are simple vari- itative or quantitative platelet disorders. Rarely, patients may
ant Ph chromosome translocations, whereas other patients show manifestations of leukostasis (increased blood viscosity
have complex variant translocations involving two or more secondary to a very high WBC count) with vaso-occlusion, such
chromosomes in addition to chromosome 22 (e.g., t(9;11;22)). as cerebrovascular accident, myocardial infarct, venous throm-
On occasion, the Ph chromosome may be “masked” by the bosis, priapism, visual disturbances, and pulmonary insuffi-
presence of a larger piece of chromatin material translocated to ciency. In the later stages, patients may occasionally present
the abnormal chromosome 22 from one of the other chromo- showing effects of basophilia with increased serum histamine,
somes involved in the rearrangement, giving a normal sized, including pruritus, diarrhea, and refractory peptic ulcer disease.
but abnormal, chromosome 22.*° The CML clone is geneti- On rare occasions, the presenting sign of a patient with CML is
cally unstable, and may accumulate additional mutations as it a granulocytic sarcoma (chloroma), a soft tissue tumor com-
progresses to accelerated and blast phase. posed of immature myeloid cells, so named because of the char-
acteristic green color of the tumor mass when exposed to air.
CML is characterized by three phases:
Clinical Features
1. Chronic phase
Patients with CML may be asymptomatic or symptomatic
2. Accelerated phase
at presentation. It is common for the disease to be discovered
3. Blast phase
incidentally during a routine physical examination, or routine
hematologic evaluation of an asymptomatic patient.” When Most patients (85%) are diagnosed in the chronic phase. Dur-
symptoms do occur, the most common complaints are general ing this phase, the disease may remain stable for several years
malaise; fullness in the upper abdomen with early satiety, and and is usually responsive to conventional chemotherapy. Pro-
loss of appetite related to splenomegaly and hepatomegaly; gression of CML to accelerated phase or blast phase, in the
complaints resulting from a hypermetabolic state such as night absence of therapy, usually occurs 3 to 5 years after onset.’
376 Chapter 17 Chronic Myeloproliferative Disorders I: Chronic Myelogenous Leukemia
Soe
We.
Sas of Chronic.
. Myelogenous Leukemia
Peaokeal Blood Bone Marrow
Neutrophilic leukocytosis Myeloid hyperplasia
with immature forms Blasts < 10% (in chronic
Basophilia/eosinophilia phase)
Thrombocytosis Minimal/no dysplasia
Anemia Increased megakaryocytes
Blasts < 10% (in Myelofibrosis (mild/
chronic phase) moderate)
Decreased LAP score Monocytes usually <3%
Increased lactate
dehydrogenase Genetic Studies
Increased uric acid Philadelphia chromosome
Increased vitamin B,,/ positive (90-95%) Figure 17-7 mA bone marrow aspirate from a patient with CML
transcobalamin BCR-ABL-positive in chronic phase shows granulocytic hyperplasia with orderly matu-
a 99%) ration through segmented neutrophils. Blasts are not significantly
LAP = fenocyte alkalineees
increased. A basophil is at upper left.
Prognosis
degree of neutrophilic leukocytosis and basophilia, while There are significant differences in clinical aggressiveness and
CML can present with increased platelet count, and occasion- prognosis between patients presenting with CML in chronic,
ally increased RBC count. The LAP score will be decreased accelerated, and blast phases. In addition, there can be signifi-
in CML and may sometimes be decreased in IMF as well, cant survival differences between patients presenting in the
while it is usually increased in PV. When the Ph chromosome same phase. Several systems have been proposed to predict
or the BCR-ABL gene is present, CML can be diagnosed with prognosis in CML patients. The system in widest use currently
confidence; otherwise, it may be better to diagnose chronic
is the Sokol score. The Sokol score takes into account age,
myeloproliferative disease, unclassifiable, and re-evaluate the splenomegaly, platelet count (platelets >700,000/uL), and blast
patient frequently. count to stratify patients into three prognostic groups with
median survivals of approximately 2.5, 3.5, and 4.5 years.
CHRONIC NEUTROPHILIC LEUKEMIA Although the Sokol score was derived by studying patients
Chronic neutrophilic leukemia (CNL) is a rare but related treated with older therapies in the 1970s and 1980s, recent stud-
chronic myeloproliferative syndrome that is characterized by ies suggest that it remains largely accurate in predicting out-
splenomegaly, peripheral blood neutrophilia, and neutrophilic come in patients treated with modern therapeutic regimens.
hyperplasia of the bone marrow.*° The absolute neutrophil Several factors may predict early blast transformation in
count in the peripheral blood is usually greater than CML. These poor prognostic indicators include the presence
25,000/uL, and bands are present, but the percentage of of karyotypic abnormalities in addition to a single Ph chromo-
immature myeloid cells (metamyelocytes, myelocytes, some or BCR-ABL gene; hepatosplenomegaly; thrombocy-
promyelocytes, and blasts) is usually low. Basophilia and topenia (less than 150,000/uL) or thrombocytosis (more than
eosinophilia are not seen. Unlike CML, CNL never shows a 500,000/uL); extreme leukocytosis (more than 100,000/uL);
Ph chromosome, and LAP score is usually increased.’ CNL peripheral blood blasts greater than 1% or bone marrow blasts
must be distinguished from CML and from reactive neu- greater than 5%; and peripheral blood basophils above
trophilia, which is far more common. 15%.*°* The tumor suppressor gene p53 is altered in 20% to
30% of CML patients in blast crisis. Mutations in the p53
CHRONIC EOSINOPHILIC LEUKEMIA gene may have prognostic significance because these muta-
Chronic eosinophilic leukemia (CEL) is another related syn- tions may induce drug resistance by interfering with normal
drome that is characterized by a clonal expansion of programmed cell death (apoptotic) pathways in leukemic
eosinophil precursors with increased peripheral blood CML cells.*”
eosinophils of greater than 1500/uL.*! There is no Ph chromo-
some, and blasts are less than 20% in blood and bone marrow.
If there is no evidence of clonality in the eosinophil line and Treatment
blasts are not increased, the term “idiopathic hypere-
osinophilic syndrome” is preferred.°’ Tissue damage occurs The treatment for CML has evolved hand in hand with increased
as a result of infiltration by eosinophils with release of understanding of the underlying molecular pathogenesis. Before
cytokines. CEL must be distinguished from CML and from the mid-1980s, patients with CML in chronic phase were treated
reactive eosinophilia. with oral chemotherapy in the form of hydroxyurea or
380. Chapter 17 Chronic Myeloproliferative Disorders |: Chronic Myelogenous Leukemia
busulfan, intended to reduce leukocytosis, thrombocytosis, and However, in a multicenter randomized trial in unrelated mar-
splenomegaly. Although such therapy frequently resulted in row donor transplantation, T-cell depletion did not reduce the
hematologic remission, Ph chromosome remained detectable in incidence of chronic GVHD or improve the survival in
blood and marrow cells, and the onset of blast phase was not patients who developed chronic GVHD.” In addition, long-
delayed.* term outcome of patients given transplants of mobilized blood
Interferon-alpha was the first agent to produce cytoge- or bone marrow reported that patients with advanced chronic
netic as well as hematologic remission in CML. When used in myeloid leukemia had higher leukemia-free survival rates
combination with low-dose cytarabine, it was able to induce with peripheral blood stem cell (PBSC) transplants than bone
complete hematologic response in 69% of patients and major marrow (BM) transplants.”? Leukemia-free survival rates
cytogenetic remission (defined as =66% normal metaphases) were lower for patients in the first chronic phase after PBSC
in 35% of patients.°° Although complete cytogenetic remis- transplants in comparison to BM transplants due to higher
sions (absence of Ph chromosome) were rare (13%), those rates of late transplant-related mortality.” Allogeneic stem
who obtained major cytogenetic remission had significant cell transplantation still remains the only cure for CML; how-
prolongation of survival as well as a lengthened time to blast ever, impressive results of imatinib in newly diagnosed
phase. One study showed that patients who achieved a com- patients has made imatinib the first line of treatment.’' The
plete or major cytogenetic remission exhibited a 72% 10-year effects of imatinib mesylate treatment before allogeneic trans-
survival rate.°’ However, the majority of patients, including plantation for CML is now being investigated.”
those who developed complete cytogenetic remissions, still
had molecular evidence of the BCR-ABL gene by polymerase
chain reaction (PCR) testing. Fade 3 alas: lod
Imatinib mesylate (Gleevec®), the first drug to be devel-
oped against a specific molecular target, has significantly
changed the treatment and prognosis for CML. Imatinib directly A 58-year-old white man presented to his physician for eval-
inhibits the mutant tyrosine kinase activity of the BCR-ABL uation of leukocytosis found incidentally on a complete
fusion gene, and is utilized to treat all phases of CML. In blood count performed at a health fair. He had no current
chronic phase CML patients, imatinib produces up to a 95% complaints, but when questioned, reported feeling of full-
complete hematologic response rate, complete cytogenetic ness in the left upper quadrant of his abdomen. He reported
responses in up to 81% of patients, and improved survival no recent fevers, night sweats, or weight loss. He worked as
rates.°? Patients who achieve complete cytogenetic response a supervisor in an automobile assembly plant. On physical
show 10-year survival rates of 78%.°' In the accelerated phase, examination, he was found to have splenomegaly, with the
one study reports a hematologic response rate of 82% and a spleen tip barely palpable below the costal margin. No
hepatomegaly or lymphadenopathy was found.
major cytogenetic response of 24%.°° However in blast phase,
Complete blood count (CBC) showed WBC 51,200/uL,
the overall hematologic response rate decreases to 55% in the
hemoglobin 13.1 g/dL, hematocrit 38.4%, mean corpuscular
myeloid variant and 70% in the lymphoid variant, with duration
volume (MCV) 90.7 fL, and platelet count 610,000/uL.
of response that is shorter than in the chronic phase.°? Whether WBC differential showed 64% segmented neutrophils, 8%
imatinib can cure patients with CML remains to be determined, bands, 5% lymphocytes, 4% monocytes, 4% eosinophils,
but it is likely that the attainment of cytogenetic remission in the 5% basophils, 2% metamyelocytes, 7% myelocytes, 1%
majority of patients receiving imatinib will translate into signifi- promyelocytes, and 1 nucleated RBC. LAP score was
cantly prolonged overall survival. Some patients’ disease has, over decreased.
time, become refractory to treatment with imatinib; however, A bone marrow aspirate and biopsy were performed, and
newer kinase receptor inhibitors have already been developed that showed a markedly hypercellular bone marrow with marked
can overcome imatinib resistance and restore cytogenetic remis- granulocytic hyperplasia and moderate megakaryocytic hyper-
sion, for example Dasatinib (BMS-354825).%° Imatinib will be plasia. Blasts constituted 6% of the cells present. Bone marrow
cytogenetics performed showed 46,XY,t(9;22) (q34.1;q11.2);
the mainstay of therapy for patients who are not candidates for
that is, a Philadelphia chromosome was present.
bone marrow transplant, or for whom no suitable donor can be
Treatment was begun with interferon-alpha and low-dose
found.
cytarabine with good hematologic response. However, the
At present, allogeneic bone marrow transplantation with patient subsequently had difficulty tolerating the treatment
an HLA-matched related or unrelated donor offers the only because of fever and malaise, and was temporarily lost to
proven cure for CML. Survival is improved if the transplant follow-up.
is performed earlier in the course of the disease, and in the He returned 18 months later complaining of fatigue and
chronic phase. Transplantation-related mortality is high fevers. Physical examination showed increasing splenomegaly
(14%), mostly due to graft-versus-host disease (GVHD) and as well as hepatomegaly. At this time, his CBC showed WBC
infection, and outcome is worse in patients older than the age 98,800/uL, hemoglobin 11.6 g/dL, hematocrit 34.9%, MCV
of 40.*’ Long-term survival rates as high as 78% have been 89.0 fL and platelet count 1,070,000/uL. WBC differential
showed 48% segmented neutrophils, 8% bands, 3% lympho-
reported.*’ Moreover, survival rates using unrelated matched
cytes, 4% monocytes, 3% eosinophils, 21% basophils, 3%
donors are approaching the rates seen with related donors.®
metamyelocytes, 8% myelocytes, 2% blasts, and 2 nucleated
The incidence of acute GVHD can be reduced by depleting
continued
the donor marrow of T-lymphocytes using various techniques.
Chapter 17 Chronic Myeloproliferative Disorders |: Chronic Myelogenous Leukemia 38|
RBCs. A repeat bone marrow aspirate again showed marked 6. When the above patient returned after being lost to
granulocytic and moderate megakaryocytic hyperplasia, now follow-up for 18 months, he was in:
with 14% blasts. a. Chronic phase
Treatment was begun with imatinib mesylate (Gleevec®), b. Accelerated phase
which had recently become available. The patient’s WBC c. Blast phase
count decreased and symptoms disappeared within a few d. Jersey City
weeks. He subsequently achieved complete hematologic 7. The worst prognosis in patients with CML is associated
and complete cytogenetic remission. RT-PCR analysis of the with:
patient’s bone marrow specimens continued to show a. Thrombocytosis
the presence of BCR-ABL fusion gene product, but he b. Blast phase
remained asymptomatic and in complete hematologic remis- c. The presence of the Ph chromosome
sion 3 years later. d. Splenomegaly
QUESTIONS ANSWERS
1. The translocation that results in the Philadelphia chro-
mosome is between chromosomes:
a. 21 and 22
b. 9 and 22
c. 8 and 14
d. 9 and 21
2. The blast phase of CML is: Sl
OS
see
ANAS
@)
>@n
i
a. Followed by the chronic phase and the accelerated
phase DISCUSSION
b. Seen only in atypical CML
c. The terminal phase of CML, characterized by The Philadelphia chromosome, the hallmark of CML, is
increased blasts (>20%) in the blood and bone marrow formed by the translocation of chromosomes 22 and
d. The earliest phase seen in typical CML 9, which results in the formation of a fusion gene
3. All of the following favors a diagnosis of CML over (BCR-ABL) and subsequent activation of the ABL tyrosine
leukemoid reaction except: kinase which results in uncontrolled myeloproliferation.
a. Absence of basophils and eosinophils in the peripheral Aside from the Philadelphia chromosome, splenomegaly
blood and a low LAP score within myeloblasts through neutrophils
b. A low LAP score in the peripheral blood favors a diagnosis of CML rather
c. Ph chromosome than a leukemoid reaction. Blast crisis in CML is the termi-
d. An enlarged spleen nal phase of CML, characterized by increased numbers of
4. The only proven curative treatment for CML at this blasts in the bone marrow and peripheral blood.
time is: Although it is clear that Gleevec® has prolonged the
a. Combination interferon-alpha with low-dose chronic phase of CML, blast crisis still occurs. Blast crisis
cytarabinel remains refractory to standard chemotherapy approaches.
b. Imatinib mesylate (Gleevec®) Allogeneic bone marrow transplant offers the best chance
c. Bone marrow transplantation for cure when performed during the chronic phase.
d. CML is not curable
5. At the molecular level, the aberrant conjoining of
genetic material from chromosome 9 and chromosome
22 results in a fusion gene called:
a. BCR-ABL gene
b. JAK2 gene
C.j03
d. The blast transformation gene
continued
382 Chapter 17. Chronic Myeloproliferative Disorders |: Chronic Myelogenous Leukemia
Orestiand
i. What is the M:E ratio in patients with CML (chronic ~c. Viruses
myelogenous leukemia)? d. All of the above
a. 1:10 7. Which of the following myeloproliferative disorders is
bales characterized by a decreased LAP score?
esOzt a. CML
aoe b. IMF
i) . What is the chromosomal abnormality in CML? CET
a. t(8;14) d. PV
Dat.22) 8. Most patients (85%) are diagnosed in which phase of
ceitiel2) CML?
d. Trisomy 12 a. Accelerated phase
. Which of the following is not consistent with leukemoid b. Chronic phase
reaction? c. Blast phase
a. High WBC count d. All of the above
b. Associated with a bacterial infection 9. The blast phase of CML is defined by what percent of
c. Presence of Philadelphia chromosome blasts found in the peripheral blood or bone marrow?
d. High LAP a. 50%
. All of the following are characteristics of chronic b. 10%
leukemias except: c. 80%
a. Insidious onset d. 20%
b. Mature leukemia cells 10. Which of the following is characteristic of the acceler-
c. Found in adults ated phase of CML?
d. Found in all ages a. Persistent thrombocytosis or thrombocytopenia
. Which phase of CML carries the worst prognosis and is b. Blasts greater than 5% in the peripheral blood or
generally unresponsive to treatment? bone marrow
a. Chronic c. Basophilia greater than 20%
b. Accelerated d. All of the above
c. Blast
d. Refractory See answers at the back of this book.
. Environmental factors that are associated with an
increased risk of developing leukemia include:
a. Ionizing radiation
_b. Chemicals and drugs
m The bone marrow in CML shows hypercellularity related a CML must be distinguished from the other chronic
to a marked granulocytic hyperplasia and a moderate myeloproliferative disorders. It must also be distinguished
megakaryocytic hyperplasia. Pseudo-Gaucher cells and from atypical chronic myelogenous leukemia, which
marrow fibrosis may also be present. lacks the BCR-ABL fusion gene.
m The Philadelphia chromosome (an abnormal chromosome = Imatinib mesylate (Gleevec) currently provides an effec-
22) results from a reciprocal translocation between chro- tive treatment for CML, which can produce long-term
mosomes 9 and 22, and contains the BCR-ABL fusion remissions. However, the only proven cure for CML is
gene. The product of this gene is a protein with tyrosine allogeneic bone marrow transplantation.
kinase activity that is the direct cause of the granulocytic
proliferation that characterizes CML. The BCR-ABL
fusion gene is almost invariably present in CML.
REFERENCES acute lymphoblastic leukemia. Cancer chronic myeloid leukemia. Cancer Genet
Genet Cytogenet 155(1):89, 2006. Cytogenet 173(2):97, 2007.
l . Dameshek, W: Some speculations on the . Quintas-Cardama, A, and Cortes, JE: . Sandberg, AA: The Chromosomes in
myeloproliferative syndrome. Blood
Chronic myeloid leukemia: Diagnosis Human Cancer and Leukemia. Elsevier,
O72 ooie and treatment. Mayo Clin Proc 81:973, New York, 1980.
. Goldman, JM: A unifying mutation in
2006. . Eaves, CJ, and Eaves, AC: Stem cell
chronic myeloproliferative disorders. . Faderl, S, et al: The biology of chronic kinetics. Baillieres Clin Haematol
N Engl J Med 352:1744, 2005. myeloid leukemia. N Engl J Med 10:233, 1997.
. Jaffee, ES, et al (Eds): Pathology and 341:164, 1999. . Gordon, MY, et al: Adhesive defects
Genetics of Tumours of Haematopoietic . Maloney, W: Radiation leukemia revis- in chronic myeloid leukemia. Curr
and Lymphoid Tissues, [ARC Press, ited. Blood 70:905, 1987. Top Microbiol Immunol 149:151,
Washington, DC, 2001. . Askoy, M, et al: Leukemia in shoework- 1989.
. Randolph, TR: Chronic myelocytic ers exposed chronically to benzene. . Strife, B, and Clarkson, A: Biology of
leukemia — Part I: History, clinical pre- Blood 44:837, 1974. chronic myelogenous leukemia: Is dis-
sentation, and molecular biology. Clin . Baikic, AG, et al: Cytogenetic studies in cordant maturation the primary defect?
Lab Sci 18(1):38, 2005. familial leukemias. Austral Ann Med Semin Hematol 25:1, 1988.
. Virchow, R: Weisses Blut. Froriep’s
IQEC, INES), Ooi) . Clarkson, B, and Strife, A: Cytokinetic
Notizen 36:151, 1845. considerations relevant to development
20. Jacobs, FM, et al: Chromosome abnor-
. Bennett, JH: Two cases of disease and malities in human cancer: Report of a of a successful therapeutic strategy in
enlargement of the spleen, in which patient with chronic myelocytic chronic myelogenous leukemia (CML).
death took place from the presence of leukemia and his nonleukemic monozy- Leuk Lymphoma 11 (Suppl) 1:101,
purulent matter in the blood. Edinburgh gotic twin. Cancer 19:869, 1966. 1993,
Med Surg J 64:413, 1845. Pb Rudduck-Sivaswaren, C, et al: Evidence . Gishizky, ML, and Witte, ON: Initiation
~— .Virchow, R: Weisses Blut und for deletion of 9q as a two-step process of deregulated growth of multipotent
Milztumoren. Med Z 16:9, 1847. in chronic myeloid leukemia. Clin Genet progenitor cells by bcr-abl in vitro. Sci-
oo. Wintrobe, MM: In Greer JP (Ed): 68(5):461, 2005. ence 256:836, 1992.
Wintrobe’s Clinical Hematology, ed 11. 2D: Batista, DA, et al: BCR/ABL rearrange- 34. Amos, TA, et al: Apoptosis in chronic
Lippincott Williams & Wilkins, ment in two cases of Philadelphia chro- myeloid leukemia. Br JHaematol
Philadelphia, 2004. mosome negative chronic myeloid DSK 7, WES,
\O. Nowell, P, and Hungerford, D: A minute leukemia: Deletion on the derivative 35) Verfaillie, CM: Biology of chronic myel-
chromosome in human chronic granulo- chromosome 9 may or may not be pre- ogenous leukemia. Hematol Oncol Clin
cytic leukaemia. Science 132:1497, 1960. sent. Cancer Genet Cytogenet North Am 12:1, 1998.
. Rowley, JD: A new consistent chromoso- 163(2):164, 2005. 36. Gordon, MI, et al: Altered adhesive
mal abnormality in chronic myelogenous 23. Melo, J, et al: The abl-bcr fusion gene is interactions with marrow stroma of
leukaemia identified by quinacrine fluo- expressed in chronic myeloid leukaemia. haematopoietic progenitor cells in
rescence and Giemsa staining. Nature Blood 81:158, 1993. chronic myeloid leukaemia. Nature
243:290, 1973. . Daley, GQ, et al: Induction of chronic 328:342, 1987.
ile Bartram, CR, et al: Translocation of myelogenous leukemia in mice by the . Oshimura, M, et al: Variant Ph transloca-
c-abl oncogene correlates with the p210bcr/abl gene of the Philadelphia tions in CML and their incidence,
presence of a Philadelphia chromosome chromosome. Science 247:824, 1990. including two cases with sequential lym-
in chronic myeloid leukemia. Nature . Loriaux, M, and Deininger, M: Clonal phoid and myeloid crises. Cancer Genet
BOC QTM MISS: cytogenetic abnormalities in Philadelphia Cytogenet 5:187, 1982.
2, Shtivelman, E, et al: Fused transcript of chromosome negative cells in chronic 38. Anelli, L, et al: A chronic myelocytic
abl and ber in chronic myelogenous myeloid leukemia patients treated with leukemia case bearing deletions on the
leukemia. Nature 315:550, 1985. Imatinib. Leuk Lymp 45(11):2197, 2004. three chromosomes involved in a vari-
. Zagaria, A, et al: Molecular cytogenetics 20. Pane, F, et al: Neutrophilic chronic ant. Cancer Genet Cytogenet 148(2):137,
characterization of deletions on der(9) in myeloid leukemia: A distinct disease 2004.
chronic myelocytic leukemia. Cancer with a specific molecular marker. Blood . Rabinowitz, I, and Larson, RS: Chronic
Genet Cytogenet 167(2):97, 2006. 88:2410, 1996. [Erratum Blood 89:4244, myeloid leukemia. In Greer JP, et al
. Costa, D, et al: Studies of complex 1997] (Eds): Wintrobe’s Clinical Hematology.
Ph translocations in cases with chronic . Gorusu, M, et al: On the genesis and Lippincott Williams & Wilkins,
myelogenous leukemia and one with prognosis of variant translocations in Philadelphia, 2004, p 2234.
384 Chapter 17 Chronic Myeloproliferative Disorders |: Chronic Myelogenous Leukemia
40. Spiers, A: Clinical manifestations of Sh Kantarjian, HM, et al: Important thera- leukemia with the Philadelphia chromo-
chronic granulocytic leukaemia. Semin peutic targets in chronic myelogenous some. N Engl J Med 344:1038, 2001.
Oncol 22:380, 1995. leukemia. Clin Cancer Res 13(4):1089, 64. Hampton, T: Looking beyond imatinib:
. Baccarani, M, et al: Evolving concepts 2007. Next line of targeted drugs for CML
in the management of chronic myeloid . Silver, RT, et al: Treatment of the chronic
nNHES, shows promise. JAMA 295:369, 2006.
leukemia: recommendations from an phase of chronic myeloid leukemia with 65. Hochhas, A, et al: Dasatinib induces
expert panel on behalf of the European an intermittent schedule of recombinant notable hematologic and cytogenetics
LeukemiaNet. Blood 108(6):1809, 2006. interferon alfa-2b and cytarabine. Leuk responses in chronic-phase chronic
. Kantarjian, IM, et al: Chronic myeloge- Lymphoma 44:39, 2003. myeloid leukemia after failure of ima-
nous leukemia in blast crisis: Analysis of ah HehlImann, R, et al: Randomized com- tinib therapy. Blood 109(6):2303, 2007.
242 patients. Am J Med 83:445, 1987. parison of busulfan and hydroxyurea in 66. Deininger, M, et al: The development of
. O'Malley, D, and Vardiman, J: Chronic chronic myelogenous leukemia: Prolon- imatinib as a therapeutic agent for
myelogenous leukemia and related disor- gation of survival by hydroxyurea. chronic myeloid leukemia. Blood
ders. In Kjedsberg, C, et al (Eds): Practi- Blood 83:398, 1993. 105(7):2640, 2005.
cal Diagnosis of Hematologic Disorders: . O’Brien, SG, et al: Imatinib compared GyE Radich, JP, et al: HLA-matched related
2 Malignant Disorders. ASCP Press, with interferon with low-dose cytarabine hematopoietic cell transplantation for
American Society for Clinical Pathology, for newly diagnosed chronic-phase chronic phase CML using a targeted
Chicago, 2006, pp 539-560. myeloid leukemia. N Engl J Med busulfan and cyclophosphamide prepara-
44. Armitage, J: Atlas of Clinical Hematol- 348:994, 2003. tive regimen. Blood 102:31, 2003.
ogy. Lippincott Williams & Wilkins, 57. Bonifaz, F, et al: Chronic myeloid 68. Davies, S, et al: Equivalent outcomes in
Philadelphia, 2003. leukemia and interferon-alpha: A study patients with chronic myelogenous
5. Dosik, H, et al: Acquired lipidosis: of complete cytogenetic responders. leukemia after early transplantation of
Gaucher cells and ‘blue cells” in chronic Blood 98:3074, 2001. phenotypically matched bone marrow
granulocytic leukemia. Semin Hematol . Lee, M, et al: Detection of minimal
Nn(oe) from related or unrelated donors. Am J
SEOS), MTR. residual disease by polymerase chain Med 110:339, 2001.
46. Castro-Malispina, H, and Moore, MA: reaction in Philadelphia chromosome- 69. Pavletic, SZ, et al: Influence of T-cell
Pathophysiological mechanisms operat- positive chronic myelogenous leukemia depletion on chronic graft-versus-host
ing in the development of myelofibrosis; following interferon therapy. Blood disease: Results of a multicenter random-
role of megakaryocytes. Nouv Rey Fr TSE), MIS. ized trial in unrelated marrow donor
Hematol 24:221, 1982. Se). Kantarjian, H, et al: Hematologic and transplantation. Blood 106(9):3308, 2005.
47. Specchia, G, et al: Derivative chromo- cytogenetic responses to imatinib mesy- 70. Schmitz, N, et al: Long-term outcome of
some 9 deletions in chronic myeloid late in chronic myelogenous leukemia. patients given transplants of mobilized
leukemia are associated with loss of N Engl J Med 346:645, 2002. blood or bone marrow: A report from the
tumor suppressor genes. Leuk 60. Kantarjian, H, et al: Imatinib mesylate International Bone Marrow Transplant
Lymphoma 45(4):689, 2004. therapy improves survival in patients Registry and the European Group for
48. Huntly, BJP, et al: Double jeopardy from a with newly diagnosed Philadelphia Blood and Marrow Transplantation.
single translocation; deletion of the deriva- chromosome-positive chronic myeloge- Blood 108(13):4288, 2006.
7
tive chromosome 9 in chronic myeloid nous leukemia in the chronic phase. — . Apperley, JF: Managing the patient with
leukemia. Blood 102(4):1160, 2003. Cancer 98:2636, 2003. chronic myeloid leukemia through and
49. Randolph, TR: Chronic myelocytic 6 i . Kantarjian, H, et al: Complete cytoge- after allogeneic stem cell transplantation.
leukemia-part II: Approaches to and netic and molecular responses to Hematology Am Soc Hematol Educ Pro-
molecular monitoring of therapy. Clin interferon-alpha-based therapy for gram 1:266, 2006.
Lab Sci 18(1):49, 2005. chronic myelogenous leukemia are asso- WP. Oehler, VG, et al: The effects of imatinib
50. Vardiman, JW, et al: The World Health ciated with excellent long-term progno- mesylate treatment before allogeneic
Organization (WHO) classification of the sis. Cancer 97:1033, 2003. transplantation for chronic myeloid
myeloid neoplasms. Blood 100(7):2292, . Talpaz, M, et al: Imatinib induces leukemia. Blood 109(4):1782, 2007.
2002. durable hematologic and cytogenetic
5 _ . Sims, KL: Peripheral eosinophilia and responses in patients with accelerated
diagnosis of hypereosinophilic syn- phase chronic myeloid leukemia: Results
drome. Lab Med 37:440, 2006. of a phase 2 study. Blood 99:1928, 2002.
. Tefferi, A: Blood eosinophilia: A new . Druker, B, et al: Activity of a specific
paradigm in disease classification, diag- inhibitor of the BCR-ABL tyrosine kinase
nosis, and treatment. Mayo Clin Proc in the blast crisis of chronic myeloid
80:75, 2005. leukemia and acute lymphoblastic
Chronic
Myeloproliferative
Disorders II
Polycythemia Vera, Essential
Thrombocythemia, and Idiopathic
Myelofibrosis
Kathrina Chua, MD
Meyer R. Heyman, MD
Introduction to OBJECTIVES
Myeloproliferative
Disorders . At the end of this chapter, the learner should be able to:
Historical Perspective . Describe the origin of myeloproliferative disorders.
—"
Case Study 2
Case Study 3
Table 18-1
Idiopathic Polycythemia Essential
Myelofibrosis Vera Thrombocythemia
chronic eosinophilic leukemia (CEL)/hypereosinophilic syn- Study Group (PVSG) in 1967.’ Unfortunately, the group was
drome (HES) and unclassifiable myeloproliferative diseases. In disbanded when they were unable to obtain funding. However,
addition, a category of myelodysplastic/myeloproliferative dis- the data they generated are still used to make decisions regard-
eases was created to encompass juvenile myelomonocytic ing the management of PV.
leukemia, atypical chronic myeloid leukemia (lacking the 9;22
translocation) and chronic myelomonocytic leukemia.’ A dis- Definition
cussion of these rare additional entities is beyond the scope of
this chapter. The reader is referred to the WHO Classification PV, polycythemia, or polycythemia rubra vera is a chronic
of Tumours of Haematopoietic and Lymphoid Tissues.” The abnormality of the hematopoietic stem cell characterized by
2008 WHO classification has replaced the term CMPD with uncontrolled proliferation of erythroid, granulocytic, and
“myeloproliferative neoplasms (MPN)”. In addition, the 2008 megakaryocytic cells. PV should be differentiated from sec-
WHO diagnostic criteria for the traditional BCR-ABL negative ondary polycythemia, in which only the erythrocytes are
CMPDs (PV, ET, IMF) and the CEL/HES CMPDs have been increased in number and from relative erythrocytosis, where the
revised. Table 18—2 compares the 2001 and 2008 WHO classi- increase in hematocrit is secondary to a decrease in plasma vol-
fications of MPN. ume. PV is compared with secondary (hypoxic erythrocytosis)
and relative erythrocytosis in Table 18-3. The updated PVSG
criteria for the diagnosis of PV include the following: (1) an
Polycythemia Vera increase in red cell mass (>36 cc/kg in men and >32 cc/kg
Historical Perspective in women), (2) absence of causes of secondary polycythemia,
(3) splenomegaly, (4) thrombocytosis, (5) leukocytosis, and
Vaquez in 1892 and Osler in 1903 were the first to describe (6) normal or decreased erythropoietin levels (Table 18—-4).*
polycythemia vera (PV) as a chronic disease characterized by The WHO revised the criteria for the diagnosis of PV in
cyanosis, polycythemia, and splenomegaly.** The accompany- 1999, with the addition of other criteria including bone mar-
ing increase in granulocytic and megakaryocytic precursors was row panmyelosis with erythroid and megakaryocytic prolifer-
described by Turk a year later in 1904.° It was not until 1935 that ation and erythropoietin independent endogenous erythroid
marrow fibrosis was added to the hematologic findings by colony growth both in vivo and in vitro.* The discovery of the
Hirsch.° Wasserman formally founded the Polycythemia Vera genetic abnormality (JAK2 exon 12 mutations) in almost all
Table
18-2 World Health Organization Classification a
2001 28
Hype
Stayea , ad
i leiaeih adbof
Clinical Features
Cyanosis Present May be present
Heart or lung disease Absent Present Absent
Splenomegaly Present in 75% Absent Absent
Hepatomegaly Present in 35% Absent Absent
Laboratory Features
Red cell mass Increased Increased Normal
Erythropoietin Decreased (rarely normal) Increased (rarely normal) Normal
Arterial O, saturation Normal Decreased Normal
Leukocyte count Increased in 80% Normal Normal
Platelet count Increased in 50% Normal Normal
NRBCs, poikilocytes Often present Absent Absent
LAP Increased in 70% Normal Normal
Bone marrow Hypercellular; increased Increased erythropoiesis Normal
erythropoiesis and myelopoiesis;
increased megakaryocytes; fibrosis
Serum vitamin B,, Increased in 75% Normal Normal
Culture studies Autonomous, erythroid proliferation EPO-dependent colony Not applicable
formation 4
NRBCs 2 peclenrea red blood oe EPO ==eee Tape epee alkalineHela
of the patients with PV, led to the revisions found in the 2008 predominance in men.’ PV is seen in all age groups, includ-
WHO diagnostic criteria for PV. Table 18—5 compares the ing young adults and occasionally children.'’ Although rare,
2001 and 2008 WHO criteria for the diagnosis of PV. there have been reports of familial incidence of PV.'' The
disease also appears to be more common in Jewish individu-
Incidence als of Eastern European descent.'*
Al. Elevated RBC mass >25% above mean normal Major Criteria:
predicted value, or Hb >19.5 g/dL in men, 16.5 g/dL 1. Hgb >18.5 g/dL (men)
in women >16.5 g/dL (women)
or
A2. No cause of secondary erythrocytosis, including: Hgb or Het >99th percentile
Absence of familial erythrocytosis of reference range for age,
No elevation of erythropoietin because of: sex or altitude of residence
Hypoxia (arterial po, 92%) or
High oxygen affinity hemoglobin Hgb >17 g/dL (men),
Truncated erythropoietin receptor or >15 g/dL (women)
Inappropriate erythropoietin production by a tumor if associated with a sustained
increase of =2 g/dL from baseline
A3. Splenomegaly that cannot be attributed to
A4. Clonal genetic abnormality other than Ph chromosome correction of iron deficiency
or BCR-ABL fusion gene in marrow cells or
AS. Endogenous erythroid colony formation in vitro Elevated red cell mass
>25% above mean normal
B1. Thrombocytosis >400 * 10°/L predicted value
BEV iB Ce De al O2/le
B3. Bone marrow biopsy showing panmyelosis with promi- 2. Presence of JAK2V617F
nent erythroid and megakaryocytic proliferation or similar mutation
B4. Low serum erythropoietin levels
Minor Criteria:
Diagnose PV when Al + A2 and any other category A are 1. BM trilineage myeloproliferation
present or when Al + A2 and any two of category B are 2. Subnormal serum erythropoietin level
present. 3. EEC growth
n
IMF (35% to 57%) depending on the progressio
a pharmacological inhibitor of the Janus kinase, as well as
75%), and
of the disease. '620!
short interfering RNAs (siRNA) against JAK2. JAK2 is a
cytoplasmic tyrosine kinase encoded by a gene on the short
arm of chromosome 9. A unique mutation in the JAK2 gene Clinical Features
(found in greater than 90% of PV patients) leads to a pheny-
lalanine to valine substitution at position 617 (V617F) and PV is a chronic disease that usually has an insidious onset.
to constitutive activation of the JAK2 kinase, which results Many patients are asymptomatic and are diagnosed incidentally
in activation of the downstream pathways STATS, PI3K, on routine blood work drawn for other evaluations. Patients may
AKT (also known as protein kinase B), mitogen activated present with thrombosis and/or bleeding secondary to erythroid
protein kinase (MAPK), and extracellular signal-regulated expansion, hyperviscosity, and elevated platelets. Symptoms
kinase (ERK), all of which are implicated in erythropoietin may include headache, epistaxis, ischemic or hemorrhagic
signaling.”' It is likely that activation of these downstream stroke, angina, myocardial infarction, or claudication (cramping
pathways as a result of the JAK2 mutation allows for or pain in leg muscles). Patients with underlying atherosclerosis
the erythropoietin independent production of red cells as are most likely to experience these symptoms. Thrombosis is the
seen in PV.*° most common complication in PV, and occurs in one-third of
Discovery of this mutation may allow for the design of the patients.*? Unusual sites of venous thrombus formation,
an inhibitor of Janus kinase, which could result in pharmaco- including hepatic vein, mesenteric venous, and portal vein
logic control of the erythroid, myeloid, and platelet prolifera- thrombosis are common complications of untreated PV. Hepatic
tion characteristic of PV. This acquired mutation was found vein thrombosis, leading to Budd—Chiari syndrome, has been
by various investigators, not only in the majority of patients reported in up to 10% of PV patients in one series that evaluated
with PV (65% to 97%), but also in patients with ET (23% to 140 PV patients.
390 Chapter 18 Chronic Myeloproliferative Disorders Il: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis
In one review, up to 30% of PV patients had, in decreas- are measures of concentration, it is important to separate
ing order of frequency, headache, weakness, pruritus, dizzi- hemoconcentration from a true increase in red cell mass. The
ness, and sweating. Pruritus, especially noted after a warm correlation between Hct and red cell mass at hematocrits
bath, is thought to be secondary to cutaneous mast cell between 42 and 55 is poor, and in some cases determination
degranulation causing the release of histamine, fibrinolytic of the red cell mass by chromium hemodilution is required.
factors, or prostaglandins, and is almost diagnostic of PV.” This test is not always available in many centers. Particular
Erythromelalgia (erythema and painful burning in the hands attention to patients on diuretics or others at risk for hemo-
and feet) is common in PV and is thought to be secondary to concentration (i.e., diabetics, elderly) is necessary to avoid
platelet thromboxane secretion.*° Characteristically, these inappropriate diagnosis. Attention must also be paid to the
symptoms are ameliorated by the administration of low-dose mean corpuscular volume (MCV), because in PV, the absence
aspirin (81 mg). Other complications of PV include gout and of iron may slow but not stop the continued production of
an increased risk for peptic ulcer disease. Acute gouty arthri- microcytic red cells. A normal hematocrit with profound
tis occurs in 5% to 10% of patients with PV and is secondary microcytosis is virtually diagnostic of PV patients with iron-
to increased nucleoprotein turnover. Peptic ulcer disease was deficient erythropoiesis secondary to gastrointestinal blood
thought to possibly be due to increased histamine release loss, which is frequent in patients with PV (Fig. 18-1).
from basophils. Patients with increasing splenomegaly may Assays for serum erythropoietin levels are now readily
develop early satiety (fullness) and abdominal pain symptoms available and are accurate and reproducible. They can be
provoked by splenic infarction. There is a marked increase in helpful in separating PV from other causes of polycythemia.
perioperative morbidity and mortality due to increased post- In PV erythropoietin levels are characteristically low. In one
operative hemorrhage and thrombosis in patients operated on study, the sensitivity and specificity of serum erythropoietin
without proper control of red cell mass. below the reference range of normal for the diagnosis of PV
Splenomegaly is the most common physical finding in was 64% and 92%, respectively. When patients were tested
PV (50% to 80%). However, in keeping with other myelopro- on two occasions the sensitivity increased to 72%. Some
liferative disorders, the manifestations of splenomegaly, patients with normal levels initially were found to have low
myeloid metaplasia, and myelofibrosis are variably expressed values on repeat testing. Therefore, when erythropoietin lev-
at diagnosis and throughout the course of the disease. Most els are low, one can be relatively certain of the diagnosis of
commonly at the time of initial presentation, the degree of PV. Interestingly, the serum erythropoietin level was found
extramedullary hematopoiesis is usually mild, and marrow to be low even when hemoglobin levels were normal after
fibrosis is most often minimal. In the polycythemic phase of venesection.””
the disease, modest hepatomegaly may be observed as well as Characteristically, at presentation there is increased red cell
facial plethora (ruddy cyanotic complexion) secondary to production in intramedullary sites. The erythrocytes are normo-
increased vascular congestion. Hypertension can occur in cytic, normochromic, and have a normal life span. As the dis-
50% of patients, again secondary to vascular congestion. ease progresses, extramedullary ineffective hematopoiesis leads
Patients may also have skin excoriations (abrasions), optic to an increasing anisocytosis and poikilocytosis, as well as
fundi vessel engorgement, and gouty arthritis and tophi. How- shortened red cell life span secondary to splenic sequestration.
ever, 15% to 20% of patients transform to a spent phase with Many patients demonstrate the microcytosis and hypochromia
progressive anemia and increasing splenomegaly. The spent associated with iron deficiency, with low serum iron, decreased
phase of PV is associated with increased marrow fibrosis and mean corpuscular volume (MCV), and decreased mean corpus-
extramedullary hematopoiesis primarily in the liver and cular hemoglobin concentration (MCHC) occurring in about
spleen. The spent phase of PV is often referred to as post poly-
cythemic myeloid metaplasia and myelofibrosis, and it can be
indistinguishable from idiopathic myeloid metaplasia and
ER
0:5,
myelofibrosis both clinically and by laboratory findings. The
spent phase is characterized by constitutional symptoms (fever,
night sweats, and weight loss), worsening hepatosplenomegaly
a
e
secondary to increased extramedullary hematopoiesis,
and resultant portal hypertension with ascites, variceal hemor- % rs
rhage, and portal systemic encephalopathy. The risk of
leukemic transformation in PV at 20 years was found to be
approximately 15%.*°
Laboratory Findings
7. Se
The most diagnostic laboratory findings in PV is an elevated 2
RBC count, hemoglobin (Hgb), and hematocrit (Hct) with
ee
accompanying moderate thrombocytosis (60% of cases) and
leukocytosis (40% of cases). The Het is often higher than Figure 18-1 @ Peripheral blood smear seen in polycythemia vera.
58% in men and higher than 52% in women. Because these Note hypochromia and increased cellularity. (magnification x 400)
Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis Bed |
one-half of patients. This iron deficiency is attributed to a shift platelets can occasionally be found. The bone marrow charac-
of iron into the expanding erythroid mass and also to gastroin- teristically shows panmyelopathy with erythroid, myeloid,
testinal blood loss, possibly aggravated by platelet dysfunction. and megakaryocytic hyperplasia, in contrast to secondary
The reticulocyte count is usually normal, and only rarely are erythrocytosis where only erythroid hyperplasia is evident
immature erythrocytes found in the peripheral blood. (Fig. 18-2). The megakaryocytes are often increased in
Relative and absolute granulocytosis occurs in approxi- number and are atypical, with deeply lobated nuclei. They are
mately two-thirds of the patients. The elevation in the total often arranged around marrow sinusoids or in a paratrabecu-
WBC count is usually moderate, with counts in the range of lar ___location. In addition to the striking increase in the number
12 to 25 X 10°/L, as per the PVSG criteria. However, because of megakaryocytes, these cells are often increased in size.
only the neutrophils are increased in PV, the total WBC may Marrow iron stores, demonstrated by Prussian blue staining,
not accurately reflect disease activity. Therefore, newer crite- are reduced or absent. This decrease results from the shift in
ria have replaced the total WBC count with an absolute neu- iron into the expanded red cell mass. Stainable iron may be
trophil count > 10 x 10°/L.*8 Occasionally, basophilia and entirely absent in patients with chronic blood loss or after
eosinophilia are apparent, and a few metamyelocytes, myelo- multiple phlebotomies. Fibrosis is rare (less than 5%) early in
cytes, and even more immature cells may occasionally be the course of the disease, but may increase to 20% at 10 to
seen on examination of the peripheral smear. 15 years, and can reach up to 50% after 20 years with progres-
Thrombocytosis is present at the time of diagnosis in sion to the spent phase. Serial biopsies performed over a
about one-half of the patients with PV. The platelet count is period of many years showed progressive increase in reticulin
most often moderately elevated, with counts between 450 and deposits even before the spent phase develops. As the disease
800 < 10°/L, but in about 5% of the patient’s platelet count runs its course, cellularity usually decreases, although
exceeds 1000 < 10°/L. Platelet life span may be shortened in megakaryocytosis may persist.
proportion to the extent of pooling in the spleen. Morpholog- The leukocyte alkaline phosphatase (LAP) score is ele-
ical alterations of platelets include the presence of giant vated in PV (97% of cases), in contrast to CML in which the
platelets as well as deficient granulation. Most patients with LAP score is decreased (Fig. 18-3). Uric acid, LDH, and
PV form spontaneous megakaryocytic colonies. Platelet vitamin B,, levels can be elevated as well, secondary to high
aggregation studies may be abnormal but are poorly predic- cellular turnover. Two of the three vitamin B,,—binding pro-
tive of the risk of thrombosis or hemorrhage and are not rou- teins, transcobalamin I and III, are frequently elevated in PV,
tinely done as part of the evaluation of PV. In most instances, as in other MPDs. Transcobalamin HI is the binding protein
the bleeding time is normal despite in-vitro platelet functional most commonly elevated in PV, whereas transcobalamin I is
abnormalities. Moreover, in general, the bleeding time is a predominantly increased in chronic myelogenous leukemia.
poor predictor of spontaneous or surgical bleeding. These increased serum values can be attributed to the exces-
The prothrombin time (PT), activated partial thrombo- sive granulocyte turnover seen in myeloproliferative disor-
plastin time (APTT), thrombin time (TT), and fibrinogen lev- ders. Furthermore, the unsaturated B,,-binding capacity
els are generally normal in patients with PV. When performing (UB,,BC) is increased in approximately 75% of patients.
coagulation tests on PV patients, the anticoagulant-to-blood Unfortunately, neither LAP score nor vitamin B,, studies are
ratio must be maintained at a 1:9 ratio. When the hematocrit is sensitive or specific enough to be used as diagnostic tests in
high as in the case of erythrocytosis, the plasma volume is PV and are not included as major or minor criteria in newer
decreased relative to the increase in packed red cells. This classifications and diagnostic algorithms. Hyperuricemia and
results in an increase in the amount of citrate anticoagulant rel-
ative to the smaller amount of plasma per unit volume of
blood. This may result in prolongation particularly of the
APTT and require a reduction in the amount of citrate in the
specimen tube. The clinician should alert the lab to this possi-
bility. The following is a formula to correct the amount of cit-
rate anticoagulant to use when the Hct is greater than 55”:
V(100—Het)
s 2s
9=
a
(595—Hect)
The RBCs in the peripheral smear are generally normo- Figure 18-2 lf Bone marrow showing panhyperplasia in poly-
cytic, normochromic. Occasionally, basophilia and immature cythemia vera. Note increased number of megakaryocytes (arrows).
granulocytes may be seen, but not circulating blasts. Giant H&E stain (low power)
Myelofibrosis
392 Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic
Differential Diagnosis
The differential diagnosis between primary (PV) and secondary
polycythemia may not always be straightforward. The arterial
oxygen saturation should be >92% as indicated in the PVSG
criteria. Patients with arterial oxygen saturations below 92%
should be suspected of having secondary polycythemia related
to hypoxia. Secondary polycythemia may be appropriate (sec-
ondary to hypoxia from chronic obstructive pulmonary disease,
Figure 18-3 ™ Leukocyte alkaline phosphatase (LAP) stain of right to left cardiac shunts, and hemoglobin with increased
peripheral blood showing increased activity in polycythemia vera oxygen affinity) or inappropriate as with erythropoietin secre-
(red staining). LAP negative stain in CML (bottom).
tion secondary to renal cysts, or hypernephroma. Please refer to
Table 18-3 for features that differentiate PV from secondary
polycythemia and relative erythrocytosis. A discussion of the
uricosuria are found in 40% of patients with PV at the time of
causes and management of secondary polycythemia is beyond
presentation. This reflects the increased synthesis and degra-
the scope of this chapter and the reader is referred to other
dation of cellular nucleotides and can be seen in other hyper-
reviews. Unlike secondary polycythemia, PV is a_pan-
proliferative marrow disorders. Most patients with elevated
myelopathy and is usually associated with thrombocytosis and
uric acid levels remain asymptomatic, but uncommonly,
leukocytosis.
clinical gout may develop. A low to normal erythrocyte
sedimentation rate (ESR) is commonly present in PV patients.
The increased Het, as well as the elevated ratio of red cell Treatment
membrane to plasma fibrinogen and globulins, may account
for this finding. The cornerstone of the treatment of PV is to provide sympto-
Peripheral blood assays for erythropoietin independent matic relief, minimize long-term complications, such as
EEC growth in vitro (as part of the PVSG criteria) can be thrombosis and hemorrhage, and to avoid increasing the risk
helpful in confirming the diagnosis, but are not readily avail- of leukemic transformation.*° The most common treatment
able. Another assay that is promising for the diagnosis of PV modality utilized in PV is phlebotomy. Reduction of blood
is the measurement of PRV-] mRNA in granulocytes. PRV-/ volume (usually one unit of whole blood—450 cc), can be
is a novel member of the urokinase-type plasminogen activa- performed weekly or even twice weekly in younger patients
tor receptor superfamily and is overexpressed in mature gran- to control symptoms. The Het target range is less than 45%
ulocytes in greater than 90% of PV patients and correlates for men, less than 42% for women, and less than 37% for
strongly with EEC growth in vitro.*°In one study using quan- pregnant women in the third trimester.*’” Occasionally more
titative reverse transcriptase-polymerase chain reaction urgent cytoreduction may be required as in patients with fre-
(RT-PCR) for PRV-1 mRNA, the assay had a very high sensi- quent transient cerebral ischemic attacks or unstable angina
tivity and specificity for the diagnosis of PV.*° The throm- pectoris. More frequent phlebotomy with smaller volumes
bopoietin receptor (c-Mp/) is essential for hematopoietic stem may be helpful in this situation. Removal of the red cells by
cell survival and differentiation including megakaryocyte and continuous flow centrifugation using some of the newer
Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis 393
pheresis machines allows for very rapid reduction of the red aggregation (via its inhibition of platelet cyclic AMP phospho-
cell mass with conservation of plasma volume and may be diesterase activity), has also been used in PV primarily for the
very helpful in such situations. Phlebotomy will inevitably control of thrombocytosis. In one study, 0.5 mg to 1.0 mg four
result in iron deficiency and reactive thrombocytosis. The lat- times a day reduced the platelet count by 50%, or less than
ter should not be mistaken as evidence of progressive disease. 600,000/mm°.7 Imatinib mesylate has been reported anecdo-
After achieving iron deficiency, phlebotomies often may be tally to reduce phlebotomy requirements.*°
required less frequently, at every 3 to 6 months. There Low-dose aspirin reduces the rate of thrombosis and
remains considerable controversy over the need for and effi- cardiovascular deaths in patients with PV receiving standard
cacy of the use of myelosuppressive therapy in PV. Although phlebotomy and supportive care. A European study randomized
myelosuppressive therapy (radiation and chemotherapy) may 519 PV patients (with no indication for anticoagulation and no
decrease the number of phlebotomies required, decrease the pre-existing clear indication or contraindication to aspirin) to
platelet count and perhaps control splenomegaly, it is unclear low-dose aspirin (100 mg daily) versus placebo. The aspirin-
whether such treatment will decrease the morbidity of PV treated group had 60% fewer major thrombosis and cardiovas-
(thrombosis and hemorrhage), delay or prevent the onset of cular deaths (3.2% versus 7.9%) after a 3-year follow-up. The
the spent phase, or prolong survival. Moreover, the early use therapeutic benefit was independent of the platelet count. Low-
of radiation and alkylating agents that cause DNA damage dose aspirin can also effectively control erythromelalgia and
have clearly been associated with an increased risk of other vasomotor symptoms. Higher doses (500 to 900 mg/day)
leukemic transformation. An early PVSG_ study, which increased the bleeding risk without any added benefit.“
randomized 400 patients to either phlebotomy alone, phle-
botomy plus chlorambucil, or phlebotomy plus radioactive
phosphorus (*’P), revealed that phlebotomy alone without Essential Thrombocythemia
cytoreductive therapy was associated with an increased risk
for thrombosis, especially in the elderly or those with a prior Historical Perspective
history of thrombosis.** On the other hand, patients who
The earliest descriptions of essential thrombocythemia was
received myelosuppressive therapy had a higher incidence of
made by di Guglielmo in 1920 and by Epstein and Goedel in
secondary malignancies including acute myeloid leukemia
1934 who referred to it as hemorrhagic thrombocythemia.*?°
(AML), lymphoma, and gastrointestinal and skin cancers.**
In retrospect, they likely identified thrombocytosis that was
As a result of this study, radioactive phosphorus and alkylat-
secondary to other disorders. It was not until 1960 that essen-
ing agents such as chlorambucil are no longer used in the
tial thrombocythemia was established as a separate disease
treatment of PV. This study, however, was biased by the fact
entity on a clinicopathologic basis.*”
that many of the patients were not phlebotomized to levels
(less than 45%) that are now known to be safer in PV. Thus
the increased thrombotic risk found in patients treated with Definition
phlebotomy alone in this trial may have been more likely the
result of inadequate phlebotomy. A relationship between the Essential thrombocythemia (ET) is a chronic myeloprolifer-
risk of thrombosis and the extent of thrombocytosis has not ative disorder characterized by marked thrombocytosis asso-
been determined in PV.*° ciated with abnormal platelet function and an increased risk
Currently, cytoreductive therapy is usually reserved for of thrombosis and hemorrhage. It was the last of the MPDs
high-risk patients who are older (greater than 60 years), have to be identified as a distinct entity (since thrombocytosis can
thrombotic risk factors; have had prior thrombotic events; be seen in CML, PV, and IMF), and is frequently thought
have increased platelet counts; or who are symptomatic with of as a diagnosis of exclusion without any defining cytoge-
thrombosis, hemorrhage, hypercatabolic symptoms, severe netic abnormalities. Diagnostic criteria that define ET was
pruritus, and painful splenomegaly. Hydroxyurea has been the proposed by the PVSG in the mid-1970s. These guidelines
traditional cytoreductive agent used in PV. Hydroxyurea can include:
reduce the thrombosis rate, normalize the platelet count and . Platelet count in excess of 600 x 10°/L (and generally
_—
spleen size, and ameliorate hypercatabolic symptoms. How- greater than 1000 x 10°/L)
ever, treatment with hydroxyurea has been associated with an . Megakaryocytic hyperplasia in the bone marrow
increased incidence of AML.*° However, there still remains . Absence of identifiable causes of reactive thrombocytosis
considerable controversy as to whether hydroxyurea increases . Absence of the Ph chromosome
the rate of leukemic transformation in PV. The available stud- Hemoglobin no higher than 13 g/dL or normal red cell mass
ies are contradictory and may be biased by patient selection. If . Absence of significant marrow fibrosis
there is a risk, it is likely considerably less than that of alkylat- . Presence of stainable iron in the marrow, or failure to
YAAK
WY
ing agents. Subcutaneous injections of interferon alpha can respond to oral iron supplements (Table 18-—6).**
also control blood counts, splenomegaly, and hypercatabolic
symptoms. However, side effects such as fever, fatigue, The WHO published a revised set of diagnostic criteria for
depression, anorexia, nausea and vomiting can limit its ET, incorporating the exclusion of certain chromosomal
usefulness.*! Anagrelide, an oral imidazoquinazoline deriva- abnormalities in 2002.* Table 18-7 compares the 2001 and
tive that decreases platelet production and inhibits platelet 2008 WHO diagnostic criteria for ET.
394 Chapter 18 Chronic Myeloproliferative Disorders Il: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis
2008
“Diagnosis of essential thrombocythemia requires meeting all four major criteria for the 2008 WHO diagnostic criteria.
Source: Modified from Tefferi, A, and Vardiman, JW: Classification and diagnosis of myeloproliferative neoplasms: The 2008
World
Health Organization criteria and point-of-care diagnostic algorithms. Leukemia advance online publication 20 September 2007;
doi: 10.1038/sj.leu.2404955
Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis 395
ET.’** As mentioned earlier, the JAK2 gene mutation has thrombocytosis and platelet functional defects all contribute
been found in 30% to 50% of patients with ET. Essential to the thrombohemorrhagic complications.
thrombocythemia has been divided into two groups based on Other signs and symptoms that have been observed in
the presence of endogenous erythroid colony growth and the this disease are recurrent abortions, fetal growth retardation,
presence of the JAK2 mutation. Those with EEC and the pruritus, gout, and priapism. Modest splenomegaly is present
JAK2 mutation are believed by some to have a greater risk of in approximately 40% of patients with ET.*’ Splenic atrophy
symptomatic hemorrhage and thrombosis.* resulting from splenic vascular thrombosis and silent infarc-
tions occur in up to 20% of patients.
Clinical Features
Laboratory Findings
With the introduction of automated instruments that routinely
perform whole blood platelet counts, asymptomatic patients The peripheral blood smear in ET will characteristically show
with coincidental high platelet counts are being discovered marked thrombocytosis (>600,000/uL) and platelet anisocyto-
more frequently, especially younger individuals. Approxi- sis with platelets ranging in size from tiny to large giant
mately two-thirds of patients are asymptomatic at diagnosis, platelets. Other abnormal morphological findings in the periph-
with the remaining one-third presenting with hemorrhagic or eral blood may include microthrombocytes, platelet aggregates,
vaso-occlusive symptoms, or both. The most common clinical abnormally granulated platelets, and megakaryocytic cytoplas-
complications in ET include thrombosis (11% to 25%) and mic fragments. A megakaryocyte fragment in the peripheral
hemorrhage (3.6% to 37%).°° blood is shown in Figure 18-4. Platelet anisocytosis, when pre-
In most instances bleeding is mild and manifestations are sent, correlates with an elevation of the platelet distribution
primarily mucocutaneous (epistaxis and ecchymoses); how- width (PDW). Enumeration of platelets in whole blood counters
ever, life-threatening hemorrhage may occur after accidental can be problematic due to variations in platelet size and sponta-
trauma or, rarely, after surgery. Bleeding of the gastrointesti- neous platelet aggregation. Large platelets and particularly
nal tract as well as esophageal varices bleeding has also been platelet aggregates may not be counted as platelets resulting in
reported. significant underestimation of the true platelet counts. Review
Thrombosis is the other major manifestation of ET and of peripheral smears for the presence of platelet aggregates is
is caused by intravascular clumping of sludged, hyperaggre- mandatory to avoid errors in platelet counting.
gable platelets. Thrombosis can occur in arterial, venous, or A mild normocytic, normochromic anemia may be present
microcirculatory locations. The most common types of major in up to 50% of patients although the hemoglobin value is not
thromboses include cerebrovascular accidents (stroke and usually less than 10 g/dL. Recurrent mucosal or gastrointestinal
transient ischemic attacks), myocardial infarction, and periph- bleeding leads to iron-deficiency anemia. The MCV and MCHC
eral arterial occlusion. Lower extremity deep venous throm- are decreased, with a microcytic, hypochromic blood picture
bosis and pulmonary embolism account for the majority of becoming apparent on examination of the peripheral smear.
venous events. Intra-abdominal venous thrombosis, including Erythrocyte morphological findings reflective of hyposplenism
mesenteric and hepatic vein thrombosis as seen in PV, is also (which occurs in the occasional patient with splenic infarction
common. Microcirculatory occlusions, causing headache, and atrophy) include the presence of Howell—Jolly bodies,
paresthesia, erythromelalgia (localized painful redness, burn- Pappenheimer bodies (siderotic granules), target cells, and
ing, and “pins-and-needles” tingling sensation), and acral acanthocytes.
cyanosis are often seen in ET. Erythromelalgia of the toes,
feet, and occasionally fingers is a characteristic vaso-occlusive
symptom and may progress to acral cyanosis and/or necrosis.
The involvement of the hands and feet may simulate a diabetic
neuropathy. Erythromelalgia, just as in PV, is a source of con- ~ Oo ote F~
v.
Leukocytosis is present in about one-third of patients, As mentioned earlier, 23% to 57% of patients with ET
with WBC counts rarely exceeding 50 X 10°/L. Neutrophilia carry a JAK2 gene mutation.”! The finding of the JAK2 muta-
is observed in the majority of patients with elevated WBC tion supports the existence of more than one type of etiology
counts, but a mild eosinophilia or basophilia, or both, may of this disease.2! Red cell mass determinations and erythro-
occasionally be seen. Rarely, nucleated red cells, and imma- poietin levels may be necessary in patients with borderline
ture granulocytes may also be evident. The LAP score is hematocrits to rule out PV.
variable, but most commonly is normal.
The bone marrow in patients with ET demonstrates a
marked increase in the megakaryocytic component. The
Differential Diagnosis
megakaryocytes are typically larger than normal and may be The differential diagnosis of ET from reactive thrombocytosis
dysplastic in appearance with abundant cytoplasm and hyper- can be difficult, and ET often remains a diagnosis of exclusion.
lobulated nuclei, usually occurring in loose clusters or dispersed Essential thrombocythemia must be differentiated from the
in the marrow (Fig. 18-5). There may be increased erythroid various causes of reactive thrombocytosis (Table 18-8). The
and granulocytic proliferation without dysplasia or increased features differentiating ET from reactive thrombocytosis are
myeloblasts. Significant marrow fibrosis points against a diag- summarized in Table 18-9. Thrombocytosis is often present in
nosis of ET, per PVSG criteria (see Table 18-6). Stainable iron patients with a wide variety of inflammatory, malignant, infec-
is normal to slightly decreased in most cases, and increased reti- tious, and trauma-associated conditions. Reactive thrombocy-
culin content is often seen. Marrow karyotype is generally nor- tosis, even when persistently present for weeks or months, is
mal; however, the deletion of the long arm of chromosome usually well tolerated in these patients and is usually unasso-
21 (21q_) has been reported in some patients. ciated with thrombosis or hemorrhage. Bone marrow exami-
Platelet function studies reveal a variety of abnormalities nation may not allow a distinction between ET and reactive
in some patients. Abnormal platelet aggregation to epineph- thrombocytosis and should be avoided when alternative etiolo-
rine, collagen, adenosine diphosphate (ADP), and ristocetin gies are readily apparent. The platelet count in secondary
are quite frequent. Studies have demonstrated both normal (reactive) thrombocytosis seldom exceeds 1000 X 10°/L, and
bleeding times (even in the case of patients with hemorrhagic commonly falls in the range of 500 to 750 X 10°/L; however,
tendencies) and prolonged bleeding times. Reduced platelet extreme thrombocytosis may also be present in reactive condi-
factor 3 (PF3), reduced platelet adhesion, low protein S levels, tions. Platelet morphology and function are generally normal
and nucleotide storage pool defects have all been reported in in chronic reactive states compared to the variety of abnormal-
association with ET. Although qualitative functional platelet ities seen in MPDs.
abnormalities have been described, they are insufficiently ET must also be differentiated from other chronic MPDs
reproducible to be useful in differential diagnosis. These with associated thrombocytosis and from the myelodysplastic
abnormalities are not useful in predicting thrombosis or hem-
syndromes (see Chap. 19) in which the platelet count can be
orrhage. In addition, the severity of thrombocytosis does not markedly elevated. When the platelet count is persistently
necessarily correlate with the presence, absence, or severity of greater than 600 X 10°/L and the bone marrow demonstrates
symptoms. Falsely elevated serum potassium and phosphorus
predominant megakaryocytic hyperplasia, the diagnosis of
levels will sometimes be seen due to platelet lysis in uncen-
ET should be investigated. Because there are no unique clin-
trifuged serum specimens.
ical, hematologic, or histopathologic findings in this disease,
it is by nature a diagnosis of exclusion. At presentation, the
hemoglobin level is normal or mildly decreased. In some
patients a determination of red cell mass may be necessary to
rule out PV with increased platelets. To ensure that masked
PV has not been overlooked in those patients having a normal
or decreased red cell mass as a result of iron deficiency, bone
Acute hemorrhage
Postsplenectomy and Hemolytic anemia
hyposplenism Myelodysplastic diseases
Postoperative Graft-versus-host disease
Malignancy Vitamin E deficiency
Chronic inflammatory disorders Hyperadrenalism
Figure 18-5 @ Essential thrombocythemia, bone marrow. Note Chronic infection Rebound recovery from
increased megakaryocytes. (From Hyun, BH: Morphology of Blood Iron-deficiency anemia thrombocytopenia
and Bone Marrow, American Society of Clinical Pathologists, Work- Drug-induced Exercise
shop 5121, Philadelphia, 1983, with permission.)
Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis 397
Treatment
Essential Reactive The therapeutic approach to ET depends on a number of factors,
Thrombocytosis Thrombocytosis including the patient’s age and childbearing potential, the height
Platelet count Frequent Infrequent of the platelet count, and, most importantly, the presence and
> 1,000,000 duration of symptoms. Treatment for ET is usually reserved for
Leukocytosis Frequent Can be present in high-risk patients who are 60 years of age or older, have a prior
reactive states history of thrombosis, and cardiovascular risk factors such as
Anemia Normocytic, Microcytic,
hypertension, hypercholesterolemia, and smoking. Low-risk
normochromic hypochromic
(secondary to patients are younger than 60 years, with no previous history of
iron deficiency) thrombosis, a platelet count of less than 1,500,000/uL, and no
Splenomegaly < 50% Not present cardiovascular risk factors. Intermediate—risk patients are those
Clustering Common Not present who do not qualify for either the low-risk or high-risk groups.**
megakaryocytes
Careful monitoring, without therapy specifically aimed at
in bone marrow
Bone marrow 20% Not present lowering the platelet count, is generally advocated for asympto-
fibrosis matic low-risk patients with extreme thrombocytosis. Young
patients with few or no symptoms do not require treatment
unless surgery is indicated or childbirth is imminent. In these
cases, plateletpheresis or a brief period of myelosuppressive
marrow aspirate with iron stains may be helpful. In addition, therapy may be useful in controlling the elevated platelet count.
if bone marrow aspirate is refused, a 1-month trial of iron Asymptomatic young patients should receive aspirin, which has
therapy should be administered. The response to iron therapy been shown to be beneficial in a randomized trial.“
should be carefully menitored, and if a rise in the Hct level Treatment of high-risk patients (older than age 60, with
and red cell volume is observed, the patient should be evalu- cardiovascular risk factors, or younger symptomatic patients)
ated for evidence of PV, evidence of blood loss, or both. is indicated to control hemorrhage and manitestations of
Chromosomal analysis of the bone marrow should be thrombosis and to control the progressive megakaryocyte pro-
performed to ensure that the Ph chromosome is not present. In liferation. Thrombotic events are far more common than
those patients with CML and thrombocytosis in whom the hemorrhagic events. It is interesting to note that the degree of
Ph chromosome is not demonstrable by standard metaphase thrombocytosis does not correlate with the risk of thrombosis.
cytogenetics, other findings are important for supporting the Acute hemorrhage and occlusive events, which occur in
diagnosis of CML. These include: myeloid hyperplasia, low approximately 30% of patients, indicate the necessity for
LAP score, the presence of BCR-ABL gene rearrangements by immediate therapy. Plateletpheresis achieves a dramatic
molecular techniques and moderate to marked splenomegaly. reduction in the platelet count in a matter of hours; however,
Early IMF is often associated with extreme thrombocy- this reduction is transient and therefore inadequate for long-
tosis. However, there is usually marked splenomegaly, a term control. In addition, the procedure is not cost-effective
leukoerythroblastic blood picture, teardrop poikilocytosis, and involves a difficult process if done four to five times
and the characteristic myelofibrotic involvement of the bone per week.
marrow (increased reticulin and collagen fibrosis). In ET, less Chemotherapy should be initiated in addition to platelet-
than one-third of the biopsy area demonstrates fibrosis. pheresis whenever thrombohemorrhagic complications
Other markers of the acute phase response such as develop, age is greater than 60, and the platelet count is more
interleukin-6 (IL-6), plasma fibrinogen, and C-reactive pro- than 1.5 x 10°/L. A number of effective myelosuppressive
tein are elevated in the acute phase response to infection, agents have been administered in the past (**P, Busulfan), but
trauma and inflammation, and may help in differentiating their use has been associated with an increased potential for
reactive thrombocytosis from ET. induction of acute leukemia. Common treatment options
Myelodysplastic syndromes associated with thrombocy- include cytoreductive agents such as hydroxyurea and anagre-
tosis usually present with a more severe degree of anemia that lide. The antimetabolite hydroxyurea is generally considered
is often macrocytic in appearance as compared with that seen to be the drug of choice because of its efficacy. Hydroxyurea,
in patients with ET. In addition, the presence of either cytoge- compared to no treatment, was shown to reduce the risk of
netic abnormalities (5q-, deletion 7, trisomy 8), or ringed thrombosis in high-risk patients from 24% to less than 4% at
sideroblasts in the bone marrow denotes a myelodysplastic a median follow-up of 27 months.* Reported incidence rates
syndrome as the cause of the associated thrombocytosis rather of leukemic conversion, when hydroxyurea is used alone in
than essential thrombocythemia (see Chap. 19). ET can reach up to 5%. Hydroxyuea should be administered
As previously noted, some patients with ET demonstrate at a daily dosage of 15 mg/kg of body weight, with adjust-
erythropoietin-independent endogenous erythmoid colony ment according to response. In 90% of patients, the platelet
398 Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis
count will be decreased to less than 600 X 10°/L in 2 to rebound thrombocytosis is inevitable without concomitant
6 weeks. A lower continuous dosage is required to maintain aggressive myelosuppression. If surgery is indicated, the platelet
disease control, because once this treatment is halted the count should be controlled preoperatively by use of platelet-
platelet count rises again. Common side effects of hydrox- pheresis or myelosuppression, or both. Platelet concentrates may
yurea are dose-related, reversible leukopenia, macrocytic red rarely be needed to provide normal functional platelets.
cell changes and anemia, and rarely, fever, rash, and leg ulcers.
The drug anagrelide is also used to treat thrombocytosis.
It is considered an excellent drug for long-term use, because
Idiopathic Myelofibrosis
it does not have mutagenic or leukemogenic potential. Ana- Historical Perspective
grelide can cause significant reduction in the platelet count, as
well as a reduction in both major and minor thromboembolic Idiopathic myelofibrosis (IMF) or idiopathic myeloid meta-
complications.°! Bleeding complications were common only plasia with myelofibrosis, was initially described by Heuck in
in those patients who were taking anagrelide along with low 1879.° He described the case of a 24-year-old butcher who
dose aspirin. Other side effects include headaches, palpita- had been afflicted with severe fatigue for 1 year. On examina-
tions, fluid retention, and diarrhea (which in 15% of patients tion, severe anemia, leukocytosis with myeloid immaturity,
are intolerable).°* However, a recent randomized trial compar- and marked hepatosplenomegaly were noted. The patient sur-
ing hydroxyurea with anagrelide showed that hydroxyurea vived for only 2 years and, on autopsy, was demonstrated to
with aspirin was superior to anagrelide with aspirin in pre- have severe osteosclerosis and extramedullary hematopoiesis.
venting thrombotic and hemorrhagic events in ET patients at Heuck thus concluded, based on the unique features of the
high risk for vascular events. case, that myelofibrosis with myeloid metaplasia was distinct
Alpha interferon has also been advocated for treatment in from leukemia. IMF was formally included as one of the
ET. This agent exerts an inhibitory effect on the growth of myeloproliferatives disorders in 1951.
megakaryocyte progenitors that correlates clinically with a
marked decrease in platelet count following treatment with Definition
interferon. Alpha interferon has also been shown to have an
overall response rate of 88% in ET patients. Aside from control- Chronic idiopathic myelofibrosis (CIMF) is characterized by
ling thrombocytosis and leukocytosis, reversal of splenomegaly the classic triad of findings of (1) fibrosis of the marrow, at first
was also noted.“ Treatment doses of 1 to 5 million units/day patchy and later widespread, that may or may not be accompa-
exert a dose-dependent inhibitory effect on thrombocytopoiesis. nied by bony sclerosis; (2) extramedullary hematopoiesis or
However, the treatment also causes moderate reduction of gran- myeloid metaplasia of the spleen and liver, giving rise to mod-
ulocyte levels and is associated with significant side effects, erate to marked splenomegaly and hepatomegaly; and
which include fatigue, depression, anorexia, weight loss, fevers (3) leukoerythroblastosis and teardrop poikilocytosis of the
and nausea. As with PV, these side effects have precluded wide- peripheral blood. The features of myelofibrosis as defined by
spread use of this agent in treating ET. However, it has been the American PVSG are described in Table 18-10.
effective and safe in pregnant patients with ET, where other CIMF, like the other myeloproliferative disorders, is a
agents such as hydroxyurea are contraindicated. clonal myeloproliferative disease. This has been demon-
Pipobroman (an oral piperazine derivative that functions as strated in female G6PD heterozygotes and also using other
an alkylating agent), is used widely in Europe for the treatment X chromosome-linked polymorphisms. It is characterized
of PV and ET. Although not available in the United States, pipo- by the proliferation of mainly granulocytes and megakary-
broman was found to be as effective as hydroxyurea. Pipobro- ocytes leading to the characteristic finding of bone marrow
man is an alkylating agent and thus may be associated with an fibrosis.
increased risk of leukemic transformation in ET. CIMF is known by at least 20 synonyms. Some of
Low-dose aspirin (100 mg/day) has been effective in pre- the most frequently employed are agnogenic myeloid
venting thrombosis in ET, with an acceptable risk for bleeding, metaplasia, myelosclerosis, osteosclerosis, chronic erythroblas-
if applied to patients with a platelet count <1000 x 10°/L and/or tosis, aleukemic myelosis, and chronic or primary myelofibro-
absence of bleeding history.°° An important role for aspirin may sis. The term myelofibrosis with myeloid metaplasia (MMM),
be found in patients with recurrent miscarriages, because this or just idiopathic myelofibrosis (IMF) for brevity, highlights the
treatment does not carry the risk of teratogenicity. In patients essential features of fibrosis and extramedullary hematopoiesis.
with markedly elevated platelet counts (1520 10°/L) the bind- CIMF can be divided into two stages, according to the
ing of high molecular weight von Willebrand multimers to the WHO criteria: a prefibrotic stage (p-CIMF) and a fibrotic stage
platelets produces in effect a type II von Willebrand’s disease (f-CIMF).* The classic triad of clinical findings is absent or
and its attendant bleeding risks.°’ Therefore aspirin should be minimal in the prefibrotic stage (hypercellular phase) of early
avoided in ET patients with platelet counts of this magnitude, CIMF. The p-CIMF stage is characterized by a granulocytic
until levels are reduced by myelosuppressive therapy. Occasion- and megakaryocytic myeloproliferation, decreased number of
ally patients with ET with life- or organ-threatening symptoms erythroid precursors, lack of fibrosis, and abnormalities in
such as transient cerebral ischemia, unstable angina, or impend- megakaryopoiesis characterized by atypical megakaryocytes.”
ing gangrene may require urgent lowering of the platelet count. Clinically the prefibrotic (p-CIMF) stage of CIMF mimics
Although platelet pheresis may be helpful, in this instance the early stages of ET and PV. The fibrotic (f-CIMF) stage is
Chapter 18 Chronic Myeloproliferative Disorders Il: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis 399
Pathogenesis
Recent evidence has indicated that IMF originates at the level
characterized by fibrosis of the marrow, extramedullary of the CD34* hematopoietic stem cell.”* As mentioned previ-
hematopoiesis (splenomegaly), and leukoerythroblastosis of ously, hematopoiesis in IMF, as in the other MPDs is clonal
the peripheral blood.* The Updated Cologne criteria has been and clonal chromosomal abnormalities were found in as many
used for the diagnosis of IMF as well as for staging purposes as 57% of patients with IMF, in one series.’ The most com-
(Table 18—11).° mon ones reported include: 13q—, 20q—, +8, +9, 12p—, and
or IMFDiagnosis
andStaging
Clinical Criteria
Al. No preceding or allied subtype of myeloproliferative disorders, CML, or myelodysplastic syndromes
A2. Early clinical stages
Normal hemoglobin or anemia, grade I: hemoglobin =12 g/dL
Slight or moderate splenomegaly on palpation or >11 cm on ultrasound scan or CT
Thrombocythemia, platelet count >400,000/,L
A3. Intermediate clinical stage
Anemia grade II: hemoglobin =10 g/dL
Definitive leukoerythroblastic blood picture and/or teardrop erythrocytes
Splenomegaly
No adverse signs (age >70 years, hemoglobin <10 g/dL, myeloblasts > 2% in peripheral blood, pronormoblasts > 2% in peripheral
blood, leukocytosis > 20,000/L, thrombocytopenia < 300,000/L, severe constitutional symptoms, massive splenomegaly,
cytogenetic abnormalities)
A4. Advanced clinical stage
Anemia grade IL: hemoglobin <10 g/dL
One or more adverse signs (age > 70 years, hemoglobin <10 g/dL, myeloblasts > 2% in peripheral blood, pronormoblasts
> 2% in peripheral blood, leukocytosis >20,000/L, thrombocytopenia <300,000/L, severe constitutional symptoms,
massive splenomegaly, cytogenetic abnormalities)
Pathologic Criteria
B1. Megakaryocytic and granulocytic myeloproliferation and relative reduction of erythroid precursors. Abnormal clustering and
increase in atypical giant-sized megakaryocytes containing cloud-like lobulated nuclei and definitive maturation defects
Source: Reprinted froth ene we F, and Barosi, G:Payslonbee withRipelold metaplasia: Diagnosis, prognostic
factors and staging. Semin Oncol 32:395, 2005, with permission from Elsevier.
400. Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis
abnormalities of chromosomes | and 7.” In a 2006 study, the hematopoiesis, splenomegaly and sometimes hepatomegaly is
JAK2 (V617F) mutation (described previously) was reported present. Splenomegaly related to IMF is present in 85% to 99%
in 56% of the patients with IMF.” Granulocytes of patients of patients at diagnosis and can be massive in about 10%.”
with CIMF had the JAK2 (V617F) mutation which correlated Symptoms due to splenomegaly include left upper quadrant
with the activation of cell signaling.'° This granulocyte acti- pain, early satiety and even left shoulder pain. Palpable
vation leads to abnormal trafficking of CD34* cells with an hepatomegaly is present in 40% to 70% of the cases
increased CD34 count in the peripheral blood.’ Granulocytes (Fig. 18-6). Although hepatomegaly is found in about 50% of
from CIMF are functionally activated as indicated by the patients, it is not generally excessive, but may be accompanied
increased values for LAP. LAP scores are significantly higher by mild to moderate jaundice, ascites, or both. Portal hyperten-
in myeloproliferative disorders than in reactive conditions. sion may develop as a result of increased splanchnic flow due
There is a direct relationship between JAK2 (V617F) muta- to splenomegaly and/or intrahepatic obstruction associated
tion and circulating CD34* cells and fibrosis. CD34* cell with extramedullary hematopoiesis in the liver*® (Fig. 18-7).
counts increase exponentially as the mutant alleles exceed Symptomatic patients complain of fever, anorexia,
50%. In one study, patients carrying the JAK2 (V617F) muta- weight loss, night sweats, pruritus, and bone pain which
tion in their granulocytes had higher PB CD34* cell counts represent the metabolic consequences of myelofibrosis.*!
than individuals without the mutation.’° This study indicated Extramedullary hematopoiesis can occur in almost any organ,
that the JAK2 (V617F) mutation may constitutively activate and may manifest itself as lymphadenopathy, pleural or peri-
granulocytes which mobilizes CD34 cells.’° Transition from cardial effusion, or ascites.**** The central nervous system
heterozygosity to homozygosity for the JAK2 (V167F) muta- can also be involved with increased intracranial pressure,
tion in granulocytes triggers significant CD34* cell mobiliza- altered sensorium, motor and sensory impairment, and even
tion. This results in higher CD34* cell count, which correlates cord compression due to extramedullary hematopoiesis in the
with progression to marrow fibrosis.’°
Unlike PV and ET which are characterized by hyperpro-
liferation, IMF is characterized by ineffective erythropoiesis
and ineffective megakaryopoiesis, suggesting that additional
genetic abnormalities may play a role in the pathogenesis of
IMF. Fibroblasts, however, are not part of the clonal process
and do not share the same clonal chromosomal abnormalities
supporting the theory that bone marrow fibrosis is a sec-
ondary reaction.’
A number of cytokines are involved in the pathogenesis
of fibrosis in IMF. These are all secreted from platelets and
megakaryocytes, although in some instances may also be
secreted from other cellular or stromal sources. These include
transforming growth factor B (TGF-B), platelet-derived
growth factor (PDGF), and basic fibroblast growth factor
(bFGF). TGF-B may possibly be the most important of the
three.” It is capable of promoting secretions of type I and type Figure 18-6 ™ Hepatosplenomegaly, a characteristic finding in
III collagen. Elevated levels of PDGF are found in all patients patients with idiopathic myelofibrosis with myeloid metaplasia
with IMF and are released from platelets and megakaryocytes (IMF/MM).
undergoing intramedullary death. PDGF stimulates fibroblast
proliferation and collagen secretion. bFGF moderates
megakaryocyte-stromal cell interaction resulting in prolifera-
tion of stromal cells such as fibroblasts.
Thrombopoietin promotes megakaryocyte growth and
development. However, neither mutations in the thrombopoietin
receptor nor autocrine stimulation of thrombopoietin has been
shown to play a role in the pathogenesis of CIMF.
Clinical Features
epidural space.** Extramedullary hematopoiesis can occur some cases of IMF, hemolysis is autoimmune in nature, and
within the pulmonary arterial circulation resulting in severe may be DAT positive with deposition of immunoglobulin
pulmonary hypertension. Rapidly developing extramedullary IgG, IgM, or complement on the erythrocyte surfaces.
hematopoiesis may occur in the liver with abnormal liver Hypochromic, microcytic anemia may occasionally occur
function, jaundice, portal hypertension, and hepatic failure.* secondary to gastrointestinal bleeding from peptic ulcer disease.
Symptoms of anemia such as weakness, pallor, lethargy, Megaloblastic anemia with macrocytes and occasionally hyper-
and dyspnea on exertion are common. Approximately 10% of segmented neutrophils, resulting from relative folate deficiency,
patients present with a serious bleeding diathesis secondary may occur due to increased folate utilization.
to thrombocytopenia or thrombocytosis, qualitative platelet The leukocyte count is variable in IMF. In about 50% of
defects, or coagulation abnormalities. Symptoms of bleeding cases the white blood cell (WBC) count exceeds 10.0 x
may be as minor as petechiae or ecchymoses, or as serious as 10°/L; in approximately 35%, the WBC count is normal; and
esophageal variceal bleeding. Infection may ensue related to in nearly 15%, the WBC count is below normal. Marked
immune deficiency, even when neutropenia is absent. leukocytosis (WBC greater than 30 x 10°/L) is initially pre-
Increased cellular turnover of nucleic acids can lead to hyper- sent in 11% to 13% of patients. Conversely, leukopenia can be
uricemia and secondary gout. Osteosclerosis, characterized by a seen in 8% of patients. As the disease evolves, the leukocyte
diffuse or patchy increase in bone density and increased promi- level declines, with an increase of immature myeloid cells
nence of bony trabeculae on radiologic studies, can be seen in dominating the peripheral blood picture, but not to the degree
IMF as well, and may be associated with severe bone pain.” seen in acute myelocytic leukemia. Rarely leukocytosis may
Various scoring systems involving the hemoglobin level, be extreme and may be mistaken for CML. The LAP score is
white count, constitutional symptoms and number of circulat- typically normal or moderately increased. Serum levels of
ing blasts have been used for prognosis in IMF. An example vitamin B,, are increased, but not to the degree found in
of this is the Lille System (Table 18—12).*° untreated CML. Eosinophilia and basophilia may be found
frequently as in other myeloproliferative diseases.
The platelet count may be normal, elevated, or decreased.
Laboratory Findings In approximately 50% of IMF patients, platelet counts range
Normocytic normochromic anemia is present in 50% to 90% of from 450 to 1000 X 10°/L at the time of diagnosis. Occasionally
patients at the time of diagnosis. Twenty-five percent of counts in excess of | million are present. As the disease pro-
patients have severe anemia with hemoglobin less than 8 g/dL. gresses, thrombocytopenia becomes increasingly prevalent.
The cause of the anemia in IMF is multifactorial and results Giant dysplastic platelets are often conspicuous, and megakary-
from additive effects of bone marrow failure, autoimmune
ocytic fragments or even dwarf megakaryocytes may be present
hemolysis, ineffective or dyserythropoiesis, and hypersplenism in the peripheral blood (Fig. 18-10). The disturbed platelet mor-
phology is reflected in abnormal platelet physiology with abnor-
(dilutional anemia).
With increasing disease, the morphological changes mal platelet functions present in as many as 50% of patients.
Spontaneous platelet aggregation may also occur. The bleeding
become increasingly abnormal, and the classic leukoerythro-
blastic blood picture appears (Fig. 18-8). This includes the time (a measure of platelet number and function) is prolonged in
up to 20% of patients, but correlates poorly with the risk of
appearance of abundant nucleated red cells, immature granulo-
bleeding. Although both thrombosis and bleeding may occur in
cytes, and teardrop shaped red cells in the peripheral blood
IMF, bleeding is particularly common, especially after splenec-
(Fig. 18-9). Improvement or even normalization of red cell
tomy. In patients who have undergone splenectomy, the number
morphology after splenectomy supports a causative relationship
of immature WBCs, poikilocytes, and morphologically abnor-
between splenomegaly and red cell morphology.
mal platelets increase because the filtering function of the spleen
Shortened red cell survival may result from ineffective ery-
is no longer present.
thropoiesis and hemolysis. Severe hemolytic anemia develops
Bone marrow aspirations are usually unsuccessful (“dry
in 15% of cases and is evidenced by marked reticulocytosis. It
tap”) secondary to myelofibrosis. The reticulin and collagen
is generally direct antiglobulin test (DAT) negative; however, in
fibrosis requires a needle biopsy for diagnosis. The bone mar-
row biopsy is hypercellular with an increase in neutrophils,
and atypical megakaryocytes in the prefibrotic phase.
Approximately 25% of patients present in the prefibrotic
stage. A left-shift may be observed without significant
increase in myeloblasts. Erythroid precursors are easily
found, but overall erythropoiesis is decreased. Megakary-
ocytes are morphologically abnormal with variations in size
Hemoglobin < 10 g/dL and “cloud-like” or “balloon-shaped” lobulations of the
Leukocyte count < 4000/wL or > 30,000/L
nuclei. Many naked megakaryocyte nuclei are seen as well.*’
Presence: scores | point
As the name implies, reticulin fibrosis is minimal in the
Median survival: Score 0: 93 months
Score |: 26 months prefibrotic phase. In the fibrotic phase, the bone marrow
Score 2: 13 months cellularity is normal or decreased. Areas of hematopoiesis
are separated by regions of loose connective tissue or fat.
402. Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis
Figure 18-8 @ Leukoerythroblastosis, teardrop poikilocytosis, and abnormal platelet morphology associated with idiopathic myelofibrosis.
A. Leukoerythroblastosis. Note the myeloblast at the large arrow and the numerous nucleated red blood cells at the small arrows. B. Teardrop
poikilocytosis. C. Dwarf megakaryocyte (or micromegakaryocyte). This pathologic alteration of a megakaryocyte may be found in any of the
myeloproliferative disorders. Although often difficult to distinguish from cells of other lineages, observation of the marked cytoplasmic granu-
larity and further comparison of this cytoplasm to that of other platelets present on the peripheral smear will aid in identification. D. Dwarf
megakaryocyte. The cell at the pointer displays cytoplasmic blebs or budding, which is another characteristic of a micromegakaryocyte. Also
note the giant platelets present on this peripheral blood smear.
Reticulin fibrosis can be prominent with small islands of suggest occult disseminated intravascular coagulation (DIC).
residual hematopoietic precursors. The marrow sinuses can be Extramedullary hematopoiesis in the liver may result in hepatic
increased in size and number with characteristic intrasinu- dysfunction which certainly contributes to the coagulation
soidal hematopoiesis. Atypical megakaryocytes often occur in abnormalities (occult DIC, enhanced fibrinolysis) seen in IMF.
clusters, sheets or within dilated sinuses.*’ Thickening of the Hyperuricemia and elevated liver enzymes are found in
bony trabeculae (osteosclerosis) may be seen (Fig. 18-11). one-third of IMF patients.
Platelet dysfunction can cause troublesome hemostatic Cytogenetic abnormalities are present in approximately
complications in IMF patients. In addition, patients occasionally 30% of cases at diagnosis. The rate increases to approximately
demonstrate prolonged prothrombin and thrombin times, as 90% after leukemic transformation. Chromosomal abnormali-
well as elevated levels of fibrin degradation products and ties are less frequent in younger patients, which may explain
reduced levels of factors V and VIII. These features their better prognosis. Common findings include 13q—, 20q-,
Figure 18-9 @ Teardrop-shaped cells (arrows): peripheral blood in Figure 18-10 ™ Micromegakaryocyte found on the peripheral
a patient with myelofibrosis. blood smear of a patient with essential thrombocythemia.
Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis 403
IMF must be distinguished from other diseases within the Secondary to Nonmalignant Conditions
spectrum of the CMPDs, as well as differentiated from fibro- Granulomatous disorders Osteoporosis
sis secondary to infiltrative disorders (see Tables 18-1 and Sarcoidosis Vitamin D deficiency
Tuberculosis Systemic lupus
18-13). Clinically, the prefibrotic (p-CIMF) stage of CIMF Histoplasmosis erythematosis
mimics ET. Toxic exposure to chemicals Systemic sclerosis
However, CML is the diagnosis considered most fre- Hypo- and hyperparathyroidism
quently in the differential diagnosis of IMF. In chronic cases
404 Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis
protein, is easily differentiated from IMF. Myelodysplasia advanced disease with marrow failure and hypersplenism, as
(MDS) with marrow fibrosis can be difficult to differentiate they can be very sensitive to even smal] doses. However, poten-
from IMF. However the marked splenomegaly, extramedullary tial leukemogenicity is again a concern. Interferon alpha
hematopoiesis, and osteosclerosis are indications of IMF. has also been reported to afford a hematologic response in
The WHO has recently changed the designation of IMF to pri- hyperproliferative patients.” Etanercept, a tumor necrosis
mary myelofibrosis (PMP). The 2008 WHO diagnostic criteria factor-alpha (TNF-a) blocker, was shown in a pilot study to
for PME can be found in Table 18-14. palliate constitutional symptoms in IMF, which can be severe
and lead to cachexia (debilitating wasting away).’' Many
patients with severe anemia require transfusion support.
Treatment Approximately one-third respond to treatment with androgens
(fluoxymesterone and oxymetholone), which may be supple-
As with PV and ET, symptomatic treatment of IMF usually
mented initially with corticosteroids usually not to exceed
involves the use of cytotoxic agents to control thrombocytosis,
30 days. Chronic androgen use may be associated with fluid
leukocytosis, and symptomatic splenomegaly. Hydroxyurea
retention, abnormal liver function tests, and an increase in
on average doses of 500 mg twice a day, can aid in reducing
thromboembolic events. Men should be checked for occult
spleen size and control of thrombocytosis and leukocytosis.*”
prostate cancer before use, and women must be appraised of the
However, the benefit is usually temporary. Treatment with
virilizing effects. The use of erythropoietin has not generally
hydroxyurea may be associated with worsening of anemia,
been successful in patients with IMF, although some regressions
which in some instances, can be ameliorated by the concomitant
have been reported in those with low erythropoietin levels. Rare
use of erythropoietin. Early reports that hydroxyurea could
patients with IMF have developed autoimmune hemolytic
cause reversal of marrow fibrosis by suppressing megakary-
anemia which may respond to steroids and, occasionally,
ocyte production and releasing fibrinogenic cytokines, have not
cyclosporine. Thalidomide has also been reported to have
been confirmed in large clinical trials or clinical practice.*?
beneficial effects in some patients with regard to improvement
Doses of hydroxyurea must be carefully titrated in patients with
in anemia and thrombocytopenia, perhaps through its anti- TNF
activity. However, paradoxically, some patients have developed
increased myeloproliferation. Trials using the thalidomide
Beare The2008World derivative lenalidomide are in progress. This drug has been
shown to have immunomodulatory and antiangiogenic proper-
ties that are more potent than thalidomide and have an improved
side effect profile. The drug inhibits TNF-a, induces T-cell pro-
liferation and upregulates [L-10. Its antitumor effects may be
related to its antiangiogenic activity. Activity of the small mole-
cule receptor tyrosine kinase inhibitor SU5416 (Sugen) of vas-
Major Criteria
cular endothelium growth factor receptor 2 (VEGFR-2), c-KIT,
. Megakaryocyte proliferation and atypia’ accompanied by and FLT3, has been minimal in patients with MPD including
either reticulin and/or collagen fibrosis, or in the IMF.” The receptor tyrosine kinase inhibitor, imatinib mesylate
absence of reticulin fibrosis, the megakaryocyte changes
(Gleevec) has not been efficacious in improving anemia. Mini-
must be accompanied by increased marrow cellularity,
granulocytic proliferation and often decreased erythro- mal to moderate activity has been seen in reducing
poiesis (i.e., pre-fibrotic PMF). splenomegaly. In some trials, however, an increase in platelet
. Not meeting WHO criteria for CML, PV, MDS, or other count has been seen. Overall, the activity of imatinib in IMF has
myeloid neoplasm not been impressive. Trials are in progress using the farnesy]l
. Demonstration of JAK2V617F or other clonal marker or transferase inhibitor, Zarnestra.
no evidence of reactive marrow fibrosis
Splenectomy is an option for patients with symptoms
Minor Criteria related to splenomegaly, such as left upper quadrant discomfort,
transfusion-dependent anemia, refractory thrombocytopenia,
1. Leukoerythroblastosis
2. Increased serum LDH hypercatabolic symptoms, or portal hypertension.?* However,
3. Anemia the operative mortality can be significant, and postoperative
4. Palpable splenomegaly thrombocytosis can occur leading to postoperative thrombosis
* Diagnosis of primary prelonnea. (PMF) requires meeting all
and decreased survival.”> Blast transformation following
three major criteria and two minor criteria. splenectomy has been reported as well in a series of patients
* Small to large megakaryocytes with an aberrant nuclear/cytoplasmic with IMF.” Splenic irradiation can provide transient benefit and
ratio and hyperchromatic and irregularly folded nuclei and dense clus-
tering. LDH = lactate dehydrogenase; MDS = myelodysplastic syn-
symptomatic relief for patients who are poor surgical candi-
drome; PV = polycythemia vera dates.” However, it must be done carefully in low doses because
Source: Modified from Tefferi, A, and Vardiman, JW: Classification it can occasionally be associated with severe and irreversible
and diagnosis of myeloproliferative neoplasms: The 2008 World
pancytopenia. Allogeneic stem cell transplantation remains the
Health Organization criteria and point-of-care diagnostic algorithms.
Leukemia advance online publication 20 September 2007; doi: only curative treatment modality for IMF at present; unfortu-
10.1038/s}.leu.2404955 nately, it is often precluded by the patients advanced age, other
comorbidities, and lack of available donors.”
Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis 405
Oi estiomns i
t. What is the origin of MPDs? 9. When is myelosuppression advocated in patients with PV?
a. Fibroid infiltration of major organs a. If the patient is less than 20 years old —
b. Neoplastic transformation of multipotential stem cells b. When thrombosis-associated risk factors are present
c. Widespread deterioration of cellular function c. If the patient shows signs of glossitis
d. Splenic sequestration of normal blood cells d. If the patient has failed iron therapy
. Which of the following is not a characteristic of a 10. Which features help to distinguish secondary erythrocy-
chronic MPD? tosis and relative erythrocytosis from PV?
a. Extramedullary hematopoiesis a. Absence of splenomegaly; normal leukocyte, platelet,
b. Possible termination into acute leukemia and LAP levels
c. Cytogenetic abnormalities b. Presence of splenomegaly; increased leukocyte,
d. Hypoplasia of bone marrow platelet, and LAP levels
. The Philadelphia chromosome involves: c. Presence of hepatomegaly; decreased leukocyte,
a. (9322) platelet, and LAP levels
b. The BCR and c-ABL genes d. Presence of both splenomegaly and hepatomegaly;
c. A 210-kD protein possessing increased tyrosine kinase increased leukocytes and platelets; decreased LAP score
activity ih What is the safest and least expensive treatment for
d. All of the above patients with polycythemia vera?
. What is the predominant abnormal erythrocyte morphol- a. High altitude
ogy ‘associated with idiopathic myelofibrosis? b. Decrease of iron levels
a. Schistocytes c. Therapeutic phlebotomy
b. Ovalocytes d. Decrease of erythropoietin levels
c. Teardrop cells . What condition is defined by a platelet count greater
d. Target cells than 600 < 10°/L, megakaryocytic hyperplasia, absence
. Which features of chronic myelogenous leukemia are the of Ph chromosome, and hemoglobin of 13 g/dL or more
most important characteristics that distinguish it from (or normal red cell mass)?
myelofibrosis? a. Essential thrombocythemia
a. Presence of increased platelets and fibroblasts b. May—Hegglin anomaly
b. Decreased erythrocytes with abnormal morphology c. Acute myelogenous leukemia
c. Increased leukocytes with hypercellular bone marrow d. Polycythemia vera
d. Low LAP score and presence of Ph chromosome . The thrombosis seen in patients with essential thrombo-
. Which of the following factors does not cause fibroblast cythemia is a result of which of the following?
proliferation? a. Protein C deficiency
a. CSE b. Marked fibroblast proliferation
b. TGF-B c. Intravascular clumping of sludged hyperaggregable
c. bFGF platelets
d. PDGF d. Splenic sequestration of platelets
. What are the laboratory findings in PV? . What condition is not characteristically associated with
a. Decreased hematocrit; increased RBCs and granulo- reactive thrombocytosis?
cytes; decreased platelets a. Acute hemorrhage
b. Increased hematocrit; increased RBCs, granulocytes, b. Aplastic anemia
and platelets c. Chronic inflammatory disorders
c. Normal hematocrit; normal RBCs; increased granulo- d. Iron-deficiency anemia
cytes and platelets 1 Which of the following chronic MPDs is associated with
d. Increased hematocrit; increased RBCs; decreased the best prognosis?
granulocytes and platelets a. CML
. What is the expected erythropoietin value in PV? b. IMF
a. Normal Cyn
b. Increased oR Sih
c. Decreased
See answers at the back of this book.
— Cr :
Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis 409
-~ SUMMARY
@ Familial polycythemia is the result of mutations in the
erythropoietin receptor (EPo-R) or mutations in the oxy-
gen sensing pathway regulating erythropoietin production
MYELOPROLIFERATIVE DISEASE (Chuvash polycythemia).
w Myeloproliferative disorders (MPDs) arise from a malig- ESSENTIAL THROMBOCYTOSIS
nant transformation of a single multipotential stem cell
m The platelet count is markedly elevated in essential
that is committed to differentiation of granulocytes,
thrombocythemia (ET), often to more than 1000 10°/L.
monocytes, erythrocytes, and platelets.
m Hemorrhage and thrombosis caused by dysfunctional
m= MPDs are grouped together because of shared characteris-
platelets, splenomegaly, erythromelalgia, and neurologic
tics, the most important being panhyperplasia of the bone
manifestations are clinical features of ET.
marrow, extramedullary hematopoiesis, bone marrow fibro-
sis, and predilection for leukemic transformation. # ET must be differentiated from the many causes of reac-
tive thrombocytosis.
POLYCYTHEMIA VERA
gw Symptomatic patients or high-risk patients should receive
mw Elevation of the hematocrit (above 58% in males and
cytoreductive therapy with hydroxyurea.
above 52% in females) is the most important hallmark of
polycythemia vera (PV). g All patients with platelets < 1,000,000/uL should receive
aspirin if they have no antecedent history of bleeding.
w Elevated red cell mass, splenomegaly, decreased erythro-
poietin, normal arterial oxygen saturation, and increased IDIOPATHIC MYELOFIBROSIS
leukocyte alkaline phosphatase (LAP) are other important w Important features of idiopathic myelofibrosis (IMF) are
features of PV. anemia with teardrop poikilocytosis, leukoerythroblastic
w 90% of patients with PV have a mutation (V617F) in the blood picture, marked bone marrow fibrosis, splenomegaly,
JAK2 gene resulting in activation of downstream path- and variable but often elevated platelet counts.
ways (STATS). a Cytokines secreted by megakaryocytes and platelets stim-
w Approximately 15-20% of patients with PV will enter the ulate bone marrow fibroblastic proliferation in IMF.
spent phase with marrow fibrosis, worsening splenomegaly w Giant, bizarre platelets and micromegakaryocytes (dwarf
and pancytopenia. megakaryocytes) may be seen in the peripheral blood in
w Treatment of PV involves phlebotomy or use of cytotoxic IMF.
myelosuppressive agents, or a combination of both. m Progressive disease is manifested by extramedullary
SECONDARY POLYCYTHEMIA hematopoiesis with increasing splenomegaly, hepatomegaly,
weight loss, cachexia, portal hypertension, and pan-
m Increased secretion of erythropoietin has been implicated
cytopenia.
as the stimulus responsible for all cases of secondary ery-
throcytosis. g@ Hydroxyurea may be helpful in controlling splenomegaly,
leukocytosis, and thrombocytosis but does not alter the
m The arterial oxygen saturation is often decreased in
natural history of the disease.
patients with secondary erythrocytosis.
w Splenic radiotherapy and splenectomy may be helpful in
# Relative erythrocytosis occurs when there is depletion in
carefully selected patients.
circulating plasma volume (causing increased hematocrit
but normal red cell mass), and it is often seen in patients w Allogeneic bone marrow transplant remains the only cura-
with dehydration. tive treatment.
410 Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis
REFERENCES and role in leukemic transformation. polycythemia vera. Curr Hematol Rep
Blood 108(10):3548, 2006. 4:224, 2005.
il Dameshek, W: Some speculations on the . Tefferi, A: JAK2 mutations in poly- 35: Copelan, EA, and Balcerzak, SP: Sec-
myeloproliferative syndrome. Blood cythemia vera—molecular mechanisms ondary polycythemia. In Wasserman,
6/29 SIE and clinical applications. N Engl J Med LR, et al (Eds): Polycythemia Vera and
i . Vardiman, J, et al: The World Health 356:444, 2007. the Myeloproliferative Disorders.
Organization (WHO) classification of the . Scott, L, et al: JAK2 exon 12 mutations in Philadelphia, WB Saunders, 1995, p 195.
myeloid neoplasms. Blood 100:2292, polycythemia vera and idiopathic erythro- . Di Torrette, O, et al: Polycythemia vera:
2002. cytosis. N Engl J Med 356(5):459, 2007. The natural history of 1213 patients fol-
. Vaquez, H: Sur une forme speciale de . Campbell, P, and Green, AR: The myelo- lowed for 20 years. Ann Intern Med
cuanose d’accompanant d’hypergloblie proliferative disorders. N Engl J Med IDS:056 81095:
excessive et persistante. C R Soc Biol 355:23, 2006. . Spivak, J: The optimal management of
(Paris): 4:384 and Supp] Nute Buil Mem . Schaefer, A: Molecular basis of the diag- polycythaemia vera. Br JHaematol
Soc Med Hop Paris, 12:60, 1892. nosis and treatment of polycythemia vera 116:243, 2002.
. Osler, W: Chronic cyanosis, with poly- and essential thrombocythemia. Blood 38. Berk, P, et al: Treatment of polycythemia
cythemia and enlarged spleen: A new 107:4214, 2006. vera: A summary of clinical trials con-
clinical entity. Am J Med Sci 126:197, 22s Wehmeier, A, et al: Incidence and clini- ducted by the Polycythemia Vera Study
1903. cal risk factors for bleeding and throm- Group. In: Wasserman, LR, et al (Eds),
. Turk, W: Beitrage zur Kenntnis des botic complications in myeloproliferative Polycythemia Vera and the Myeloprolif-
Symptomenbildes Polyzythamie mit disorders. A retrospective analysis of 260 erative Disorders. WB Saunders,
Milztumor und Zyanose. Wien Klin patients. Ann Hematol 63:101, 1991. Philadelphia, 1995.
Wochenschr 17:153, 1904. . Anger, B, et al: Budd-Chiari syndrome . Tefferi, A, and Spivak, J: Polycythemia
. Hirsch, R: Generalized osteosclerosis and thrombosis of other abdominal ves- vera: Scientific advances and current
with chronic polycythemia vera. Arch sels in the chronic myeloproliferative practice. Semin Hematol 42:206, 2005.
Pathol 19:91, 1935. diseases. Klin Wochenschr 67:819, 1989. 40. Fruchtman, S, et al: From efficacy to
— . Wasserman, L: Polycythemia Vera Study
24. Berlin, N: Diagnosis and classification of safety—A Polycythemia Vera Study
Group: A historical perspective. Semin the polycythemias. Semin Hematol Group report on hydroxyurea in patients
Hematol 23:193, 1986. DEBI). HIS. with polycythemia vera. Semin Hematol
oo. Pearson, T: Evaluation of diagnostic cri-
25. Steinman, H, et al: Polycythaemia rubra 34:17, 1997.
teria in polycythemia vera. Semin Hema- vera and water-induced pruritus: Blood 41. Gilbert, H: Long-term treatment of
tol 38:21, 2001. histamine levels and cutaneous fibri- myeloproliferative disease with
\O . Tefferi, A: Polycythemia vera: A compre- nolytic activity before and after water interferon-alpha-2b: Feasibility and
hensive review and clinical recommen- efficacy. Cancer 83:1205, 1998.
challenge. Br JDermatol 116:329, 1997.
dations. Mayo Clin Proc 78:174, 2003. . Michiels, J: Erythromelalgia and vascu- 42. Silverstein, M, et al: Anagrelide, a ther-
. Passamonti, F, et al: Polycythemia vera lar complications in polycythemia vera. apy for thrombocythemic states: Experi-
in young patients: A study on the long- Semin Thromb Hemost 23:441, 1997. ence in 577 patients. Anagrelide Study
term risk of thrombosis, myelofibrosis . Messinezy, M, et al: Low serum Group. Am J Med 92:69, 1992.
and leukemia. Haematologica 88:13, erythropoietin—A strong diagnostic . Cortes, J, et al: Results of imatinib
2003. criterion of primary polycythemia vera mesylate therapy in patients with refrac-
. Sokol, L, et al: Primary familial poly- even at normal haemoglobin levels. Clin tory or recurrent acute myeloid
cythemia: A frameshift mutation in the Lab Haematol 17:217, 1995. leukemia, high-risk myelodysplastic syn-
erythropoietin receptor gene and
28. Michiels, J, and Juvonen, E: Proposal for drome, and myeloproliferative disorders.
increased sensitivity of erythroid progen- revised diagnostic criteria of essential Cancer 97:2760, 2003.
itors to erythropoietin. Blood 86:15, thrombocythemia and polycythemia vera 44. Landolfi, R, et al: Aspirin for the control
1995. by the thrombocythemia vera study of platelet activation and prevention of
. Chaiter, Y, et al: High incidence of
group. Semin Thromb Hemost 23:339, thrombosis in essential thrombocythemia
myeloproliferative disorders in Ashke- OOF and polycythemia vera: Current insights
nazi Jews in northen Israel. Leuk Lym-
29. Ingram, GIC, and Hills, M: The pro- and rationale for future studies. Semin
phoma 7:251, 1992.
thrombin test: effect of varying citrate Thromb Hemost 32:25, 2006.
. Fialkow, P: Use of glucose-6-phosphate
concentration. Thrombos Hemost 45. Di Guglielmo, G: Erithroleucemia
dehydrogenase markers to study human
36:230, 1976. e piastrinemia. Folia Med 1:36, 1920.
myeloproliferative disorders. Haematol
. Klippel, S, et al: Quantification of PRY-1 46. Epstein, E, and Goedel, A: Hamorrhagis-
Blood Transfus 23:53, 1979.
mRNA distinguishes polycythemia vera che thrombozythamie bei vaskularer
. Briere, J, and el-Kassar, N: Clonality
from secondary erythrocytosis. Blood Schrumpfmilz. Virchows Arch Pathol
markers in polycythemia and primary
102:3569, 2003. Anat 292:233, 1934.
thrombocythaemia. Baillieres Clin
. Bock, O, et al: Thrombopoietin receptor 47. Gunz, F: Hemorrhagic thrombocythemia:
Haematol 11:787, 1998.
(Mpl) expression by megakaryocytes in A critical review. Blood 15:706, 1960.
. Takefumi, I, et al: Involvement of vari-
myeloproliferative disorders. J Pathol 48. Murphy, S, et al: Experience of the Poly-
ous hematopoietin - cell lineages by the
203:609, 2004. cythemia Vera Study Group with essen-
JAK2‘°' mutation in polycythemia
. Moliterno, A, et al: Impaired expression tial thrombocythemia: A final report on
vera. Blood 108:3128, 2006.
if the thrombopoietin receptor by diagnostic criteria, survival, and
. Lippert, E, et al: The JAK2%°'* mutation
platelets from patients with poly- leukemic transition by treatment. Semin
is frequently present at diagnosis in
cythemia vera. N Engl J Med 338:572, Hematol 34:29, 1997.
patients with essential thrombocythemia
1998. 49. Mesa, R, et al: Population-based inci-
and polycythemia vera. Blood 108:1865,
. Diez-Martin, J, et al: Chromosome stud- dence and survival figures in essential
2006.
ies in 104 patients with polycythemia thrombocythemia and agnogenic
. Campbell, P, et al: Mutation of JAK2 in
vera. Mayo Clin Proc 66:287, 1991. myeloid metaplasia: An Olmsted County
the myeloproliferative disorders: Timing,
. Andrieux, J, and Demory, J: Karyotype Study, 1976-1995. Am J Hematol 61:10,
clonality studies, cytogenetic associations,
and molecular cytogenetic studies in 1999.
Chapter 18 Chronic Myeloproliferative Disorders II: Polycythemia Vera, Essential Thrombocythemia, and Idiopathic Myelofibrosis Al]
50. Fialkow, P, et al: Evidence that essential 66. Van Genderen, P, et al: Prevention and agnogenic myeloid metaplasia. Case
thrombocythemia is a clonal disorder treatment of thrombotic complications in report and review of the literature. Acta
with origin in a multipotent stem cell. essential thrombocythaemia: Efficacy Haematol 89:104, 1993.
Blood 58:916, 1981. and safety of aspirin. Br JHaematol . Yusen, RD, and Kollef, MH: Acute respi-
Dk. El-Kassar, N, et al: Clonality analysis of MINIS), WIN. ratory failure due to extramedullary
hematopoiesis in essential thrombo- 67. Budde, U, et al: Elevated platelet count hematopoiesis. Chest 108:1170, 1995.
cythemia: Advantages of studying as a cause of abnormal von Willebrand 84. Lundh, B, et al: Intracranial myeloid
T lymphocytes and platelets. Blood factor multimer distribution in plasma. metaplasia in myelofibrosis. Scand J
S9M2ZS5 1997: Blood 82:1749, 1993. Haematol 28:91, 1982.
2s Bacher, U, et al: Conventional cytoge- 68. Heuck, G: Zwei Falle von Leukamie mit 85. Fedeli, G, et al: Extramedullary
netics of myeloproliferative diseases eigenthumlichem Blut-resp Knochen- hematopoiesis involving the esophagus
other than CML contribute valid infor- marcksbend. Virchows Arch (Pathol in myelofibrosis. Am J Gastroenterol
mation. Ann Hematol 84:250, 2005. Anat) 78:475, 1879. ssl SIZ, ISI0).
SS: Kiladjian, J, et al: Study of the throm- 69. Cervantes, F, and Barosi, G: Myelofibro- 86. Dupriez, B, et al: Prognostic factors in
bopoietin receptor in essential thrombo- sis with myeloid metaplasia: Diagnosis, agnogenic myeloid metaplasia: A report
cythemia. Leukemia 11:1921, 1997. prognostic factors, and staging. Semin on 195 cases with a new scoring system.
54. Taksin, A, et al: Autonomous megakary- Oncol 32:395, 2005. Blood 88:1013, 1996.
ocyte growth in essential thrombo- 70. Bonduel, M, et al: Familial idiopathic 87. Thiele, J, et al: Bone marrow
cythemia and idiopathic myelofibrosis is myelofibrosis and multiple heman- histopathology in myeloproliferative
not related to a c-mpl mutation or to an giomas. Am J Hematol 59:175, 1998. disorders-current diagnostic approach.
7
autocrine stimulation by Mpl-L. Blood _ . Bosch, X, et al: Toluene-associated Semin Hematol 42;184, 2005.
OSA Sa9993 : myelofibrosis. Blut 58:219, 1989. 88. Reilly, J: Cytogenetic and molecular
55): Bench, A, and Pahl, H: Chromosomal . Anderson, R, et al: Myelofibrosis with abnormalities in agnogenic myeloid
abnormalities and molecular markers in myeloid metaplasia in survivors of the metaplasia. Semin Oncol 32:359, 2005.
myeloproliferative disorders. Semin atomic bomb in Hiroshima. Ann Intern 89. Lofvenberg, E, and Wahlin, A: Manage-
Hematol 42:196, 2005. Med 60:1, 1964. ment of polycythaemia vera, essential
56. Elliott, M, and Tefferi, A: Thrombosis . Xu, M, et al: The constitutive mobiliza- thrombocythaemia and myelofibrosis
and haemorrhage in polycythaemia vera tion of bone marrow-repopulating cells with hydroxyurea. Eur JHaematol
and essential thrombocythaemia. Br J into the peripheral blood in idiopathic 41:375, 1988.
Haematol 128:275, 2005. myelofibrosis. Blood 105:1699, 2005. 90. Wickramasinghe, S, et al: Alpha-interferon
ah Picardi, M, et al: Measurement of spleen 74. Tefferi, A, et al: Cytogenetic findings in primary idiopathic myelofibrosis.
volume by ultrasourfd scanning in and their clinical relevance in myelofi- Lancet ii:1524, 1987.
patients with thrombocytosis: A prospec- brosis with myeloid metaplasia. Br J Oil, Steensma, D, et al: Etanercept, a soluble
tive study. Blood 99:4228, 2002. Haematol 113:763, 2001. tumor necrosis factor receptor, palliates
58. Tefferi, A: Risk-based management in . Bellanne-Chantelot, C, et al: Genetic and constitutional symptoms in patients with
essential thrombocythemia. ASH Educa- clinical implications of the Val617Phe myelofibrosis with myeloid metaplasia:
tion Program Book. Hematology 172, JAK2 mutations in 72 families with Results of a pilot study. Blood 99:2252,
IIE), myeloproliferative disorders. Blood 2002.
sy). Cortelazzo, S, et al: Hydroxyurea for 108(1):346, 2000. Py. Giles, F, et al: Phase II study of
patients with essential thrombocythemia 70. Steensma, D: JAK2 V617F in myeloid SU5416—a small-molecule, vascular
and a high risk of thrombosis. N Engl J disorders: Molecular diagnostic tech- endothelial growth factor tyrosine-kinase
Med 332:1132, 1995. niques and their clinical utility. JMD receptor inhibitor—in patients with
60. Sterkers, Y, et al: Acute myeloid 8(4):397, 2006. refractory myeloproliferative diseases.
leukemia and myelodysplastic syn- Whe Reeder, T, et al: Both B and T lympho- Cancer 97:1920, 2003.
dromes following essential thrombo- cytes may be clonally involved in . Tefferi, A, et al: Splenectomy in myelofi-
cythemia treated with hydroxyurea: High myelofibrosis with myeloid metaplasia. brosis with myeloid metaplasia: A single
proportion of cases with 17p deletion. Blood 101:1981, 2003. institution experience with 223 patients.
Blood 91:616, 1998. 78. Martyre, M, et al: Transforming growth Blood 95:2226, 2002.
6 jak. Steurer, M, et al: Anagrelide for throm- factor-beta and megakaryocytes in the 94. Barosi, G, et al: Splenectomy and risk of
bocytosis in myeloproliferative disor- pathogenesis of idiopathic myelofibrosis. blast transformation in myelofibrosis
ders. Cancer 101:2239, 2004. Br JHaematol 88:9, 1994. with myeloid metaplasia. Italian Cooper-
62. Storen, E, and Tefferi, A: Long-term use SD. Barosi, G: Myelofibrosis with myeloid ative Study Group on Myeloid with
of anagrelide in young patients with metaplasia: Diagnostic definition and Myeloid Metaplasia. Blood 91:3630,
essential thrombocythemia. Blood prognostic classification for clinical 1998.
97:863, 2001. studies and treatment guidelines. J Clin WB). McFarland J, et al. Palliative irradiation
63. Harrison, C, et al: Hydroxyurea com- Oncol 117:2954, 1999. of the spleen. Am J Clin Oncol 26:178,
pared with anagrelide in high-risk essen- 80. Wanless, IR, et al: Hepatic vascular dis- 2003.
tial thrombocythemia. N Engl J Med ease and portal hypertension in poly- 96. Papageorgiou S, et al. Allogeneic stem
Spsinaos ADO). cythemia vera and agnogenic myeloid cell transplant. Bone Marrow Transplant
64. Elliott, M, and Tefferi, A: Interferon- metaplasia. Hepatology 12:1166, 1990. 38:721, 2006.
alpha therapy in polycythemia vera and 8 . Koch, CA, et al: Nonhepatosplenic
essential thrombocythemia. Semin extramedullary hematopoiesis: Associated
Thromb Hemost 23:463, 1997. diseases, pathology, clinical course, and
65. Tefferi, A, et al: A clinical update in treatment. Mayo Clin Proc 78:1223, 2003.
polycythemia vera and essential throm- 82. Knobel, B, et al: Ectopic medullary
bocythemia. Am J Med 109:141, 2000. hematopoiesis as a cause of ascites in
Chapter
| q Myelodysplastic
Syndromes
Giovanni D’Angelo, FCMLS
Luigina Mollica, MD, FRCP(C), PhD
Josée Hébert, MD, FRCP(C)
Lambert Busque, MD, FRCP(C)
Introduction OBJECTIVES
Epidemiology—Etiology— At the end of this chapter, the learner should be able to:
Pathogenesis
MDS: Clonal Proliferative 1. Provide an overview of the clonal nature and stem cell origin of myelodysplastic
Diseases syndromes (MDS).
Ineffective Hematopoiesis . Master the criteria of the French-American—British and World Health Organization
iw)
Genetic Anomalies (WHO) classifications.
Biologic Characteristics of
Disease Progression 3. Recognize the morphologic features of MDS on examination of blood and bone
marrow smears.
Morphological
Characteristics of Blood 4. Differentiate the different subtypes of MDS based on their characteristic laboratory
and Bone Marrow features.
Definitions of Specific
5. Distinguish MDS from acute myeloid leukemia on examination of blood and bone
Morphological
marrow smears.
Characteristics
Lineage Dysplasias 6. Know the ringed sideroblast values in MDS.
Classification of MDS 7. Differentiate between primary and secondary MDS.
Subtypes
8. Know the International Prognostic Scoring System (IPSS) and the value of Wilms
FAB Classification Tumor gene (W717) expression.
Refractory Anemia
Refractory Anemia with 9. Recognize a myelodysplastic/myeloproliferative disorder.
Ringed Sideroblasts 10. Describe the therapeutic regimen that is most likely to achieve long-term survival.
Refractory Anemia with
Excess Blasts
Refractory Anemia with
Excess Blasts in
Transformation
Chronic Myelomonocytic
Leukemia
WHO Classification
Laboratory Features
Cytochemistry
Bone Marrow Histology
Cytogenetics and Molecular
Abnormalities
Immunology
412
Chapter 19 Myelodysplastic Syndromes 413
Apoptosis is characterized by three sequential phases: factor, GM-CSF) are diminished in MDS. Therefore, the
(1) initiation, (2) commitment, and (3) execution.'* A variety balance between growth-stimulatory and growth-inhibitory
of factors can initiate apoptosis, such as chemotherapy, radio- cytokines may favor apoptosis in MDS. As Fas antigen and
therapy, corticosteroids, and growth factor deprivation. It may Fas-ligand are also abnormally elevated on the surface of MDS
also occur as a direct effect of cytotoxic T-cell activity. Apop- cells, the Fas apoptotic pathway may also be important in the
tosis can be initiated by inhibitory cytokines such as tumor pathogenesis of MDS.
necrosis factor-alpha (TNF-a), transforming growth factor- Although increased apoptosis is a key biologic feature of
beta (TGF-B), and by the action of Fas-ligand on its receptor MDS, it is still not clear if it is central to the pathogenesis of this
FAS/CD95 (Fig. 19-2). Proteins belonging to the Bcl-2 family disease or merely an indication of ineffective hematopoiesis
are involved in the regulation of the commitment phase. Some caused by other genetic anomalies of the involved clone. The
of these are pro-apoptotic (Bax, Bak, Bad, Bcl-XS, Bik), investigation of the cause and mechanisms of apoptosis in MDS
whereas others are anti-apoptotic (Bel-2, Bcl-XL, Mcl-1). If may provide insight into its pathogenesis and lead to novel
the balance of Bcl-2 family protein activities favors apopto- therapeutic approaches.
sis, a family of cysteine proteases called caspases will be
activated, leading to cell death after a series of molecular
cleavages affecting the cytoskeleton, cytosol, and nuclear Genetic Anomalies
proteins. This will lead to a pathognomonic phenotype char- The most common genetic anomaly associated with MDS is
acterized by chromatin condensation, oligodimerization of the loss of genetic material. The cytogenetic analysis of
DNA, and nuclear disintegration. © patients with MDS is characterized mainly by chromosomal
Yoshida first proposed that the ineffective hematopoiesis deletions, involving part of or a whole chromosome (see the
seen in MDS was caused by an abnormally high rate of later discussion of cytogenetics). This is in sharp contrast to
intramedullary apoptosis.'” This hypothesis was tested by sev- the acute leukemias, where chromosomal translocations pre-
eral groups using different methods (such as in situ end labeling dominate. The acute leukemias appear to follow the model of
(ISEL), nick-end labeling (TUNEL), and flow cytometry), all of oncogene activation, resulting in the transformation of the
which confirmed an elevated apoptotic rate in MDS patients cell of origin, whereas the deletional pattern found in MDS
compared to normal controls. Apoptosis seems to affect CD34+ favors a tumor suppressor gene model. In this model, the lack
progenitors as well as maturing cells. The degree of apoptosis is of both copies (one from each parental chromosome) is nec-
more pronounced in the early phases of the disease, compared essary to lead to carcinogenesis. One allele may be mutated
to more advanced or proliferative stages. Several lines of and the other deleted.*' The best described examples of this
evidence suggest a major role for inhibitory cytokines in the model are found in retinoblastoma and the Li-Fraumeni syn-
initiation of apoptosis in MDS. TNF-a is elevated in the bone drome (p53).” It is also possible that haploinsufficiency (the
marrow of MDS patients, and correlates with the degree of presence of only one active allele of a gene) alone may be suf-
apoptosis.”? TGF-B has also been shown to be elevated in MDS, ficient to cause a malignant hematological process in some
and may also play an important initiating role.*° Certain growth- cases. Hereditary haploinsufficiency for the gene RUNX/
stimulatory cytokines endowed with anti-apoptotic activity (AMLI) has been recently found to be associated with the devel-
(such as granulocyte/macrophage-monocyte colony-stimulating opment of acute leukemia in a family.* These data suggest
that MDS patients are missing key genes essential for maintain-
Fas and Fas ligand up-regulation ing normal hematopoiesis and that, with time, this leads to the
T lymphocyte
accumulation of more severe and numerous genetic anomalies
associated with disease progression and deterioration of the
hematological phenotype. To date, no single gene has been
found to be commonly deleted in MDS patients, but the identi-
fication of such genes is the focus of research of several groups.
the proliferation of abnormal cells.*? MDS is thus one of the best specific and nonspecific esterases may be used in the investiga-
examples of the multistep pathogenesis of neoplasia for hema- tion of MDS. The review of these materials allows the distinc-
tological cancers. The accumulation of different genetic alter- tion between MDS subtypes based on established criteria. MDS
ations changes the phenotype of the cell from normal to clonal are most often suspected in patients presenting with anemia
and dysplastic, and further changes confer a full-blown malig- (usually macrocytic or normochromic), with or without addi-
nant phenotype, leading to the development of acute leukemia. tional cytopenias. A vast array of laboratory tools is available
to help the physician to establish the diagnosis of each type
Morphological Characteristics of Blood ofLMDS 2:
and Bone Marrow BLASTS
Definitions of Specific Morphological Some types of myelodysplastic syndromes are diagnosed
Characteristics according to the number of blasts in the bone marrow at
initial diagnosis. There are two types of blast, according to
At the laboratory level, the diagnosis of a MDS is made by the the FAB classification: type I refers to a myeloblast of
careful morphological study of blood and bone marrow smears variable size, without any azurophilic granules or Auer rods
using the Wright-Giemsa stain, and of the bone marrow biopsy (Fig. 19-3); type II refers to a myeloblast that is slightly
specimen using hematoxylin and eosin (H & E) and reticulin larger and contains few azurophilic granules (1 to 20)
stains (Table 19-1). The iron content of both marrow aspirate (Fig. 19-4); Goasguen and associates*® have proposed a type
and biopsy specimens is revealed by the use of Prussian blue II myeloblast, containing more than 20 azurophilic granules
(Perl’s stain).*® Other special stains, such as myeloperoxidase with a basophilic cytoplasm and absence of the Golgi zone
(MPO), Sudan black B (SBB), periodic acid—Schiff (PAS), and similar type I and II myeloblasts.
Erythroid/Dyserythropoiesis
Myeloid/Dysgranulopoiesis
Thrombopoiesis/Dysmegakaryopoiesis
SIDEROBLASTS
A type I sideroblast refers to a normal sideroblast containing
one to four cytoplasmic iron-containing granules, normally
accounting for 15% to 50% of bone marrow erythroblasts.
A type II sideroblast is considered abnormal, harboring 5 to
10 granules, scattered throughout the cytoplasm. Type III is
synonymous with a ringed sideroblast, having more than
10 granules that cover at least one-third of the nuclear rim or
forming a complete ring around the nucleus."
Lineage Dysplasias
DYSERYTHROPOIESIS
PERIPHERAL BLOOD Anemia is present in at least 90% of
cases and is most often macrocytic or normocytic with a
decreased reticulocyte number. Erythrocyte morphological on i ie 4
characteristics that may be encountered are macrocytosis, Figure 19-6 m RAEB (peripheral blood): anisomacrocytosis, neu-
anisopoikilocytosis, basophilic stippling, a dual red blood cell trophil and macrothrombocyte (arrow).
population (normochromic, hypochromic), Pappenheimer bod-
ies, dacryocytes (teardrop cells), fragmented cells, elliptocytes,
Howell—Jolly bodies, and acanthocytes (Figs. 19-5 to 19-8).
ee
Figure 19-8 mAn erythrocyte with basophilic stippling (A), one Figure 19-10 @An erythroblast with basophilic stippling (A), and
with a Howell-Jolly body (B), and two erythroblasts connected by two erythroblasts connected through internuclear bridging (bone
internuclear bridging (peripheral blood) (©). marrow) (8B).
DYSGRANULOCYTOPOIESIS
PERIPHERAL BLOOD Neutropenia is found in almost 60% of
patients, although neutrophilia may be encountered occasion-
ally. Granulocytes show variable degrees of hyposegmentation
with bilobulation (pseudo-Pelger—Huet anomaly) or monolobu-
lation (pseudo-Stodtmeister anomaly),** abnormal chromatin,
or, rarely, atypical hypersegmentation (Figs. 19-15 to 19-17). In
the cytoplasm, one may find variable degrees of hypogranula-
tion, persistent basophilic zones (pseudo-Dohle bodies), or,
more rarely, hypergranulation with larger granules than usual
(Fig. 19-18). Combined nuclear and cytoplasmic dysplasia is
detected in more than 90% of cases.
Inna
a i
Figure 19-19 @ RAEB (bone marrow): monolobular (dwarf) Figure 19-22 ™ Sq- syndrome (bone marrow): monolobulated
megakaryocyte. micromegakaryocytes.
Chapter 19 Myelodysplastic Syndromes 421
FAB Classification
MDS are classified into five distinct subtypes. The distinction
between these subtypes of MDS relies essentially on the study
Figure 19-25 @ RA (bone marrow): hypogranular megakaryocyte. of the bone marrow aspirate stained with Wright—Giemsa and
Prussian blue. The distinctive hematologic features of each
MDS subtype are summarized in Table 19-2.
Refractory Anemia
Refractory anemia (RA) is the most difficult MDS to recog-
nize and diagnose. The diagnosis rests mainly on the sub-
jective identification of qualitative abnormalities involving
one or more bone marrow hematopoietic cell lineages, in a
patient suspected of having RA. Anemia is present in more
than 90% of patients. It is usually normochromic and
macrocytic, but in some cases, erythrocytes may be normo-
cytic, with a variable degree of morphological abnormali-
ties, usually more subtle than those found in other
subgroups. Cytopenias are common, with a white blood cell
count less than 3.9 * 10°/L and a platelet count below
130 X 10°/L. Morphological abnormalities such as dys-
granulopoiesis or dysmegakaryopoiesis are more subtle
than in other types of MDS. The bone marrow is usually
normocellular or hypercellular. Infrequently, it may be
Figure |9-24 @ RA (bone marrow): detached nuclei megakaryocyte. slightly hypocellular or may present a variable degree of
fibrosis. Blast cells, type I and type II, represent less than
5% of all nucleated cells. Erythroid hyperplasia is relatively
common, with mild dyserythropoiesis and occasional
ringed sideroblasts (less than 15% of nucleated red blood
cells). Cytogenetic studies are crucial in the diagnosis of
this particular subtype of MDS in view of the absence
of quantitative criteria for its diagnosis. Indeed, the finding
of a clonal chromosomal abnormality in such a setting con-
firms the diagnosis. Approximately 50% of patients are
found to have clonal abnormalities. The diagnosis may also
be supported by abnormal bone marrow culture results or
an X-inactivation clonality assay showing a clonal pattern
in myeloid cells in the presence of a normal somatic tissue
control. In the absence of a clonal chromosomal abnormal-
ity, the establishment of this diagnosis based solely on
single lineage dysplasia should be made with great caution.
Isolated dyserythropoiesis on blood or bone marrow smears
may be a feature shared by a variety of hematological
disorders, including relatively benign diseases with no
potential to progress to acute leukemia and bearing no rela-
Figure 19-25 mRA (bone marrow): vacuolated immature
megakaryocyte. tionship to the MDS.
422 Chapter 19 Myelodysplastic Syndromes
mainly on the presence of more than | X 10°/L monocytes in the The most significant amendments of the WHO classifi-
peripheral blood and the findings of lineage dysplasia in the cation (relative to the FAB classification) are the following:
peripheral blood or bone marrow, or both, as in other MDS.
1. RA is now subdivided according to the presence or absence
of multilineage dysplasia. The presence of dysplasia in at
WHO Classification**® least 10% of the progeny from two or more myeloid cell
lines defines a new subtype of MDS called RCMD. This
The FAB classification of MDS was revisited by WHO in con- subtype of MDS has less than 5% blasts in marrow, no
junction with the European Association of Haematopatholo- Auer rods, and no peripheral monocytosis. The subdivision
gists and the Society for Haematopathology.* The WHO of RA according to level of lineage dysplasia is based on
classification preserves many of the concepts and definitions the worse prognosis of patients in which more than the ery-
of the FAB classification, but incorporates other data to throid progenitors are affected.
improve clinical relevance.** Several studies have validated the tO. RARS is also subdivided according to the presence or absence
improved clinical significance of the WHO classification of of multilineage dysplasia. The presence of multilineage dys-
MDS.***° The WHO classification defines eight subtypes of plasia defines another subtype of MDS called RCMD-RS.
MDS: refractory anemia (RA), refractory anemia with ringed 3. The 5q— syndrome becomes a specific category with the 5q
sideroblasts (RARS), refractory cytopenia with multilineage deletion (q21—q32) as the sole cytogenetic anomaly, fewer
dysplasia (RCMD), refractory cytopenia with multilineage dys- than 5% blasts in the marrow, no Auer rods, and normal or
plasia and ringed sideroblasts (RCMD-RS), refractory anemia elevated platelet counts.
with excess blasts-1 (RAEB-1), refractory anemia with excess 4. RAEB is subdivided into RAEB-1 for patients with 5% to
blasts-2_ (RAEB-2), myelodysplastic syndrome-—unclassified 9% bone marrow blasts, and RAEB-2 for patients with
(MDS-u), and MDS associated with isolated del (5q). The dis- 10% to 19% bone marrow blasts.
tinctive hematologic features of each MDS subtype are summa- 5. RAEB-t is eliminated and all patients are considered to
rized in Table 19-3. have acute leukemia.
si
Figure 19-35 @ RA (peripheral blood, Kleihauer—Betke staining): Figure 19-36 @ RAEB (bone marrow, reticulin stain, magnification
positive RBCs, HbF. 400): severe fibrosis.
Chapter 19 Myelodysplastic Syndromes 427
«
tendency for a long leukemia-free survival. The remaining
(Fig. 19-37). The cytogenetic anomalies reported in sec- patients have an intermediate prognosis.”
ondary MDS are related to the type of therapy that patients
THE S5q- SYNDROME
have previously received. More than 90% of patients treated
with an alkylating agent show anomalies of chromosome 5 or In the mid-1970s, van den Berghe® described patients with an
7, or both.*' Patients treated with agents of the epipodophyl- acquired interstitial deletion of the long arm of chromosome 5
lotoxins class demonstrate anomalies (mainly translocations) (Fig. 19-39). These patients had specific clinical features that
involving the MLL gene located at chromosome band 1|1q23.°? included a macrocytic anemia, normal or elevated platelet
count, mild leukopenia, and, in the bone marrow, monolobular
Most chromosome abnormalities are not specifically associ-
ated with any FAB subgroup; however, the 5q— anomaly or dwarf megakaryocytes and erythroid hypoplasia. Patients
is documented more frequently in RA,» as is del(20q). Mono- are usually classified as having RA according to the FAB clas-
sification and are, in contrast to other MDS patients, predomi-
somy 7 is rarely seen in RA.* Complex karyotypes (three
nantly females. The 5q— syndrome is associated with a good
or more chromosomal abnormalities within the same cell)
prognosis with a median survival of approximately 8 years.
The deleted DNA segment lies between bands 5q13 and 5q33
e % ~~ ”
4 & 1 4 13 a a ee hy * % % 4 te
tamy ; 4 4 :
o- eb4 —<.§ a
v 2
8
g
i
to
ee
23
# a
O84
7
5 eA. 2 $8 eG LFed
1 2 34 emo : 1 2 3 4 5
% r a i P ’ "i
6 7 8 9 iL): ee is
x
7 $2 af «Ee OKs 4
6 i 8 9 10 11 12
A » 4
a * i s+
‘¥ oo
ff ay
i
13 14 65 ¢? 16 17 18
J & 4 ae <a
NS 13 14 15 16 1 1
a
; a ¥ 21 22 y;
19 20 Soy
ne ee a8 ae § g
19 20 21 22 X V4
Figure 19-57 @ Karyotype GTG banding, resolution 450 bands,
showing complex clonal abnormalities involving at least 8 chromo- Figure 19-38 @ Karyotype (GTG banding) showing the
somes within the same cell, including del(5)(q13q33), —7, t(5;12)(q33;p13) translocation and a loss of chromosome 7 in a
del(20)(q11.2). case of CMML.
428 Chapter 19 Myelodysplastic Syndromes
Fy ‘ ¥
é ¥, 4 ¥ 44 a
x eS ¥ i
‘ & om
2 3 4 5
~ * a¢
. 4 wo, % ¥ Ge ¥ ®.
‘ 4 id mS ss 3% wo Ff
6 7 8 9 10 11 12 X
x -e RZ
3 * A . $ «@ $
13 14 15 16 ily 18
ne eh 8s
19 20 21 22 NV.
6. @-: LJ & | Bo
$3 10 4
=~ 17 ~-22
W -16
16 17
Figure 19-42 m RAEB-2, spectral karyotyping (SKY): complex karyotype with numerous chromosomal translocations and trisomy 22.
In the rare cases in which a lymphoblastic proliferation is Marrow cells pass through all stages of maturation to become
found, the immunophenotypic pattern is consistently of the morphologically recognizable mature cells. In vitro cultures
early B type (CD19+, CD10—, TdT+). RAEB and RAEB-t of bone marrow cells from individuals with MDS have pro-
show a higher frequency of myeloblastic and megakaryoblastic vided information about the nature of the pathophysiologic
immunologic markers, whereas in RA, RARS, and CMML, a defect. In general, the ability of hematopoietic progenitor
granulomonocytic phenotype predominates. Hybrid lymphoid- cells to form colonies is found to be either reduced or
myeloid cells or cells coexpressing granulomegakaryocytic absent.’° Aberrant growth patterns are mostly observed in the
antigens are also frequently observed. At the molecular level, more severe subgroups of MDS, such as RAEB and RAEB-t.
rearrangements of the immunoglobulin H ({gH) and the T-cell A notable exception is CMML, which is distinguished by
antigen receptor gamma (TCR-y) and -delta (TCR-5) genes increased colony growth.’° Investigators have tried to corre-
can be found. late these growth patterns with the propensity to evolve to
Patients with type I and II blasts expressing the H blood acute leukemia, dividing them into leukemic and non-
group antigen” have a poor prognosis, as opposed to those leukemic types. Given the lack of standardization and results
expressing a purely myeloid phenotype. Expression of surface that are often conflicting, no firm conclusions can be drawn
antigens from monocytes and granulocytes may vary consider- about growth patterns with respect to either prognosis or
ably, with either loss or gain of specific or nonspecific antigens. leukemic potential in individual patients with MDS at this
Lymphopenia is frequently observed, with a decrease of CD4+ time. In vitro bone marrow culture from patients with MDS is
cells and a slight increase of CD8+ cells.”' It has been reported a valuable complementary tool to be considered together with
that an increased number of CD8+ cells at initial diagnosis in cytogenetics and other laboratory tests.
RA and RARS is associated with a decreased incidence of acute
non-lymphoblastic leukemia (ANLL) transformation. Although
natural killer lymphocytes are decreased, antibody-dependent Cellular Dysfunction
cellular cytotoxicity is conserved. Genetic mutations or other abnormalities such as chromoso-
Fewer B-lymphocyte abnormalities have been reported. mal deletions, translocations, or gene amplification are
B-cell Epstein-Barr virus (EBV) receptors are decreased in responsible for a vast array of functional abnormalities within
number.’”? Immunoglobulin synthesis may be abnormally reg- specific cell lineages.
ulated. Polyclonal hypergammaglobulinemia is observed in
approximately 30% of patients with MDS; 12% have a mono- RED BLOOD CELLS
clonal gammopathy, whereas 13% are hypogammaglobuline- Several metabolic abnormalities may be encountered, the
mic. Polyclonal hypergammaglobulinemia is observed in most frequent being acquired pyruvate kinase deficiency and
more than 60% of patients with CMML.” Autoantibodies have increased level of HbF. Acquired hemoglobin H, increased
been reported in 22% of all patients with MDS, but they are a expression of the i antigen, loss of ABO blood group antigens,
more frequent occurrence (present in more than 50% patients) unmasking of the Tn antigen, and increased sensitivity to
in CMML. Platelet antibodies are present in 55% of the cases complement (positive serum acid or sugar water test) have
studied, and erythrocyte autoantibodies in 46%. The direct also been reported.””*° At times, loss of erythrocyte mem-
antiglobulin test has been found positive in 8% of the cases brane proteins such as decay accelerating factor (DAF)
where IgG! and C3d were detected on the erythrocytes.” CD55, and glycosyl phosphatidyl] inositol (GPI)-linked anti-
gen CD59, may also be observed. Concomitantly, such loss
Evaluation of Progenitor Cell Growth may be observed on neutrophils and other cells, thereby over-
lapping with paroxysmal nocturnal hemoglobinuria.
in Semisolid Media
Several investigators have studied in vitro growth patterns WHITE BLOOD CELLS
from MDS patients. There is a specific pattern of colony Abnormal granulocyte function is frequently observed
clusters that grow in culture from normal progenitor cells. with decreased cytoplasmic granules and myeloperoxidase
430 Chapter 19 Myelodysplastic Syndromes
deficiency. Alkaline phosphatase and a-naphthol acetate these patients have been previously exposed to epipodophyllo-
esterase isoenzyme activities may also be decreased in granu- toxins and anthracyclines, which target DNA topoisomerase if
locytes. Chemotaxis, adhesion, phagocytosis, and microbicidal often in combination with other drugs including alkylating
capacity may be impaired.*'** These functional abnormalities agents. DNA topoisomerases are a class of enzymes important
are not necessarily related to cellular hypogranulation. Abnor- in various DNA transactions such as replication, transcription,
mal phagocytosis, chemotaxis, or bactericidal activities in and recombination.?! Most of the balanced translocations
neutrophils have been closely associated with chromosome involve 11q23 and different partners such as chromosomes 4, 6,
7 anomalies—monosomy 7 and del(7q). These specific dys- 9, or 19. Usually the MLL gene (located at 11q23) is rearranged.
functions are associated with a high frequency of bacterial Balanced translocations involving the gene RUNX1I(AMLI) at
infections. In these situations, the GP130 molecule, produced 21q22 are also reported in this setting. Patients with these
by a gene located on chromosome 7, is secreted in inadequate translocations usually have a short latency between exposure
amounts, and is responsible for the major defect in chemotaxis and development of sMDS, and tend to present with acute
and phagocytosis.*? leukemia with monocytic features. The sMDS caused by topoi-
somerase II inhibitors respond better to treatment than those
caused by alkylating agents.
PLATELETS
Platelet aggregation, adhesion, and other platelet functions
are also frequently impaired in MDS.™ Other laboratory find- Myelodysplastic Syndromes in Children
ings that may be found in MDS include abnormal vitamin B,,
and folate levels, increased serum ferritin and transferrin sat- MDS occurs rarely in children, and most cases are of the
uration, and increased muramidase (in serum and urine of RAEB and RAEB-t subtypes. Children are most often symp-
patients with CMML). tomatic with fever, pallor, and asthenia.”* Unlike adults, chil-
dren more commonly present normocytic, normochromic
anemia rather than macrocytic anemia. Several syndromes
Secondary Myelodysplastic Syndromes such as Fanconi’s anemia, Down’s syndrome, Kostmann’s
agranulocytosis, and Shwachman’s syndrome are associated
MDS are said to be secondary when they occur after signifi-
with an increased risk of development of acute myeloblastic
cant exposure to chemotherapy, radiotherapy, or other toxic
leukemia in children and should be investigated.”*°* Mono-
agents.°!*°8° The term “secondary myelodysplastic syn-
somy 7 syndrome and juvenile chronic myeloid leukemia
dromes” (SMDS) seems more appropriate than the often-seen
usually present with leukocytosis and hepatosplenomegaly
“therapy-related MDS” because exposure to toxic agents is
and are considered myeloproliferative disorders rather
also responsible for sMDS. The laboratory findings, clinical
than MDS.”
manifestations, and evolution resemble those of de novo MDS,
but sMDS show a higher frequency of clonal chromosomal
abnormalities and a greater tendency to early leukemic transfor- Clinical Features
mation, particularly if a complex karyotype is present. The prog-
nosis is generally poorer in sSMDS than in de novo MDS. Most commonly, the presenting symptoms are those attributable
There has been an increase in the number of patients diag- to progressive bone marrow failure. Clinical manifestations
nosed with sMDS in recent years, which is likely attributable develop in relationship to the degree of anemia, neutropenia, or
to the increased usage of intensive chemotherapy protocols. thrombocytopenia. Elderly patients with anemia may present
Between 5% and 10% of patients submitted to chemotherapy symptoms of cardiac failure, such as dyspnea on exertion.
(with or without radiotherapy) as preparative regimen for autol- Fatigue and weakness may also be noted, particularly in patients
ogous stem cell transplantation will develop sMDS.*’*’ Most with RAEB and RAEB-t. Symptoms related to infection and
cases are causally related to previous therapy with alkylating hemorrhage may be encountered in patients with MDS and
agents, almost all of which have been shown to be leuke- severe bone marrow failure. Patients without excess numbers of
mogenic. The leukemogenic potential of alkylating agents is blasts in the bone marrow may remain asymptomatic for a long
enhanced further by the use of radiotherapy. The period of period of time. Rarely, patients may present systemic symptoms
latency between exposure to alkylating agents and development such as infection, arthralgias, weight loss, fever, and cutaneous
of sMDS is usually 6 to 8 years, and patients usually have a poor vasculitis.
response to therapy with the exception of allogeneic bone Abnormal physical findings are neither prominent nor
marrow transplantation. Recently, the use of the epipodophyllo- specific. The spleen may be palpable in 20% of patients, and
toxins etoposide and tenoposide has been shown to be leuke- the liver may be enlarged in approximately 10%. Enlarged
mogenic if administered in combination with cisplatinum, lymph nodes are not usually present. Compared to the other
doxorubicin, or alkylating agents.”° In patients with MDS and a types of MDS, in CMML, splenomegaly, hepatomegaly, lym-
history of previous exposure to alkylating agents, cytogenetic phadenopathy, and nodular cutaneous leukemic infiltrates are
studies usually show unbalanced cytogenetic aberrations, pri- more common.”°*’ The overall clinical picture of CMML is
marily —7, —5, 5q—, and 7q—. Recently, an increasing number of more closely related to the myeloproliferative disorders such
patients with sMDS have been observed with balanced chromo- as CML, polycythemia vera, essential thrombocytosis, and
somal translocations. Studies have revealed that the majority of primary myelofibrosis.
Chapter 19 Myelodysplastic Syndromes 431
RARS
RA
RAEB
CMML
RAEB-t
10 20 30 40 50 60 0 10 20 30 40 50 60
Survival in months MDS subtypes Leukemic progression in %
Figure 19-45 m Survival and leukemic progression for each subgroup of MDS. Data pooled from several series with a total of 3554 patients.
432 Chapter 19 Myelodysplastic Syndromes
Karyotype:
“Good” = normal, —y, del (5q) only, del (20q) only
“Poor” = complex (= 3 abnormalities) or chromosome 7 anomalies
“Intermediate” = other abnormalities
Cytopenias = Hb < 100 g/L; neutrophils < 1.5 X 10°/L; platelets < 100 x 10°/L
*This group is recognized as AML in the WHO classification.
Sources: From Greenberg, P, et al: International scoring system for evaluating prognosis in myelodysplastic syndromes.
Blood 89:2079, 1997; and Heany, ML, and Golde, DW: Myelodysplasia (review article). N Engl J Med 340:1649, 1999.
documentation of the JAK2V617F mutation will orient the t(9;22) chromosome abnormality by standard cytogenetics,
diagnosis toward a myeloproliferative disorder.'’ molecular studies by FISH or PCR techniques may be neces-
sary. CMML with the t(5;12)(q33;p13) and other chromoso-
mal translocations involving tyrosine kinase genes give rise to
MDS with Features of Chronic a clinical picture that is closely related to CML.'°* According
Myelogenous Leukemia to the WHO, MDS falling into this category should be classi-
fied as myelodysplastic/myeloproliferative diseases.
Because CMML may present with a very high WBC count
and a significant number of immature myeloid cells in the
peripheral blood, this disease may mimic CML. Cytogenetic MDS versus Acute Erythroid Leukemia
studies are of major importance to distinguish between these
two entities. The finding of t(9;22) is diagnostic of CML. MDS are often difficult to differentiate from acute erythroid
Because 5% to 10% of patients with CML may not exhibit the leukemia when the total population of marrow erythroblasts
exceeds 50% of all nucleated cells. In that situation, the num-
AML FAB M2 with less
ber of blasts may not exceed the minimal requirement of 20%
than 30% blasts (WHO) or 30% (FAB) of all nucleated cells. FAB and WHO
AML FAB M6 when
have suggested that to establish the correct diagnosis, the
MDS with PNH features erythroblasts are >50% number of blasts should be calculated as a percentage of non-
erythroid cells (NEC). If the percentage exceeds 20% (WHO)
or 30% (FAB) of NEC, the diagnosis is AML of acute ery-
throid leukemia (WHO) or AML M6 (FAB) rather than MDS
(Fig. 19-45).
Treatment
Allogeneic stem cell transplantation is the only therapeutic
MDS with hypocellular marrow MDS with fibrosis modality with the potential to cure MDS. However, as most
MDS with thrombocytosis patients with MDS are elderly, they are not amenable to
transplantation and are therefore incurable. As there is signif-
Figure 19-44 @ Overlap between MDS and other hematologic
disorders. AA = aplastic anemia; MPD = myeloproliferative icant prognostic heterogeneity among MDS patients, the
disorder; PNH = paroxysmal nocturnal hemoglobinuria; AML = IPSS is an invaluable tool that helps tailor therapy to the pre-
acute myeloid leukemia. dicted prognosis. In general, elderly patients in the IPSS low
Chapter 19 Myelodysplastic Syndromes 433
Ques tions
1.Which type of anemia is most common in MDS? : Which of the following agents may lead to secondary
~ a. Microcytic, hypochromic MDS (sMDS)?
b. Normocytic, hypochromic a. Hydrocortisone
c. Macrocytic, normochromic b. Alkylating agents
d. Dimorphic c. Irradiated blood components
2. Which of the following morphological abnormalities are d. Folic acid
helpful in the diagnosis of MDS? . Which of the following 1s most likely to affect prognosis
CO
chromosomes 11, 12, and 20. There are no chromosomal @ Secondary myelodysplastic syndromes (SMDS) occur after
abnormalities specifically associated with any given sub- significant exposure to chemotherapy, radiotherapy, or other
group of MDS. However, the Sq— anomaly is more frequent
toxic agents. The laboratory findings, clinical manifesta-
in RA. tions, and evolution resemble those of de novo MDS, except
for a higher frequency of chromosomal abnormalities and a
m The 5q— syndrome has specific clinical features that greater tendency to early leukemic transformation.
include macrocytic anemia, normal or elevated platelet
@ MDS occurs rarely in children. RAEB and RAEB-t
count, mild leukopenia, and in the bone marrow,
(AML) are the most frequent.
monolobular or dwarf megakaryocytes and erythroid
hypoplasia. Patients are predominantly female. w Clinical manifestations of patients with MDS are related to
the degree of anemia, neutropenia, or thrombocytopenia.
m Cell surface antigens of peripheral blood cells, bone mar-
row, or biopsy specimens can be studied by flow cytome- # Progenitor cell growth in semisolid media is dramatically
try or immunocytochemistry via alkaline phosphatase decreased in MDS, with the exception of CMML.
antialkaline phosphatase (APAAP) and other techniques. # Several factors influence the prognosis in MDS. The most
w Generally, the ability of hematopoietic progenitor cells to significant are cytogenetic abnormalities, severity of
form colonies in vitro is decreased or absent. Aberrant cytopenias, age of the patient, and percentage of blasts.
growth patterns are mostly observed in RAEB and @ The International Prognosis Scoring System (IPSS) helps
RAEB-t. In CMML, increased colony growth is noted. determine the prognosis of individuals with MDS.
# Functional abnormalities of red blood cells, white blood mAt the present time, allogenic hematopoietic stem cell
cells, and platelets are frequently observed in MDS. transplantation is the only curative treatment for MDS.
REFERENCES itil. Busque, L, and Gilliland, DG: Clonal 20. Shetty, V, et al: Measurement of apopto-
evolution in acute myeloid leukemia. sis, proliferation and three cytokines in
fo . Bennett, JM, et al: Proposals for the
Blood 82:337, 1993. 46 patients with myelodysplastic syn-
classification of thé myelodysplastic 2. Tricot, G, et al: The myelodysplastic dromes. Leuk Res 20:891, 1996.
syndromes. Br JHaematol 51:189, syndromes: different evoiution patterns De Knudson, AG, Jr: Mutation and cancer:
1982. based on sequential morphological and statistical study of retinoblastoma. Proc
tO. Bennett, JM: Classification of the
cytogenetic investigations. Br JHaema- Natl Acad Sci USA 68:820, 1971.
myelodysplastic syndromes. Clin
(Koll SOI), 1k5), . Santibanez-Koref, MF, et al: p53
Haematol 15:909, 1986. 13 . Michiels, JJ, et al: Bleeding prophylaxis germline mutations in Li-Fraumeni syn-
Oo. Jaffe, E, et al: World Health Organiza- i
for major surgery in patients with type drome. Lancet 338:1490, 1991.
tion Classification of Tumours: Pathol- 2 von Willebrand disease with an . Liu, L, et al: Inhibition of thrombin
ogy and Genetics of Tumours of intermediate purity factor VIII-von by antithrombin HI and heparin
Haematopoietic and Lymphoid Tissues. Willebrand factor concentrate Haemate P. cofactor II in vivo. Thromb Haemost
Lyon, France: IARC Press, 2001.
Blood Coagul Fibrinolysis 15:323, 2004. 73:405, 1995.
ee a ae ENA Incidence eRe 14. Kereveur, A, et al: Production of tissue fac- . Carter, G, et al: Genetic lesions in
pice epidemiological ESOS tor pathway inhibitor in cardiomyocytes preleukemia. Crit Rev Oncogenet 3:339,
myelodysplastic syndromes, Br J and its upregulation by interleukin-1. 1992.
Becpiatol G28, 1272. Thromb Haemost 86:1314, 2001. . Quesnel, B, et al: Methylation of the
Nn
En, ee uses ee oo 15. Gerritsen, WR, et al: Clonal analysis of pI5(INK4b) gene in myelodysplastic syn-
EOC Neu, SSE JULES ae myelodysplastic syndrome: Monosomy 7 dromes is frequent and acquired during
control study and correlation with is expressed in the myeloid lineage, but disease progression. Blood 91:2985, 1998.
clinicobiological findings. Br JHaematol not in the lymphoid lineage as detected 26. Dacie, J, and Lewis, SM: Practical
10S Tee a : : by fluorescent in situ hybridization. Haematology, ed 6. Churchill Living-
. Almhanna, K, et al: Sideroblastic anemia Blood 80:217, 1992. stone, Edinburgh, 1984, p 107.
after bariatric surgery. Am J Hematol 16. Tokunaga, F, et al: Cellular basis for pro- ile Hamblin, TJ, and Oscier, DG: The
81:155, 2006. tein C deficiency caused by a single myelodysplastic syndrome—a practical
. Aksoy, M, and Erdem, S: Followup amino acid substitution at Arg15 in the guide. Hematol Oncol 5:19, 1987.
study on the mortality and the develop- gamma-carboxyglutamic acid ___domain. . Hast, R, et al: Diagnostic significance
ment of leukemia in 44 pancytopenic J Biochem Tokyo 120:360, 1996. of dysplastic features of peripheral
patients with chronic exposure to 7 . Busque, L, et ai: Nonrandom X-inactivation blood polymorphs in myelodysplastic
benzene. Blood 52:285, 1978. patterns in normal females: Lyonization syndromes. Leuk Res 13:173, 1989.
. Strom, SS, et al: Risk factors of ratios vary with age. Blood 88:59, . Kouides, PA, and Bennett, JM: Morphol-
myelodysplastic syndromes: A case- 1996. ogy and classification of myelodysplastic
control study. Leukemia 19:1912, 2005. . Parker, JE, and Mufti, GJ: Ineffective syndromes. Hematol Oncol Clin North
. Mecucci, C, and Van den Berghe, H: haemopoiesis and apoptosis in Am 6:485, 1992.
Cytogenetics. Hematol Oncol Clin North myelodysplastic syndromes. Br J 30. Goasguen, JE, et al: Prognostic implica-
Am 6:523, 1992. Haematol 101:220, 1998. tion and characterization of the blast cell
10. Willems, GM, et al: Transient high affin- . Yoshida, Y: Hypothesis: Apoptosis may population in the myelodysplastic syn-
ity binding of tissue factor pathway be the mechanism responsible for the drome. Leuk Res 15:1159, 1991.
inhibitor-factor Xa complexes to nega- premature intramedullary cell death in Sul. Hast, R: Sideroblasts in myelodysplasia:
tively charged phospholipid membranes. the myelodysplastic syndrome. their nature and clinical significance.
Biochemistry 37:3321, 1998. Leukemia 7:144, 1993. Scand J Haematol Suppl 45:53, 1986.
438 Chapter 19 Myelodysplastic Syndromes
ws)bo. Lindriz, EA: Pelger-Huet anomaly, the 47. Yoshida, Y, et al: Refractory myelodys- 5 in malignant myeloid diseases. Proc
Stodtmeister type. Atlas of Haematology, plastic anaemias with hypocellular bone Natl Acad Sci USA 90:5484, 1993.
ed 2. Sandoz, Medyczne Wydawnictow marrow. J Clin Pathol 41:763, 1988. 62. Horrigan, SK, et al: Delineation of a
Multimedialne, Gdansk, Poland, 2001, . Mitelman, F: An International System minimal interval and identification of
plate 143B, p 82 for Human Cytogenetic Nomenclature. 9 candidates for a tumor suppressor gene
55) Jakobsen, PE, et al: Ballooning platelets S. Karger, Basel, 1995. in malignant myeloid disorders on 5q31.
in the myelodysplastic syndromes, MDS. . Gersen, L, and Keagle, MB: The Princi- Blood 95:2372, 2000.
Eur JHaematol 53:175, 1994. ples of Clinical Cytogenetics. Humana 63. Cvirn, G, et al: Effects of alpha(2)-
34. Bennett, JM: A comparative review of Press, Totowa, NJ, 1999. macroglobulin and antithrombin on
classification systems in myelodysplastic 50. Heim, S, and Mitelman, F: Chromosome thrombin generation and inhibition in
syndromes, MDS. Semin Oncol 32:S3, abnormalities in the myelodysplastic syn- cord and adult plasma. Thromb Res
2005. dromes. Clin Haematol 15:1003, 1986. 101:183, 2001.
is)nN . Swirsky, DM, et al: 8;21 translocation in 5 _—. Le Beau, MM, et al: Clinical and 64. Martinez-Ramirez, A, et al: Cytoge-
acute granulocytic leukaemia: Cytologi- cytogenetic correlations in 63 patients netic profile of myelodysplastic syn-
cal, cytochemical and clinical features. with therapy-related myelodysplastic dromes with complex karyotypes: An
Br JHaematol 56:199, 1984. syndromes and acute nonlymphocytic analysis using spectral kar-yotyping.
. Gale, RP, and Cannani, E: The molecu- leukemia: Further evidence for charac- Cancer Genet Cytogenet 153:39, 2004.
lar biology of chronic myelogenous teristic abnormalities of chromosomes 65. Barouk-Simonet, E, et al: Role of multi-
leukaemia. Br JHaematol 60:395, 1985. no. 5 and 7. J Clin Oncol 4:325, 1986. plex FISH in identifying chromosome
Sih. Golub, TR, et al: Fusion of PDGF recep- . Pedersen-Bjergaard, J, and Philip, involvement in myelodysplastic syn-
tor beta to a novel ets-like gene, fel, in P: Balanced translocations involving dromes and acute myeloid leukemias
chronic myelomonocytic leukemia with chromosome bands 11q23 and 21q22 with complex karyotypes: A report on
t(5;12) chromosomal translocation. Cell are highly characteristic of myelodys- 28 cases. Cancer Genet Cytogenet
77:307, 1994. plasia and leukemia following therapy 157:118, 2005.
38. Vardiman, JW, et al: The World Health with cytostatic agents targeting at DNA- 66. Trost, D, et al: Molecular cytogenetic
Organization (WHO) classification of the topoisomerase I. Blood 78:1147, 1991. profiling of complex karyotypes in pri-
myeloid neoplasms. Blood 100:2292, Se). Van den Berghe, H, et al: The 5q— mary myelodysplastic syndromes and
2002. anomaly. Cancer Genet Cytogenet acute myeloid leukemia. Cancer Genet
3h). Navarro, I, et al: Classification and 17:189, 1985. Cytogenet 165:51, 2006.
scoring systems in myelodysplastic syn- 54. Yegneswaran, S, et al: Protein S alters 67. Martinez-Ramirez, A, et al: Analysis
dromes: A retrospective analysis of the active site ___location of activated pro- of myelodysplastic syndromes
311 patients. Leuk Res 30:971, 2006. tein C above the membrane surface. A with complex karyotypes by high-
40. Schmalzl, F: The value of cytochemical fluorescence resonance energy transfer resolution comparative genomic
investigations in the diagnosis of the study of topography. J Biol Chem hybridization and subtelomeric CGH
myelodysplastic syndromes. In EASON, MINT. array. Genes Chromosomes Cancer
Schmalzl, F, and Mufti, GJ (eds), SD: Hasle, H, et al: Myelodysplastic syn- 42:287, 2005.
Myelodysplastic Syndromes. Springer- drome, juvenile myelomonocytic 68. McKee, PA, et al: Molecular structural
Verlag, New York, 1992, p 44. leukemia, and acute myeloid leukemia studies of human factor VIII. Ann NY
4 fk. De Pasquale, A, and Quaglino, associated with complete or partial Acad Sci 240:8, 1975.
D: Enzyme cytochemical studies in monosomy 7. European Working Group 69. Dumas, JJ, et al: Crystal structure of
myelodysplastic syndromes. In on MDS in Childhood, EWOG-MDS. the wild-type von Willebrand factor
Schmalzl, F, and Mufti, GJ (Eds): Leukemia 13:376, 1999. Al-glycoprotein Ibalpha complex
Myelodysplastic Syndromes. Springer- 56. Lai, JL, et al: Myelodysplastic syn- reveals conformation differences with
Verlag, New York, 1992, p 51. dromes and acute myeloid leukemia with a complex bearing von Willebrand
42. Seo, IS, et al: Myelodysplastic syn- 17p deletion. An entity characterized by disease mutations. J Biol Chem
drome: Diagnostic implications of specific dysgranulopoiesis and a high 279:23327, 2004.
cytochemical and immunocytochemical incidence of P53 mutations. Leukemia 70. Koeller, U, et al: Immunological phe-
studies. Mayo Clin Proc 68:47, 1993. CESTAO), iS). notyping of blood and bone marrow
43. Tricot, G, et al: Prognostic factors in the SM. Testoni, N, et al: 3q21 and 3q26 cyto- cells from patients with myelodysplas-
myelodysplastic syndromes: Importance genetic abnormalities in acute tic syndromes. In Schmalz], F, and
of initial data on peripheral blood myeloblastic leukemia: biological Mufti, GJ (eds), Myelodysplastic
counts, bone marrow cytology, trephine and clinical features. Haematologica Syndromes. Springer-Verlag,
biopsy and chromosomal analysis. Br J 84:690, 1999. New York, 1992, p 60.
Haematol 60:19, 1985. 58. Apperley, JF, et al: Response to imatinib 7 —_. Knox, SJ, et al: Studies of T-lymphocytes
44. Delacretaz, F, et al: Histopathology of mesylate in patients with chronic myelo- in preleukemic disorders and acute
myelodysplastic syndromes. The FAB proliferative diseases with rearrangements nonlymphocytic leukemia: In vitro
classification, proposals applied to bone of the platelet-derived growth factor recep- radiosensitivity, mitogenic responsive-
marrow biopsy. Am J Clin Pathol tor beta. N Engl J Med 347:481, 2002. ness, colony formation, and enumeration
87:180, 1987. Sy). Greenberg, PL: Risk factors and their of lymphocytic subpopulations. Blood
. Rios, A, et al: Bone marrow biopsy in relationship to prognosis in myelodys- 61:449, 1983.
myelodysplastic syndromes: Morpholog- plastic syndromes. Leuk Res 22(Suppl TWP, Anderson, RW, et al: Lymphocyte abnor-
ical characteristics and contribution to 1):S3, 1998. malities in preleukemia—I. Decreased
the study of prognostic factors. Br J 60. Van den Berghe, H, et al: Distinct NK activity, anomalous immunoregula-
Haematol 75:26, 1990. haematological disorder with deletion of tory cell subsets and deficient EBV
46. Tricot, G, et al: Bone marrow histology in long arm of no. 5 chromosome. Nature receptors. Leuk Res 7:389, 1983.
myelodysplastic syndromes. I. Histologi- 251:437, 1974. 1: Mufti, GJ, et al: Immunological abnor-
cal findings in myelodysplastic syndromes 6 jae. Le Beau, MM, et al: Cytogenetic and malities in myelodysplastic syndromes.
and comparison with bone marrow molecular delineation of the smallest I. Serum immunoglobulins and autoan-
smears. Br JHaematol 57:423, 1984. commonly deleted region of chromosome tibodies. Br JHaematol 63:143, 1986.
Chapter 19 Myelodysplastic Syndromes 439
74. Sokol, RJ, et al: Erythrocyte autoanti- transplantation for lymphoid malignan order the test and how to interpret the
bodies, autoimmune haemolysis, and cies. J Clin Oncol 12:2527, 1994. results. Leuk Res 30:739, 2006.
myelodysplastic syndromes. J Clin 90. Pedersen-Bjergaard, J, and Philip, P: 108. Wadleigh, M, et al: After chronic myel-
Pathol 42:1088, 1989. Two different classes of therapy-related ogenous leukemia: Tyrosine kinase
75. Richert-Boe, KE, and Bagby, GC, Jr: In and de-novo acute myeloid leukemia? inhibitors in other hematologic malig-
vitro hematopoiesis in myelodysplasia: Cancer Genet Cytogenet 55:119, 1991. nancies. Blood 105:22, 2005.
Liquid and soft-gel culture studies. Hema- . Liu, LF: DNA topoisomerase poisons 109. Kushner, J, et al: Secondary iron over-
tol Oncol Clin North Am 6:543, 1992. as antitumor drugs. Annu Rev Biochem load. American Society for Hematology
70. Everson, MP, et al: Interleukin-6 and Dessall, UNIS). Education Program Book, Orlando, FL,
granulocyte-macrophage colony- . Blank, J, and Lange, B: Preleukemia in 2001, p 47.
stimulating factor are candidate growth children. J Pediatr 98:565, 1981. 110. Sasahara, Y, et al: Deficient activity
factors for chronic myelomonocytic . Linman, JW, and Bagby, GC, Jr: The of von Willebrand factor-cleaving
leukemia cells. Blood 74:1472, 1989. preleukemic syndrome, hemopoietic protease in patients with Upshaw-
H.. Gumina, RJ, et al: Characterization of dysplasia. Cancer 42:854, 1978. Schulman syndrome. Int J Hematol
the human platelet/endothelial cell adhe- . Kleihauer, E: The preleukemic 74:109, 2001.
sion molecule-! promoter: Identification syndromes, hematopoietic dysplasia in Lh. Hellstrom-Lindberg, E, et al: Treatment
of a GATA-2 binding element required childhood. Eur J Pediatr 133:5, 1980. of anemia in myelodysplastic syn-
for optimal transcriptional activity. . Sieff, CA, et al: Monosomy 7 in child- dromes with granulocyte colony-
Blood 89:1260, 1997. hood: A myeloproliferative disorder. stimulating factor plus erythropoietin:
78. Salmon, C: Blood groups changes in Br JHaematol 49:235, 1981. Results from a randomized phase I
preleukemic states. Nouv Rev Fr 90. Copplestone, JA, et al: Monocytic skin study and long-term follow-up of 71
Hematol Blood Cells 17:211, 1976. infiltration in chronic myelomonocytic patients. Blood 92:68, 1998.
PD. Hauptmann, G, et al: “False positive” leukaemia. Clin Lab Haematol 8:115, 2. Molldrem, JJ, et al: Antithymocyte
acidified serum test in a preleukemic 1986. globulin for patients with myelodys-
dyserythropoiesis. Acta Haematol 59:73, D7. da Silva, MA, et al: Extramedullary plastic syndrome. Br JHaematol
1978. disease in myelodysplastic syndromes. MSIE), Oi.
80. Liaw, PC, et al: Patients with severe Am J Med 85:589, 1988. 1S Saunthararajah, Y, et al: HLA-DR15
sepsis vary markedly in their ability to 98. Mufti, GJ, et al: Myelodysplastic syn- (DR2) is overrepresented in myelodys-
generate activated protein C. Blood dromes: A scoring system with prognostic plastic syndrome and aplastic anemia
104:3958, 2004. significance. Br JHaematol 59:425, 1985. and predicts a response to immunosup-
8 i . Martin, S, et al: Defective neutrophil oY), Kerkhofs, H, et al: Utility of the FAB clas- pression in myelodysplastic syndrome.
function and microbicidal mechanisms in sification for myelodysplastic syndromes: Blood 100:1570, 2002.
the myelodysplastic disorders. J Clin investigation of prognostic factors in 237 114. Biesma, DH, et al: Immunosuppressive
Pathol 36:1120, 1983. cases. Br J Haematol! 65:73, 1987. therapy for hypoplastic myelodysplastic
82. Boogaerts, MA, et al: Blood neutrophil 100. Yunis, JJ, et al: Refined chromosome syndrome. Cancer 79:1548, 1997.
function in primary myelodysplastic analysis as an independent prognostic Ils). Baccarani, M, et al: Low dose arabi-
syndromes. Br J Haematol 55:217, 1983. indicator in de novo myelodysplastic nosyl cytosine for treatment of
i. Gyger, M, et al: Childhood monosomy 7 syndromes. Blood 67:1721, 1986. myelodysplastic syndromes and sub-
syndrome. Am J Hematol 13:329, 1982. 101. Aul, C, et al: Primary myelodysplastic acute myeloid leukemia. Leuk Res
84. Lintula, R, et al: Platelet function in syndromes: Analysis of prognostic fac- Tess), Mss).
preleukaemia. Scand J Haematol 26:65, tors in 235 patients and proposals for an 116. Raza, A, et al: Thalidomide produces
1981. improved scoring system. Leukemia transfusion independence in long-
85. Iurlo, A, et al: Cytogenetic and clinical Ea, WS. standing refractory anemias of patients
investigations in 76 cases with therapy- . Botella, LM, et al: Transcriptional acti- with myelodysplastic syndromes. Blood
related leukemia and myelodysplastic vation of endoglin and transforming 98:958, 2001.
syndrome. Cancer Genet Cytogenet growth factor-beta signaling compo- 117. Appelbaum, FR, and Anderson,
43:227, 1989. nents by cooperative interaction J: Bone marrow transplantation for
86. Pedersen-Bjergaard, J, et al: Therapy- between Sp| and KLF6: Their potential myelodysplasia in adults and children:
related myelodysplasia and acute role in the response to vascular injury. When and who? Leuk Res 22(Suppl 1):
myeloid leukemia. Cytogenetic charac- Blood 100:4001, 2002. Sao MOOK.
teristics of 115 consecutive cases and 3. Goasguen, JE, et al: Prognostic factors 118. Fenaux, P, et al: Aggressive chemother-
risk in seven cohorts of patients treated of myelodysplastic syndromes—a sim- apy in adult primary myelodysplastic
intensively for malignant diseases in the plified 3-D scoring system. Leuk Res syndromes. A report on 29 cases. Blut
Copenhagen series. Leukemia 7:1975, 14:255, 1990. SHIM, NOS.
ISEB). 104. Greenberg, P, et al: International scor- 9} Wiyjermans, PW, et al: Continuous infu-
87. Stone, RM, et al: Myelodysplastic syn- ing system for evaluating prognosis in sion of low-dose 5-aza-2°-deoxycytidine
drome as a late complication following myelodysplastic syndromes. Blood in elderly patients with high-risk
autologous bone marrow transplantation 89:2079, 1997. myelodysplastic syndrome. Leukemia
for non-Hodgkin’s lymphoma. J Clin 105. Cilloni, D, et al: Significant correlation 11(Suppl 1):S19, 1997.
Oncol 12:2535, 1994. between the degree of W7/ expression 120. Kantarjian, H, et al: Decitabine
. Miller, JS, et al: Myelodysplastic syn- and the International Prognostic Scoring improves patient outcomes in
drome after autologous bone marrow System Score in patients with myelodys- myelodysplastic syndromes: Results of
transplantation: An additional late com- plastic syndromes. J Clin Oncol 21:1988, a phase III randomized study. Cancer
plication of curative cancer therapy. 2003. 106:1794, 2006.
Blood 83:3780, 1994. 106. Pagliuca, A, et al: Myelofibrosis in pri-
89. Darrington, DL, et al: Incidence and char- mary myelodysplastic syndromes: A
acterization of secondary myelodysplastic clinico-morphological study of 10
syndrome and acute myelogenous cases. Br JHaematol 71:499, 1989.
leukemia following high-dose chemora- 107. Tefferi, A, and Pardanani, A: Mutation
diotherapy and autologous stem-cell screening for JAK2V6/7F: When to
Chapter
Chronic Lymphocytic
Leukemia and Related
Lymphoproliferative
Disorders
Laurel D. Holmer, MEd, MT(ASCP)SH
Carlos E. Bueso-Ramos, MD, PhD
Introduction OBJECTIVES
Chronic Lymphocytic At the end of this chapter, the learner should be able to:
Leukemia/Small
Lymphocytic Lymphoma 1. List general features of chronic lymphocytic leukemia.
Etiology and 2. Name laboratory methods used to study lymphocytes in lymphoproliferative disorders.
Pathophysiology
Immunologic Features and 3. List diagnostic criteria of chronic lymphocytic leukemia.
Methods for Studying 4, Describe treatment for chronic lymphocytic leukemia.
Lymphocytes
Clinical Features 5. Explain differential diagnostic criteria that are used to characterize lymphoproliferative
Laboratory Features disorders.
Chromosomal Abnormalities
Clinical Course, Prognostic
Factors, and Staging
Treatment
Differential Diagnosis
Case Study 1
Case Study 2
440
Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders 44]
failed programmed cell death (PCD), also called apoptosis.* accompanied by a large fraction of CLL cells in S/G,, phase
In essentially all self-renewing tissues, new cell production is of the cell cycle.
normally offset by a commensurate amount of cell destruction Expression of some of these cell cycle regulatory fac-
through PCD. Imbalances in the activities of opposing genes tors, e.g., cdk4, cyclin E, and cyclin D2 as well as other cell-
that either promote or block physiologic apoptosis can, there- cycle dependent factors such as nucleoside diphosphate
fore, slow or halt the rate of cell turnover, creating a selective kinase NM23-F2 in CLL lymphocytes led to the hypothesis
survival advantage for a particular clone that permits expan- that CLL cells are not quiescent but are recruited to the cell
sion at the expense of its normal neighbors.* cycle and blocked in its early G, phase. These observations
Most B-CLL cells have been reported to contain high show that the accumulation of CLL lymphocytes is due not
levels of the antiapoptotic protein BCL2.? The mechanisms only to defective apoptosis but also to an abnormality of the
responsible for the high amounts of BCL2 observed in more regulation of cell proliferation.
than 80% of B-CLL cells remain unknown, but only rarely do Additional infiltration of the lymph nodes and spleen by
they involve rearrangements of the BCL2 gene as a result of the malignant lymphocytes occurs in 50% of patients,
chromosomal translocations.'° whereas cutaneous invasion occurs in 5% of patients.'° As the
Although the pathogenesis of the disease remains largely bone marrow becomes more extensively infiltrated by the
unknown, previous investigations have focused on the defec- leukemic clone, marrow replacement results in anemia,
tive apoptosis of the malignant cells that seems to play an thrombocytopenia, and neutropenia (Fig. 20-2). Organ infil-
important role in disease progression and chemotherapy resis- tration can lead to massive adenopathy with splenomegaly,
tance.'' There is additional evidence that the deregulation of hypersplenism, and subsequent peripheral cytopenias. An
cell cycle regulatory genes may contribute to the expansion of increased tendency for hemorrhage further contributes to ane-
the malignant clone. CLL consists not only of resting lympho- mia and compromises hemostasis.
cytes, but also of proliferating leukemia cells, which have Patients with CLL have significantly impaired immuno-
been des¢ribed in proliferation centers in the lymph node and logic activity. Hypogammaglobulinemia is found in approxi-
the bone marrow. This proliferative compartment is thought mately 50% of patients with CLL. The deficiency in Ig leads to
to be important for disease progression.'*!? infections with a variety of agents. Bacterial infections, espe-
Cell cycle progression in mammalian cells is governed cially of the respiratory tract, urinary tract, and skin, as well as
by interactions between cyclins, cyclin-dependent kinases viral infections such as herpes zoster and herpes simplex are
(CDKs) and cyclin-dependent kinase inhibitors. Individual common and dramatically contribute to patient morbidity and
cyclins act at different phases of the cell cycle by binding to mortality (Fig. 20-3). Autoimmunity is a phenomenon that is
suitable cdks and allowing the subsequent activation of the frequently seen in CLL, with 15% to 35% of patients develop-
cyclin-cdk complexes. The D-type cyclins (D1, D2, and D3) ing autoimmune hemolytic anemia at some time during the
regulate the G, phase progression by binding to and stimulat- course of the disease.'® Antibodies produced against red blood
ing the activities of their catalytic partners: cdk4 and cdk6. cells and detected with the direct antiglobulin (Coombs) test
Kinases cdk4 and cdk6 phosphorylate pRB protein. This may precede, simultaneously occur with, or follow the devel-
process leads to inactivation of pRB and release of a tran- opment of CLL. Red cell aplasia is a rare occurrence.'’ Autoan-
scription factor E2F, which activates transcription of the tibodies to platelets and neutrophils may also develop and lead
components of the DNA replication machinery, thereby com- to immune thrombocytopenic purpura (ITP) and neutropenia.
mitting the cell to S phase of the cell cycle. A family of pro- The production of autoantibodies coupled with marrow crowd-
teins termed cdk inhibitors exerts the inhibitory effect on the ing and hypersplenism can lead to low peripheral platelet and
cdks or cyclin—cdk complexes. Two classes of these inhibitors
have been described so far. The first one (p15, p16, and p19)
is specific for pRB kinases, 1.e., cdk4 and cdk6, the second
one (p21, p27, and p57) has a broader range of activity and
inhibits most of the known cdk-cyclin complexes. D-type
cyclins regulate early G, progression and are thought to be
growth factor sensors linking extracellular signals to the cell
cycle. In contrast to normal B-cells, cyclin D2 mRNA but not
cyclin D3 mRNA is overexpressed in CLL cells.'* Expression
of D-type cyclins is needed to activate cyclin-dependent
kinases and mediate phosphorylation of target proteins.
Recent research has indicated that cyclin D2 and D3 are
both key regulatory cyclins in CLL cells with cdk4 as
the corresponding catalytic partner.'° Phosphorylation of the
retinoblastoma (RB) protein identifies the restriction point in
G, of the cell cycle. The expression of RB protein is altered
in CLL. However, when an appropriate second signal is pro- Figure 20-2 @ Photomicrograph of bone marrow aspirate smear
vided, the proliferative hyporesponsiveness of CLL cells can from a patient with CLL. Note monotonous appearance of mature-
be overcome and rapid phosphorylation of the RB protein is appearing lymphocytes with condensed nuclear chromatin.
Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders 443
Massive adenopathy
Impaired Splenomegaly
immunologic
activity
Hypersplenism
esi
Hemorrhage
Hypogammaglobulinemia
|
Infection
Thrombocytopenia
Neutropenia
Figure 20-4 m The pathophysiology of CLL. The three major processes that typically interact are marrow replacement by long-lived lympho-
cytes, hypersplenism, and autoimmunity. ITP = immune thrombocytopenic purpura.
444 Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders
Immunoperoxidase and flow cytometry Vator Piticrentiation antigens on B cells siaionT ak usingit anes antibodies
(see Table 20-3)
Molecular probes Rearrangements of the B-cell Ig and T-cell receptor genes
Cytogenetics Consistent chromosomal abnormalities such as t(8;14)-B-ALL, Burkitt’s lymphoma;
t(14;18)—follicular lymphoma; trisomy 12—CLL, SLL; t(11;14)—mantle cell
lymphoma; t(4;11)-ALL-L2
Cytochemistry Tartrate-resistant isoenzyme 5 of acid phosphatase in hairy cells
Localized alpha-naphthol acetate esterase positivity in Golgi area of T cells, i.e., T-ALL,
Sézary cell, T-CLL
Electron microscopy Nuclear and cytoplasmic ultrastructure such as nuclear whorls in Sézary cells and
_Tibosomal lamellar cytoplasmic aggregates iin hairy cells
Immunophenotypic marker expression and gene differentiation. Studies have shown, however, that under certain
rearrangement during normal B-cell ontogeny is shown in in vitro conditions such as stimulation with phorbol ester, typi-
Figure 20-5. The malignant B lymphocytes of CLL do not cal CLL cells can undergo transformation to more mature levels
progress normally to the final stages of B-cell development, of B-cell development.*??' Genetic recombination in pre-B cells
namely the formation of plasma cells, but rather appear to be in the bone marrow generates an immature B cell that expresses
developmentally arrested at an earlier B-lymphocyte stage of a functional Ig on the surface, IgM or IgM and IgD, which then
“Table 20-3 Cluster Differentiation (CD) Markers and Clinical Application ate a
Markers (CD Designation) — Monoclonal Antibodies _ Clinical Application —
B Cells
CD5 Ole eent B-CLL, some NHL, T lymphomas
CD9 BA-2 (p24) Pre-B
CD10 J5, BA-3 (CALLA) Lymph progenitor, CALL, some NHL
CD19 B4, Leul2 CALEB Clik, Bapebat Cle
CD20 B1, Leul6 CALL, B-CLIE B-PLIc Hel
@p22) B3, Leul4 Late B cells, hairy cells
CD23 CD23.1 (BU38), Leu20 B-CLL, activated B cell
CD24 BA-1 Most B cells
@D25 TAC (IL-2 receptor) HEE
CD43 Leu22 B-CLL, mantle cell lymphoma
CD79 CD79 B cells
CD103 HML-1 Hairy cells
T Cells
CD1 T6 some T-€LIE, 1-PLL EALD
GD? T11, LeuS, 9.6 (E rosette) T-CLL, T-PLL, Sézary cells, LGL, ATLL
Gps eed! T-CELL, T-PLL, Sézary cells, IM
CD4 T4, Leu3 T-PLL, Sézary cells, IM, ATLL
CD5 JUL, ieewmil, MOA T-CLL, T-PLL, Sézary cells, ATLL
CD7 3A1, Leu9 T-PLL
CD8 T8, Leu2 T-CLL, some LGL, IM
CD25 Tac (IL-2 receptor) ATLL
CD57 Leu7 (HNK1) LGL
Other Cells
CD38 T10, Leul7 Plasma cells, germinal center B cells,
cortical thymocytes
CD52 _CAMPATH- 1H All lymphoid cellsand monocytes
B-CLL= B-Giee chronicfoipraplion teleukemia; NHL = non-Hodgkin’sEe iyniphonaneCALL = common acute
lymphocytic leukemia; B-PLL= B-lineage prolymphocytic leukemia; HCL= hairy-cell leukemia; T-ALL= T- lineage
acute lymphoblastic leukemia; LGL= large granular lymphocytosis (T-gamma lymphocytosis): ATLL = adult T-cell
leukemia/lymphoma; IM = infectious mononucleosis; HNK = human natural killer.
Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders 445
B-CELL ONTOGENY exits the bone marrow as a virgin (mature) B cell (antigen-
independent phase) and migrates to the secondary lymphoid tis-
sues. The normal B cell may undergo stimulation by antigen
(antigen-dependent phase) and enter the lymphoid follicle. At
p rearrangement this point in differentiation, the somatic hypermutation mecha-
nism is activated, introducing point mutations into the variable
K rearrangement
(V) region genes. In the presence of limiting antigen, there will
i rearrangement be subsequent selection of V gene sequences that provide
Cytoplasmic immunoglobin increased affinity for antigen. After antigen-driven B-cell prolif-
eration, B cells can initiate two pathways of maturation. Some
Surface immunoglobin
B cells revert back to small lymphocytes to generate memory
HLA-DR B cells, whereas others undergo terminal differentiation into
CD19 long-lived plasma cells.**** The postulated normal B-cell devel-
CD24 opment is shown in Figure 20-6.
The level of somatic mutation in CLL is low.* This has
CD10
led to the conclusion that the cell of origin has not encoun-
CD20 tered antigen. The subset that remains unmutated, perhaps
CD21 derived from naive B cells, appears prone to accumulate the
CD22
trisomy 12 abnormality. In contrast, the mutated subset of
CLL, perhaps derived from more mature B cells, appears
CD23
more prone to abnormalities of 13q14.**
Figure 20-5 @ Hypothetical scheme of marker expression and The characteristic immunophenotype for B-CLL, is
gene rearrangement during normal B-cell ontogeny. (From Pui, CH, expression of faint or low-density slg with kappa (Kk) or
et al: Clinical and biologic relevance of immmunologic marker stud- lambda (\) light-chain restriction and expression of B-cell-
ies in childhood acute lymphoblastic leukemia. Blood 82[2]:344, associated antigens (CD19, CD20, CD79a), CD5, CD23,
1993, with permission.)
CD43, and faint CD11c (Table 20-4). A typical example of a
«
B-CLL immunophenotype is shown in Figure 20-7.
PREFOLLICULAR FOLLICULAR :
Lymphoblasts Mature
“Virgin”
B cell
Centrocytes_
2aved cell
SPs s)
tes
Memory B
cells
Lymph Node
Figure 20-6 ™ Postulated B-lymphocyte development scheme. Lymphoblasts undergo several maturation steps in the microenvironment of
the bone marrow (BM), giving rise to mature (virgin or naive) lymphocytes. Naive B lymphocytes enter blood vessels (BV) to reach lymph
nodes and populate the mantle zone of the lymphoid follicles. These cells differentiate into centroblasts (large, noncleaved follicular center
cells) in the dark zone of the germinal center. Centroblasts undergo rearranged-immunoglobulin gene hypermutation and develop into
centrocytes (cleaved follicular center cells). Centrocytes that bind antigen on the surface of the follicular dendritic cells survive, whereas those
that fail to bind die by apoptosis. Surviving centrocytes in the light zone of the germinal center may differentiate into monocytoid B cells,
which populate the marginal zone of the lymphoid follicle, and plasma cells, which reenter the bone marrow and memory cells. Memory cells
actively recirculate between the blood, lymph, and lymphoid organs.
446 Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders
Of particular interest is the unique expression of CD5 in are necessary. Aberrant loss of T-cell-associated antigens, such
B-CLL. Expression of CD5 is normally seen on T lympho- as CD3, CD5, and CD7, or the predominance of specific T-cell
cytes and on the majority of B cells of early ontogeny such as subsets, either CD3+CD4+, CD3+CD8+, or CD31CD4+
cord blood cells; however, normally less than 20% of B cells CD8-+, in blood or bone marrow suggests the need for T-cell
in adult human peripheral blood express CD5. Although receptor molecular studies. A typical example of a T-CLL/
CD5-negative B-CLL does exist,*’° it accounts for fewer T-PLL immunophenotype is shown in Figure 20-9.
than 10% of all B-CLL cases. The precise functional role of When the conventional immunophenotypic techniques
the CD5 molecule is unknown. CDS has been shown to phys- fail to reveal the nature of the lymphoid neoplasm, molecular
ically associate with the antigen-specific receptor complex probe technology using DNA probes can often contribute to
present on both T and B lymphocytes.” Studies are ongoing the diagnosis and classification of malignancy by detecting
to determine whether differences in antigen expression and gene rearrangements that occur in lymphocytes. On a molec-
gene rearrangement are associated with variation in clinical ular level, Ig heavy and light chains are rearranged. The
course.*°?*? As mentioned earlier, T-CLL/T prolymphocytic rearrangement of heavy-chain Ig genes occurs first and is
leukemia is rare but can be distinguished from B-CLL on the followed by rearrangement of light-chain genes. The order of
basis of differentiation antigens. Immunophenotypic marker this rearrangement proceeds from mu () to k to \ and is the
expression and gene rearrangement during normal T-cell earliest detectable commitment to B-cell development.*!
ontogeny is shown in Figure 20-8. Monoclonality rather than polyclonality (the presence of a
The demonstration of slg in B-CLL was mentioned earlier mixture of k- and \-bearing B lymphocytes) is a feature of
and is, in fact, the classic marker for B cells. The detection of a many malignancies, including CLL; however, it is not, per se,
predominance of either k or \ light chains by the B cells indi- indicative of malignancy. Analogous to the detection of Ig
cates monoclonality’? In contrast, no immunophenotypic gene rearrangements in B cells, is the ability of molecular
markers for T-cell clonality are available, and molecular studies probes to detect rearrangement patterns of the genes coding
Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders 447
1118YW.001 x 1118YW.003 for the T-cell receptor (TCR), the antigen-specific surface
1000 SAE i Alen reas
molecule characteristic of T cells.** With the use of Ig and
= TCR gene probes to detect gene rearrangements, the unusual
:
a
oO
;
Ww
a
case of CLL that cannot be diagnosed and classified by mor-
phology and cell markers can now be characterized. It must
wo oO
wn
be stressed that it is extremely important that the final diagno-
oO sis of any lymphoproliferative disorder be made as a result of
40° ae10' 10° 10° 104 10° 10' 10% 10% 104 composite information from clinical data in addition to mor-
CD45 PerCP CD19 FITC phological, histologic, and immunologic analysis.
4° 1118YW.008 =° 1118YW.004 Although CLL is generally characterized by an elevated
~
white blood cell count in which there are abundant lympho-
cytes to analyze, if the number of neoplastic cells is low, a
os on
T-CELL ONTOGENY
TCR 6 rearrangement
TCRy rearrangement
TCRB rearrangement
TCR @ rearrangement
CD7
CD5
CD2
CD1
CD4
CD8
CD3
TCR (af or y 8) Cytoplasmic |=iFay
Figure 20-8 @ Hypothetical scheme of marker expression and gene rearrangement during T-cell ontogeny. TCR = T-cell receptor.
(From Pui, CH, et al: Clinical and biologic relevance of immmunologic marker studies in childhood acute lymphoblastic leukemia. Blood
82[2]:344, 1993, with permission.)
448 Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders
1068BR.001 1060BR.003
1000
800
=o
as
Dn oO
= 10°
9g
OSs
CD5
PE
a
Oo o
N oe
a
10° 10! 10° 10° 10 CD19 FITC
CD45 PerCP
Be 4060ADD.002 1060ADD.003
[o}
w
a
oO
Q
oO PE
CD3
10° 10°
CD4 FITC CD8 FITC
1060ADD.004 1060ADD.005
PE
CD3 PE
CD4
10° 10 40° 10
CD7 FITC CD7 FITC
Figure 20-9 @ Flow cytometric analysis of T-prolymphocytic leukemia (T-PLL). Leukemic peripheral blood lymphocytes are gated by CD45-side
scattered analysis. Comparison of CD5 versus CD19 shows a predominance of CD5-positive cells (upper panel). Comparisons of CD3 versus CD4
and CD3 versus CD8 demonstrate a predominance of CD4+ T lymphocytes (middle panel). CD4+ T lymphocytes also express CD7 antigen
(lower panel).
males as females. Unlike acute leukemia, the signs and symp- gastrointestinal tract, prostate, and gonads.** CLL has also
toms of CLL develop gradually, and the onset of the disease been reported to occur simultaneously with acute myeloblastic
is difficult to pinpoint. In fact, it is not unusual for the leukemia (AML).*°
disease to be accidentally discovered during the course of a
routine visit to a physician. The duration of a relatively asymp-
tomatic phase of CLL is extremely variable. Unexplained
Laboratory Features
absolute and persistent lymphocytosis; cervical, supraclavicu-
lar, or axillary lymphadenopathy; and splenomegaly are the The requirements for the diagnosis of CLL have undergone
earliest signs of CLL. The clinical course is indolent, but as the revision since earlier criteria were established by Rai and col-
disease progresses, chronic fatigue, recurrent or persistent leagues*” in 1975 and Binet and colleagues in 1981.*!4? The
infections, and easy bruising are the consequences of anemia, International Workshop on Chronic Lymphocytic Leukemia‘
neutropenia, B-cell immunologic dysfunction, and throm- recommends a minimum peripheral blood B-cell lymphocytosis
bocytopenia. Hepatomegaly may accompany splenomegaly. of 5000 cells/uL (5 x 10° cells/L) along with a 30% lymphocy-
Dermatologic manifestations such as nodular and diffuse tosis of the bone marrow, consisting of morphologically mature-
skin infiltrations, erythroderma, exfoliative dermatitis, and appearing lymphocytes. This guideline is similar to criteria
secondary skin infections may occur. Leukemic lymphocytes established by the National Cancer Institute-sponsored working
may invade unusual locations such as the scalp, orbits, subcon- group. A new classification and terminology for the lympho-
junctivae, gums, pharynx, pleura and lung parenchyma, cytic neoplasms, including CLL, has been proposed.*®
Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders 449
T cells were discussed earlier in the section entitled Immuno- Clinical Course, Prognostic Factors,
logic Features and Methods for Studying Lymphocytes. and Staging
Immune dysfunction within the proliferating B cells is
indicated by the presence of hypogammaglobulinemia or The overall median survival for CLL is currently 4 to 5 years;
hypergammaglobulinemia and monoclonal gammopathy. The 50% of patients are living 5 years after diagnosis, whereas
frequent expression of autoantibodies in CLL contributes to a 30% have a 10-year survival. CLL can be an indolent disease
variety of autoimmune phenomena, including those leading to with an asymptomatic presentation and may not require any
anemia and thrombocytopenia. treatment until progressive lymphocytosis of the peripheral
blood and marrow, lymphadenopathy, splenomegaly, anemia,
neutropenia, thrombocytopenia, autoimmune phenomena,
and infection develop. This may be as late as 10 to 15 years
Chromosomal Abnormalities
after the initial diagnosis. In contrast to those with an indolent
The most common chromosomal abnormality in B-CLL is course of disease, approximately 20% of patients with CLL
an extra chromosome 12, called trisomy 12 (in 15% to 20% have a very aggressive clinical course that progress rapidly
of cases),°!°* which may occur alone or together with dele- from initial diagnosis and results in death within | to 2 years.
tions or translocations of chromosome 13q14 (Gin 25% of The wide variation seen among patients is not fully under-
cases). The 13q14 deletions represent early clonal aberra- stood, but clinical and pathologic data have been used to try
tions and suggest the presence of a tumor suppressor gene to predict the CLL patient’s prognosis and identify various
whose loss or inactivation may be crucial to development stages and risk groups.
of CLL. Most of the 13q14 deletions are undetectable at The Rai system,*° the Binet system,*' and the International
the cytogenetic level but are detectable in more than 50% Workshop on CLL system* are the three major staging systems
of the CLL cases with the use of molecular probes for the developed for CLL; however, only the Rai system is widely
13q14 région.** used in the United States. The Rai and Binet staging systems,
Genetic abnormalities, including the most common along with median survival for each system by stage, are shown
chromosomal abnormalities and gene rearrangements, are in Table 20-5. Staging systems for CLL do not consistently pre-
listed in Table 20-4.°'° The presence of multiple chromoso- dict whether a patient’s clinical course is more likely to be indo-
mal abnormalities in B-CLL has been implicated as a poor lent or progressive. The most reliable predicting factors for
prognostic indicator. None of the proto-oncogenes involved indolent CLL are blood lymphocyte doubling time (LDT)
in the pathogenesis of other mature B-cell malignancies, greater than 12 months*’ and a nondiffuse pattern of bone mar-
including BCL2, BCL6, PAX5, and c-MYC, show primary row lymphocyte infiltration,*® along with a Rai stage of 0, I, or
alterations in CLL.°° II. A short LDT (less than 12 months), a diffuse lymphocyte
Binet System
A Two or fewer node-bearing regions, no it
anemia or thrombocytopenia (Hgb > 10 g/dL,
platelets > 100 x 10°/L)
B Three or more node-bearing regions + no 5
anemia or thrombocytopenia
GE Anemia and/or thrombocytopenia 2
independent of regions involved
Parameter, ae
Source: Bink JL, et al: Perspective on the use of new DAienoene oe in the treatment ofeee oes here
Blood 107:859, 2006.
infiltration of the bone marrow (see Fig. 20-11A), a Rai stage of the original CLL or an independent disease; these include
III or IV, an elevated level of serum 8,-microglobulin, elevated cytogenetic analysis, immunoglobulin gene rearrangement by
level of soluble CD23 in serum, and the presence of chromoso- Southern blot analysis, and anti-idiotypic antibodies. Mole-
mal abnormalities, are associated with a more progressive clin- cular studies have shown that the development of Richter’s
ical course and shorter survival duration.®*“ In previous, mostly syndrome in CLL may represent either the identical clone of
retrospective analyses, the parameters shown in Table 20-6 cells present in the preceding CLL or a different malignant
have shown promising results. The first four parameters of clone.” In addition, p53, a tumor suppressor gene that is fre-
Table 20-6 have proven prognostic value independent of the quently mutated in a variety of human cancers, has been dis-
clinical stage in several studies. However, the methods for mea- covered in 15% of CLL patients and in 40% of patients with
suring some of these parameters (ZAP-70, Fig. 20-11B, serum Richter’s syndrome.’ The close association of p53 with
markers, cytogenetics, and mutational status of immunoglobu- transformation of CLL into a very aggressive lymphoma may
lins) have yet to be fully standardized and/or may not be readily also be a prognostic indicator for resistance to chemotherapy
feasible in most clinical laboratories. Further, the prognostic by interference with normal programmed cell death
value of some of the more easily accessible markers (apoptotic) pathways in tumor cells.*.”°
(e.g., B.-microglobulin) has not been as well established as that
of immunoglobulin mutation status or certain cytogenetic Treatment
abnormalities (e.g., 20p—).
Several types of transformation in B-CLL have been Some patients diagnosed with CLL do not require immediate
described: prolymphocytoid transformation, which is rela- treatment; however, when the signs and symptoms of pro-
tively low grade and slowly progressive; Richter’s syndrome gressive disease appear, it is time to begin therapeutic inter-
(diffuse large-cell lymphoma), which is rapidly progressive vention. Major physical and clinical signs and symptoms
and accounts for about 5% of all deaths in CLL®*’; and acute identify advancing disease. These include progressive mar-
leukemia. Transformation to acute Jeukeinia is unusual in row failure with resulting anemia; thrombocytopenia and
CLL. Most patients with CLL die with residual leukemia and neutropenia; progressive lymphocytosis; progressive lym-
usually succumb to infection or a cause totally unrelated to phadenopathy; enlarging spleen; autoimmunity (autoimmune
their CLL, such as cardiovascular disease.®* The onset of the hemolytic anemia or ITP); and increased susceptibility to
terminal transformation of CLL is suggested when there is a infection and persistent constitutional symptoms such as
proliferation of a new population of lymphoid cells, namely, night sweats, fever, and weight loss.
larger cells with immature-appearing morphological features, Conventional treatment for CLL is chemotherapy. Com-
a finer nuclear chromatin pattern, and a prominent nucleolus. binations of chemotherapeutic agents are used for patients
This morphological transformation is often accompanied by with disease that is refractory to conventional therapy or those
the appearance of complex chromosomal changes that were with advanced disease.°'”*’? The initial management of
not present earlier or are in addition to the commonly present patients with CLL according to Rai staging groups is summa-
trisomy 12. The proliferation of a more malignant clone of rized in Table 20-7.
cells is accompanied by an increasing resistance to therapy Radiation therapy is an alternative or adjunctive therapeu-
and an exceptionally poor prognosis.°* tic approach in treating CLL.’* Leukemic masses in enlarged
A variety of techniques is available to help determine lymph nodes and in the spleen may respond to focused, local
whether the transformation represents a clonal evolution of irradiation to relieve discomfort or eliminate obstruction. As
452 Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders
with chemotherapy, irradiation also has its detrimental effects, between 5 X 10° and 10 X 10° cells/L, the presence of both
especially in terms of causing life-threatening neutropenia. bone marrow infiltration of more than 30% lymphocytes and
Splenectomy is recommended in patients with massive B-CLL immunophenotype (dim CD20+, CD19+/CD5+,
splenomegaly and autoimmune hemolytic anemia or autoim- CD23+, FMC7-, and weak surface intensity Ig) is necessary
mune thrombocytopenia resulting from splenic pooling that is for the diagnosis of CLL.
uncontrollable by chemotherapy.” The distinction between CLL and ALL is easily made in
In addition to chemotherapeutic intervention, the use most instances based on morphological differences of the pro-
of high-dose intravenous gammaglobulin therapy prevents liferating cell population (see Fig. 20-12A to L). The differ-
major bacterial infections,*’*' and the immunosuppressant ence between the smoother nuclear chromatin pattern of the
cyclosporine, a fungal metabolite, aids in the prevention or lymphoblast in ALL and the heavy condensation of nuclear
treatment of red cell aplasia, both of which can be manage- chromatin in the CLL lymphocyte is readily appreciated when
ment problems in patients with CLL. examining an appropriate monolayer area or feather-like
Experimental therapies are also being studied. They edge of a well-stained blood smear. Lymphoblasts show
include not only new drugs but also the use of the monoclonal positive expression of the nuclear enzyme terminal deoxynu-
antibodies.°****° cleotidyl transferase (TdT). B-CLL cells are TdT-negative.
Bone marrow transplantation (autologous and _allo- B-Prolymphocytic leukemia (B-PLL) is characterized by a
geneic) is being explored as a possible curative therapy for predominance of circulating prolymphocytes (greater than
patients with aggressive CLL, especially in patients younger 55%, usually greater than 70%). Prolymphocytes are larger,
than 50 years of age. 87°? 9° less mature-appearing cells than the typical lymphocytes seen
in CLL, with moderately condensed nuclear chromatin and a
Differential Diagnosis prominent vesicular nucleolus (see Fig. 20—-12C). The clinical
and laboratory features that make PLL a distinct lymphopro-
As previously outlined in Table 20—1, CLL is a lymphoprolifer- liferative disorder include extreme leukocytosis (often greater
ative disorder that must be differentiated from other malignant than 100 X 10° cells/L) and prominent splenomegaly without
or reactive lymphoid proliferations. The differential diagnosis lymphadenopathy.**°°°* As in all disorders accompanied by
includes: acute lymphoblastic leukemia (ALL); prolymphocytic leukocytosis, morphological detail of the predominating
leukemia (PLL)**°°; non-Hodgkin’s lymphomas in leukemic cell may not be appreciated unless appropriate areas of
phase, especially mantle cell lymphoma (MCL); small cleaved- well-stained blood and bone marrow smears are examined.
cell lymphoma (SCCL); hairy cell leukemia (HCL); Sézary syn- Prolymphocytes may be seen in patients with CLL but
drome; T-cell large granular lymphocytic leukemia; and reactive account for less than 10% of the circulating cells (see
lymphocytosis (Fig. 20—12A). The morphological and immuno- Fig. 20-12D). When 11% to 55% prolymphocytes are pre-
logic characteristics of these lymphoproliferative disorders are sent, a mixed-cell type of CLL, designated CLL/PLL is diag-
shown in Table 20-8. In addition to these disorders, other hema- nosed.” This category includes patients with prolymphoid
tologic malignancies may be confused with CLL. These include transformation. Most cases of PLL are B cell in nature
adult T-cell leukemia/lymphoma (ATLL) and Waldenstrom’s (B-PLL) as demonstrated by strong CD20 and sIg (in contrast
macroglobulinemia.”” to weak CD20/sIg in CLL), and reactivity with the B-cell
The diagnosis of CLL requires a sustained absolute lym- markers CD19 and CD22.'
phocytosis of mature-appearing lymphocytes in the absence For cases of T-cell CLL or PLL, in addition to
of other causes. The diagnosis of CLL is established when the immunophenotype two cytochemical techniques are helpful
peripheral blood lymphocyte count is 10 X 10° or more in distinguishing T cells from B cells, namely, the e-naphthol
cells/L (which is typically the case), lymphocyte infiltration acetate esterase (ANAE) stain and the acid phosphatase (AP)
of the bone marrow is more than 30% lymphocytes of all reaction.'*' The typical T-lymphocyte pattern with ANAE and
nucleated cells, and the circulating lymphocytes have a AP is one large, localized dot of reaction product positivity
B-CLL immunophenotype. When lymphocyte counts are (Fig. 20-13).
A. Chronic lymphocytic leukemia (CLL). B. Acute lym-
Figure 20-12 ™ Periph eral blood smears of various lymphoproliferative disorders.
prolymphocyte. E. Small lymphocytic lymphoma (SLL) in
phoblastic leukemia (ALL). C. Prolymphocytic leukemia (PLL). D. CLL with occasional
G. Hairy-cell leukemia (HCL). H. Sézary syndrome. |. Adult T-cell leukemia/lymphoma.
leukemic phase. F. Small cleaved-cell lymphoma (SCCL).
mononucleosis with atypical lymphocytes. L. Plasma cell dyscrasia.
J. T-gamma lymphocytosis with large granular lymphocytes. K. Infectious continued
454 Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders
Small lymphocytic lymphoma (SLL) is the nodal counter- lymphoma is shown in Tables 20-4 and 20-8. In contrast to
part of B-CLL.* It is a diffuse non-Hodgkin’s lymphoma B-CLL, the cells in MCL lack expression of the CD23 surface
characterized by neoplastic transformation of small B lym- antigen.*°!° A typical example of the MCL immunopheno-
phocytes (Fig. 20-14). type is shown in Figure 20-15. The bright slg expression and
Mantle cell lymphoma arises from the centrocytes of the positivity for CD19, CD20, CDS, and FMC7 are characteris-
mantle zone of lymphoid follicles (see Fig. 20-6). In the tic. This immunophenotype also distinguishes MCL from
lymph node, MCL usually exhibits a diffuse pattern with follicular lymphoma, which are CD10+ and CD5— (see
complete effacement of lymph node architecture.** It can also Table 20-4). In most cases, a distinctive chromosomal
form a nodular or mantle zone pattern. MCL is composed of translocation t(11;14)(q13;q32) may occur.'°?'® It involves
a homogeneous population of small- to intermediate-sized the Ig heavy-chain locus on chromosome 14 and the BCL/
neoplastic lymphoid cells, usually slightly larger than normal locus on the long arm of chromosome 11. The hybrid gene is
lymphocytes. The nuclear characteristics and cytology of the associated with overexpression of the cyclin DI (PRAD /)
cells are variable: round to clefted nuclear contour, and mRNA and protein. This chromosomal translocation can be
clumped to disperse and blastoid chromatin. detected in MCL (70%) by Southern blot analysis or (30% to
MCL involves the peripheral blood in up to 25% of 45%) by PCR. Unlike most other B-cell neoplasms, MCL is
cases, and the peripheral blood count may exceed 200 x 10° cyclin D1+ (72% to 100%) as demonstrated using immuno-
cells/L. Under these circumstances, morphological recogni- histochemical staining on sections of formalin-fixed tissue.!°°
tion of MCL and its distinction from CLL/SLL are difficult. Small cleaved-cell lymphoma is also a non-Hodgkin’s
Identification of these two entities 1s important because MCL lymphoma consisting of B lymphocytes that may be nodular
is considered to have a poor prognosis, with a median survival (follicular) or diffuse in distribution*s (Fig. 20-16) and can
of 3 to 5 years. Bone marrow involvement may also occur progress to a leukemic phase.'”’ The circulating cells of SCCL
with or without peripheral lymphadenopathy. MCL shows are morphologically characterized by nuclei that are irregular
diffuse or focal bone marrow involvement (paratrabecular in shape and that demonstrate irregular clefts, notches, or
and nonparatrabecular). folds that may traverse the entire width of the nucleus (see Fig.
Neoplastic lymphoid cells in MCL cells are exclusively 20-12F). These abnormal lymphocytes typically have very
of B-cell lineage. The common immunophenotype of this scanty cytoplasm and were formerly called “lymphosarcoma
oe
a)
WY
Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders 4
quourmoid
A00ydur
S][29
Suepunqy
Avur
posuapue]
€dD 8d)40
aanORaY
aj
aj AsoydurA|
snonoojuy
|,
Jo/pue
jeordAyy
A]
SS]
-U09 jewiou
Ajjetouas oAeyorrydoinze
sopnueis
saponoea vydo
(I-MNH)
Jojdsoo1
sIsoopNUOUO;]nq
Sole, Ari
aq
(anesol
ose]
/pueortydoinze
sis
RUTUeS
¢€dD 8d)
(TOT)
24
Ajaie1opopy
sojonova
L$d9
TGDq)
10
Adoydus]
a JuBpunge
YMquoutwioidsopnueis
juoutuo.id
d31e7] pasuapuog
Iepnuels
BULUIE)-[,
sisoyA00ydus’] Iepnsoy juourwuold
JON ase] IPL
TY
[[99-poarajo seUoYyduiA]
Aueul spjoy
¢d)D €d)
7d) $d)
evuoydury
Ayueos
juourtuo.d JON
[S951
NSO]
‘Te[
ALVZIC Arezasg
[Jao
IUOIPUAS SUIUIe}S-yIeVG SOLIvA,
udd-V’1H
‘10}d9991
|[99-q jasqns
JAY SUIOS
SEMOICCa®)
S[S
[feuls
SUONS
Seed
S1e[NSOLUI
Z-"T])
Apustys
SUIPOJ
JOS
quourtuolidJON
ABUL
=
pasuapuos
Ajoyerapoul suonoafoid dG)
vc/0C/61
€OIGD
“OeL
Yd-VTH
TOH [joo
Arey UL]
0} Iejnsay
0} ayeIJOpopy
UIM SOLIRA,
(VTIVO)
BUONS
UNM
‘
OIGO+
JepnsoLy
vrurayne] onAdoydurAjoid = JJg ‘enuayney] onAdoydurcy] otuoIyS = TIO ‘erusyne] onsepqoydurAyT ajnov = [TV
_{euouqy,,
Ud-VTH Vd/0C/61d)D
Ayueos
SOLIBA,
[]90-g
3Is
ayAd0yduidoig
JON
JIP.LOSIC] aAd0ydurdy
* ‘sya
‘sayoiou
SPIOF
(enesol
TOS posuopuog OIdGO=
(%07)
€dO vd) dD
8d)0+
JUSUTUIOIg
snooudso.1a}0y
Ie[NSIY
199-L V1
€7/07/61G0 7d
0} Td TADq)
Ld)
AyuRos *SOLIVA, [199-4
snosuasouloy
Is
Ajaye1opoyy
uonejndod
‘WS]) (3]
ud-VTHSd)
Ne,
«[[@4 19990S,,
“SOLIVA,
Ayueos
:posuapuoZ,
3Is
snosussouloy
(%86)
AIM (%7)
TAD
A) €dO8d)
(eneso1
T10
1-2
yse|qoydwi
IepNsay juourwioid
JON [PL
TD
JUSUTUIOIg
UONRLICA
T1v
uowu0;)
IM SUIOS
/Puno! —%S1)
UI
ueumy
[eanyeu
JOT[PY
"][99
=
610) LPL
SSOLIVA
Aqueos
uonendod
[[90-], Ayreq
‘sare, TIV
ISLIIIVIVYD
dZIS
pur
258
[eAo %0C
J°
‘TIV
A}
St
[[99-[,
(ad
AT
(%0L)
Ud-VIH
(WTIVO)
OIdD
0cd)D
our
unewo1ys
TIV
surdjnuapy
uuse{doyAZ
SUNeUTUIOpaIg
Jeo[onN
[[99 adAq
JOyIeUL
[[2D IZIS
JUBIYTUSIS
10 uloned aTyoid
reaponN
adeys TTOOTONNY dISOTOUNTILT
456 Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders
1365WH.008 1365WH.008
wi
oO
= Lo
a
a (6)
o
t
oO
”
iri
CD19 FITC
1365WH.011 1365WH.015
Ww Ww
o a
o oO
N
a N
ra
oO (6)
10° 4
. FMC-7 FITC CD10 FITC
= 1365WH.012 1365WH.013
iS
Ww Ww
o a
2 QD
Q a
S) oO
10
KAPPA FITC LAMBDA FITC
Figure 20-15 @ Flow cytometric analysis of mantle cell lymphoma in leukemia phase. Leukemic marrow lymphocytes are gated by CD45-
scattered analysis. The plot of CD5 versus CD19 demonstrates dual positive neoplastic lymphocytes. In contrast to CLL, the neoplastic lympho-
cytes in mantle cell lymphoma show positive staining for FMC7 but no staining of CD23. Cyclin D1 immunohistochemical stain was positive.
marked variation in the size of the cells, ranging from that of Chronic T-cell large granular lymphocytic leukemia
a small lymphocyte to that of a large monocyte. Nucleoli are (LGL) has the morphological distinction of persistent circu-
typically inconspicuous but, when present, may be prominent lating lymphocytes that have abundant pale blue cytoplasm
and cause confusion with prolymphocytes. The clinical with azurophilic granules (see Fig. 20-12/). These granular
course of ATLL can be acute, with a high white cell count and lymphocytes usually constitute 50% to 95% of the circulating
survival less than | year; chronic, with a lower white cell white cells. Large granular lymphocytic leukemia was first
count and survival of more than | year; or “smoldering,” with described in patients with CLL of T-cell origin''® then subse-
a normal white cell count and low numbers of abnormal T quently in patients with chronic neutropenia.''’ Three distinct
lymphocytes. As seen in T-PLL and Sézary syndrome, ATLL clinical syndromes are now described in patients with an
cells show reactivity with T-cell-associated antigens (CD2, increased number of circulating LGL cells.''’ When LGL car-
CD3, and CD5); most are CD4+, CD25+ but usually lack ries a phenotype of T-LGL leukemia (a clonal proliferation of
CD7. Rare CD8+ cases have been reported. Mutation in the CD3+ LGL), then chronic neutropenia and autoimmunity,
tumor suppressor gene p53 is seen in 30% to 50% of patients especially rheumatoid arthritis, are characteristic.!!-'?!
with ATLL.'» Natural killer LGL leukemia is characterized by a clonal
—_
458 Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders
$45
and multiorgan involvement.'”? The majority of patients with
21 % ba increased numbers of CD3— cells do not have features of
3," NK-LGL leukemia but rather demonstrate a more indolent
clinical course.'?! Because quantitative abnormalities of LGL
ee"
eat are fairly common and their presence in peripheral blood may
represent a transient reactive phenomenon associated with
viral infections, it is important to perform immunophenotyp-
ing and molecular studies, and to correlate these data with the
clinical picture. Oral low-dose methotrexate has been shown
to be an effective treatment for some patients with LGL.'*°
Reactive (atypical) lymphocytosis is self-limiting, rarely
exceeds 5 X 10° cells/L, and is most commonly caused by a
viral infection such as infectious mononucleosis, viral hepati-
tis, and cytomegalovirus in adults and Bordetella pertussis in
Figure 20-16 @ Small cleaved-cell lymphoma (SCCL), lymph node. children.' The large reactive lymphocytes that characterize
viremia are polyclonal and T cell in origin. Abundant cyto-
plasm that may vary in degree of basophilia from very pale to
deep blue is the most prominent feature of the reactive lym-
phocyte. These cells often have an irregular nuclear outline
resembling a monocyte, and the nuclear chromatin is mostly
coarse (see Fig. 20—k2K). Reactive B-cell lymphocytosis is
rare. See Chapter 15, which discusses infectious mononucle-
osis and other causes of reactive lymphocytosis.
Plasma cell dyscrasias, namely Waldenstrom’s
macroglobulinemia,’’ multiple myeloma, and plasma cell
leukemia, may be associated with the presence of abnormal
circulating plasma cells (see Fig. 20-12L). Plasma cells are
characterized by abundant basophilic cytoplasm, an eccentric
nucleus with clumped nuclear chromatin, and a prominent
perinuclear clear zone. Plasma cells are end-stage B lympho-
cytes with the aforementioned characteristic morphology and
distinctive immunologic markers, namely, the presence of
monoclonal cytoplasmic Ig and expression of CD38. Plasma
cell disorders are covered extensively in Chapter 22.
Figure 20-17 ® Hairy-cell leukemia (HCL), bone marrow aspirate. The significant clinical, morphological, and immunophe-
notypic features of CLL, as well as treatment, prognosis, and
differential diagnosis, are shown in Table 20-9.
In summary, B-cell small lymphocytic disorders are
clonal diseases that usually present in an indolent fashion in
older patients. The diagnosis is often made incidentally from
a persistent lymphocytosis. B-cell CLL is a clonal accumula-
tion of mature-appearing B lymphocytes caused by failed
apoptosis, exhibiting monoclonal sIg. An unusual feature is
expression of the CD5 membrane antigen seen on mature T
lymphocytes. The B lymphocytes accumulate slowly in the
bone marrow, blood, spleen, liver, and lymph nodes. About
half of the patients with B-CLL exhibit hypogammaglobu-
linemia and an increased susceptibility to infection. Red cell
aplasia, immune-mediated anemia, and thrombocytopenia are
also common.
The course of CLL disease depends on the leukemia
burden, which can be assessed by clinical staging systems.
Lymphoproliferative disorders caused by T lymphocytes
are rare. T-prolymphocytic and ATLL are rapidly progressive
Figure 20-18 @ Tartrate-resistant acid phosphatase (TRAP) stain of diseases with more aggressive clinical courses. Morphologi-
peripheral blood showing positivity in hairy cell and no staining in cal and immunophenotypic studies, and positive HTLV-1
neutrophilic. serology, are helpful in characterizing these entities.
Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders 459
t2 1414WH.012 =iS
lop)
2
Ww
W aq
o See
= a
Q oO
) 5
©
oa oO -
5 if 3 4 P 10° 10! 102 103 ©6104
10 10 10 10 10 CD103 FITC
CD22 FITC
= 1414WH.015 a 1414WH.013
[o) ‘
So
el ‘
=
ae Ae
~
2
ae a
2a = a
oO
oO
3 ©
o (49)
a
oe 3°
2°
o
a
oO
© —
B g =
200
CD25 FITC
Figure 20-19 m Flow cytometric analysis of hairy-cell leukemia. Large mononuclear cells in leukemic bone marrow are gated by CD45-side
scattered analysis (left lower panel). The plots of CD22 versus CD11¢ and CD20 versus CD103 demonstrate predominance of dual positive B cells
(upper panel). The plots for both CD20 and 2X show X light-chain clonality (middle panel). The B cells are also reactive with anti-CD25
(anti-interleukin-2 receptor) (lower right-sided histogram).
460 Chapter 20. Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders
ALL = acute lymphoblastic leukemia; PLL = prolymphocytic leukemia; SLL = small lymphocytic lymphoma; SCCL =
small cleaved-cell lymphoma; HCL = hairy-cell leukemia; LGL = large granular lymphocytosis; ATLL = adult T-cell
leukemia/lymphoma.
ie
examination, however, disclosed retroperitoneal lym-
phadenopathy. Laboratory workup showed marked leuko-
cytosis (182 x 10° cells/L), thrombocytopenia (71 xX
A 64-year-old man with no significant past medical 10° cells/L), and anemia (hemoglobin, 8.1 g/dL). Periph-
history was admitted to the hospital with mild upper eral blood and bone marrow aspirate smears were per-
abdominal pain, malaise, anorexia, weight loss, and formed. More than 60% of the leukocytes counted in the
low-grade fever of 1-month’s duration. He denied having peripheral blood and 85% of all nucleated cells counted in
a cough, dyspnea, nausea, vomiting, jaundice, or recent the bone marrow specimen were described as “medium to
viral infection. His vital signs were essentially unremark- large atypical cells with distinct rim of cytoplasm, round
able except for a mild fever of 37.8° C. Physical nuclear contour, dispersed nuclear chromatin, and a single
examination revealed marked splenomegaly with no prominent nucleolus.”
hepatomegaly or peripheral lymphadenopathy. Radiologic
continued
©), estions
1. The ceils of CLL are morphologically identical to those of: ¢. The neoplastic lymphocytes in the blood or bone mar-
a. ALL Z = row are large with fine chromatin and prominent
b. Small lymphocytic lymphoma nucleoli.
c. Infectious mononucleosis Directions: Each item below contains one or more correct
d. Sézary syndrome an swers. Use the following letters to answer questions
. Surface immunoglobulin is the most reliable surface
i) 7 through 9.
marker for: a. If 1, 2, and 3 are correct
a. T lymphocytes , b. If 1 and 3 are correct
b. Plasma cells c. If 2 and 4 are correct
c. B lymphocytes d. If 4 is correct
d. Histiocytes e. If 1, 2, 3, and 4 are correct
Uo . Cells that demonstrate a positive reaction with the tar- . Which of the following findings would point to a diag-
trate-resistant acid phosphatase (TRAP) stain are most nosis of CLL?
likely: 1. Clonal proliferations of B lymphocytes
a. T lymphoblasts of ALL PACDIOT CDsicer CD23=
b. Atypical lymphocytes of a viral infection SR CDI9F ECMailee eD23en
c. Large granular lymphocytes of T-gamma lymphopro- 4, Ph chromosome—positive
liferative disorder . Which of the following clinical and laboratory manifesta-
d. Hairy cells of hairy-cell leukemia tions would point to a diagnosis of hairy-cell leukemia?
4, A mutated tumor suppressor gene found in a variety of 1. Pancytopenia
human cancers, including CLL is: 2. Splenomegaly
a, p53 3, D200 CD102Cm2a-
b. MDM2 4. Prominent generalized lymphadenopathy
c. HLA-DR . The poorest prognosis for patients with CLL is associ-
d. CD4 ated with which of the following features?
5. The immunophenotype that best describes mantle cell 1. Anemia
lymphoma is: 2. Splenomegaly
[email protected], FMC7 +, cyclin DI+ 3. Thrombocytopenia
bHGDS PaCD194— CD23-—, EMC] +, cyclin DI— 4. White cell count greater than 15,000 cells/uL
Peel) se eto tC D237, hMC/—, cyclin DI+ Directions: Choose the one best answer:
GeO DSee eC DIO smc 2am eMC Tt, cyclin DI+ 10 . The workup step(s) include:
1G D5a CD19. CD25c5,.kMCi+, cyclin DI+ a. Cytochemical staining, including myeloperoxidase and
6. In chronic lymphocytic leukemia: nonspecific esterase, and TdT
a. The absolute lymphocyte count is usually equal to or b. Immunohistochemical study of trephine biopsy sections
exceeds 5000 X 10° cells/L. c. Immunophenotypic study by flow cytometry
b. Hemoglobin level, platelet count, and absolute number d. Cytogenetic studies
of neutrophils may be normal or elevated. e. All of the above
5
46 Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders
. Immunophenotypic study using flow cytometry shows 12. Cytochemical stains (myeloperoxidase, nonspecific
negative T-cell markers. However, neoplastic cells esterase) and TdT performed on samples of bone mar-
express bright CD19, CD20, and weak CD5 and show row aspirate smear were negative. The most likely
monotypic kK light-chain restriction. Cyclin D1 diagnosis is:
immunohistochemical stain is negative. These cells will a. Chronic myelocytic leukemia, accelerated phase
also express: b. Typical chronic lymphocytic leukemia
a. CD103 c. Acute lymphoblastic leukemia
b. Strong FMC7 d. Chronic lymphocytic leukemia/prolymphocytic
Cleh?3 leukemia
d. Dim surface immunoglobulin io) . B-cell prolymphocytic leukemia
e. GD38
. Melo, JV, et al: Splenic B cell lymphoma Lippincott Williams & Wilkins, lymphocytic leukemia. Leukemia
with circulating villous lymphocytes: Philadelphia 2001, p 307. 14(8):1414, 2000.
differential diagnosis of B cell 35: Crescenzi, M, et al: Thermostable DNA . Rowley, JD, and Testa, JR: Chromosome
leukaemias with large spleens. J Clin polymerase chain amplification of abnormalities in malignant hematologic
Pathol 40(6):642, 1987. t(14;18) chromosome breakpoints and diseases. Adv Cancer Res 36:103, 1982.
20. Conley, CL, et al: Genetic factors predis- detection of minimal residual disease. 2. Knuutila, S, et al: Trisomy 12 in B cells
posing to chronic lymphocytic leukemia Proc Natl Acad Sci USA 85(13):4869, of patients with B-cell chronic lympho-
and to autoimmune disease. Medicine 1988. cytic leukemia. N Engl J Med 314(14):
(Baltimore) 59(5):323, 1980. 36. Stetlet-Stevenson, M, et al: Detection of 865, 1986.
2 —_. Caligaris-Cappio, F, et al: Lineage rela- occult follicular lymphoma by specific . Que, TH, et al: Trisomy 12 in chronic
tionship of chronic lymphocytic DNA amplification. Blood 72(5):1822, lymphocytic leukemia detected by fluo-
leukemia and hairy cell leukemia: stud- 1988. rescence in situ hybridization: analysis
ies with TPA. Leuk Res 8(4):567, 1984. . Spier, CM, et al: Chronic lymphocytic by stage, immunophenotype, and mor-
. Berek, C, and Milstein, C: Mutation drift leukemia in young adults. Am J Clin phology. Blood 82(2):571, 1993.
and repertoire shift in the maturation of Pathol 84(5):675, 1985. . Kalachikov, S, et al: Cloning and gene
the immune response. Immunol Rev . Johnson, LE: Chronic lymphocytic mapping of the chromosome 1|3q14
96:23, 1987. leukemia. Am Fam Physician 38(6):167, region deleted in chronic lymphocytic
23. Gray, D: Immunological memory. Annu 1988. leukemia. Genomics 42(3):369, 1997.
Rev Immunol 11:49, 1993. 3). Conlan, MG, and Mosher, DF: Concomi- . Gaidano, G, et al: Analysis of alterations
24. Oscier, DG, et al: Differential rates of tant chronic lymphocytic leukemia, acute of oncogenes and tumor suppressor
somatic hypermutation in V(H) genes myeloid leukemia, and thrombosis with genes in chronic lymphocytic leukemia.
among subsets of chronic lymphocytic protein C deficiency. Case report and Am J Pathol 144(6):1312, 1994.
leukemia defined by chromosomal review of the literature. Cancer 63(7): . Croce, CM, and Klein, G: Chromosome
abnormalities. Blood 89(11):4153, 1398, 1989. translocations and human cancer. Sci Am
1997. 40. Rai, KR, et al: Clinical staging of 252(3):54, 1985.
. Ikematsu, W, et al: Surface phenotype chronic lymphocytic leukemia. Blood . Dohner, H, et al: Chromosome aberra-
and Ig heavy-chain gene usage in 46(2):219, 1975. tions in B-cell chronic lymphocytic
chronic B-cell leukemias: expression of 4 — . Binet, JL, et al: A new prognostic classi- leukemia: Reassessment based on molec-
myelomonocytic surface markers in fication of chronic lymphocytic leukemia ular cytogenetic analysis. J Mol Med
CDS- chronic B-cell leukemia. Blood derived from a multivariate survival TIN 2669 19993
83(9):2602, 1994. analysis. Cancer 48(1):198, 1981. 58. Athan, E, et al: bcl-1 rearrangement.
. Shapiro, JL, et al: CD5- B-cell lympho- 42. Binet, JL, et al: A clinical staging system Frequency and clinical significance
proliferative disorders presenting in for chronic lymphocytic leukemia: Prog- among B-cell chronic lymphocytic
blood and bone marrow. A clinicopatho- nostic significance. Cancer 40(2):855, leukemias and non-Hodgkin’s lym-
logic study of 40 patients. Am J Clin NOW phomas. Am J Pathol 138(3):591, 1991.
Pathol 111(4):477, 1999. 43. Chronic lymphocytic leukemia: Recom- 5), Dyer, MJ, et al: BCL2 translocations in
. Kishimoto, T: Leukocyte Typing VI: mendations for diagnosis, staging, and leukemias of mature B cells. Blood
White Cell Differentiation Antigens. response criteria. International Workshop 83(12):3682, 1994.
Garland, New York, 1998, p 125. on Chronic Lymphocytic Leukemia. Ann 60. Miyashita, T, and Reed, JC: Bcl-2 onco-
. Newman, RA, et al: Phenotypic markers Intern Med 110(3):236, 1989. protein blocks chemotherapy-induced
and BCL-1 gene rearrangements in 44. Cheson, BD, et al: National Cancer apoptosis in a human leukemia cell line.
B-cell chronic lymphocytic leukemia: A Institute-sponsored Working Group guide- Blood 81(1):151, 1993.
Cancer and Leukemia Group B study. lines for chronic lymphocytic leukemia: 6 _ . Campos, L, et al: Effects of BCL-2 anti-
Blood 82(4):1239, 1993. Revised guidelines for diagnosis and sense oligodeoxynucleotides on in vitro
. el Rouby, S, et al: p53 gene mutation in treatment. Blood 87(12):4990, 1996. proliferation and survival of normal mar-
B-cell chronic lymphocytic leukemia is 45. Jaffe, ES, et al: World Health Organiza- row progenitors and leukemic cells.
associated with drug resistance and is tion Classification of Tumors: Pathology Blood 84(2):595, 1994
independent of MDRI/MDR3 gene and Genetics of Tumors of Haematopoi- 62. Korsmeyer, SJ, et al: Immunoglobulin
expression. Blood 82(11):3452, etic and Lymphoid Tissues. [ARC Press, gene rearrangement and cell surface
1993. Lyon, France, 2001. antigen expression in acute lymphocytic
30. Molica, S, et al: CD11c expression in 46. Rozman, C, et al: Bone marrow histo- leukemias of T cell and B cell precursor
B-cell chronic lymphocytic leukemia. logic pattern—the best single prognostic origins. J Clin Invest 71(2):301, 1983.
Blood 81(9):2466, 1993. parameter in chronic lymphocytic . Faguet, GB: Chronic lymphocytic
. Korsmeyer, SJ: Antigen receptor genes leukemia: a multivariate survival analy- leukemia: an updated review. J Clin
as molecular markers of lymphoid neo- sis of 329 cases. Blood 64(3):642, 1984. Oncol 12(9):1974, 1994.
plasms. J Clin Invest 79(5):1291, 1987. 47. Montserrat, E, et al: Lymphocyte dou- 64. Borthakur, G, et al: Chronic lymphocytic
. Knowles, DM: Immunophenotypic bling time in chronic lymphocytic leukemia and associated disorders. In
markers useful in the diagnosis and clas- leukaemia: Analysis of its prognostic Kantarjian HM, Wollf RA, and Koller
sification of hematopoietic neoplasms. In significance. Br JHaematol 62(3):567, CA (eds), MD Anderson Manual on
Knowles, DM (ed): Neoplastic 1986. Medical Oncology. McGraw-Hill,
Hematopathology. Lippincott Williams 48. Molica, S, et al: Prognostic value of New York, 2006, p 39.
& Wilkins, Philadelphia, 2001, p 93. enhanced bone marrow angiogenesis in 65. Binet, JL, et al: Perspectives on the use
Bo) Foroni, L, et al: Rearrangement of the early B-cell chronic lymphocytic of new diagnostic tools in the treatment
T-cell receptor delta genes in human leukemia. Blood 100(9):3344, 2002. of chronic lymphocytic leukemia. Blood
T-cell leukemias. Blood 73(2):559, 1989. 49. Aguayo, A, et al: Angiogenesis in acute 107(3):859, 2006.
34. Pan, L, et al: Antigen receptor genes: and chronic leukemias and myelodysplas- 66. Galton, DA: Terminal transformation in
Structure, function, and genetic analysis tic syndromes. Blood 96(6):2240, 2000. B-cell chronic lymphocytic leukaemia.
of their rearrangements. In Knowles, 50. Kini, AR, et al: Increased bone Bone Marrow Transplant 4 (Suppl 1):
DM (ed): Neoplastic Hematopathology. marrow angiogenesis in B cell chronic 156, 1989.
464 Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders
67. Tsimberidou, AM, and Keating, MJ: leukemia. European Study Group of 96. Schlette, E, et al: Mature B-cell
Richter syndrome: Biology, incidence, CAMPATH-1IH Treatment in Chronic leukemias with more than 55% prolym-
and therapeutic strategies. Cancer Lymphocytic Leukemia. J Clin Oncol phocytes. A heterogeneous group that
103(2):216, 2005. 15(4):1567, 1997. includes an unusual variant of mantle
68. Foon, KA, et al: Chronic lymphocytic . Bowen, AL, et al: Subcutaneous cell lymphoma. Am J Clin Pathol
leukemia: New insights into biology and CAMPATH-1H in fludarabine-resistant/ 115(4):571, 2001.
therapy. Ann Intern Med 113(7):525, relapsed chronic lymphocytic and Sie Mansoor, A, et al: Cytogenetic findings
1990. B-prolymphocytic leukaemia. Br J in lymphoplasmacytic lymphoma/
69. Foon, KA, et al: Genetic relatedness of Haematol 96(3):617, 1997. Waldenstrom macroglobulinemia. Chro-
lymphoid malignancies. Transformation 84. Pangalis, GA, and Griva, E: Recombi- mosomal abnormalities are associated
of chronic lymphocytic leukemia as a nant alfa-2b-interferon therapy in with the polymorphous subtype and an
model. Ann Intern Med 119(1):63, 1993. untreated, stages A and B chronic lym- aggressive clinical course. Am J Clin
70. Matolesy, A, et al: Molecular genetic phocytic leukemia. A preliminary report. Pathol 116(4):543, 2001.
demonstration of the diverse evolution of Cancer 61(5):869, 1988. 98. Lampert, I, et al: The histopathology of
Richter’s syndrome (chronic lympho- . Rozman, C, et al: Recombinant alpha prolymphocytic leukaemia with particu-
cytic leukemia and subsequent large cell 2-interferon in the treatment of B lar reference to the spleen: A compari-
lymphoma). Blood 83(5):1363, 1994. chronic lymphocytic leukemia in early son with chronic lymphocytic
Tile Imamura, J, et al: p53 in hematologic stages. Blood 71(5):1295, 1988. leukaemia. Histopathology 4(1):3,
malignancies. Blood 84(8):2412, 1994. 86. Kay, NE, et al: Evidence for tumor 1980.
. Gaidano, G, et al: p53 mutations in reduction in refractory or relapsed OO} Melo, JV, et al: The relationship
human lymphoid malignancies: associa- B-CLL patients with infusional between chronic lymphocytic
tion with Burkitt lymphoma and chronic interleukin-2. Nouv Rev Fr Hematol leukaemia and prolymphocytic
lymphocytic leukemia. Proc Natl Acad 30(5-6):475, 1988. leukaemia. III. Evaluation of cell size
Sci USA 88(12):5413, 1991. 87. Dighiero, G, et al: Chlorambucil in indo- by morphology and volume measure-
13. Wattel, E, et al: p53 mutations are asso- lent chronic lymphocytic leukemia. ments. Br J Haematol 64(3):469, 1986.
ciated with resistance to chemotherapy French Cooperative Group on Chronic 100. Jennings, CD, and Foon, KA: Recent
and short survival in hematologic malig- Lymphocytic Leukemia. N Engl J Med advances in flow cytometry: Applica-
nancies. Blood 84(9):3148, 1994. 338(21):1506, 1998. tion to the diagnosis of hematologic
74. Fenaux, P, et al: Mutations of the p53 88. O’Brien, SM, et al: Rituximab dose- malignancy. Blood 90(8):2863, 1997.
gene in B-cell chronic lymphocytic escalation trial in chronic lymphocytic 10 onal. Catovsky, D, and Costello, C: Cyto-
leukemia: a report on 39 cases with cyto- leukemia. J Clin Oncol 19(8):2165, 2001. chemistry of normal and leukaemic
genetic analysis. Leukemia 6(4):246, 89. O’Brien, SM, et al: Results of the flu- lymphocytes: A review. Basic Appl
1992. darabine and cyclophosphamide combi- Histochem 23(4):255, 1979.
. Meloni, G, et al: Chronic lymphocytic nation regimen in chronic lymphocytic 102. Swerdlow, SH, et al: Centrocytic
leukemia: From palliative therapy to leukemia. J Clin Oncol 19(5):1414, 2001. lymphoma: A distinct clinicopathologic
curative intent. Haematologica 90. Michallet, M, et al: HLA-identical sib- and immunologic entity. A multiparame-
83(7):660, 1998. ling bone marrow transplantation in ter study of 18 cases at diagnosis and
76. Hansen, MM, et al: CHOP versus pred- younger patients with chronic lympho- relapse. Am J Pathol 113(2):181, 1983.
nisolone + chlorambucil in chronic lym- cytic leukemia. European Group for 103. Medeiros, LJ, et al: Association of
phocytic leukemia (CLL): preliminary Blood and Marrow Transplantation and bel-1 rearrangements with lymphocytic
results of a randomized multicenter the International Bone Marrow Trans- lymphoma of intermediate differentia-
study. Nouv Rey Fr Hematol plant Registry. Ann Intern Med tion. Blood 76(10):2086, 1990.
30(5-6):433, 1988. 124(3):311, 1996. 104. Williams, ME, et al: Genotypic character-
ale O’Brien, S, et al: Results of fludarabine Oe Khouri, IF, et al: Allogeneic blood or ization of centrocytic lymphoma:
and prednisone therapy in 264 patients marrow transplantation for chronic lym- Frequent rearrangement of the chromo-
with chronic lymphocytic leukemia with phocytic leukaemia: Timing of transplan- some 11 bel-1 locus. Blood 76(7):1387,
multivariate analysis-derived prognostic tation and potential effect of fludarabine 1990.
model for response to treatment. Blood on acute graft-versus-host disease. BrJ 105. Coiffier, B, et al: Mantle cell lym-
82(6):1695, 1993. Haematol 97(2):466, 1997. phoma: A therapeutic dilemma. Ann
78. Richards, F, Jr, et al: The control of . Provan, D, et al: Eradication of poly- Oncol 6(3):208, 1995.
chronic lymphocytic leukemia with merase chain reaction-detectable chronic 106. Swerdlow, SH, et al: Expression of
mediastinal irradiation. Am J Med lymphocytic leukemia cells is associated cyclin D1 protein in centrocytic/mantle
64(6):947, 1978. with improved outcome after bone mar- cell lymphomas with and without
vo: Yam, LT, and Crosby, WH: Early splenec- row transplantation. Blood 88(6):2228, rearrangement of the BCL//cyclin D1
tomy in lymphoproliferative disorders. 1996. gene. Hum Pathol 26(9):999, 1995.
Arch Intern Med 133(2):270, 1974. . Khouri, IF, et al: Transplant-lite: induc- 107. Lambrechts, AC, et al: Translocation
80. Bunch, C: Immunoglobulin replacement tion of graft-versus-malignancy using (14;18)-positive cells are present in the
in chronic lymphocytic leukaemia. Nouv fludarabine-based nonablative circulation of the majority of patients
Rev Fr Hematol 30(5-6):419, 1988. chemotherapy and allogeneic blood with localized (stage I and I) follicular
81. Intravenous immunoglobulin for the pre- progenitor-cell transplantation as treat- non-Hodgkin’s lymphoma. Blood
vention of infection in chronic lympho- ment for lymphoid malignancies. J Clin 82(8):2510, 1993.
cytic leukemia. A randomized, controlled Oncol 16(8):2817, 1998. 108. Mintzer, DM, and Hauptman, SP: Lym-
clinical trial. Cooperative Group for the 94. Galton, DA, et al: Prolymphocytic phosarcoma cell leukemia and other
Study of Immunoglobulin in Chronic leukaemia. Br J Haematol 27(1):7, non-Hodgkin’s lymphomas in leukemic
Lymphocytic Leukemia. N Engl J Med 1974. phase. Am J Med 75(1):110, 1983.
319(14):902, 1988. . Katayama, I, et al: B-lineage prolympho- 109. Yam, LT, et al: Tartrate-resistant acid
. Osterborg, A, et al: Phase II multicenter cytic leukemia as a distinct clinicopatho- phosphatase isoenzyme in the reticulum
study of human CD52 antibody in previ- logic entity. Am J Pathol 99(2):399, cells of leukemic reticuloendotheliosis.
ously treated chronic lymphocytic 1980, N Engl J Med 284(7):357, 1971.
Chapter 20 Chronic Lymphocytic Leukemia and Related Lymphoproliferative Disorders 465
110. Visser, L, et al: Monoclonal antibodies ILS) Loughran, TP, Jr: Clonal diseases of 129. Orchard, JA, et al: ZAP-70 expression
reactive with hairy cell leukemia. Blood large granular lymphocytes. Blood and prognosis in chronic lymphocytic
74(1):320, 1989. SCD EI, WCE). leukaemia. Lancet 363(9403):105,
JUL. Catovsky, D, et al: Hairy cell leukemia 120. Scott, CS, et al: Disorders of large 2004.
(HCL) variant: An intermediate disease granular lymphocytes and natural killer- . Keating, M, et al: The serum B2-
between HCL and B prolymphocytic associated cells. Blood 83(1):301, microglobulin (B2m) level is more
leukemia. Semin Oncol 11(4):362, 1994. powerful than stage in predicting
1984. . Shastri, KA, and Logue, GL: Autoim- response and survival in chronic lym-
i Matutes, E, et al: The immunopheno- mune neutropenia. Blood 81(8):1984, phocytic leukemia (CLL) [abstract].
type of splenic lymphoma with villous 1993. Blood 86:606a, 1995.
lymphocytes and its relevance to the . Tefferi, A, et al: Chronic natural killer 13 fk. Hallek, M, et al: Serum beta(2)-
differential diagnosis with other B-cell cell lymphocytosis: A descriptive clini- microglobulin and serum thymidine
disorders. Blood 83(6):1558, 1994. cal study. Blood 84(8):2721, 1994. kinase are independent predictors of
3. Flandrin, G, and Brouet, JC: The . Loughran, TP, Jr, et al: Treatment of progression-free survival in chronic
Sézary cell: cytologic, cytochemical, large granular lymphocyte leukemia lymphocytic leukemia and immunocy-
and immunologic studies. Mayo Clin with oral low-dose methotrexate. Blood toma. Leuk Lymphoma 22(5-6):439,
Proc 49(8):575, 1974. 84(7):2164, 1994. 1996.
114. Bottaro, M, et al: Heteroduplex analysis . Dohner, H, et al: Genomic aberrations ee Reinisch, W, et al: Soluble CD23 reli-
of T-cell receptor gamma gene and survival in chronic lymphocytic ably reflects disease activity in B-cell
rearrangements for diagnosis and moni- leukemia. N Engl J Med 343(26):1910, chronic lymphocytic leukemia. J Clin
toring of cutaneous T-cell lymphomas. 2000. Oncol 12(10):2146, 1994.
Blood 83(11):3271, 1994. WS. Hamblin, TJ, et al: Immunoglobulin . Sarfati, M, et al: Prognostic importance
1S: Shimoyama, M: Diagnostic criteria and V genes and CD38 expression in CLL. of serum soluble CD23 level in chronic
classification of clinical subtypes of Blood 95(7):2455, 2000. lymphocytic leukemia. Blood
adult T-cell leukaemia-lymphoma. 126. Damle, RN, et al: Ig V gene mutation 88(11):4259, 1996.
A report from the Lymphoma Study status and CD38 expression as novel 134. Ibrahim, S, et al: CD38 expression as
Group (1984-87). Br JHaematol prognostic indicators in chronic lym- an important prognostic factor in B-cell
79(3):428, 1991. phocytic leukemia. Blood 94(6):1840, chronic lymphocytic leukemia. Blood
116. Sakashita, A, et al: Mutations of the 1999, 98(1):181, 2001.
p53 gene in adult T-cell leukemia. Wie Rassenti, LZ, et al: ZAP-70 compared
Blood 79(2):477, 1992. with immunoglobulin heavy-chain gene
ITA Brouet, JC, et al: Chronic lymphocytic mutation status as a predictor of disease
leukaemia of T-cell origin. Immunolog- progression in chronic lymphocytic
ical and clinical evaluation in eleven leukemia. N Engl J Med 351(9):893,
patients. Lancet 2(7941):890, 1975. 2004.
118. McKenna, RW, et al: Chronic lympho- 128. Crespo, M, et al: ZAP-70 expression as
proliferative disorder with unusual clin- a surrogate for immunoglobulin-
ical, morphologic, ultrastructural and variable-region mutations in chronic
membrane surface marker characteris- lymphocytic leukemia. N Engl J Med
tics. Am J Med 62(4):588, 1977. 348(18):1764, 2003.
Chapter 4 ;
The Lymphomas
Dan M. Hyder, MD
Introduction OBJECTIVES
Hodgkin Lymphoma At the end of this chapter, the learner should be able to:
Etiology and Pathogenesis
Pathology 1. List distinguishing features for nodular lymphocyte predominant, mixed cellularity,
Clinical Features lymphocyte-rich, lymphocyte depletion, and nodular sclerosis Hodgkin lymphoma.
Diagnostic Evaluation and 2. Describe the morphological and immunophenotypic features of Reed—Sternberg cells.
Stagin
i ah and Prognosis 3. List distinguishing features for the four stages of Hodgkin lymphoma.
Non-Hodgkin Lymphoma 4. Describe the classification criteria for non-Hodgkin lymphomas by the World Health
Etiology and Pathogenesis Organization classification.
se ee 5. Describe tests that may be needed to provide a differential diagnosis for lymphomas.
aging
Treatment and Prognosis 6. Describe the prognosis and treatment options for Hodgkin lymphoma and the more
Case Study common categories of non-Hodgkin lymphomas.
466
Chapter 21 TheLymphomas 467
Pathology
The cytological hallmark of HL is the presence of an unusual
giant cell, the Reed—Sternberg cell. The features of this cell
include large size (up to 45 um in diameter), abundant aci-
dophilic cytoplasm, multinucleated or polylobated nucleus, and
gigantic (more than 5 um in diameter), inclusion-like nucleoli
(see Fig. 21-1). There 1s often clearing of the chromatin around
the macronucleoli, resulting in a distinct halo effect. Formerly,
it was necessary to identify at least one of these “diagnostic”
Reed-—Sternberg cells before a primary diagnosis of HL could be
Figure 21-1 ™ Reed-Sternberg cell (arrows). Prototypic cell of made; however, with current immunophenotyping techniques,
Hodgkin lymphoma (HL)is characterized by large size, abundant this is no longer a requirement. The identification of Reed—
eosinophilic cytoplasm, multinucleation, and inclusion-like Sternberg (or Reed—Sternberg—like) cells, however, is not a
macronuclei. sufficient condition for the diagnosis of HL. Reed—Sternberg—
like cells are commonly seen in a variety of benign and
malignant conditions other than HL (Table 21-1). The diag-
Hodgkin lymphoma (HL), with particular attention to certain nosis of HL should be based on finding Reed—Sternberg
ribonucleic acid (RNA) tumor viruses and Epstein-Barr virus cells (morphologically or immunophenotypically) in the
(EBV). Conclusive experimental evidence, however, is lack- proper cellular, stromal, and clinical setting. Many nondiag-
ing. Certain epidemiologic data, such as the occasional occur- nostic variants of Reed—Sternberg cells have been described
rence of several cases of HL within a small locale and brief (Fig. 21-2A through D). Observation of these cells is help-
space of time (time-space clustering), suggest an infectious ful in suggesting the possibility of HL and in the subclassi-
etiology. Other epiderhiologic studies, however, have failed to fication of HL.
support an infectious mode of transmission. Several classification schemes for HL are in use; how-
Although an infectious mode of transmission is unlikely, ever, they all share the fact that they are primarily based on
an infectious cofactor would appear to be operational in the growth pattern and cellular composition. Clinicians com-
pathogenesis of HL. The probable infectious agent is EBV, monly utilize the Rye classification which is a simplification
although cytomegalovirus and herpesvirus 6 have also been of the Lukes and Butler classification.’”* Recent advances in
implicated. EBV nucleic acids can be demonstrated in the understanding the molecular defects in HL indicates that there
lesions of approximately 80% of cases of HL, and EBV anti- are two pathologically distinct forms of HL, namely NLUPHL
gens such as latent membrane protein can be detected in and CHL.’ The classical group may be further subdivided
approximately 50% of the cases. The mechanism by which a based on cellular composition. The recently proposed World
virus such as EBV promotes the development of HL is Health Organization (WHO) classification of HL (Table 21-2)
unclear; however, it is postulated that a preceding state of incorporates this new information.'°
immunosuppression permits an abnormal immune response
to an EBV infection. The resulting genetic errors in the NODULAR LYMPHOCYTE-PREDOMINANT HODGKIN
immunoregulatory cells (T and B lymphocytes) are hypothe- LYMPHOMA
sized to lead to the development of HL. NLPHL 1s a relatively uncommon (5% of cases) but patho-
The current view of HL is that it is truly a malignant logically and clinically distinct variety of HL. The growth
(clonal) proliferation. The malignant cells are the Reed—
Sternberg cell and its variants, which usually represent only a
small percentage of the total cells in a HL lesion. Despite the
prevailing view that the Reed—Sternberg cell is the malignant
cell of HL, the origin of this cell has been difficult to estab-
lish. Kinship to almost every cell of the mononuclear phago-
cytic and lymphoreticular system has been proposed for the
Reed-Sternberg cell. Based on recent investigations, including
single-cell molecular techniques, the vast majority of cases of ¢ Viral infection, e.g., infectious mononucleosis
¢ Anticonvulsant-induced lymphadenopathy
both NLPHL and CHL are clonal proliferations of B cells.
e Epithelial and stromal malignancies
The neoplastic cells of NLPHL appear to correspond to ger- e Melanoma
minal center B centroblasts with productive rearrangement e Various lymphomas and leukemias
(clonal) of the IgH gene.® A germinal center B-cell origin of ¢ Myeloproliferative disorders
the neoplastic cells of CHL is also suggested by recent data;
468 Chapter 21 The Lymphomas
Figure 21-2 ® A. Mononuclear Hodgkin cell (arrows). Variant RS cell with single monolobed nucleus and inclusion-like macronucleolus.
B. L&H (popcorn) cell (arrows). Characteristic cell of nodular lymphocytic-predominant Hodgkin lymphoma (NLPHL). C. Lacunar cell. Large
cell with artifactual clear space (lacuna) due to formalin fixation that surrounds the nucleus. Wisps of cytoplasm are visible in the space.
D. Pleomorphic Reed-Sternberg cell. Large cell with bizarre multinucleated/multilobated nucleus.
pattern is generally characterized by a vague nodularity numbers of a characteristic, though nondiagnostic, Reed—
although in some cases it may be well developed Sternberg variant referred to as an L&H or popcorn cell
(Fig. 21-3). It is questionable whether a purely diffuse form (see Fig. 21-2B). This cell has a variable amount of
of NLPHL exists. The cellular milieu is composed of a mix- pale staining cytoplasm, a convoluted nucleus prosaically
ture of small, normal-appearing lymphocytes, benign histi- referred to as popcorn-shaped, and an indistinct nucleolus.
ocytes, rare or absent Reed—Sternberg cells, and variable Phenotypic and genotypic studies suggest that L&H
cells are closely related to proliferating germinal center
cells (centroblasts). The immunophenotype of the neoplas-
tic L&H cells of NLPHL is distinct from the neoplastic
' Table 21-2 WHO Classification
cells of CHL (Table 21-3). Plasma cells, eosinophils, fibro-
of Hodgkin Lymphoma sis, and necrosis are usually absent. The histological
features of NLPHL must be distinguished from various
Frequency (%)
reactive conditions such as infectious mononucleosis
Nodular Lymphocyte-Predominant 5 and progressive transformation of germinal centers as
Classic Hodgkin Lymphoma well as certain subcategories of CHL and NHL. Such differ-
Nodular sclerosis, grades I and II 60-80 entiation may require special phenotypic and genotypic
Lymphocyte-rich 5
studies.
Mixed cellularity 15-30
Lymphocyte depletion =|
Source: From Harris, NL: Hodgkin’s disease: Classification and CLASSIC HODGKIN LYMPHOMA
differential diagnosis. Mod Pathol 12:159, 1999, with permission Classic HL is morphologically defined by the observation of
classic Reed—Sternberg cells in the appropriate cellular and
Chapter 21 TheLymphomas 469
stromal background. It is the background that determines the MIXED CELLULARITY HODGKIN LYMPHOMA As its
subcategory of classic HL to which the lesion belongs. name implies, mixed cellularity Hodgkin lymphoma (MCHL)
is characterized by a heterogeneous mixture of cells, including
NODULAR SCLEROSIS HODGKIN LYMPHOMA Nodular lymphocytes, histiocytes, plasma cells, eosinophils, Reed—
sclerosis (NS) Hodgkin lymphoma (NSHL) is the most com- Sternberg cells, and Reed—Sternberg variants (Fig. 21-6).
mon subtype of HL, ‘representing 60% to 80% of HL cases. Classic Reed—Sternberg cells and mononuclear variants are
The cardinal histological features of NSHL are the presence usually readily apparent. MCHL represents 15% to 30% of
of birefringent collagenous sclerosis, classic Reed—Sternberg cases of HL.
cells, and a distinctive Reed—Sternberg variant called a lacunar In addition to the cellular milieu, there is usually an
cell. The sclerosis observed in NSHL is different from that increase in the background stroma in the form of a disorderly,
seen in the other subtypes of HL. It is found in the form of noncollagenous fibrosis distinct from that seen in NSHL.
well-organized bands of collagen that subdivide the tissue Small areas of necrosis are commonly present. In some cases,
into distinct nodules (Fig. 21-4). Within the nodules is a vari- clusters of epithelioid histiocytes may be numerous, making
able mixture of lymphocytes, classic Reed—Sternberg cells, distinction from Lennert’s lymphoma (a T-cell lymphoma
lacunar cells, plasma cells, eosinophils, and neutrophils. The with high content of epithelioid histiocytes) difficult. The
lacunar cells often form distinct collections within the central lymphocytes of Lennert’s lymphoma are cytologically atypi-
region of the nodule (“grouped lacunars”), and these collec- cal as opposed to the small, uniform features of the lympho-
tions may be associated with focal necrosis (Fig. 21-5). The cytes in all forms of HL, including MCHL. There is often
lacunar cell is best identified in formalin-fixed tissue sections only partial involvement of nodes involved by MCHL. This is
Antibody TCRLBL
CD30 rs
++4 +++
+/— aahe
4+ t+
+ = all cases positive; +/— = majority of cases positive; —/+ = minority of cases positive; — = all cases negative.
NLPHL = nodular lymphocyte predominant Hodgkin lymphoma; CHL = classic Hodgkin lymphoma; TCRLBL = T-cell
rich large B-cell lymphoma; DLBCL = diffuse large B-cell lymphoma; ALCL = anaplastic large cell lymphoma.
470. Chapter 21 The Lymphomas
HISTOLOGIC PROGRESSION
The concurrent evaluation of multiple sites of involvement
with HL usually demonstrates similar histology at each site.
On the other hand, with the passage of time, reevaluation
often reveals histologic progression in the sequence: LRHL to
MCHL to LDHL. Although it is often stated that NSHL does
not undergo such progression but remains NSHL, there may be
progression over time within the histologic spectrum of NSHL.
That is, cases of NSHL initially rich in lymphocytes may
undergo a progressive decrease in the number of lymphocytes,
with concomitant increase in the number of Reed—Sternberg
cells and Reed—Sternberg variants equivalent to that seen in the
etc
pe 4 S ®
Sy 2. non-NS types. This progression, however, maintains the over-
ae. 28. all nodular sclerosing pattern of NSHL. Eventually a stage of
Figure 21-6 @ Mixed cellularity Hodgkin lymphoma. Easily identi- extreme cellular depletion may be reached, referred to as oblit-
fied Reed—Sternberg cells and RS variants are seen admixed with erative total sclerosis, in which few cellular elements, includ-
small lymphocytes, plasma cells, eosinophils, and histiocytes. ing Reed—Sternberg cells, are found.
Chapter 21 Thelymphomas 47]
Clinical Features staging would alter the therapy. The results of the staging
evaluation are used to assign the patient to staging groups.
In the United States, HL accounts for approximately 15% of The most widely utilized staging scheme is the Cotswold’s
newly diagnosed cases of lymphoma. It is estimated that revision of the Ann Arbor classification (Table 21—5).'* These
8,190 new cases of HL will be diagnosed in the United States staging schemes may be used with the data obtained from
in 2007 (SEER Cancer Statistics, NCI). The incidence of HL either clinical or pathologic staging, or both. In addition to the
exhibits a bimodal distribution, with peaks between the ages anatomical extent of the disease implicit in the staging cate-
of 15 and 35 and in the over-50 age group. gories, the disease is further classified based on the presence
Most patients with HL present with a complaint of non- or absence of specific symptoms (Table 21-6). The letter “A”
painful lymph node swelling. Each subtype of HL is associ- (asymptomatic) or “B” (symptomatic) is then appended to the
ated with rather characteristic, but not totally specific, clinical appropriate stage number.
features. NLPHL demonstrates a male predominance and is The clinical utility of the Cotswold’s staging scheme
generally a disease of younger patients. The disease is usually stems from the predictable behavior of HL. HL is a lymph
localized at presentation to one peripheral node or node group. node-based disease and rarely, if ever, starts in extranodal
Despite excellent long term survival, patients with NLPHL sites. It spreads by the lymphatic route in an orderly and pre-
experience an increased frequency of late relapses compared dictable pattern to contiguous lymph nodes. Only late in the
with CHL and progression to a large B cell lymphoma occurs disease course when hematogenous spread may occur is a
in 3% to 5% of cases. NSHL shows a female predominance and more disorderly pattern seen.
is usually associated with cervical, scalene, or supraclavicular
adenopathy. An anterior mediastinal mass is often present.
Most patients with NSHL are asymptomatic on presentation. Treatment and Prognosis
MCHL and LDHL are most often seen in symptomatic patients
The selection of therapy is directed by the results of the staging
with widely disseminated disease. Symptoms include fever,
evaluation rather than the specific histologic subtype. In general,
drenching night sweats, and weight loss. Extranodal involve-
multiagent chemotherapy with or without involved field radia-
ment is common in these two subcategories of HL.
tion is the mode of treatment. With appropriate therapy, the
10-year survival for stages I and IT HL now exceeds 80%.
Diagnostic Evaluation and Staging Ten-year survival for stages III and IV HL has been improved
with aggressive chemotherapy and now approaches 70%.'?
The diagnosis of HL requires tissue biopsy and microscopic
evaluation. Because of the complexities involved in accurate
diagnosis and subclassification, adequate tissue should be Non-Hodgkin Lymphoma
obtained at the time of initial biopsy for routine light micro-
scopic studies as well as for ancillary studies, such as Virchow (1858)'* and Billroth (1871)!° were the first to
immunophenotypic and genotypic analysis, if these are found employ the terms lymphoma and malignant lymphoma. The
to be necessary. distinction between the two major categories of malignant
Following a tissue diagnosis of HL, the patient should be lymphoma (1.e., HL and NHL) was suggested in 1893 by
appropriately staged to determine the extent of disease and Dreschfield'® and Kundrat.'’ It is estimated that 56,390 cases
permit selection of appropriate therapy. Clinical staging of NHL were diagnosed in 2005.
should be performed on all newly diagnosed patients with
HL. The elements of clinical staging are listed in Table 21-4. Etiology and Pathogenesis
Pathologic staging requires exploratory laparotomy with
multiple node samplings and splenectomy. Except at certain It seems certain that a prerequisite for the development of
institutions, routine pathologic staging is not performed and is lymphoma is damage to the regions of the genetic code that
reserved for those cases in which the results of pathologic regulate the growth and reproduction of cells of the immune
Definition
Involvement of a single lymph node region or lymphoid structure (e.g., spleen, thymus, Waldeyer’s ring) :
Involvement of two or more lymph node regions on the same side of the diaphragm (the mediastinum is a single site:
hilar lymph nodes are lateralized); the number of anatomical sites should be indicated by suffix (e.g.. II)
Involvement of lymph node regions or structures on both sides of the diaphragm
With or without splenic, hilar, celiac, or portal nodes
With paraaortic, iliac, or mesenteric nodes
Involvement of extranodal site(s) beyond those designated E
E = Involvement of a single extranodal site, or contiguous or proximal to known nodal site of disease. 7.
Source: Jaffe, ES, et al: World Health Organization Classification of Tumours Pathology & Genetics, Tumours of Haematopoietic
and Lymphoid Tissues. IARC Press, Lyon, France, 2001.
Trisomy 12 ? B-CLL/SLL
Del 13q14 ? B-CLL/SLL
t(9;14) PAX5-IgH Lymphoplasmacytoid cell lymphoma
t(11;14) BCLI-IgH (Prad 1) Mantle cell lymphoma
t(14;18) IgH-BCL2 Follicle center lymphoma
t(11;18) API2-MALT1 Marginal zone lymphoma (extranodal)
Trisomy 3 2 Marginal zone lymphoma (extranodal)
t(8;14) c-myc—IgH Burkitt lymphoma
t(2;8) k/c-myc Burkitt lymphoma
(8322) c-myc! Burkitt lymphoma
t(3;14) BCL6-IgH Diffuse large B-cell lymphoma
Inv 14 (ql1;q32) TCR-TCL1 T-Cell Pele
t(2:5) ALK-NPM Systemic anaplastic large cell
Tso(7q) 2 Hepatosplenic T-cell lymphoma
ont er nesses
ence DASA mam EN
B-CLL = B-cell chronic lymphocytic‘leukemia; SLL = small cell lymphocytic lymphoma; IgH = immunoglobulin heavy chain locus;
BCLI = B cell lymphoma | gene; BCL2 = B cell lymphoma 2 gene; ALK = kinase gene; NPM = nucleolar protein gene; PLL = prolymphocytic
leukemia; AP/2 = apoptosis inhibitor gene; MALT] = mucosa-associated lymphoid tissue gene; c-myc = c-myelocytomatosis oncogene;
BCL6 = B cell lymphoma 6 gene; TCR = T-cell receptor gene; TCL/ = T-cell leukemia | gene; PAXS = paired homeobox-5 gene.
c-myc
8 14 t(8;14)(q24;q32)
In 1994 the International Lymphoma Study Group pub- A conceptual understanding of the common classifica-
lished the Revised European-American Classification of tion schemes requires knowledge of the morphological
Lymphoid Neoplasms (REAL), which utilized clinical, ontogeny of normal lymphocytes. Although it is customary to
morphological, immunophenotypic, and genotypic features to discuss the lymphomas with reference to one of the classifica-
subclassify the lymphomas.” The utility of this more “biolog- tion schemes, taking a more conceptual approach will facili-
ically correct” classification scheme has been subsequently tate understanding the relationship between the various
verified.*! The principles of REAL classification were incor- lymphomas as well as the various classification schemes.
porated and updated in the World Health Organization classi- The lymphocytic lymphomas may be conceptualized as a ma-
fication of tumors of lymphoid tissues (Table 21-9) and is lignant population of lymphocytes arrested at a particular
currently the most widely utilized classification scheme.'° stage (morphological, genotypic, phenotypic, or functional)
B-Cell Neoplasms
Precursor B-cell neoplasm
¢ Precursor B lymphoblastic leukemia/lymphoma
Mature B-cell neoplasms
¢ Chronic lymphocytic leukemia/small lymphocytic lymphoma
¢ B-cell prolymphocytic leukemia
¢ Lymphoplasmacytic lymphoma
¢ Splenic marginal zone lymphoma
¢ Hairy cell leukemia
e Plasma cell myeloma
* Solitary plasmacytoma of bone
¢ Extraosseous plasmacytoma
* Extranodal marginal zone B-cell lymphoma of mucosa-associated lymphoid tissue (MALT-lymphoma)
¢ Nodal marginal zone B-cell lymphoma
° Follicular lymphoma
¢ Mantle cell lymphoma
° Diffuse large B-cell lymphoma
¢ Mediastinal (thymic) large B-cell lymphoma
¢ Intravascular large B-cell lymphoma
¢ Primary effusion lymphoma
¢ Burkitt lymphoma/leukemia
B-cell proliferations of uncertain malignant potential
¢ Lymphomatoid granulomatosis
¢ Post-transplant lymphoproliferative disorder, polymorphic
T-Cell and NK-Cell Neoplasms
Precursor T-cell neoplasms
¢ Precursor T lymphoblastic leukemia/lymphoma
¢ Blastic NK cell lymphoma*
Mature T-cell and NK-cell neoplasms
*¢ T-cell prolymphocytic leukemia
° T-cell large granular lymphocytic leukemia
° Aggressive NK cell leukemia
¢ Adult T-cell leukemia/lymphoma
* Extranodal NK/T-cell lymphoma, nasal type
¢ Enteropathy-type T-cell lymphoma
° Hepatosplenic T-cell lymphoma
* Subcutaneous panniculitis-like T-cell lymphoma
¢ Mycosis fungoides
¢ Sézary syndrome
* Primary cutaneous anaplastic large cell lymphoma
* Peripheral T-cell lymphoma, unspecified
¢ Angioimmunoblastic T-cell lymphoma
¢ Anaplastic large cell lymphoma
T-cell proliferation of uncertain malignant potential
¢ Lymphomatoid papulosis
Chapter 21 TheLymphomas 475
Hodgkin Lymphoma
* Nodular lymphocyte predominant Hodgkin lymphoma
* Classical Hodgkin lymphoma
* Nodular sclerosis classical Hodgkin lymphoma
* Lymphocyte-rich classical Hodgkin lymphoma
¢ Mixed cellularity classical Hodgkin lymphoma
* Lymphocyte-depleted classical Hodgkin lymphoma
Histiocytic and Dendritic-Cell Neoplasms
Macrophage/histiocytic neoplasm
* Histiocytic sarcoma
Dendritic cell neoplasms
* Langerhans cell histiocytosis
¢ Langerhans cell sarcoma
* Interdigitating dendritic cell sarcoma/tumour
* Follicular dendritic cell sarcoma/tumour
¢ Dendritic cell sarcoma, not Otherwise ee
of lymphocyte maturation. Thus, we would expect to see transformed into secondary follicles containing a germinal
lymphomas that express the attributes of normal lymphoid center surrounded by a crescent of B cells referred to as the
cells found in each of the various normal lymphoid compart- mantle zone. The germinal center is populated with centro-
ments; that is, the precursor (bone marrow), intrafollicular, cytes (resting B cells), centroblasts (proliferating B cells),
mantle zone, marginal zone, and interfollicular compart- dendritic cells, and histiocytes/macrophages. The paracor-
ments. As illustrated in Figures 21-9 and 21-10, each of the tex (inner or deep cortex) is occupied mostly by T cells. The
lymph node compartments is occupied by morphologically paracortex also contains specialized vessels called high
characteristic cells. These cells are also immunophenotypi- endothelial venules which are the point of entry of circulat-
cally and genotypically distinct. ing lymphocytes into the lymph node parenchyma. The
The three major anatomical regions of a lymph node medulla contains a mixture ofT cells, B cells, macrophages,
are the cortex, paracortex and medulla. The cortex, also and plasma cells.
referred to as the outer cortex, is populated primarily by The NHLs may arise from any of these normal cellular
B lymphocytes along with macrophages, plasma cells, and counterparts. B-cell lymphomas are by far the most common
reticular cells. The B cells are mostly organized into dis- (85% to 90%). T-cell/natural killer (T/NK) cell lymphomas
tinct nodules referred to as primary and secondary follicles. make up the majority of the remainder (10% to 15%) whereas
Primary follicles are composed predominantly of small true histiocytic and dendritic cell “lymphomas” are distinctly
B cells. On antigenic stimulation the primary follicles are uncommon (<1%).
Germinal center
(follicular center)
Mantle zone
N iM uh
Paracortex a
Efferent =) Cortex
ONTOGENY
B Lymphocytes T Lymphocytes
@ Early B-cell
Mature virgin B-cell
rtex
PERIPHERAL BLOOD Medulla
T lymphoblast
Mature virgin T cells : g, HYMUS
Primary follicle
Marginal es
zone cell Mature virgin PERIPHERAL
Monocytoid B cell Mature virgin T cells BLOOD
Plasmacytoid
A lymphocyte B
Figure 21-10 @ Maturation (ontogeny) of (A) B lymphocytes and (B) T lymphocytes in relation to the pertinent anatomical compartments.
B-CELL LYMPHOMAS (Fig. 21-11A and B). Whereas the mantle zone is well devel-
B-cell lymphomas may be subdivided into precursor and mature oped in the benign secondary follicle, it is absent or attenu-
subtypes. Precursor B-cell lymphoma is distinctly uncommon ated in malignant follicles. The normal polarization of
(<1% of NHLs) whereas mature B-cell lymphomas represent the germinal center into light and dark zones is lost in malig-
approximately 90% of all lymphoid neoplasms diagnosed nant follicles. Tingible body macrophages, which are usually
worldwide. frequent in benign secondary follicles, are usually absent in
malignant follicles.
MATURE B-CELL LYMPHOMAS Mature B-cell lymphomas Follicular lymphomas are clonal proliferations of follicular
are malignant (clonal) proliferations of B cells that correspond center cells (centrocytes and centroblasts) (Table 21-10). Cen-
to stages of B-cell ontogeny from naive B cells to mature trocytes (cleaved cells) are small to medium size cells
plasma cells. Plasma cell myeloma, because of its unique clin- (6 tol5 um diameter) with scant cytoplasm, slightly to distinctly
ical presentation and treatment, is discussed in Chap. 22. irregular nuclei (angulated, clefted), and indistinct nucleoli.
Mature B-cell lymphomas are most common in developed Centroblasts (large noncleaved cells) are 20 to 40 um in diame-
countries (United States, Europe, Australia, and New Zealand) ter and have a modest amount of pyranophilic (RNA-rich)
and predominate in older adults (median age 60 years), except cytoplasm, round to oval vesicular nuclei, and small but distinct
for Burkitt lymphoma and mediastinal large B-cell lymphoma. nucleoli. In the WHO classification the number of centroblasts
Within the broad category of mature B-cell lymphoma, two per high-power microscopic field (hpf = 0.159 mm‘”) is used to
subtypes (follicular lymphoma and diffuse large B-cell lym- grade the follicular lymphomas: grade | = 0 to 5 centroblasts/
phoma) account for the majority of cases. hpf; grade 2 = 6 to 15 centroblasts/hpf; and grade 3 =
>15 centroblasts/hpf (Fig. 21-12A and B). Grading is
Follicular B-cell Lymphoma correlated with clinical outcome although the strength of the
The follicular lymphomas by definition exhibit at least a par- correlation has been subject to great debate.” Most cases show
tial follicular growth pattern and are composed of a mixture a predominance of centrocytes (grade | and 2), which are rest-
of the two principal cell types found in the germinal center, ing B cells. This may explain the indolent behavior of most
namely centrocytes (cleaved cells) and centroblasts (large follicular lymphomas.
noncleaved cells). The follicular growth pattern may be vague Although all follicular lymphomas demonstrate at least a
as observed by routine light microscopy. partial follicular growth pattern, diffuse growth areas are
The follicles of a follicular lymphoma bear a resem- often present and may predominate. The percentage of follic-
blance to the benign secondary follicles of a reactive lymph ular growth should be included in the pathologic assessment.
node; however, subtle differences aid the pathologist in rec- Lymphomas derived from follicular center cells are, by
ognizing the malignant follicle of a follicular lymphoma definition, B-cell lymphomas. The expected immunophenotype
Chapter 21 The Lymphomas 477
CLL = chronic lymphocytic leukemia; SLL = small cell lymphocytic lymphoma; CB/CC = centroblastic/centrocytic.
478 — Chapter 21 The Lymphomas
—/+ = less than 50% of cases positive; +/— = more than 50% of cases positive; CLL = chronic lymphocytic leukemia;
SLL = small lymphocytic lymphoma.
immunoglobulin heavy-chain gene on chromosome 14 (see irregular nuclei, and inconspicuous nucleoli (Fig. 21—-13A).
Table 21-8). This translocation is detectable in 70% to 95% of Most often, MCL has a diffuse or vaguely nodular pattern;
cases of follicular lymphoma. however, occasionally a distinct mantle zone pattern is
Follicular center lymphomas accounts for approximately observed. In this form the malignant cells are distributed as an
40% of adult NHLs in the United States. The disease most expanded mantle zone surrounding benign (polyclonal) ger-
commonly affects adults and is usually widespread at diagno- minal centers. This pattern of MCL must be distinguished
sis (Table 21-12). Although the disease course is usually from mantle zone hyperplasia.
indolent, it is not often curable with current therapy (median The immunophenotype of MCL aids in differentiating
survival is 6 to 8 years). Most studies indicate that grade 3 fol- this subcategory of lymphoma from other morphologically
licular lymphomas are more aggressive than grades | and 2. similar lymphomas. Characteristically, the malignant cells
Consequently grade 3 follicular lymphoma ts typically treated express monoclonal slg, B-cell-associated antigens, CD5, and
more aggressively. Progression with time from follicular lym- CD43, but are negative for CD23, CDI 1c, and usually CD10
phoma to a more aggressive, diffuse large-cell pattern occurs (see Table 21—11).'' This immunophenotype corresponds to
in approximately 40% of cases (Table 21—13); however, spon- the normal CD5-positive, CD23-negative B cell of the inner
taneous remissions of follicular lymphoma have also been follicle mantle.* Most cases of MCL demonstrate a unique
described. chromosomal translocation, t(11;14), involving the BCL/
proto-oncogene on chromosome 11 and the immunoglobulin
Mantle Cell Lymphoma heavy-chain gene on chromosome 14 (see Table 21-8). This
Lymphomas composed of lymphocytes with the morphologi- translocation results in the overexpression of the cell cycle
cal and immunophenotypic features of mantle cells have only protein cyclin D1, which may be detected in the nuclei of
recently been accepted as a distinctive subcategory of NHL.” MCL by immunocytochemical techniques (see Fig. 21—-13B).
The cytological features of mantle cell lymphoma (MCL) are MCL is a disease of older adults with high male-
somewhat variable, creating difficulty in differentiating on to-female ratio (see Table 21-12). The disease is usually wide-
histologic grounds mantle cell lymphoma from other low- spread at diagnosis, with lymph nodes and spleen being the
grade NHLs. The cells of MCL are most commonly small- most common sites of involvement. MCL has a moderately
to medium-sized lymphocytes with scant pale cytoplasm, aggressive course (3- to 5-year median survival), but behaves
similarly to other low-grade lymphomas in that it is generally been described and are associated with a more aggressive
not curable with current therapies. A peculiar extranodal clinical course.”°
presentation of the disease in the intestine is referred to as
lymphomatous polyposis (Fig. 21-14). Several histologic Marginal Zone Lymphoma
variants of MCL (pleomorphic, large cell, and blastoid) have The marginal zone of lymph nodes is a poorly defined anatomi-
cal compartment.*’ However, in other lymphoid tissues (i.e.,
spleen and Peyer’s patches), the marginal zone is_ better
defined.** The typical marginal zone cell is a centrocyte-like
cell (small cleaved cell) with more abundant pale-staining
cytoplasm than a true centrocyte. Under antigenic stimulation,
these marginal zone B cells are believed, by some investigators,
capable of differentiation into monocytoid (parafollicular)
B ceils and plasma cells (see Fig. 21-10). Monocytoid B cells
are medium-sized lymphocytes morphologically resembling
monocytes that are found predominantly in the lymphoid
sinuses and paracortex of some reactive lymph nodes but not in
normal lymph nodes. They are particularly common in the
reactive lymphadenopathies associated with human immun-
odeficiency virus (HIV) infection and toxoplasmosis.
re: * ee! : ;
Small Lymphocytic Lymphoma/Chronic Lymphocytic
pete. base
Leukemia
Small lymphocytic lymphoma (SLL) is the tissue equivalent
of chronic lymphocytic leukemia (CLL). SLL and CLL are
considered to be different clinical expressions of a single
disease process and therefore are combined in the WHO clas-
sification. The growth pattern of SLL is diffuse, although a
pseudofollicular pattern may be observed (Fig. 21—18A).
These pseudofollicles, also called growth centers, do not have
cS
"ries the cytological or histological features of true neoplastic fol-
AS licles. In the early phase of nodal involvement, the neoplastic
—
Se:
Pee cells may be confined to the interfollicular areas with sparing
of the normal follicles. Later, there is total effacement of
lymph node architecture. The majority of the neoplastic cells
8 are small, uniform lymphocytes with scant cytoplasm, round
Se
cmeeceses:obecl
ray
Ps
ame .t nucleus, clumped chromatin, and absent to small nucleoli; that
‘s PEAT Be
Pi
is, they appear cytologically similar to normal small lympho-
"exes
Dae
Cae
== e- -& 7 “J A
Spee
Sites3
este
e
Fy he
SLL may show evidence of plasmacytoid differentiation,
en, 3 including the presence of cytoplasmic immunoglobulin and a
small amount of circulating monoclonal immunoglobulin in
the peripheral blood. These cases are phenotypically and
clinically similar to small lymphocytic lymphoma and as such
should be distinguished from true lymphoplasmacytic lym-
See eae
phoma (see later discussion). Diagnostic difficulty may also
viae paestee ct occur when there is overgrowth of the lymphoma by the
23) atest Sa 3 Botes
br atiteen WES
larger cells of the growth centers (paraimmunoblastic and
prolymphocytoid transformation).
The normal counterpart cell to the malignant cell of SLL
is believed to be a recirculating naive B lymphocyte. In addi-
tion to the expression of B-cell-associated antigens (CD19,
CD20, and CD22), with sensitive techniques, low-density
monoclonal sIg is detectable. SLL is aiso positive for CD5,
CD23, and CD43 but lacks CD10 (see Table 21-11). A small
number of cases are weakly positive for CDI Ic.
Approximately 80% of cases of SLL will exhibit clonal
cytogenetic abnormalities including trisomy 12 (20%),
del 13q14 (50%), and del 11q22-23 (20%). The presence of
these cytogenetic abnormalities is associated with prognosis.
The 13q14 abnormality is associated with improved survival
whereas the other abnormalities predict for worse outcome.
Recently hypermutation of the immunoglobulin gene variable
region has been shown to correlate with improved survival.*’
Expression of the ZAP-70 protein has been shown to provide
the same prognostic information as the more complicated
mutation analysis.
Small lymphocytic lymphoma/chronic lymphocytic
leukemia is a disease of older adults and is usually wide-
spread at diagnosis. The disease, although indolent in
behavior, is considered incurable with standard therapy.
Improvement in disease-free survival has been demon-
strated with purine nucleoside analogues such as fludara-
bine. Transformation to a high grade lymphoma (3%) or
HL (0.5%) can occur.**
Lymphoplasmacytic Lymphoma
A phenotypically and clinically distinct form of lymphoma of
small lymphoid cells exists in which the cells exhibit plasma-
cytoid features and a phenotype distinct from typical small
lymphocytic lymphoma.*” Specifically, they express mono-
clonal surface and cytoplasmic immunoglobulin, usually of
the IgM class; are positive for B-cell-associated antigens
(CD19, CD20, CD22, and CD79a); but lack CD5 and CD10
(see Table 21-11). Fewer than 50% of cases are positive for
CD43. The normal counterpart of this lymphoma is believed
to be a recirculating CDS-negative cell, which has been stim-
ulated to differentiate into plasma cells. Approximately 50%
of cases of lymphoplasmacytic lymphoma will demonstrate
t(9;14), involving the gH and PAXS genes (see Table 21-8).
Clinically, these lymphomas usually correspond to the syn-
Figure 21-18 8A. Small lymphocytic lymphoma. Diffuse efface- drome of Waldenstrém’s macroglobulinemia. Lymphoplas-
ment of lymph node by proliferation of small lymphocytes. Pale macytic lymphoma is a disease of older adults and most
areas are growth centers (arrows). B. Cytological detail of small
commonly involves lymph nodes, spleen, and bone marrow.
lymphocytes of SLL. Cells have scant cytoplasm with uniform nuclei,
coarsely clumped chromatin and indistinct nucleoli. C. Growth A monoclonal gammopathy of the IgM type is often present
center of SLL is composed of prolymphocytes and paraimmunoblasts, and may be associated with hyperviscosity symptoms. The
cells that are larger than the typical small lymphocyte of SLL challenge to the pathologist is to differentiate this lymphoma
Chapter 21 TheLymphomas 483
from other low-grade lymphomas that can show plasma cell A subset of DLBCLs express CD5 (10%) or CD10 (25% to
differentiation. Similar to other low-grade lymphomas, this 50%). CD5-positive DLBCL may arise de novo or from an
disease is indolent but incurable, and large-cell lymphoma underlying low-grade CD5 positive B-cell lymphoma (SLL or
transformation may occur (see Table 21-13). MCL). The absence of BCL-1 expression rules out a blastoid
variant of MCL. The expression of CD10 and BCL-6 by a
Diffuse Large-Cell Lymphoma DLBCL suggests the possibility of transformation from an
Diffuse large B-cell lymphoma (DLBCL) represents the most underlying follicular lymphoma. The t(14;18) translocation
frequently diagnosed category of NHL worldwide (30%). By characteristic of follicular lymphoma can be found in 20% to
definition, DLBCL exhibit a diffuse growth pattern and are 30% of cases of DLBCL.
composed of clonal B cells of large size (nuclear diameter equal DLBCL occurs in a broad age range including children
to or greater than normal macrophage nucleus). Large-cell lym- although the median age is in the seventies. The disease may
phomas in general exhibit diverse morphologic features; how- present as nodal or extranodal disease. The gastrointestinal
ever, attempts to subclassify these large cell lymphomas based tract is the most common extranodal site. A distinct clinical
on morphology proved to be nonreproducible.*” Consequently subtype of DLBCL is mediastinal (thymic) large B-cell lym-
all diffuse large cell lymphomas composed of B cells were phoma. This subtype usually presents as a large anterior
grouped by the WHO into a single category of DLBCL. mediastinal mass in a young adult female. DLBCL is an
On morphological grounds DLBCL consist of a mixture aggressive lymphoma if untreated; however, cure is possible
of cells with centroblastic, immunoblastic, and anaplastic/ in many patients with multiagent chemotherapy.
pleomorphic features (Fig. 21-19). One morphologic type,
most commonly centroblastic, may predominate. In rare cases Burkitt Lymphoma
of DLBCL referred to as T-cell/histiocyte rich DLBCL Burkitt lymphoma (BL), first described in 1958 by Dennis
(TCRLBL) there is a predominance of non-neoplastic T cells Burkitt,” is endemic to Africa but also accounts for approxi-
and/or histiocytes with fewer than 10% large neoplastic B mately one-third of non-African pediatric lymphomas (sporadic
cells. These cases may be easily confused with NLPHL. BL) and a high percentage of lymphomas in immunocompro-
Most cases of DLBCL arise de novo; however, transfor- mised patients, particularly patients with acquired immunodefi-
mation from a lower grade B-cell lymphoma, particularly ciency syndrome (AIDS).
SLL, MCL, MZL, follicular lymphoma, or NLPHL may also The monotonously uniform cells of Burkitt’s lymphoma
occur. Utilizing gene expression analysis two types of are medium-sized cells with uniformly round nuclei, multiple
DLBCL lymphoma appear to exist, a germinal center B-like small nucleoli, and a modest amount of intensely basophilic
(centroblastic) DLBCL and an activated B-like (immunoblas- cytoplasm (Fig. 21—20A). Lipid vacuoles are readily evident
tic) DLBCL.*! Patients with germinal center B-like gene in the cytoplasm on smears or imprints of the tissue. The
expression have a better overall survival than those with acti- proliferation rate of Burkitt’s lymphoma is very high
vated B-like expression. (see Fig. 21—20B), and the estimated volume doubling time of
By definition DLBCL is a clonal proliferation of B cells. this lymphoma is the highest of any tumor (approximately
The expected immunophenotype include expression of pan- 1 day). Programmed tumor cell death (apoptosis) is also very
B-cell antigens, e.g., CD19, CD20, CD22, CD79a; however, high, and a starry sky pattern of tingible-body macrophages is
one or more of these pan-B-cell antigens may be absent. usually evident owing to phagocytosis of the apoptotic debris.
Variant histologies are recognized and include BL with plas-
macytoid differentiation and atypical BL characterized by
cells more pleomorphic than those of classical BL.
As previously discussed, Burkitt’s lymphoma is cytoge-
netically characterized by a t(8;14) in the majority of cases or
less often by t(2;8) or t(8;22). In the endemic (African) form
of this disease, the breakpoint on chromosome 14 involves
the heavy-chain joining region, suggesting an early B-cell
origin of the Burkitt cell. However, in the nonendemic form,
the breakpoint is in the heavy-chain switch region, consistent
with origin at a latter stage (possibly follicular center cell) of
B-cell development. The endemic and immunodeficiency-
related cases are also associated with a high frequency of
tumor cell incorporated EBV genomes. The immunopheno-
type of Burkitt’s lymphoma is surface IgM-positive, B-cell
antigen-positive, CD10+, CDS5—, CD23-, Bcl-2—, and TdT-.
The male-to-female ratio for Burkitt’s lymphoma is
approximately 2.5 to 1. The facial bones, particularly the jaw,
are the most common sites of involvement for the African
Figure 21-19 ™ Diffuse large B-cell lymphoma. This common
(endemic) variety (Fig. 21-21). In the nonendemic variety, an
lymphoma is composed of a mixture of large B cells that exhibit
centroblastic, immunoblastic, and pleomorphic morphology. abdominal mass is the most common presenting pattern. The
484 Chapter 21 The Lymphomas
distal ileum, cecum, and mesentery are the most frequent sites
of involvement. Other less common sites of involvement “ ie * >
include the kidneys, ovaries, and breasts. Burkitt’s lymphoma AY. oy ae gs
a
is a highly aggressive and rapidly fatal disease if untreated;
however, this disease is potentially curable and prognosis
correlates with tumor bulk (see staging).
¥°
Reh
y©
Intravascular Large B-Cell Lymphoma %
ye EE EMAL Vlas 8
Ok Oo Dee Pee
Figure 21-26 @ A. Systemic anaplastic large cell lymphoma. Figure 21-27 @ A. Angioimmunoblastic T-cell lymphoma. Mixture
Pleomorphic cells that vary in size and shape. “Hallmark” cells are of lymphocytes, some of which are atypical, dendritic cells, and
readily apparent (arrows). B. Detailed view of several “hallmark” thick-walled high endothelial venules. B. Clusters of medium to
cells (arrows) showing kidney or horseshoe-shaped nuclei and large cells with clear cytoplasm (arrows) are characteristic of
juxtanuclear eosinophilic inclusion-like zone. angioimmunoblastic T-cell lymphoma.
disorders are associated with evidence of hyperimmunity (fever, involve the skin; however, this is seldom an isolated or present-
skin rash, hypergammaglobulinemia) and often develop after ing finding in B-cell lymphoma. Hodgkin’s lymphoma rarely
exposure to certain drugs and infectious agents. There is a high involves the skin in the form of direct extension from adjacent
frequency of evolution of these clinicopathological entities that involved lymph nodes. The category of cutaneous T-cell lym-
run the gamut from benign (possibly premalignant) disorders to phoma includes a broad group of dysplastic and frankly malig-
aggressive, rapidly fatal lymphomas. The affected lymph nodes nant T-cell proliferations with a predilection for infiltration of
of angioimmunoblastic T-cell lymphoma are partially effaced the skin.* Disorders within the umbrella of this category
by a mixture of atypical lymphoid cells, follicular dendritic include mycosis fungoides, Sézary syndrome, lymphomatoid
cells, and thick-walled high endothelial venules (Fig. 21—27A). papulosis, and primary cutaneous anaplastic large-cell lym-
The lymphoid cells are predominantly CD4 positive with phoma. Mycosis fungoides and Sézary syndrome, two related
admixed CD8 positive T cells.” Clusters of CD21+ follicular disorders, are characterized by infiltration of the dermis and
dendritic cells are aberrantly located and most often found sur- epidermis by malignant T cells with a peculiar cerebriform
rounding the high endothelial venules (see Fig. 21-27B). Clonal nucleus. Thin sections of well-fixed paraffin-embedded material
T-cell receptor rearrangement is detectable in approximately or plastic-embedded sections are required to appreciate this
75% of cases. Frank angioimmunoblastic T-cell lymphoma usu- nuclear detail. In 90% of cases, these cells exhibit a mature
ally follows an aggressive clinical course. “helper” phenotype (pan-T+, CD4+, CD8-) and in 10%, a
mature suppressor phenotype (pan-T+, CD4-, CD8+). In
Primary Cutaneous T-Cell Lymphoma many cases, mycosis fungoides progresses through three clini-
Primary involvement of the skin by lymphoma is usually of the cal phases. In the premycotic (erythroderma) phase, lasting
T-cell type. B-cell lymphomas may occasionally secondarily from 6 months to 50 years, the T-cell infiltrate produces a
488 Chapter 21. The Lymphomas
ce
Figure 21-28 @ A. Myosis fungoides. Plaque lesion of MF showing rs se, Me. eeu. : a
band-like infiltrate (arrows) of dermis by atypical lymphocytes that
extent into the overlying epidermis. B. Pautrier microabscess. Figure 21-30 @ Sézary cells in the peripheral blood from a patient
Cluster of atypical lymphocytes within the epidermis. with Sé€zary syndrome.
Chapter 21 TheLymphomas 489
Lymphoma Characteristics
Extranodal T/NK lymphoma, nasal type*? Angiocentric lymphoma that predominantly involves the nasal cavity,
nasopharynx, palate, skin, GI tract and testis of Asians and related racial
groups; usually NK phenotype (CD3—, CD2+, CD56+) but may be T-cell
(CD3+, CD56—). Both types are EBV positive. TCR is usually non-clonal
(CD3— type) but clonal episomal EBV; variable clinical behavior but
usually aggressive if outside nasal cavity.
Enteropathy-type™! T-cell lymphoma most commonly involving the jejuneum and ileum;
associate with celiac disease; variable morphology but usually large cells
admixed with inflammatory cells that ulcerate the mucosa and invade
bowel wall; adjacent mucosa shows enteropathy changes; phenotype
CD3+, CD5—, CD7+, CD8—/+, CD4—, CD103+; genotype clonally
rearranged TCR; prognosis is usually poor.
Hepatosplenic» Aggressive lymphoma showing infiltration of sinusoids of spleen, liver
and bone marrow by medium sized cytotoxic T-cells of the y6 type (rare
aB type). Median survival is less than 2 years. Most commonly affects
adolescent males.
Subcutaneous panniculitis-like** Cytotoxic T-cell lymphoma that preferentially infiltrates subcutaneous tissue;
usually CD3+, CD8+T,,¢ phenotype; clonally rearranged TCR; commonly
associated with hemophagocytic syndrome; aggressive disease but may
respond to therapy.
Blasic NK* Aggressive lymphoma of blastic cells with NK phenotype (CD3—, CD56+)
that has a predilection for skin; TCR is germline; poor prognosis
Mycosis fungoides* Epidermotropic T-cell lymphoma of skin slowly progressing from patches
to plaques to tumor nodules (MF); small to medium sized cerebriform
cells infiltrate the dermis and epidermis; phenotype CD3+, CD4+, CD8—
mature T-cell with clonally rearranged TCR (Figs. 21-28 and 29A and B).
Adult disease with excellent prognosis for limited disease; worse
prognosis for tumor stage/extracutaneous spread
Sézary syndrome* Generalized T-cell lymphoma with involvement of skin, lymph node and
blood; skin lesions identical to MF; phenotype similar to MF; adult
disease with aggressive behavior (10-20% 5-year survival).
Primary cutaneous ALCL” T-cell lymphoma composed of CD30+ large pleomorphic T cells confined
to skin at diagnosis (Fig. 21-30); usually solitary or localized disease
which may spontaneously regress; excellent prognosis unless
extracutaneous disease develops. A related clinical disorder is
lymphomatoid papulosis.
Lymphoma
200
Negative <—+
160
&
Al
Gy
“e EE
120
pres
80 mm
SR
acer
40
104
Figure 21-52 ™ Dendritic cell sarcoma. An atypical spindle cell
proliferation characterizes this tumor which expresses a dendritic
cell phenotype.
103
Antibo*
Melanoma
*Antibody directed again st the cytokeratin group of intermediate filament proteins found in epithelial cells.
*“Antibody directed against the 58-kD intermediate filament protein found in mesenchymal cells.
‘Antibody directed against the 200-kD leukocyte common antigen found in various hematopoietic and lymphoreticular cells.
“Antibody directed against the 25-kD protein found in selected cells of the nervous system, melanocytes, interdigitating reticulum cells,
etc. The antibodies HMB-45 and Melan A are more specifically restricted to melanocytes than anti-S100.
with PTCL and ALCL whereas NLPHL is most easily con- in a diffuse background composed predominantly of small
fused with TCRLBCL or LR-CHL. T cells. In addition, conventional PCR will often detect
The neoplastic cells of all subtypes of CHL are CD45 clonal immunoglobulin rearrangement in TCRLBCL but not
and ALK protein negative, CD30 positive, and usually CD15 in NLPHL.
positive (Fig. 21-36). They are most often negative for pan-B
and T cell antigens (see Table 21-3). PTCL and ALCL are
Staging
usually CD45 positive and express one or more pan-T-cell
antigens (CD2, CD3, CD5, CD7). Systemic ALCL also The staging evaluation of the NHLs is similar to that for HL.
expresses ALK protein (Fig. 21-37). The same staging scheme, the Cotswold modification of the
The immunophenotype of the neoplastic cells of NUPHL Ann Arbor classification (see Table 21-5), which was
and TCRLBL are virtually identical. However there are dif- designed for HL, is also used for the staging of NHLs. Because
ferences in the immunoarchitecture of the involved tissue that the natural history of HL is different from that of NHL, there
may be helpful in distinguishing these two entities. The L&H are certain problems with using the Ann Arbor classification
cells of NLPHL are generally found in a vaguely nodular in staging the NHLs. Despite these drawbacks, the Ann Arbor
background composed of CD20+ small lymphocytes. The scheme remains the staging scheme of choice. One major
individual CD20+ L&H cells are often surrounded by a exception to this statement is Burkitt’s lymphoma. In this
rosette of CD3 positive small T cells (Fig. 21-38). The lymphoma the bulk of tumor, rather than sites of involvement,
CD20+ large neoplastic cells of TCRLBCL are usually found is a primary determinant of prognosis. Consequently, a
Case Study |
Stage A Single extra-abdominal site
Stage B Multiple extra-abdominal sites A 68-year-old male auto mechanic presents to his primary
Stage C Intra-abdominal tumor care physician with a chief complaint of bilateral, painless
Stage D Intra-abdominal tumor with multiple masses in the neck region of at least 6 months’ duration. He
extra-abdominal sites has a 120 pack-year smoking history, and his family history
Stage AR Stage C disease with >90% of tumor includes a father and brother with thyroid cancer. His past
surgically removed
continued
Chapter 21 TheLymphomas 495
a cetions
1. What infectious agent is most commonly associated with 3. An asymptomatic mediastinal mass affecting a young
the pathogenesis of Hodgkin’s lymphoma? woman is a common form of presentation for which type
a. Echovirus of Hodgkin’s lymphoma?
b. Herpesvirus a. Nodular sclerosing
c. Helicobacter pylori b. Lymphocyte-predominant
d. Epstein—Barr virus c. Mixed cellularity
d. Lymphocyte depletion
. The cell characteristic of all types of classic Hodgkin’s
i)
lymphoma is the: 4. All follicular center lymphomas are composed of a mixture of:
a. Lacunar cell a. Small cleaved and small noncleaved cells
b. L&H cell b. Centrocytes (smail cleaved) and centroblasts (large
c. Reed—Sternberg cell noncleaved) cells
d. Sézary cell c. Large cleaved cells and immunoblasts
d. Prolymphocytes and paraimmunoblasts
496 Chapter 21 The Lymphomas
5. Small lymphocytic lymphoma is characterized by all of eh A lymphoma associated with infection by a type C retro-
the following except: virus 1S:
a. Indolent but incurable a. Endemic to Japan
b. Usually CD19+, CD5+, CD23+, CD10- b. Associated with hypercalcemia
c. Growth centers c. Usually rapidly fatal
d. t(14:18) d. All of the above
6. Translocations involving the BCL/ and /gH genes are 14. The immunophenotypic definition of B-cell monoclonality is:
commonly associated with which low-grade B-cell a. Clonal rearrangement of the IgH gene
lymphoma? b. Loss of B-cell antigens by the malignant cells
a. Small lymphocytic c. Expression of a single light-chain class
b. Lymphoplasmacytic d. Expression of a single heavy-chain class
c. Mantle cell . Alternatives to karyotypic analysis for the demonstration
d. Marginal zone of chromosomal translocations include:
7. Lymphomatous polyposis is a type of a. Southern blot analysis
a. Small lymphocytic lymphoma b. PCR
b. Large-cell lymphoma c. FISH
c. Mantle cell lymphoma d. All of the above
d. MALT lymphoma 16. Reed-Sternberg cells usually express the following
8. MALT lymphomas are characterized by all of the immunophenotype:
following except: a. CD45+, CD30+, CD15+
a. Extranodal involvement b. CD45-, CD30+, CD15+
b. Associated follicular hyperplasia c. CD45+, CD30+, CD15-
c. CD19+, CD5+, CD10+ phenotype d. CD45-—, CD30-, CD15+
d. t(11;18) Te All of the following are true regarding the low-grade
9. A lymphoma expressing a monoclonal B-cell phenotype non-Hodgkin’s lymphomas except:
with coexpression of CD10 and rearrangement of the a. Survival without treatment averages more than 5 years
BCL2 gene is most likely derived from: b. Disease is usually advanced at diagnosis
a. Follicular center cells c. Multiagent chemotherapy can prolong survival
b. Mantle cells d. Cure is possible in more than 50% of cases
c. Marginal zone cells 18. The diffuse aggressive lymphomas are:
d. Interfollicular cells a. Seldom curable
10. Burkitt’s lymphoma is histologically characterized by b. Rapidly fatal if untreated
which of the following? c. Best treated with a combination of surgery and
a. Uniform nuclei radiotherapy
b. High mitotic rate d. Single-agent chemotherapy is the treatment of choice
c. “Starry sky” pattern
. The staging system for Hodgkin’s lymphoma and the
d. All of the above
non-Hodgkin’s lymphomas is:
11. Lymphoblastic lymphomas are associated with all the a. Working Formulation
following except: b. Duke’s staging system
a. High frequency of developing acute lymphoblastic c. Rye staging system
leukemia d. Ann Arbor staging system
b. Mediastinal mass
20. A patient with Hodgkin’s lymphoma that is localized to
c. Usually express B-cell phenotype
lymph nodes above and below the diaphragm and is asso-
d. Potentially curable in young patients
ciated with drenching night sweats would be classified as:
12. All of the following are T-cell disorders except: a. Stage INE
a. Mycosis fungoides b. Stage ITB
b. Sézary syndrome c. Stage IIIB
c. Hepatosplenic gamma/delta lymphoma d. Stage INA
d. Burkitt's lymphoma
awe 7 eres at the back of this book. —
case
. =
Chapter 21 Thelymphomas 497
16. Dreschfield, J: Ein Beitrag Zur Lehre 2. Ngan, B-Y, et al: Monocytoid B-cell 48. Shimoyama, J, et al: Immunoblastic lym-
von den Lymphosarkomen. Dtsch Med lymphoma: A study of 36 cases. Hum phadenopathy (IBL)-like T cell lym-
Wochenschr 17:1175, 1893. Pathol 22:409, 1991. phoma. Jpn J Clin Oncol 9(Suppl): 347,
. Kundrat, H: Uber Lympho-sarkomatosis. 3) Liu, H, et al: T(11;18) is a marker for LOD
Wien Klin Wochnschr 6:211, 234, 1893. all stage gastric MALT lymphomas 49. Lee, SS, et al: Angioimmunoblastic
. Heim, S, and Mitelman, F: Cancer that will not respond to H. pylori eradi- T cell lymphoma is derived from mature
Cytogenetics. Alan R. Liss, New York, cation. Gastroenterology 122:1286, T-helper cells with varying expression
1995. 2002. and loss of detectable CD4. Int J Cancer
. Rosenberg, SA, et al: National Cancer 34. Ye, H, et al: Bcl-10 expression in normal 103(1):12, 2003.
Institute sponsored study of classifica- and neoplastic lymphoid tissue. Nuclear 50. Lee, J, et al: Extranodal nasal type
tion on non-Hodgkin’s lymphomas: localization in MALT lymphoma. Am J NK/T-cell lymphoma: Elucidating clini-
Summary and description of a working Pathol 157:1147, 2000. cal prognostic factors for risk-based
formulation for clinical usage. Cancer . Oscier, D, et al: Splenic marginal stratification of therapy. Eur J Cancer
49:2112, 1982. zone lymphoma. Blood Rev 19(1):39, 41(10:1402, 2005.
20. Harris, NL, et al: A Revised European- 2005. 5 — . Wright, DH: Enteropathy associated
American Classification of Lymphoid 36. Hammer, RD, et al: Splenic marginal T cell lymphoma. Cancer Surv 30:249,
Neoplasms: A proposal from the Interna- zone lymphoma: A distinct B-cell IN,
tional Lymphoma Study Group. Blood neoplasm. Am J Surg Pathol 10:613, ay. Belhadj, K, et al: Hepatosplenic gam-
84:1361, 1994. 1996. madelta T-cell lymphoma is a rare clini-
21 Grogan, TM, et al: A Southwest Oncol- 3h Krober, A, et al: Additional genetic copathologic entity with poor outcome:
ogy Group Perspective on the revised high-risk features such as |1q deletion, Report on a series of 21 patients. Blood
European-American Lymphoma Classifi- 17p deletion, and V3-21 usage charac- 102(13):4261, 2003.
cation. Hematol Oncol Clin North Am terize discordance of ZAP-70 and VH Soy Ghobrial, IM, et al: Clinical outcome of
IG) SOMOS mutation status in chronic lymphocytic patients with subcutaneous panniculitis-
. Miller, TP, et al: Follicular lymphomas: leukemia. J Clin Oncol 24(6):969, like T-cell lymphoma. Leuk Lymphoma
Do histologic subtypes predict outcome? 2006. 46(5):703, 2005.
Hematol Oncol Clin North Am Oct . Freedman, AS: Biology and manage- 54. Petrella, T, et al: Blastic NK-cell lym-
TG) S98 M1997: ment of histologic transformation of phomas (agranular CD4+CD56+ hema-
. Swerdlow, SH, and Williams, ME: indolent lymphoma. Hematology todermic neoplasms): A review. Am J
From centrocytic to mantle cell lym- (Am Soc Hematol Educ Program) Clin Pathol 123(5):662, 2005.
phoma: A clinicopathologic and molec- 314, 2005. 35) Whittaker, S: Biological insights into the
ular review of 3 decades. Hum Pathol . Lin, P, and Medeiros, LJ: Lymphoplas- pathogenesis of cutaneous T-cell lym-
33(1):7, 2002. macytic lymphoma/Waldenstrom phomas (CTCL). Semin Oncol
24. Inghirami, G, et al: Autoantibody- macroglobulinemia: An evolving 33(1 Suppl 3), 2006.
associated cross-reactive idiotype- concept. Adv Anat Pathol 12(5):246, 56. Abst, A, et al: Prolymphocytic leukemia.
bearing human B lymphocytes: 2005. Curr Treat Options Oncol 6(3):197, 2005.
Distribution and characterization, 40. Salar, A, et al: Diffuse large B-cell Se Lamy, T, and Loughran, TP: Clinical
including IgVH gene and CD5 antigen lymphoma: Is morphologic subdivision features of large granular lymphocyte
expression. Blood 78:1503, 1991. useful in clinical management? Eur J leukemia. Semin Hematol 40(3):185,
25. Williams, ME, and Densmore, JJ: Biol- Haemotol 60:202, 1998. 2003.
ogy and therapy of mantle cell lym- 41. Rosenwald, A, and Staudt, LM: Gene 58. Suzuki, R, et al: Aggressive natural
phoma. Curr Opin Oncol 17(5):425, expression profiling of diffuse large killer-cell leukemia revisited: Large gran-
2005. B-cell lymphoma. Leuk Lymphoma 44 ular lymphocyte leukemia of cytotoxic
26. Majilis, A, et al: Mantle cell lym- (Suppl 3), 2003. NK cells. Leukemia 18(4):763, 2004.
phoma: Correlation of clinical outcome 42. Burkitt, D: A sarcoma involving the jaws Se) Ratner, L: Adult T cell leukemia lym-
and biologic features with three histo- in African children. Br J Surg 46:218, phoma. Front Biosci 9:2852, 2004.
logic variants. J Clin Oncol 15:1664, 1958. 60. Warnke, RA, et al: Tumors of the lymph
IDI, 43. Lui, PC, et al: Intravascular lym- nodes and spleen. In: Warnke, RA, et al:
. van den Oord, J, et al: The marginal phomatosis. J Clin Pathol 56(6):468, Atlas of Tumor Pathology. Armed Forces
zone in the human reactive lymph node. 2003. Institute of Pathology, Washington, DC,
Am J Clin Pathol 86:475, 1986. 44. Boulanger, E, et al: Prognostic factors 1995, pp 233-244.
. vanKrieken, JHJM, et al: Splenic mar- and outcome of human herpesvirus 6 = . Pileri, SA, et al: Tumours of histiocytes
ginal zone lymphocytes and related cells 8-associated primary effusion lymphoma and accessory dendritic cells: An
in the lymph: A morphologic and in patients with AIDS. J Clin Oncol immunohistochemical approach to
immunohistochemical study. Hum Pathol 23(19):4372, 2005. classification from the International
20:320, 1989. 45. Maitra, A, et al: Precursor B-cell Lymphoma Study Group based on 61
DS). Maes, B, and De Wolf-Peeters, C: lymphoblastic lymphoma. A study of cases. Histopathology 41(1):1, 2002.
Marginal zone cell lymphoma—an nine cases lacking blood and bone 62. Banks, PM: Technical aspects of speci-
update on recent advances. marrow involvement and review of the men preparation and introduction to spe-
Histopathology 40(2):117, 2002. literature. Am J Clin Pathol 115(6):868, cial studies. In: Jaffe, ES (ed), Surgical
30. Isaacson, P, et al: Malignant lymphoma 2001. Pathology of the Lymph Nodes and
of mucosa-associated lymphoid tissue. 46. Reimer, P, and Hentrich, M.: Peripheral Related Organs, ed 2. WB Saunders,
Histopathology 11(5):445, 1987. T-cell lymphoma: Diagnosis and treat- Philadelphia, 1995, pp 1-21.
. Montalban, C, and Norman, F: Treat- ment. Dtsch Med Wochenschr 63. Dabbs, DJ: Diagnostic Immunohisto-
ment of gastric mucosa-associated 131(13):685, 2006. chemistry. Churchill Livingstone,
lymphoid tissue lymphoma: Helicobac- 47. Kadin, ME, and Carpenter, C: Systemic Philadelphia, 2006.
ter pylori eradication and beyond. and primary cutaneous anaplastic large 64. Kuppers, R: Molecular biology of
Expert Rev Anticancer Ther 6(3):361, cell lymphomas. Semin Hematol Hodgkin’s lymphoma. Adv Cancer Res
2006. 40(3):244, 2003. 84:277, 2002.
Chapter 21 TheLymphomas 499
(OS), Solal-Celigny, P: Increasing treatment 68. Maloney, DG: Treatment of follicular 71. Ansell, SM, and Armitage, J: Non-
options in indolent non-Hodgkin’s non-Hodgkin’s lymphoma. Curr Hematol Hodgkin lymphoma: Diagnosis and
lymphoma. Semin Oncol 29(2 Suppl 6):2, Rep 4(1):39, 2005. treatment. Mayo Clin Proc 80(8):1087,
2002. 69. Armitage, JO: Overview of national and 2005.
66. McLaughlin, P: Progress and promise in individualized therapeutic strategies for 72. Hoelzer, D, and Gokbuget N: Treatment
the treatment of indolent lymphomas. non-Hodgkin’s lymphomas. Clin of lymphoblastic lymphoma in adults.
Oncologist 7(3):217, 2002. Lymphoma 3 (Suppl 1):S5, 2002. Best Pract Res Clin Haematol 15(4):713,
67. Gandhi, MK, and Marcus, RE: Folliecu- 70. Marcus, R: Current treatment options in 2002.
lar lymphoma: Time for a rethink? Blood aggressive lymphoma. Leuk Lymphoma
Rev 19(3):165, 2005. 44 (Suppl 4):S15, 2003.
Chapter / jj
Multiple Myeloma
and Related Plasma
Cell Disorders
Ashraf Z. Badros, MD
Overview OBJECTIVES
Plasma Cell Development At the end of this chapter, the learner should be able to:
Immunoglobulin
Le Describe the normal structure and function of immunoglobulin and the significance of
Structure and Function
the M-spike.
Laboratory Recognition and
Méasurements . Describe the workup for patients suspected of plasma cell dyscrasia.
Clinical Consequences of
. List the diagnostic criteria for multiple myeloma and its variants.
Increased Monoclonal
Immunoglobulin . Compare multiple myeloma and monoclonal gammopathy of undetermined
WwW
R
500
Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders SQ]
Case Study 1
Case Study 2
Case Study 3
ari) Natural
c @ antibodies
wy NZL and IgA
B1 cell ae
“ae |
o
Marginal-
zone
ee
B cell _Y ake
Plasma @) >
ind
cell Se loM
Apoptotic |
cell
Germinal center
Proliferation
and mutation
—
Follicular
¥ B cell
Selection
and CSR
Figure 22-1 ™ Development of plasma cells; on encounter with an antigen (indicated by week 1), naive marginal-zone B cells differentiate
into plasma cells. CSR = class-switch recombination.
Heavy chain
Antigen
binding ab Heavy chain
hypervariable
regions
Carboxy terminal
Figure 22-2 ™ Structure of the basic immunoglobulin unit. V4, = variable region heavy chain; V, = variable region light chain;
C, = constant region heavy chain; C, = constant region light chain.
Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders 503
Normal serum
O2
IgG Myeloma with y spike “M” or monoclonal spike
and reduced albumin
Polyclonal
hypergammaglobulinemia
Oy, 2 B Y
Waldenstrom’s
macroglobulinemia
with IgM spike
ALB 01 02 B
Hypogammaglobulinemia
Figure 22-3 ™ Patterns of serum protein electrophoresis showing characteristic patterns of normal serum, monoclonal M-spike, polyclonal
antibody production, IgM M-spike, and the absence of antibody production seen in hypogammaglobulinemia.
from headache, dementia, disturbances of consciousness, to the abnormal immunoglobulin.'* The low immunoglobulin
stroke, and/or coma. Failure of the heart to compensate for levels lead to increased susceptibility to infections such as
widespread hypoxemia, increased intravascular volume, and sinusitis, bronchitis, and pneumonia, which is a main cause of
high pressure may lead to congestive heart failure. Although the death of multiple myeloma patients. Intravenous infusions of
correlation between serum viscosity and clinical manifestations immunoglobulin can help decrease the frequency of these
is not precise, clinical manifestations are rarely attributable to infections.'?
hyperviscosity if serum viscosity is less than 4 centipoises (CP),
CRYOGLOBULINS
(normal value =1.8 CP). Most patients are symptomatic when
Cryoglobulins are serum immunoglobulins that precipitate
serum viscosity is greater than 6 CP. Most cases of hyperviscos-
reversibly on exposure to cold. This is a rare complication
ity syndrome are seen with IgM, IgA, and IgG3 because of their
of plasma cell disorders. In cryoglobulinemic syndrome, a
molecular weight and physical properties. Plasma exchange
multitude of organs can be involved, but usually patients
(plasmapheresis) and removal of the immunoglobulin is an
suffer from painful extremities on exposure to cold, palpable
emergency therapy for symptomatic patients."
purpura, arthralgia, and fatigue. In severe cases it may result
in vasculitis and kidney damage.'* Cryoglobulins are classi-
DECREASED PRODUCTION OF NORMAL
fied into:
IMMUNOGLOBULINS
Patients with the plasma cell disorders of multiple myeloma Type I: The presence of isolated monoclonal
and Waldenstr6m’s macroglobulinemia have decreased levels immunoglobulin (IgG or IgM). This is common in
of the normal “uninvolved” immunoglobulin. The low levels Waldenstr6m’s macroglobulinemia and/or multiple
result from suppression of normal plasma cells as a response myeloma.
Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders 505
+ Normal (n = 282)
® x light-chain multiple myeloma (n = 120)
65
O Renal impairment from non-light-chain diseases (n = 31)
50
4 i light-chain multiple myeloma (n = 104)
© Nonsecretory myeloma (n = 28) Relative risk of
full progression 25
100,000 at 20 years
=) 13.6
Ss
= 10,000
2 3.6
=
i 0.5 1.0 Wes 2.0 2.5 3.0
—
7 Serum M-spike value
ee FOOO
>
@ Figure 22-5 @ Actuarial risk of full progression by serum mono-
2 clonal protein (M-protein) value at diagnosis of monoclonal
S 100 gammopathy of undetermined significance (MGUS) in persons from
i=
2 southeastern Minnesota.
s
. 10
)
(es marrow (BM) aspirate, and no end-organ damage or other
e)
o
en
o
findings of multiple myeloma such as lytic bone disease.
After completing the initial screening tests for the presence of
n
D
monoclonal protein, computed tomographic (CT) scans of the
fo} chest and abdomen, with biopsies of suspicious enlarged
ot
0.1
lymph nodes, may also be required. Reassurance for the
aN \ SS Ors patient and full explanation of the condition is crucial.
x SS Patients will have to understand that no immediate therapy is
needed. However, close monitoring initially every 3 months,
Log serum concentration of free-light-chain « (mg/L)
and if the M-spike remains stable, every 6-month intervals is
Figure 22-4 ™ Concentrations of « and } free-light-chains in sera needed for life.'’
from healthy individuals and those with myeloma. (From Bradwell,
AR: Serum test for assessment of patients with Bence-Jones myeloma.
Lancet 361:489, 2003. Reprinted with permission from Elsevier.) Multiple Myeloma
Multiple myeloma is the most common plasma cell dyscrasia,
Type II: A mixture of polyclonal immunoglobulin in affecting terminally differentiated B cells. The biology of
association with a monoclonal immunoglobulin, multiple myeloma suggests a multistep process as illustrated
typically IgM or IgA, with rheumatoid factor by the clinical progression from MGUS, to a symptomatic
activity. Type II is seen in patients with persistent multiple myeloma (Fig. 22-6). A B-cell precursor, after
viral infections, particularly hepatitis C and human immunoglobulin gene rearrangement is presumed to be the
immunodeficiency virus infections. origin of the malignant clone in multiple myeloma. The
events that determine the susceptibility of B cells to undergo
Type III: Mixed cryoglobulins consisting of polyclonal
such malignant transformation are at best speculative. Plasma
immunoglobulin that is secondary to connective
cells are generally localized in the bone marrow until late in the
tissue diseases."°
disease when the cells grow independent of the bone marrow
microenvironment, usually causing a more aggressive presen-
Monoclonal Gammopathy tation of the disease. Multiple myeloma is currently an incur-
able hematological malignancy; although complete responses
of Undetermined Significance
can be obtained with various therapies as outlined below.
Although monoclonal gammopathy of undetermined signifi- Patients’ survival varies widely— ranging from a few months
cance (MGUS) was previously named benign monoclonal to more than 15 years. The disease is characterized by the
gammopathy, 20% of these patients will develop overt malig- triad of an abnormal proliferation of plasma cells in the bone
nancy such as multiple myeloma, non-Hodgkin’s lymphoma marrow, over production of monoclonal immunoglobulin, and
(NHL), and chronic lymphocytic leukemia (CLL).'° The lytic/destructive bone disease. This clinical triad causes vari-
incidence of MGUS varies by age, sex, and race. MGUS is ous disease manifestations which include: life threatening
diagnosed in approximately 1% of individuals older than end-organ damage with renal insufficiency, anemia, immuno-
the age of 50 and up to 3% of those older than the age of 70 suppression and infections. Probably the most devastating
(Fig. 22-5). MGUS is defined by the finding of a small feature of myeloma is the osteolytic lesions, resulting in bone
M-spike in patients with normal levels of uninvolved pain, pathological fractures, spinal cord compression, and
immunoglobulin, fewer than 10% plasma cells on bone hypercalcemia.'*
506 Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders
® Incidence
® Mortality
15
0
1975 1980 1985 1990 1995 2000 1975 1980 1985 1990 1995 2000 1975 1980 1985 1990 1995 2000 1975 1980 1985 1990 1995 2000
Figure 22-7 @ SEER incidence and U.S. death rates from myeloma, by race and sex. (From SEER 9 areas and NCHS public use data file.)
Rates are age-adjusted to the 2000 U.S. standard million population by 5-year age groups. Regression lines are calculated using the Join Point
Regression program.
commonly, plasmacytomas may affect soft tissues away from the formation of new bone by osteoblasts. Bone lesions in
the bone and bone marrow. multiple myeloma present as lytic lesions as well as osteo-
porosis that manifest as pathological factures and/or spinal
BONE MARROW STROMA cord compression. Late in the disease course, many patients
The bone marrow microenvironment plays an important role in report loss of height because of vertebral collapse. Multiple
supporting the malignant plasma cells in multiple myeloma by myeloma bone disease results from dissociation of bone
promoting their growth and preventing apoptosis (Fig. 22-9). absorption and formation secondary to stimulation of osteo-
Adhesion of myeloma cells to the stromal cells induces the clasts and inhibition of osteoblasts, respectively.*° Myeloma
secretion of several cytokines (proteins that promote or inhibit cells adhere and interact with bone marrow stromal cells
cell functions) from the stromal cells, endothelial cells, and/or resulting in the production of osteoclast-activating factors,
osteoclasts as well as from the plasma cells themselves. These including interleukin (IL)-18, IL-6, and IL-3 and tumor
molecules play an important role in multiple myeloma patho- necrosis factor; these cytokines stimulate the tumor necrosis
genesis and mediate many of the disease signs such as bone factor-related induced cytokine (TRANCE). TRANCE is
destruction, tumor cell proliferation, drug resistance, and responsible for the activation and maturation of osteoclasts.
genetic instability of plasma cells. The details of each cytokine TRANCE activity is blocked by osteoprotegerin. Osteoprote-
and the various pathways involved are beyond the scope of this gerin is a cytokine and a member of the tumor necrosis
review. In brief, they include osteoclast activating factors, such factor (TNF) receptor superfamily. It is also known as
as interleukin (IL)-18, IL-6, and the tumor necrosis factor osteoclastogenesis inhibitory factor (OCIF). The balance of
family proteins, as well as vascular endothelial growth factor, TRANCE/osteoprotegerin is totally disrupted in multiple
transforming growth factor-B, and insulin-like growth factors. * myeloma patients leading to overproduction of TRANCE and
inactivation of osteoprotegerin by binding to syndecan- | that
BONE DISEASE IN MULTIPLE MYELOMA is secreted by plasma cell surface (Fig. 22-10).?’
Normal bone is a dynamic structure undergoing continuous Recently, inhibition of osteoblasts is established as a
renewal through the resorption of old bone by osteoclasts and major contributor to bone disease in myeloma. Dickkopf |
Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders
MULTIPLE MYELOMA
MALIGNANT PROLIFERATION
OF PLASMA CELLS
Figure 22-8 ™ Mechanisms of disease in multiple myeloma. Skeletal destruction, abnormal immunoglobulin production, marrow failure,
and decreased production of normal immunoglobulin all play a role.
(DKK1), an inhibitor of the Wnt signaling pathway, which is to hypercalcemia, renal insufficiency, or amyloidosis. Bone pain
crucial for osteoblast differentiation, leads to reduced bone is the most common presentation in multiple myeloma. Spinal
formation. In the Wnt signaling pathway, signaling mole- cord compression from extramedullary plasmacytoma is an
cules (Wnt proteins) regulate morphology, proliferation, acute emergency that should be diagnosed and treated promptly
motility, and cell fate. to prevent long-term disability. Magnetic resonance imaging
(MRI) or computed tomographic myelography of the entire
HYPERCALCEMIA spine must be done immediately, with appropriate follow-up
Calcium balance plays a critical role in regulation of cellular treatment by chemotherapy, radiotherapy, or neurosurgery to
function. With increased bone turn- over, calcium is released avoid permanent damage.
in the blood causing several symptoms. One of the earliest is The serum creatinine concentration is elevated in 20% of
constipation and cramping from altered motility of the intes- patients at diagnosis (>2 mg/dL).** The most common cause
tine followed by increased urination, dehydration and muscle of renal insufficiency in multiple myeloma is the precipitation
weakness. Increased calcium in the urine can lead to forma- of monoclonal light chains in the renal tubules called cast
tion of kidney stones and kidney failure. Later on, change in nephropathy. Other causes of renal dysfunction in multiple
mental status may develop, mostly confusion. The severity of myeloma include deposition of light chains in the kidney
the symptoms is dependent on both the level of calcium and parenchyma (light-chain deposition disease) and amyloid
the rate of rise. Hydration and forced diuresis and bisphos- fibrils in AL amyloidosis. Acute renal failure can occur in
phonate therapy are the main therapies for hypercalcemia (see multiple myeloma after administration of radiocontrast media
supportive care section below). during CT scans, or the use of none steroidal pain medica-
tions. In a few cases, acute renal failure may occur secondary
to hypercalcemia.
Clinical Features
Osteoclasts Increase
BMECs Angiogenesis
Bone Reabsorption
©D. Harmening
Figure 22-9 @ Bone marrow microenvironment. Effects of cytokines and transforming growth factors on the bone marrow (BM) microenvi-
ronment in patients with multiple myeloma (MM). Bone marrow stromal cells (BMSC) secrete: IL-6, VEGF, SDF-1a, and RANKL. . This IL-6
secreted in the BM microenvironment promotes the binding of MM cells to the BMSCs augmenting more secretion of IL-6 and other
cytokines from the BMSCs and MM cells. For example, TGF8, TNFa, and VEGF from malignant myeloma cells enhance IL-6 secretion from
BMSCs. The MM cells secrete IL-6, VEGF, bFGF, TNFa, TGFB, and MIP-1a. The IL-6, VEGF, bFGF, secreted by the MM cells and the VEGF from
BMSCs initiate angiogenesis in bone marrow endothelial cells (BMECs). The cytokines MIP-1a@ secreted by the MM cells and RANKL secreted
by BMSCs initiate or activate osteoclast formation, leading to bone reabsorption and bone destruction in MM patients. Osteoclasts also
secrete IL-6, inducing the growth of the malignant clone of MM cells. IL-6 = Interleukin 6; VEGF = vascular endothelial growth factor;
SDF-1a = stromal cell-derived factor alpha; RANKL = receptor activator of nuclear factor a—B ligand; bFGF = basic fibroblast growth factor;
TNF = tumor necrosis factor; TGFa = transforming growth factor beta; MIP-la = macrophage inflammatory protein alpha.
Myeloma cells
(2) IL-6 ®
TNF-B
Stroma © @ C@® va
—__ EDI SS... (> Osteoblast
TRANCE \ Fait
PTHrP U0
@)
1.
2.
MMcells adhere to stroma.
Stromal cells secrete OAFs.
Bo
a Syndecan
3. OAFs induce stroma and osteoblasts to secrete TRANCE.
4a. TRANCE is blocked by OPG; Syndecan from MM cells U
traps OPG and reduces OPG concentrations.
6) © 4b. Excess TRANCE is available to stimulate osteoclast. U0
5. Increased osteoclastic activity increases cytokine release
'e from bone matrix. U 4b)
6. These cytokines stimulate MM cell growth.
7. These cytokines also cause release of PTHrP from MM cells,
which activates stromal cells to secrete additional TRANCE.
TGF-B
FGF-1&2 e
IGF-1&ll Osteoclasts nn
PDGF Pe Osteoclast
progenitors
Figure 22-10 w Bone destruction in multiple myeloma. (From Tricot, G: New insights into role of microenvironment in multiple myeloma.
Lancet 355:248, 2000. Reprinted with permission from Elsevier.)
510. Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders
d Sympt
om s immunoglobulin in the blood, causing red blood cells to
‘Sig ns an
Table 22-2 adhere to each other. The same phenomenon results in the
increased erythrocyte sedimentation rate. Circulating plasma
cells can be occasionally seen in multiple myeloma patients
Possible Cause and it carries a poor prognosis. Later in the disease, course
replacement of the bone marrow with plasma cells may also
Bone pain (68%) Fracture, lytic lesions
Easy fatigue (62%) Anemia (73%) cause teardrop-shaped red cells and the appearance of earlier
Polyuria (30%) Hypercalcemia (13%) forms of white blood cells in the peripheral blood.
Nausea and vomiting Renal failure, hypercalcemia
Recurrent infections Low normal Ig levels CHEMISTRY STUDIES
Paraplegia Cord compression
Routine chemistry panels are essential in evaluating myeloma
Confusion (15%) Hyperviscosity, hypercalcemia
Bleeding Thrombocytopenia patients. Serum blood urea nitrogen (BUN) and creatinine mea-
Arrhythmia (7%) Amyloidosis (4%) sures the kidney function that is often affected in myeloma.
Peripheral neuropathy Amyloidosis Serum lactate dehydrogenase (LDH) is a nonspecific marker of
Fever Infection tissue breakdown and elevated levels are associated with shorter
Renal insufficiency (19%) Cast nephropathy, light chain
survival. Calcium levels are often elevated in myeloma patients,
deposition
indicating bone destruction. Calcium is primarily bound to
Source: Modified from Kyle, RA, et al: Review of 1027 patients with albumin; the unbound or free calcium is the major cause of
newly diagnosed multiple myeloma. Mayo Clin Proc 78:21, 2003.
symptoms. Calcium levels must always be interpreted with the
albumin level in mind. For each mg/dL of albumin below
normal, the serum calcium should be increased by 0.8 mg/dL.
Laboratory Studies Albumin levels can be low in multiple myeloma due to inhibi-
tion by IL-6 as well as loss in the nephrotic kidney. In either
COMPLETE BLOOD COUNT AND PERIPHERAL BLOOD
case low albumin is a major prognostic criterion in multiple
SMEAR
myeloma (see international staging system below).
The automated complete blood count (CBC) is an easy avail-
8.-Microglobulin is the light chain of the histocompati-
able assessment of bone marrow function. The most common
bility locus antigen (HLA). It is the most useful predictor of
finding in multiple myeloma is a decrease in the number of
tumor load and disease activity, and in predicting the progno-
red blood cells with no change in size (normocytic nor-
sis of multiple myeloma patients. C-reactive protein is another
mochromic anemia). On peripheral smear examination, the
nonspecific marker of disease activity, mostly reflecting the
most characteristic finding in multiple myeloma is rouleaux
activity of IL-6, an important growth factor for myeloma
formation of the red cells (stacking of red cells together like
cells. Its prognostic value is limited by the non-specificity of
coins) (Fig. 22-11). It is caused by increased amounts of
the test.
lial }
, 4
Mili,
Figure 22-12 ® Bone marrow aspirate showing atypical and Figure 22-14 m Flame cell, sometimes associated with IgA
binucleated plasma cells and Russell bodies (arrow). myeloma.
such as flame cells (large, intensely staining plasma cells) can CYTOGENETICS IN MULTIPLE MYELOMA
identify IgA subtype (Fig. 22-14). A differential count of Chromosomal studies (karyotyping) on bone marrow plasma
marrow cells (usually 500 cells are counted) is performed to cells in multiple myeloma have been slow to be recognized
establish percentages of various cell types. Patients with and implemented in clinical practice, mostly due to the low
myeloma have a variable level of plasmacytosis ranging from proliferative state of the plasma cells.
10% to 100%. This variability is partially related to the Chromosomal abnormalities can be found in 30% to 40%
pattern of bone marrow involvement in multiple myeloma, of patients. Frequently, cytogenetic abnormalities in multiple
which is often focal. Special stains are usually performed to myeloma are characterized by complex karyotypes with fre-
confirm clonality with k or ) restriction. quent iumerical and structural aberrations. *? Those with mono-
Bone marrow plasma cell labeling index (% of dividing somy 13, 13ql4 deletion and hypodiploidy have the worst
plasma cells) can differentiate between benign and malignant prognosis. Interphase fluorescence in situ hybridization (FISH)
plasma cells and is a major prognostic factor. Plasma cells detected a higher trequency of abnormalities even in patients
have a characteristic immunophenotype. In addition to stain- with normal karyotype (Fig. 22-15). Chromosomal transloca-
ing positively for cytoplasmic immunoglobulin, malignant tions involving 14q32, the site of the IgH (heavy chain) locus,
plasma cells stain positive for CD38, CD56 (neural celi adhe- is the most frequent abnormality in multiple myeloma. The
sion molecule), and CD138. They are usually negative for most common of these are translocations involving t(11;14)
surface immunoglobulin and the pan-B-cell antigen CD19; (q13;q32), t(4:14)(p16.3;q32.3), t(6;14)(p25;q32), and t(14;16)
15% to 20% will stain positively for CD20 and CD52. These (q32.3;q23) (Table 22-4). The t(4; 14) and t(14; 16) are associ-
latter markers led to the use of monoclonal antibodies (such ated with a poor prognosis (Fig. 22—16).°° In contrast, t(11;14)
as rituximab and Campath, respectively) in several clinical (q13;q32) patients have a better prognosis with up-regulation of
trials for the treatment of multiple myeloma. cyclin DI (a gene located on 11q13, which can function as an
oncogene), and lower levels of serum monoclonal proteins and
lymphoplasmacytic or small mature plasma cell morphology,
along with CD20 expression (see Table 22-4).*!
In recent years, gene expression profiling has emerged as
a useful tool to identify subgroups that follow a similar pro-
filing. These subgroups confirm the previously recognized
clinical observation of the diversity of multiple myeloma
patients. Recently, two major pathogenetic pathways for the
development of plasma cell tumors were defined: one that is
associated with hyperdiploidy and one that is characterized by
the presence of chromosome translocations involving the
immunoglobulin heavy chain locus (IgH).** While these studies
are in their infancy, this technology, as well as comparative
genomic hybridization, and studies of tumor-associated
antigens will ultimately define the critical genetic lesions in
multiple myeloma, providing new diagnostic and prognostic
indicators as well as a therapeutic paradigm for multiple
Figure 22-13 m Bone marrow biopsy sample showing replace-
ment of marrow by plasma cells. myeloma patients.
Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders
t(11;14) Cyclin DI
t(4;14) MGFR-3 and MMSET
t(6;14) Cyclin D3
t(14;16) C-MAF
CEPI7 LSI p53§CEP17 t(14;20) MAF-B
RADIOLOGICAL INVESTIGATIONS
The role of imaging in the management of multiple myeloma
includes defining the extent of bone disease, and, in the uncom-
mon cases, defines soft tissue involvement. This 1s a critical step
in the diagnosis; as well as establishing adequate interventions
to prevent fractures and cord compression, etc. Full discussion
of the various modalities is available in an excellent recent
16q23§14q32
review listed in the references.**** The following is a brief
Figure 22-15 ™@ PCs with both the normal and abnormal pattern outline.
of hybridization. The depicted PCs show (A) a cell with the normal
configuration of 2 pairs of signals for the probes localizing to the
centromere 17 (CEP17; aqua) and the 17p13.1 (LSI p53) (red) PLAIN RADIOGRAPH—X-RAY EXAMINATION Skeletal
probe. (B) A cell with deletion of 17p13.1. There are two green survey remains the standard method for radiological screen-
signals arising from the centromeric probe but only one red signal ing of suspected multiple myeloma cases. Multiple lytic or
from the p53 locus probe. (C) A normal configuration of probes punched out lesions are very suggestive of multiple myeloma
used to detect the t(14;16)(q32;q23). The locus-specific 14q32
and are present in 76% of patients at diagnosis (Figs. 22—17
probes are labeled in green, and the 16q23 probes are labeled in
red. (D) A cell with fusion of probes for 14q32 (green) and 16q23 and 22-18). X-ray exams provide a prognostic parameter for
(red). (From Fonseca, R, et al: Clinical and biologic implications of assessment of the tumor load at diagnosis in the classic
recurrent genomic aberrations in myeloma. Blood 2003;101:4570. Durie/Salmon staging system. X-ray films of the long bones
Reprinted with permission from the American help identify critical cortical thinning that may lead to patho-
Society for Hematology.)
logical fracture, allowing for preventive interventions. How-
ever, plain radiography, demonstrating lytic disease only
when at least 30% of trabecular bone substance has been lost,
p<0.001 provides an inadequate assessment of generalized osteopenia
that affect more than 25% of the patients with no lytic disease.
2 oo All others including t(11;14)
es
oOiN
(=) rep)
Survival
probability
t(4;14) or
S Ne)
t(14;16) or
—17p13
2 ro)
0 10 20 30 40 50 60 70 80 90 100110120130140150
Months
OSITRON
RJ LN EMISSION TOMOGRAPHY Positron emission
tomography (PET) imaging with 18-fluorine-fluoro-
deoxyglucose (FDG) is a promising new scanning tech-
nique under evaluation in multiple myeloma. PET scan
appears to be very sensitive in detecting occult sites of
disease and appears to be most useful in diagnosing and
follow up of nonsecretory multiple myeloma (only 1% to
2% of all myeloma patients) and extramedullary plasma-
cytoma (Fig. 22-20).
8) 7 Vala:
} 4 & 4 Y A
UNE OWL
MAG aX g |ES(
Ne) CE IMAG Magnetic resonance
imaging (MRI) produces computer-generated images based Figure 22-19 @ MRI showing compression fracture at L-2 (red
on tissue absorption of radiofrequency energy while in a arrow) and focal plasmacytoma (white arrows and circles).
514 — Chapter 22. Multiple Myeloma and Related Plasma Cell Disorders
CT Transaxial
CT Scout View
PET Transaxial
=
ve
&i
@to.
*
*
oa
rt
oO
ae
os
%
on
==
ta]
CY
vy
rye @°9
“©
Fused Transaxial
s+ n900
Figure 22-20 mPET-scan showing metabolic activity in multiple plasmacytomas throughout the skeleton.
response was approximately one and a half years. Overall sur- 27% was achieved in heavily treated relapsed and refractory
vival for patients was less than 3 years and was mostly depen- multiple myeloma patients with a median duration of
dant on the stage at diagnosis rather than therapy.*” More response of 13 months and a median overall survival of
intensive combinations with various chemotherapy drugs did 17 months. Responses to bortezomib have been rapid and
not show improvement of overall survival in comparison to are seen after one or two cycles, in most patients. A Phase
melphalan/prednisone. The introduction of dexamethasone in III trial confirmed the superiority of single-agent borte-
the VAD regimen (vincristine, doxorubicin, and dexametha- zomib over dexamethasone in terms of response rate, time to
sone), initially for relapsed patients and later for newly diag- progression, and overall survival. The additive effect of
nosed patients has been associated with increased complete dexamethasone was seen in nonresponders and in those
remission (CR) rates (5% to 10%) with no significant impact who progressed on single-agent bortezomib. The main side
on overall survival.*° effects of the velcade include peripheral neuropathy and
In the mid-1990s, high-dose chemotherapy with autolo- thrombocytopenia; both appear to be reversible on stopping
gous stem cell transplant (auto-SCT) support changed the therapy. Currently bortezomib is being evaluated in the
way we treat myeloma patients, especially those younger first-line setting, alone and in various combinations with
than age 65.*' Auto-SCT has improved the outcome in newly dexamethasone, thalidomide, doxorubicin, and with mel-
diagnosed multiple myeloma patients, with higher complete phalan with and without stem-cell transplantation, with very
remission rates (40%), longer event-free survival (4 to 5 years), encouraging results.°?°?
and improved overall survival (greater than 7 years). Many of Lenalidomide (Revlimid) is another novel therapy that
these patients had durable remission, especially the subset of has shown significant activity in relapsed multiple myeloma.
patients with no chromosomal abnormalities and normal f,,, In March 2005, two Phase III trials in relapsed and refractory
at diagnosis.*” Several studies have shown that tandem trans- myeloma patients were stopped prematurely when interim
plants can further increase the duration of remission and over- efficacy endpoint “time to disease progression” was found to
all survival.**4 be statistically better in patients receiving Revlimid plus
Allogeneic stem cell transplants have been advocated dexamethasone compared with patients receiving dexametha-
as the only curative therapy for multiple myeloma patients.* sone alone. The trials were un-blinded and Revlimid was
The role of allo-SCT has been controversial; standard con- added to patients receiving dexamethasone alone. Revlimid
ditioning allo-SCT was associated with 50% treatment has now been approved by the Food and Drug Administration
related mortality in the first 100 days. However, 30% to in June 2006 for use in relapsed multiple myeloma patients
50% of patients who survive the first year remain disease- that have received at least one prior therapy. Lenalidomide is
free at 3 to 6 years, with few well documented cases of sus- being explored in combinations with other drugs, and in the
tained molecular remissions. These durable remissions have newly diagnosed multiple myeloma patients.
been attributed to immunological effects of T cells known as Additional therapy after achievement of a maximal
graft versus myeloma effect.*° Several reports have shown response following chemotherapy or transplant (called mainte-
that a nonmyeloablative conditioning followed by allo- nance) produced conflicting results.°° Continuing conventional
geneic graft (T cells) had lower mortality and can lead to chemotherapy therapy (often referred to as consolidation) is not
prompt engraftment.*’ Despite initial success, long-term sur- beneficial following the achievement of the plateau phase, espe-
vival remains low, especially in high-risk relapsed myeloma cially in high-risk patients, as most relapse while on therapy.
patients.** Continuing conventional chemotherapy is associated with high
Radiation therapy has limited efficacy in treating sys- morbidity and adversely affects the patient’s quality of life.
temic manifestation of multiple myeloma. However, it is Interferon (IFN) maintenance results in prolonged relapse-free
highly effective in treating the localized disease process such survival by 4 to 9 months with a small gain in overall survival.*°
as painful lesions resulting from plasmacytoma, and lytic However, the drug has many side effects, and its use in
bone lesions especially in spinal cord compression or impend- myeloma is decreasing in favor of novel therapeutics mentioned
ing fractures. above.”’
In 1999, single-agent thalidomide (Thalomid) showed sig- One study investigated alternate day, oral prednisone at
nificant activity in relapsed refractory multiple myeloma with an two different dose levels (10 mg versus 50 mg) in multiple
overall response rate of 30%; few patients remain in continuous myeloma patients after they responded to induction. The
remission at 6 years. In combination with dexamethasone, higher dose of prednisone significantly improved progression-
response rates increase to approximately 50%. The combination free survival (14 months versus 5 months), and overall sur-
of thalidomide plus dexamethasone is now the main induction vival (37 months versus 26 months).** The study did not
therapy for newly diagnosed multiple myeloma patients with an include a formal evaluation of side effects. Several newer
overall response rate of 65% to 70%.*° Thalidomide/dexametha- agents are now being studied as maintenance therapies in
sone has the advantage of being an oral regimen; however it car- the plateau phase following initial chemotherapy or high-
ries a risk of neuropathy and deep venous thrombosis (15%).°°>! dose chemotherapy, including thalidomide, lenalidomide, and
The proteasome inhibitor bortezomib (Velcade) was bortezomib. Their role in this setting remains experimental.
approved in 2003 in the United States for relapsing multiple Preliminary data suggest that remission and overall survival
myeloma patients. Initial approval was based on the results may be prolonged with thalidomide/pamidronate mainte-
of the Phase II trial “SUMMIT” in which a response rate of nance after high-dose chemotherapy.°*?
Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders 517
Atypical Variants of Plasma Cell patients remaining free of disease for at least 10 years. New
Syndromes modalities for diagnosis (PET, free light chain assays, etc.)
should help further define these cases.°?
Smoldering Myeloma
These are patients who meet the criteria for diagnosis of mul- Plasma Cell Leukemia
tiple myeloma but have a low tumor mass (Durie-Salmon
stage I) with an M-protein greater than 3 g/dL and/or greater Plasma cell leukemia (PCL) is a rare variant of multiple
than or equal to 10% bone marrow plasma cells and no end- myeloma (2% to 3%), with an aggressive presentation and
organ damage from the disease (normal kidney function, short survival. It is defined as circulating peripheral blood
normal serum calcium, and no lytic bone lesions).°° These plasma cells exceeding 2000/uL, and 20% of peripheral blood
patients should not be treated as long as they are asympto- white cells (Fig. 22-21). Half of these cases are found in
matic, but they need careful follow-up as their incidence of advanced multiple myeloma, usually late in the course of the
progression to multiple myeloma is higher than that of MGUS disease, and usually herald a terminal event. The other half,
patients, and most will progress to advanced stages requiring often referred to as primary plasma cell leukemia, affect
therapy within 3 years.°' The administration of bisphospho- newly diagnosed patients. Patients present with high calcium
levels, kidney failure, and much more anemia, thrombocy-
nates to patients with smoldering disease, to delay the
progression to overt multiple myeloma, is undergoing clinical topenia, and involvement of various organs, such as the
trials. It is thought that bisphosphonates change the bone mar- liver, spleen, and lymph nodes. Treatment is the same as for
row “soil” to become an ineffective microenvironment to newly diagnosed multiple myeloma. Prognosis is poor, with
expected survival of only several months, even after autolo-
plasma cell “seeding.” The benefit of this treatment has to be
balanced against the long term risks of therapy.” gous and/or allogeneic transplantation.
Extramedullary Plasmacytoma
Extramedullary plasmacytoma (EMP) is a soft tissue plasma
cell tumor. Similar to SPB, there should be no evidence of
bone destruction or evidence of marrow plasmacytosis. EMP
must be distinguished from reactive plasmacytoma, plasma
cell granuloma, and lymphoma (MALT, marginal zone, and
immunoblastic) (see Chap. 21). EMP occurs mostly in
mucosal sinuses in the head/neck; less frequently it may
affect the gastrointestinal tract, skin, lungs, bladder, thyroid, Figure 22-21 @ Peripheral blood in plasma cell leukemia showing
testis, ovaries, and tonsils. Treatment is radiotherapy just like presence of circulating plasma cells. (From Dutcher, T: Hematology.
Solitary Plasmacytoma of the bone (SPB). The risk of conver- In: Listen, Look, and Learn. Health and Education Resources, Inc,
sion to multiple myeloma is lower, with 50% to 65% of Bethesda, MD, with permission.)
518 Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders
Waldenstrém’s Macroglobulinemia
Waldenstr6m’s macroglobulinemia is a malignant lymphopro-
liferative disorder involving small lymphocytes that exhibit
plasma cell differentiation (plasmacytoid lymphocytes); these
cells produce IgM monoclonal protein. Waldenstrém’s
macroglobulinemia cells express pan-B lymphocyte surface
antigens (CD19, CD20, and CD24), with light chain restric-
tion (mostly k). Chromosomal abnormalities are common in
Waldenstr6m’s macroglobulinemia; the most common is Figure 22-22 @ Plasmacytoid lymphocytes in marrow aspirate
from a patient with Waldenstrom’s macroglobulinemia. (From
deletion of 6q21, which is detected in 42% of Waldenstrém’s Hyun, BH: Morphology of Blood and Bone Marrow. American
macroglobulinemia patients. The bone marrow is extensively Society of Clinical Pathologists. Workshop 5121, Philadelphia,
infiltrated with lymphoid and plasmacytoid cells. Intranuclear September 7, 1983, with permission.)
Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders 519
macroglobulinemia therapy. If severe symptoms are present at Light Chain Deposition and Heavy
diagnosis, IgM may be quickly removed by plasmapheresis,
Chain Diseases
resulting in the rapid improvement in symptoms.”
Light chain deposition disease (LCDD) is similar to primary
amyloidosis; both are clonal plasma cell proliferative disor-
Amyloidosis ders, but the light chain fragments do not form amyloid fibrils
Amyloid is a protein conformation disorder that renders the and while most cases of AL-amyloidosis are related to
protein insoluble and fesults in its deposition in the extra cel- secretion, LCDD is usually k. As with AL-amyloidosis,
lular matrix of various organs. There are many precursor pro- LCDD may be associated with multiple myeloma or other
teins that may result in amyloidosis.” The most common is conditions, such as lymphoma or Waldenstr6m’s macroglob-
AL amyloid (amyloid with presence of light chain secretion); ulinemia. In the kidney, LCDD is often associated with renal
the precursor protein is derived from immunoglobulin light failure, in contrast to AL-amyloidosis, which results in excess
chain fragments. Less common is AA amyloidosis (secondary protein excretion. Treatment is directed at eliminating the
amyloidosis associated with inflammation). This usually plasma cell clone, and if the patient is young and had end-
complicates chronic diseases in which there is ongoing or stage renal disease, renal transplants may be considered.
recurring inflammation, such as rheumatoid arthritis or Heavy chain diseases (HCDs) are very heterogeneous
spondyloarthropathy; chronic infections; or periodic fever group of disorders and do not represent true plasma cell disor-
syndromes. The fibrils are composed of fragments of the ders. They are mentioned here owing to the presence of
acute phase reactant serum amyloid A. Discussion of other abnormal serum immunoglobulin component. These are
familial amyloidosis syndromes is beyond the scope of this usually related to lymphoma, chronic lymphocytic leukemia,
chapter. and marginal zone lymphoma. The B cells in these disorders
Amyloidosis is a rare and potentially fatal disease that exclusively produce monoclonal heavy chains and no light
occurs when insoluble protein fibers are deposited in tissues chains. The heavy chains are divided into three main types:
and organs, impairing their function (e.g., heart, kidneys, y, pw, and a. Patients have systemic symptoms and
liver, spleen, nervous system, and gastrointestinal tract) organomegaly. The details of these disorders are beyond the
(Fig. 22-24). Amyloidosis causes progressive loss of function scope of this book and can be found in reference 78.
of the involved organ. Amyloid patients have a wide range
of presentations depending on the organ involved. Renal
amyloid may present with asymptomatic proteinuria. Cardiac
involvement results in systolic/diastolic dysfunction leading Case Study 1
to heart failure. Mixed sensory and motor peripheral neuropa- Multiple Myeloma
thy and/or autonomic dysfunction is a prominent feature in
many patients with amyloidosis. Skin may provide clues of A 71-year-old man presented with a 6-month history of back
amyloid involvement when thickening, easy bruising, and pain. His wife noted that he was frequently confused, slept
periorbital purpura are seen. Treatment is aimed at eliminating much of the day, and made occasional nonsensical state-
the source of the abnormal precursor protein. In AL-amyloid, ments. On evaluation, he was anemic with a hemoglobin
treatment involves chemotherapy or stem cell transplantation (Hgb) level of 10 g/dL and a mean corpuscular volume
to eliminate the plasma cells, the source of the abnormal light (MCV) of 91 fL. He had a calcium level of 10.6 mg/dL
continued
Chainsy es!
520 Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders
QUESTIONS
QUESTIONS
1. What is the most likely diagnosis for this patient?
1. What major and minor criteria for multiple myeloma are
a. Multiple myeloma
met in this patient?
b. Waldenstr6m’s macroglobulinemia
2. What stage myeloma does he have?
c. Monoclonal gammopathy of unknown significance
3. What is the prognosis of this patient?
d. Non-Hodgkin’s lymphoma
4. What is the best therapy for this patient? What are the
2. Which explanation should be given to the patient?
main toxicities of such therapy?
a. You have an incurable disease, probably a cancer that
has not yet been found. I want to see you back every
ANSWERS 3 months for tests.
1. Major criteria met were (a) more than 30% plasma cells
b. Nothing of concern was found. There is nothing to
in the bone marrow; (b) IgG-k M-spike greater than worry about. Have a nice day.
3.5 g/dL. Minor criteria met were (a) lytic bone lesions; c. Your body is producing some abnormal antibodies. It
(b) low-normal immunoglobulins (see Table 22-6).
is acommon disorder that will probably never cause
2. The patient has clinical Durie Salmon stage HI and you any problems. A small percentage of patients may
international stage II. develop a more serious problem later, so I need to see
3. He will be intermediate risk with a median surival of you back in 3 months, and then periodically to see if
44 months (see Table 22-7). there has been any change.
4, Dexamethasone and thalidomide and/or lemalidomite is 3. What malignancy is likely to occur in this patient?
currently the most widely used regimen in therapy of a. Multiple myeloma
myeloma patients. The main toxicities are dexametha- b. Chronic lymphocytic leukemia
sone related such as hyperglycemia, hypertension and c. Amyloidosis
fluid retention. Thalidomide combinations have been d. Non-Hodgkin’s lymphoma
associated with increased risk of deep venous thrombo- e. All of the above
sis and neuropathy. The patients should receive monthly 4. What follow-up schedule would be appropriate?
infusions of bisphosphonates to prevent skeletal compli- a. Monthly serum protein electrophoresis.
cations such as fractures b. Bone marrow biopsy repeated every 6 months for
2 years. If there is no evidence of myeloma at that
time, the patient does not have to return anymore.
QO . Repeat the complete evaluation every 6 months.
ANSWERS QUESTIONS
1. c. Although a few patients develop multiple myeloma, 1. Which of the following is a possible cause of the
non-Hodgkin’s lymphoma, or chronic lymphocytic patient’s problems in concentrating and reading?
leukemia, most patients with MGUS are not adversely a. The calcium level is actually high when the low albu-
affected by this disorder. min level is considered.
2a€ b. The increased viscosity of the blood is causing poor
3. All of the above. MGUS patients can develop any lym- circulation through the blood vessels in the brain,
phoid malignancy. resulting in these symptoms.
4. d. Once the initial evaluation is complete, no further test- c. On exposure to cold, cryoglobulins in the patient’s
ing needs to be done unless there is a change in the pro- head precipitate and block blood vessels.
duction of the M-spike immunoglobulin. Three months is d. Osteolytic lesions in the spine.
considered a reasonable interval because of the known 2. Which statement is most correct about the patient’s
growth rate of these disorders. If the patient’s condition diagnosis?
is stable after that time interval, the period of time may a. This is a terminal disease that may end her life within
safely be increased to 6-month follow-ups for life. a year.
b. This is a chronic disorder that often requires treatment,
but she will probably live a relatively normal life.
c. This disease is rapidly fatal and does not respond to
treatment.
‘Case Study 3: d. This is a terminal disease, but the numbers of abnor-
mal cells in the bone marrow suggest that, with
Waldenstrom’s treatment, she may live another 4 or 5 years.
3. If the patient developed more severe neurologic prob-
Macroglobulinemia lems, what could be done to rapidly lower the IgM in
the blood?
A 59-year-old woman presented with a 35-pound uninten-
tional weight loss oyer the past 18 months. She had been ANSWERS
having trouble concentrating at work, had decreased energy,
|. b. Hyperviscosity syndrome is common with Waldenstrém’s
and had two frightening episodes during which she could
macroglobulinemia, and these are classic symptoms. The
not make sense of the words she was reading in a novel.
plasmacytoid lymphocytes may infiltrate nerves, meninges,
On physical examination, she was noted to be a tall, thin
and the brain and may be a less common cause of mental
woman with no enlarged lymph nodes. Her spleen was
status changes and neurologic changes. Hypercalcemia is
enlarged, and easily felt approximately 7 cm below her left
uncommon with Waldenstr6m’s macroglobulinemia, and
ribs. Her neurologic examination was normal. Blood tests
correcting the albumin to 4 g/dL (1.3 g/dL increase) would
revealed a Hgb of 9.7 g/dL, hematocrit (Hct) of 29.2%, and
result in a correction of serum calcium level of 0.8 <
MCV of 89 fL. Platelet count, white cell count, and white
1.3 = 1.04 mg/dL, totaling 9.34 mg/dL. This is within the
cell differential were normal. A chemistry panel was normal
normal range. Chilling of the scalp would not result in pre-
with the exception of a total protein level of 9.7 g/dL and a
cipitation of cryoglobulins in the brain, but might affect
low albumin of 2.7 g/dL. The calcium level was 8.3 mg/dL.
the tips of the ears and nose.
A serum protein electrophoresis was ordered and revealed a
2. d. The prognosis of patients with Waldenstrém’s
5.7 g/dL M-spike.
macroglobulinemia is associated with extent of plasma-
Immunoelectrophoresis identified the protein as an IgM-k.
cytoid lymphocytes in the bone marrow. This patient
The bone marrow was hypercellular with 23% plasmacytoid
may live for 3 to 4 years after diagnosis.
lymphocytes consistent with Waldenstr6m’s macroglobu-
3. Plasmapheresis is useful in removing IgM-rich plasma
linemia. Blood viscosity was measured and found to be
from the patient within | to 2 hours and may result in
moderately increased.
dramatic resolution of symptoms.
continued
= P19) Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders
f). esncone
r Which laboratory test(s) provide the most important prog- a. IgM M-spike and hyperviscosity
nostic information in multiple myeloma? b. Lytic:bone lesions and rouleaux
a. Hemoglobin c. Renal failure and more than 30% plasma cells
b. Albumin d. M-spike of IgM, IgG, or IgA; low-normal levels of
c. B.-microglobulin other immunoglobulins; inability to make light chains
d. All of the above 4. What does staging refer to?
bo. Which list would contain diagnostic criteria for multiple a. Bone marrow compatibility
myeloma? b. Estimate of disease severity
a. Biopsy-proven plasmacytoma and 10% to 30% plasma c. Chemotherapy regimen
cells d. Determination of specific immunoglobulin in heavy-
b. More than 30% plasma cells in bone marrow chain disease
c. Biopsy-proven plasmacytoma and M-spike present
d. Monoclonal protein and M-spike present See answers at the back of this book.
. Which of the following would be diagnostic criteria for
Waldenstr6m’s macroglobulinemia?
REFERENCES Evidence of a genetic association. Sie). Alexanian, R, et al: Treatment for multi-
Cancer Epidemiol Biomarkers Prev ple myeloma. Combination chemother-
Il. Shapiro-Shelef, M, and Calame, K: Reg-
eiW7, WED. apy with different melphalan dose
ulation of plasma-cell development.
Dale Durie, BG: The epidemiology of multi- regimens. JAMA 208:1680, 1969.
Nat Rev Immunol 5:230, 2005.
ple myeloma. Semin Hematol 38:1, 40. Barlogie, B, et al: Effective treatment of
. De Raeve, HR, and Vanderkerken, K:
2001. advanced multiple myeloma refractory to
The role of the bone marrow microenvi-
DOP Morgan, GJ: Myeloma aetiology and alkylating agents. N Engl J Med
ronment in multiple myeloma. Histol
epidemiology. Biomed Pharmacother 310:1353, 1984.
Histopatho! 20:1227, 2005.
56:223, 2002. 4]. Attal, M, et al: A prospective, random-
. Sutton, BJ: Immunoglobulin structure
. Zappasodi, P, et al: Changes in multiple ized trial of autologous bone marrow
and function: the interaction between
myeloma epidemiology in the last thirty transplantation and chemotherapy in
antibody and antigen. Curr Opin
years: a single centre experience. multiple myeloma. Intergroupe Francais
Immunol 2:106, 1989.
Eur J Cancer 42:396, 2006. du Myelome. N Engl J Med 335:91,
. Kyle, RA: Sequence of testing for mono-
. Kyle, RA, et al: Review of 1027 patients 1996.
clonal gammopathies. Arch Pathol Lab
with newly diagnosed multiple 2. Desikan, R, et al: Results of high-dose
Med. 123:114, 1999.
myeloma. Mayo Clin Proc 78:21, 2003. therapy for 1000 patients with multiple
Nn. O'Connell, TX, et al: Understanding and
. Yasui, H, et al: Novel therapeutic strate- myeloma: Durable complete remissions
interpreting serum protein electrophore-
gies targeting growth factor signalling and superior survival in the absence of
sis. Am Fam Physician 71:105, 2005.
cascades in multiple myeloma. chromosome 13 abnormalities. Blood
Oo. Myara, I: [Current role of the laboratory
Br JHaematol 132:385, 2006. 95:4008, 2000.
in the demonstration and follow up of
. Stewart, JP, and Shaughnessy, JD, Jr: . Attal, M, et al: Single versus double
monoclonal immunoglobulinopathy].
Role of osteoblast suppression in multi- autologous stem-cell transplantation for
Pathol Biol (Paris). 38:134, 1990.”
ple myeloma. J Cell Biochem 98:1, 2006. multiple myeloma. N Engl J Med
—. Bradwell, AR, et al: Serum test for
. Tricot, G: New insights into role of 349:2495, 2003.
assessment of patients with Bence Jones
microenvironment in multiple myeloma. 44. Barlogie, B, et al: Thalidomide and
myeloma. Lancet 361:489, 2003.
Lancet 355:248, 2000. hematopoietic-cell transplantation for
. Nowrousian, MR, et al: Serum free light
. Ganeval, D, et al: Proteinuria in multiple multiple myeloma. N Engl J Med
chain analysis and urine immunofixation
myeloma and related diseases. Am J 354:1021, 2006.
electrophoresis in patients with multiple
Nephrol 10(Suppl 1):58, 1990. 45. Aschan, J, et al: Graft-versus-myeloma
myeloma. Clin Cancer Res. 11:8706, . Pantou, D, et al: Cytogenetic manifesta- effect. Lancet 348:346, 1996.
2005.
tions of multiple myeloma heterogeneity. 40. Badros, A, et al: High response rate in
. Drayson, M, et al: Serum free light-chain
Genes Chromosomes Cancer 42:44, refractory and poor-risk multiple
measurements for identifying and moni-
2005. myeloma after allotransplantation using
toring patients with nonsecretory multi-
. Gertz, MA, et al: Clinical implications of a nonmyeloablative conditioning regi-
ple myeloma. Blood 97:2900, 2001. t(11;14)(q13;q32), t(4;14)(p16.3;q32), men and donor lymphocyte infusions.
. Chauveau, D, and Choukroun, G: Bence
and -17p13 in myeloma patients treated Blood 97:2574, 2001.
Jones proteinuria and myeloma kidney. 47. Crawley, C, et al: Outcomes for reduced-
with high-dose therapy. Blood.
Nephro! Dial Transplant 11:413, 1996. 106:2837, 2005. intensity allogeneic transplantation for
. Mehta, J, and Singhal, S: Hyperviscosity . Fonseca, R, et al: Clinical and biologic multiple myeloma: An analysis of prog-
syndrome in plasma cell dyscrasias. implications of recurrent genomic aberra- nostic factors from the Chronic
Semin Thromb Hemost 29:467, 2003. tions in myeloma. Blood 101:4569, 2003. Leukaemia Working Party of the EBMT.
. Kyrtsonis, MC, et al: Mechanisms of . Bergsagel, PL, and Kuehl, WM: Molecu- Blood 105:4532, 2005.
polyclonal hypogammaglobulinaemia in lar pathogenesis and a consequent classifi- 48. Lee, CK, et al: Clonal cytogenetic
multiple myeloma (MM). Med Oncol cation of multiple myeloma. J Clin Oncol changes and myeloma relapse after
KOS, 1999! 23689552005: reduced intensity conditioning allogeneic
. Mouthon, L, and Lortholary, O: Intra- transplantation. Bone Marrow Transplant
. Mulligan, ME: Imaging techniques used
venous immunoglobulins in infectious in the diagnosis, staging, and follow-up SHSM IL, ZOOS.
diseases: Where do we stand? Clin of patients with myeloma. Acta Radiol 49. Richardson, P, and Anderson, K:
Microbiol Infect 9:333, 2003. 46:716, 2005. Immunomodulatory analogs of thalido-
. Morra, E: Cryoglobulinemia. Hematol- 34. Angtuaco, EJ, et al: Multiple myeloma: mide: an emerging new therapy in
ogy (Am Soc Hematol Educ Program). Clinical review and diagnostic imaging. myeloma. J Clin Oncol 22:3212,
368, 2005. Radiology 231:11, 2004. 2004.
. Gertz, MA: Cold agglutinin disease and . Moulopoulos, LA, et al: Prognostic sig- . Zangari, M, et al: Thrombogenic activity
cryoglobulinemia. Clin Lymphoma nificance of magnetic resonance imaging of doxorubicin in myeloma patients
5:290, 2005. of bone marrow in previously untreated receiving thalidomide: Implications for
. Kyle, RA, et al: A long-term study of patients with multiple myeloma. Ann therapy. Blood 100:1168, 2002.
prognosis in monoclonal gammopathy of Oncol 16:1824, 2005. Syl. Palumbo, A, et al: Oral melphalan and
undetermined significance. N Engl J 36. Smith, A, et al: Guidelines on the diagno- prednisone chemotherapy plus thalido-
Med 346:564, 2002. sis and management of multiple myeloma mide compared with melphalan and
. Kyle, RA, and Rajkumar, SV: Mono- 2005. Br J Haematol 132:410, 2006. prednisone alone in elderly patients
clonal gammopathies of undetermined . Durie, BG, and Salmon, SE: A clinical with multiple myeloma: Randomised
significance: A review. Immuno! Rev staging system for multiple myeloma. controlled trial. Lancet 367:825, 2006.
194:112, 2003. Correlation of measured myeloma cell . Anderson, KC: Bortezomib therapy for
. Kyle, RA, and Rajkumar, SV: Multiple mass with presenting clinical features, myeloma. Curr Hematol Rep 3:65, 2004.
myeloma. N Engl J Med 351:1860, response to treatment, and survival. . Richardson, P, and Anderson, K:
2004. Cancer 36:842, 1975. Thalidomide and dexamethasone: A new
_ Jemal, A, et al: Cancer statistics, 2006. . Greipp, PR, et al: International staging standard of care for initial therapy in
CA Cancer J Clin 56:106, 2006. system for multiple myeloma. J Clin multiple myeloma. J Clin Oncol 24:334,
. Pottern, LM, et al: HLA and multiple Oncol 23:3412, 2005. 2006.
myeloma among black and white men:
524 Chapter 22 Multiple Myeloma and Related Plasma Cell Disorders
7
. Hideshima, T, et al: Current therapeutic 63. Weber, DM: Solitary bone and . Ghobrial, IM, and Witzig, TE:
—
Lipid (Lysosomal)
Storage Diseases and
Histiocytosis
Denise M. Harmening, PhD, MT(ASCP), CLS(NCA)
Catherine M. Spier, MD
Lipid (Lysosomal) Storage Diseases Lipid storage diseases have a wide clinical expression,
ranging from essentially asymptomatic to severe and inca-
The lipid storage diseases are rare, autosomally inherited disor- pacitating with early death. The aim of control in these
ders. They are known as lysosomal storage diseases because disorders has been directed at prenatal detection. The only
there is subcellular accumulation of unmetabolized material in currently effective practical therapy is enzyme replacement,
the lysosomes of various cells. Lipid or lysosomal storage dis- which has initiated a new age of treatment for genetic dis-
eases are caused by various enzyme defects (inborn errors) in orders and has improved the lives of many patients. The
lipid metabolism linked to an enzyme deficiency (Fig. 23-1). greatest controversy regarding enzyme replacement ther-
Although many different types of lipid storage disorders have apy, however, is the optimum amount and frequency of
been documented, the most widely known and well established treatment.' In addition, allogeneic bone marrow transplan-
include Gaucher’s, Niemann—Pick, and Tay—Sachs diseases tation has been used to treat some patients and can be con-
and mucopolysaccharidoses (Table 23-1). Although all ethnic sidered curative.* However, bone marrow transplantation is
groups are known to be affected by lipid storage diseases, an extremely aggressive, expensive, and high-risk therapy
there is an increased incidence of selected disorders such as considering that these conditions now have an effective and
Gaucher’s and Tay-Sachs diseases in certain ethnic groups, practical treatment in enzyme replacement therapy. The
most notably Ashkenazi Jews (Jews who trace their origin to general characteristics of lipid storage diseases are summa-
the Baltic Sea region). rized in Table 23-2.
Bos)
526 Chapter 23 Lipid (Lysosomal) Storage Diseases and Histiocytosis
GLOBOSIDE
Ceramide
Hexosaminidase A & B
(Deficiency — Sandhoff’s Disease)
a-Galactosidase
(Deficiency — Fabry’s Disease)
B-Galactosidase
(Deficiency — Lactosy! ceramidosis)
B-Glucosidase
(Deficiency — Gaucher’s Disease)
GANGLIOSIDE
Ceramide N-Acetylneuraminic acid
Hexosaminidase A
(Deficiency — Tay-Sachs)
B-Galactosidase
(Deficiency > Lactosy! ceramidosis)
B-Glucosidase
(Deficiency — Gaucher's Disease)
Figure 25-1 ™ Schematic structure of globoside and ganglioside to show site of action of the several catabolic enzymes, which, when defec-
tive, result in one of the storage diseases. (From Wintrobe, MM, et al: Clinical Hematology, ed 8. Lea & Febiger, Philadelphia, 1981, p 1341,
with permission.)
Gaucher’s Disease
Table 23-1 Lipid Storage Diseases
HISTORICAL PERSPECTIVES
Gaucher’s disease This disorder was first described in 1882 by Philippe C.
Niemann—Pick disease
Gaucher in a 32-year-old woman with an enlarged spleen.
Tay-Sachs disease
Mucopolysaccharidoses Gaucher believed that the abnormal cells found in her spleen
at autopsy were part of a primary splenic tumor. This abnormal
cell, later named Gaucher's cell, is the result of the deficiency
of the enzyme beta ()-glucocerebrosidase, which leads to an
accumulation of unmetabolized substrate glucocerebroside in
cells, predominantly the monocyte-macrophage system (the
“4 Table 23-2 General Characteristics | reticuloendothelial system; Fig. 23-2). Gaucher’s observa-
. of Lipid Storage | tions were studied further, and the entity known as Gaucher’s
Diseases disease was defined and characterized as a familial disorder at
the turn of the last century.* In 1920, another variation or type
¢ Rare, inherited autosomal-recessive disorders } of Gaucher’s disease that is characterized by neurologic
¢ Also known as lysosomal storage diseases because of involvement was first described. It was after this date that
accumulation of unmetabolized material in lysosomes
Gaucher’s disease was classified as a lysosomal storage disor-
* Caused by enzyme deficiencies in lipid metabolism
* Increased incidence of some lipid storage diseases in der resulting from an enzyme deficiency with an autosomal-
certain ethnic groups (i.e., Gaucher’s disease in Ashkenazi recessive inheritance pattern. Although Gaucher’s disease is
Jews) the most frequent lysosomal storage disease, it was not until
* Great variation in clinical expression (i.e., asymptomatic to | 1965 that the actual enzyme deficiency was identified as glu-
severe with early death) cocerebrosidase. In 1984 the gene for glucocerebrosidase was
¢ Effective therapy: enzyme replacement
cloned, and in 1991 an important breakthrough occurred with
* Most well-known and characterized: Gaucher’s disease,
Niemann-—Pick disease, Tay—Sachs disease, and the initiation of clinical trials for enzyme replacement therapy
mucopolysaccharidoses at the National Institutes of Health (NIH).? Gaucher’s disease
became the first enzyme-deficiency disorder to be successfully
Chapter 23 Lipid (Lysosomal) Storage Diseases and Histiocytosis BPA
Hepatosplenomegaly
Gaucher’s cells in the bone marrow
Increase in serum acid phosphatase
(bone crises/fractures)
Neurodegenerative course os +++ ++
Life expectancy 6-80+ years 2 years 20-40 years
Ethnic predilection Ashkenazi Jew Panethnic Swedish (Norrbottnian)
2. The moderate clinical presentation, in which the patient has than 40 genetic mutations have been described.' The muta-
hepatosplenomegaly, near-normal blood counts, and a nor- tions include both single insertional and point mutations as
mal physical appearance well as crossover mutations. All of the mutations that cause
3. The most severe form, in which patients present with mas- Gaucher’s disease have complex effects on the properties of
sive hepatosplenomegaly, significant thrombocytopenia, this enzyme.'' The normal enzyme, glucocerebrosidase (acid
anemia, and skeletal complications.'° 8-glucosidase), is a lysosomal enzyme responsible for the
degradation of the AMES SOS EMS molecule, preventing its
A few patients with the severe forms of the disease may
buildup in tissue cells.'* Protein synthesis of the normal
develop central nervous system damage, delayed sexual mat-
enzyme occurs in the endoplasmic reticulum, with transport
uration, severe wasting, and eventually die. It should be noted
to the Golgi apparatus for glycosylation and delivery to the
that clinical presentations can be misleading because some of
lysosomes of the cell. Mutations at the genetic level that code
these patients may still develop underlying skeletal complica-
for the production of this enzyme have direct effects on the
tions (Table 23-7). It is important, therefore, to perform a
catalytic activity, with decreases from 5- to 100-fold.'* In
baseline skeletal evaluation even in patients with a mild case
addition, enzyme stability and half-life activity (normal is
of the disease. The femoral head is the most common initial
60 hours) are also decreased for acid B-glucosidase as a result
bone site to be affected by the disease, and this area should be
of these mutations.'*
evaluated by magnetic resonance imaging (MRI) to assess for
avascular necrosis.”
The definitive diagnosis of Gaucher’s disease is made TYPE If: INFANTILE GAUCHER’S DISEASE (ACUTE OR
with an assay for the enzyme acid f-glucocerebrosidase MALIGNANT NEURONOPATHIC) Type If Gaucher’s disease
activity of the leukocytes. Bone marrow examination alone is is a much rarer form that occurs in infancy, and patients rarely
insufficient for confirming the diagnosis because Gaucher’s survive past the age of 2 years. Type II acute neuronopathic
cells may be present in other disorders. Gaucher’s disease is seen in all ethnic groups, although it is
The gene for the enzyme glucocerebrosidase is located uncommon in the Jewish population.” The frequency of type II
on chromosome 1q21—31. Since the characterization, cloning, disease is estimated at approximately | in 100,000.? The hall-
and sequencing of the glucocerebrosidase gene in 1984, more mark of type Il Gaucher’s disease is neurologic involvement,
Stage Bescon ®
LABORATORY DIAGNOSIS
Figure 25-3 ™ Anteroposterior radiograph of the knee shows
Peripheral blood, bone marrow, and spleen are the sites most
diffuse mottled increased density of the distal femur and proximal
tibia, characteristic of widespread bone infarction in Gaucher's frequently examined in patients with Gaucher’s disease. The
disease. The metaphyseal regions are broader than normal (arrow), peripheral blood nearly always demonstrates a moderate
resembling an Erlenmeyer flask deformity. (Courtesy of Charles S. normocytic, normochromic anemia with thrombocytopenia
Resnik, MD, Department of Diagnostic Radiology, University of because of the replacement of normal hematopoietic cells
Maryland Medical Center, Baltimore, MD.)
with Gaucher’s cells in the bone marrow. There is pooling of
blood in the enlarged spleen and some degree of ineffective
erythropoiesis, with decreased incorporation of iron in
including multiple signs such as difficulty swallowing, erythroid precursors in the bone marrow. As a result, active
opisthotonos (extreme arching of the spine), and other mani- signs of a compensated anemia such as polychromasia and
festations of brain stem involvement that are noted early in nucleated red blood cells are usually absent on the peripheral
infancy.'> The infant has difficulty in feeding and fails to grow. smear.'* Leukocytes are commonly decreased in number.
Death usually occurs before the age of 2 years. Familial inter- Platelets are also usually decreased in number as a result
marriage is frequently found in the infant’s family history. of splenic sequestration. These patients may have a bleeding
The clinical presentation of this type II disease is much tendency, with nosebleeds especially common. Gaucher’s
more uniform than that observed in type I Gaucher’s dis- cells are noted only rarely in the peripheral blood. Bone mar-
ease and is very severe. The disease exhibits a progressive row aspirates are often the first tissue in which Gaucher’s
pattern of clinical symptoms, with hepatosplenomegaly cells are detected; these cells are required for diagnosis (see
evident within the first 6 months of life, and is often discov- Fig. 23-2). They are histiocytes, 20 to 100 um in diameter,
ered by 3 months of age. The principle cause of death in found in moderate numbers and as clumps of cells in the
infants with type II Gaucher’s disease is brain stem thickest areas of the smear. One or more round to oval nuclei
damage. are present in each cell. The cytoplasm is faintly blue with
Wright’s stain and has a “crumbled tissue paper” or finely The excess is therefore stored in histiocytes, with their end
folded appearance, possibly as a result of glycolipid deposi- morphological expression identical to that of true Gaucher’s
tion. Electron microscopy has demonstrated that this appear- cells. This phenomenon is also seen in a variety of other
ance is the result of lamellar bodies stacked inside secondary disorders, including acute myelocytic leukemia, chronic lym-
phagolysosomes. These cells stain positive with periodic phocytic leukemia, plasma cell myeloma, aplastic anemia,
acid-Schiff (PAS), acid phosphatase, Giemsa, iron, Sudan idiopathic thrombocytopenic purpura, thalassemia major, and
black B, and oil red O stains because of the accumulation of rheumatoid arthritis'? (Table 23-10). The presence of Gaucher-
the unmetabolized glucocerebroside. It is important to note like cells in patients with these diseases has no known prognos-
that the presence of Gaucher’s cells is not pathognomonic for tic significance. It should be emphasized that in each of these
Gaucher’s disease, because these cells are also found in other diseases there is no deficiency of the B-glucocerebrosidase, as
lymphoproliferative disorders. there is in Gaucher’s disease, but rather an overtaxation of a
The spleen is variably enlarged, owing to the accumula- normal system.
tion of masses of Gaucher’s cells. The enlargement is com-
monly up to 10 times normal splenic weight and can cause PROGNOSIS
considerable discomfort to the patient. Other organs and sys- As previously stated, the length of survival in patients with
tems commonly affected include the liver and, in type II, the Gaucher’s disease is variable and depends on the type. The
nervous system, pituitary gland, kidneys, lung, and ovaries. adult form (type I) has the longest survival, with patients sur-
These organs contain massive deposits of Gaucher’s cells. viving commonly into adulthood. In the infantile form (type ID),
The serum acid phosphatase level is increased, and survival beyond 2 years of age is rare. Like the clinical fea-
isozyme measurement of this enzyme has shown that the tures, survival in the juvenile form (type II) is intermediate
tartrate-resistant fraction is what is increased in patients with between the first two, and patients usually live into adoles-
Gaucher’s disease. The laboratory findings in Gaucher’s dis- cence. A relatively increased risk of cancer in patients with
ease are summarized in Table 23-9. Gaucher’s disease has been reported,*? primarily because of
Although the Gaucher’s cell is associated with the an increased incidence of hematologic malignancy. Patients
disease, so-called pseudo-Gaucher’s cells have also been with Gaucher’s disease are 15 times more likely to develop a
described. They are seen in disease states with increased cel- hematologic malignancy than a healthy individual. The most
lular turnover, especially chronic myelogenous leukemia, in frequently reported hematologic malignancies are listed in
which the phenomenon was first described. In theory, the Table 23-11. This increased risk of malignancy may be asso-
increased cell turnover presents so much glycosylceramide to ciated with immunologic abnormalities found in patients
the reticuloendothelial system that its enzyme system is over- with Gaucher’s disease. These abnormalities include increased
whelmed and cannot adequately metabolize all of the material. helper-to-suppressor (T,:T;) cell ratio, decreased natural killer
Niemann-Pick Disease
This inherited form of lipid storage disease was first described
in 1914 by Niemann and subsequently by Pick in 1933.?’
Niemann-Pick disease is caused by a deficiency of the
enzyme sphingomyelinase, with a secondary accumulation of
the unmetabolized lipid sphingomyelin as well as cholesterol.
Chronic lymphocytic leukemia (CLL) Sphingomyelin is a sphingophospholipid that is a common
Hodgkin’s disease and non-Hodgkin’s lymphoma
constituent of cell membranes as well as cellular organelles.
Acute leukaemia
As a result, a deficiency of sphingomyelinase is a serious dis-
order. The general characteristics of Niemann—Pick disease
are summarized in Table 23-13.
(null) cells, polyclonal B-cell lymphocytosis, and plasmacyto- A wide variety of clinical manifestations of variable sever-
sis.°° The immunologic abnormalities described in Gaucher’s ity have been reported in patients with Niemann-Pick disease.
disease are listed in Table 23-12. These include growth retardation, hepatosplenomegaly, lym-
phadenopathy, pigmentation, and impaired neurologic functions
TREATMENT (Table 23—14).** A large number of lipid-laden giant foam cells
Before the advent of the newer treatment modality of known as Niemann—Pick cells can be found in affected tissues
enzyme replacement therapy, Gaucher’s disease was tradi- and organs (Fig. 23-4). The detection of Niemann—Pick cells in
tionally managed by supportive therapy. Total or partial patients with this disorder is essential for the diagnosis of this
splenectomy was frequently performed. In addition, trans- disease. There is an increased incidence of Niemann—Pick dis-
fusions, orthopedic procedures, and occasionally bone mar- ease in the Jewish population, especially in consanguineous
row transplantation were used in some patients. Although groups. Because of the very different clinical manifestations of
potentially curative, allogeneic bone marrow transplanta- the disease, five types, A through E, have been described.” Only
tion is an extremely aggressive and high-risk therapy. In types A, B, and C are discussed here. Type E, which is very
1991, a major advancement occurred in the treatment of rare, has been found only in adults and is characterized by a
Gaucher’s disease type I. The U.S. Food and Drug Admin- mild chronic course and a lack of neurologic manifestations.
istration approved the use of enzyme replacement therapy Types A, B, and C of Niemann—Pick disease are compared in
for this disorder. Gaucher’s disease is the first lysosomal Table 23-15.
storage disorder for which enzyme replacement therapy is avail-
able. Enzyme replacement therapy has successfully reversed CLASSIFICATION AND CLINICAL FEATURES
many of the clinical complications of this disorder, including
correcting blood counts and reducing the organomegaly that TYPE A This form is also known as infantile or classic
occurs in these patients.?!*° Niemann—Pick disease. \t is the most common form, account-
The first enzyme replacement therapy utilized was a puri- ing for up to 85% of all cases of Niemann—Pick disease. The
fied enzyme from human placenta. This enzyme, which is an onset is early in infancy and is associated with failure to
alglucerase injection, is manufactured by Genzyme Corporation thrive, difficulty feeding, and retarded physical and mental
as Ceredase and has demonstrated effectiveness by the reversal development. The skin has a waxy consistency. There 1s often
of signs and symptoms of Gaucher type I, non-neuronopathic jaundice at birth and, usually, hepatosplenomegaly with a dis-
disease.* Another form of the enzyme, the recombinant form, tended abdomen. The lymph nodes are enlarged as well.
which is also produced by Genzyme Corporation as Cerezyme, A cherry-red spot in the macula of the eye 1s found in approx-
is genetically engineered and has the advantage of being unlim- imately 50% of the affected infants. The neurologic symp-
ited in supply. In addition, the recombinant Cerezyme has the toms are more pronounced in this type of Niemann—Pick
advantage of a very low risk of transmitting any infectious agent disease than in any of the other types. Deterioration is rapid,
and also has a lower rate of patients developing IgG antibodies and survival past the age of | or 2 years is rare.
to the glucocerebrosidase enzyme.”
Niemann-Pick cells and sea-blue histiocytes (histiocytes dis- deficiency of the enzyme hexosaminidase A, with an increase
tended with blue-staining ceroid on Wright’s stain). It is of the other isoenzyme, hexosaminidase B. The gene for
believed that the sphingomyelin is gradually metabolized to hexosaminidase A is located on chromosome 15.** Inheritance
ceroid, thus generating the sea-blue histiocytes. A marrow of two abnormal alleles (one from each parent) accounts for
specimen with these findings would then need to be distin- almost all infantile Tay-Sachs cases in the Ashkenazi-Jewish
guished from the entity of sea-blue histiocytosis (see the sec- population. The severity of the disease correlates with the
tion at the end of this chapter). level of residual enzyme activity. The unmetabolized GM,
Other disorders that may cause Niemann—Pick-like cells ganglioside accumulates in almost all tissues and has its most
in the bone marrow are GM, gangliosidosis, lactosyl cerami- devastating effects within the central nervous system and
dosis, and Fabry’s disease. eye. The general characteristics of Tay-Sachs disease are
The peripheral blood is most remarkable for the vacuoles summarized in Table 23—16.*°
that may be found in lymphocytes and monocytes of a routine
peripheral blood smear (Fig. 23-5). These vacuoles are round, CLINICAL FEATURES
and from 2 to 20 may be found within one cell. Anemia and Although affected infants appear normal at birth, by 6 months
leukopenia may be present but do not usually present any of age both physical and mental deterioration are notable.
threat to the patient. Serum lipids are not usually increased. They have an exaggerated physical response to noise (the
An assay of the enzyme sphingomyelinase activity in leuko- startle reflex) beginning at age 3 to 5 months. There is a pro-
cytes and fibroblasts can also be performed. gressive loss of motor function with weakness, decreased
attentiveness to surroundings, hypotonia (diminished tone of
PROGNOSIS AND TREATMENT skeletal muscles), and poor head control between the ages
There may be a slightly longer survival in patients with the other of 6 and 10 months.” In addition, a cherry-red spot in the
types of Niemann-Pick disease, but those with type A have a macula of each eye is found; this is the most characteristic
very short life expectancy. Survival past the age of 2 years is feature of Tay-Sachs disease. The central nervous system
uncommon. Currently there is no treatment for Niemann—Pick steadily degenerates after 1 year of age. Along with the con-
disease. However, successful allogeneic bone marrow trans- tinual deterioration, there is enlargement of the head (macro-
plants have been reported for type B.'* In addition, research cephaly), seizures, and paralysis. Spasticity with hyperactive
studies have focused on finding a source of enzyme replacement reflexes, deafness, and blindness follow. The neurons are
for sphingomyelinase*' and gene therapy using retroviruses.” greatly enlarged by accumulation of the unmetabolized
ganglioside in vacuoles in the cytoplasm. In contrast to many
other lipid storage diseases, the spleen, liver, and lymph
Tay-Sachs Disease nodes are not enlarged. Feeding is poor, and death occurs by
4 years of age. It should be noted that cherry-red spots are not
Also known as GM, gangliosidosis, Tay—Sachs disease was first
pathognomonic for Tay—Sachs disease; however, this clinical
described in 1881 by the British ophthalmologist Warren Tay.
finding in a Jewish infant with the absence of organomegaly
In 1886, the New York neurologist Bernard Sachs used the
is strongly suggestive of Tay—Sachs disease.*°
term familial amaurotic infantile idiocy to describe this disor-
der. Its incidence in the Ashkenazi Jewish population is
LABORATORY DIAGNOSIS
100 times greater than that in the non-Jewish population.” It
A deficiency of hexosaminidase A is the basic cause of this
is estimated that this high-risk group has a | in 30 carrier rate.
disease. Hexosaminidase A is the enzyme responsible for
This autosomal-recessive sphingolipidosis is the result of a
hydrolyzing GM, ganglioside, the glycolipid that accumulates
in neurons. This deficiency can be demonstrated in the serum,
plasma, leukocytes, and cultured fibroblasts of infants with
Tay-Sachs disease.
(Sanfilippo’s A)
(Sanfilippo’s B)
(Sanfilippo’s C)
(Sanfilippo’s D)
(wide range of
severity)
MPS IVA Autosomal- Keratan sulfate N-Acetylgal- Normal intelligence, 3rd—6th decade
(Morquio’s) recessive Chondroitin actosamine, severe skeletal
(wide range sulfate 6-sulfate deformities,
of severity) sulfate dwarfism,
thoracolumbar
gibbus
8-Galactosidase Kyphoscoliosis, facies
similar to Hurler’s
syndrome, corneal
clouding, valvular
and coronary artery
lesions, joint
hypermobility,
genu yalgum
MPS IV B
(Morquio’s)
MPS VI Autosomal- Dermatan N-Acetylgalactosamine Similar to Hurler’s 2nd decade
(Maroteaux— recessive sulfate 4-sulfatase, syndrome, but
Lamy) (arylsulfatase B) normal intelligence,
longer survival
MPS VII Autosomal- Dermatan 8-Glucuronidase Variable from severe Variable,
(Glucuronidase recessive sulfate mental retardation 1-40 years
deficiency with dysostosis
disease) multiplex and
hepatosplenomegaly
to a milder form;
also severe neonatal
form with hydrops
fetalis
Source: Modified from Fensom, AH, and Benson PF: Recent advances in the prenatal diagnosis of the mucopolysaccharidoses.
Prenatal Diagn 14:1, 1994.
is only temporary. These individuals are abnormally short and common. Patients affected with Sanfilippo’s syndrome
have coarse facial features, with a broad, flat nose, widely (MPS III) have a more normal stature but unfortunately many
spaced eyes, and thickened tongue and lips.*® Some authors more severe neurologic problems and decreased survival.
have described their appearance as similar to that of a gar- Compared with patients with Hurler’s syndrome, those with
goyle (the carved heads sometimes found on older European Scheie’s syndrome (MPS I S) have more prominent corneal
churches). The amount of body hair is increased, dark, and clouding but less abnormality in stature, facial appearance,
especially prominent on the forehead. The skin is thickened. and mental development. Patients with Maroteaux—Lamy
Patients are mentally retarded. Clouding of the corneas of the syndrome (MPS VI) have growth and skeletal abnormalities
eyes 1s present. These individuals may have hearing loss or be but no mental retardation. In Morquio’s syndrome (MPS IV),
completely deaf. The heart is damaged, owing to the accumu- patients have numerous skeletal changes, giving a markedly
lation of mucopolysaccharides in the valves and blood abnormal physical appearance; however, there is no mental
vessels. There is a hump on the back and a prominent retardation.**
abdomen, with enlarged liver and spleen. The arms and legs
are abnormal, with contractures of many joints. In addition, LABORATORY DIAGNOSIS
the hands are very wide and the fingers shortened. An accurate enzymatic diagnosis should be established for
In Hunter’s syndrome (MPS II), the changes are similar, all suspected cases of MPS, as clinical diagnosis alone is
although not as severe. Corneal clouding is much less often impossible because of overlapping phenotypes. The
Chapter 23 Lipid (Lysosomal) Storage Diseases and Histiocytosis 5 oS)—
diagnosis of MPS can be made by performing simple striking blue color of the histiocytes after staining with
enzyme assays using leukocytes, serum, or fibroblasts.1° Wright’s or May-Griinwald-Giemsa stain gives the syndrome
The identification of heterozygotes, however, is still a dif- its name.
ficult process because of the overlays of normal and The mode of transmission has not been clearly estab-
heterozygous levels of enzyme activity. Molecular studies lished, but autosomal-recessive inheritance, with a variable
such as the cloning of complementary deoxyribonucleic degree of expression, appears most likely. Most patients
acids (cDNAs) can complement accurate enzyme assays.*” receive the diagnosis before they reach 40 years of age. The
Several specialized substrates used for the diagnosis of earlier in life the disease is found, the more severe it is
MPS are now available commercially. likely to be. Major findings on physical examination are
In contrast to findings in the other lysosomal storage splenomegaly and usually hepatomegaly. Also described, but
diseases, nonmetabolized products may be detected in the occurring less consistently, are abnormalities of the eyes,
urine of patients with MPS.*° The toluidine blue spot test or skin, and nervous system. Involvement of the lung may be
the turbidity test to detect acid mucopolysaccharides is the noted on radiographic examination. Involvement of the
initial screening test. The spot test may be unreliable, how- lymph nodes is not seen.
ever, with up to 32% false-negative test results in patients Significant laboratory findings are usually confined to
with Hurler’s syndrome reported. Also of note is that the the blood. In the peripheral blood, thrombocytopenia is found
urine of normal healthy newborn infants may give false- with great frequency. Consequently, clinical manifestations
positive results, a phenomenon that disappears by 2 weeks of such as epistaxis, gastrointestinal tract bleeding, and purpura
age. Any positive screening test result should be confirmed may be expected. However, there is no correlation of the
by lysosomal enzyme assays. degree of thrombocytopenia with the size of the spleen. Blood
An interesting but somewhat inconsistent finding in the lipid levels are normal. Abnormal liver function study results
peripheral blood of patients with MPS is the presence of large are only rarely seen.
granules in leukocytes, especially lymphocytes. These are The bone marrow aspirate is usually the site of diagnosis.
known as Alder-Reilly bodies (see Fig. 23-6). In polymor- Histiocytes of variable size (20 to 60 um) are present in
phonuclear leukocytes this needs to be distinguished from greatly increased numbers. They contain the blue-to-green
toxic granulation, but the large size of the granules in MPS staining granules that vary in size, shape, and ability to take
usually leaves little doubt. A metachromatic stain, such as up the stain (Fig. 23-7). Thus, not all cells will have the same
toluidine blue, aids in confirmation. These granules are found staining intensity. It is not currently known why the granules
with much greater regularity, however, in histiocytes and stain blue with these stains. The cells also react with the PAS,
lymphocytes in the bone marrow. Sudan black B, and acid-fast stains, but not with toluidine
blue or iron stains.”
PROGNOSIS Most patients with this syndrome do well and have
The prognosis of the MPSs varies somewhat with the type. normal life spans. Splenectomy is not always required;
Patients with Hurler’s syndrome may live only one decade, many patients never have the spleen removed. As previ-
whereas those affected with Hunter’s syndrome may live into ously mentioned, manifestations of the disease at an
their 20s.*’ The theoretical aid of enzyme replacement therapy early age may imply more severe symptoms. The general
has yet to be translated into practical results.
TREATMENT
Bone marrow transplantation has been used successfully
in treating some patients with MPS.*' More studies are
needed, however, to prove that bone marrow transplanta-
tion can be a practical treatment leading to a complete
cure. Prenatal diagnosis is still important, including first
trimester diagnosis by chorionic villus sampling, early
amniocentesis for more sensitive lysosomal enzyme assays,
use of DNA analysis for detecting mutations, and the possi-
bility of preimplantation diagnosis of early embryos after
in vitro fertilization.
Histiocytosis
Sea-Blue Histiocyte Syndrome Figure 25-7 ® Sea-blue histiocytes. Note the abnormally coarse
azurophilic granules present in neutrophils, lymphocytes, and
Although initially described in isolated case reports of young monocytes. (From Hyun, BH, et al: Bone marrow. In: Practical
adults with an enlarged spleen, the syndrome of the sea-blue Hematology. A Laboratory Guide with Accompanying Filmstrip.
histiocyte is a genetic disorder with a benign course. The WB Saunders, Philadelphia, 1975, with permission.)
538 Chapter 23 Lipid (Lysosomal) Storage Diseases and Histiocytosis
QUESTIONS
1. This case history is representative of what lipid storage
disease?
2. What further testing must be done to confirm the
diagnosis?
3. Is “effective erythropoiesis” apparent in this case? Why
Autosomal-recessive, benign genetic disorder or why not?
Striking blue histiocytes with Wright’s stain 4. Classify the anemia according to the red blood cell
Splenomegaly and hepatomegaly (RBC) indices.
Thrombocytopenia 5. What treatment is available to this patient?
Eosinophilic Children and young adults, Unifocal—skull, rib, femur most common Rare spontaneous healing;
granuloma especially males, often no most require surgical
symptoms until bone removal; occasional
fracture patients develop
recurrence later
Hand-—Schiiller— Usually <5 years old Multifocal—bones, skin, lymphoid 50%; spontaneous recovery
Christian tissue; triad of pituitary, eye, and skull 50%; recovery with
disease involvement is characteristic but chemotherapy
uncommon
Letterer-Siwe Usually <3 years old Generalized—skin, lymphoid tissue, Chemotherapy has
disease bones, +/— bone marrow; more severe improved prognosis,
and extensive than Hand—Schiiller— which was previously
Christian disease considered poor
Chapter 23 Lipid (Lysosomal) Storage Diseases and Histiocytosis 539
| y
©. stions ne=
1, Whatdefect
isfound in lipid storage diseases? 2 6. What
are the characteristics of Niemann—Pick cells?
a. Subcellular accumulation of unmetabolized material iin a. Atypical lymphocytes with large vacuoles
lysosomes b. Cytoplasm filled with lipid droplets, inconspicuous
b. Cellular accumulation of metabolites in cytoplasm nucleus
c. Protein accumulation in cellular mitochondria c. Vacuolated histiocytes or foam cells
d. Abnormal sequestration of minerals and trace ele- d. Lymphocytes with Alder—Reilly bodies
ments in cellular nuclear organelles 5 . What is the enzyme deficiency seen in Tay-Sachs disease?
2. What is the enzyme deficiency seen in Gaucher’s disease? a. Sphingomyelinase
a. Sphingomyelinase b. Hexosaminidase A
b. Hexosaminidase A c. B-Glucocerebrosidase
c. B-Glucocerebrosidase d. 8-Galactosidase
Se eOaiae iyidise 8. What are the clinical features of Tay—Sachs disease?
3. Which description best characterizes type I Gaucher’s a. Waxy, jaundiced skin; retarded physical and mental
disease? development; cherry-red spot in macula of eye
a. Found in any ethnic group; multiple neurologic signs, b. Startle reflex; blindness; macrocephaly; no enlarge-
including difficulty in swallowing and manifestations ment of liver, spleen, or lymph nodes
involving brain stem; enlargement of liver and spleen c. Abnormal facial features; deafness; increased body
b. Found primarily in Ashkenazi Jews; enlargement of hair, mental retardation; heart damage; structural
liver and spleen; anemia thrombocytopenia deformities
c. Found in northern Sweden; neurologic disorders, bone d. Splenomegaly; hepatomegaly; eye, skin, nervous
disorders, skin pigment changes system, and lung abnormalities
d. Found in Mediterranean populations; hypermetabolic 9. Which cell is found in Tay-Sachs disease, but is not
manifestations;
risers fever, lethargy, poor musculature, considered diagnostic?
bone deformities
a. Atypical lymphocytes with large vacuoles
4. What are the characteristics of Gaucher’s cells? b. Cytoplasm filled with lipid droplets; inconspicuous
a. Atypical lymphocytes with foamy cytoplasm nucleus
b. Hypersegmented neutrophils with Auer’s rods c. Vacuolated histiocytes or foam cells
c. Large, multilobed monocytes with prominent red d. Lymphocytes with Alder—Reilly bodies
granules ' : 10. Which cell may be found in MPS disorders?
d. Histiocytes with blue, folded cytoplasm a. Large, foamy histiocytes with blue or green granules
5. What is the enzyme deficiency seen in Niemann—Pick b. Neutrophils with toxic granulation
disease? c. Neutrophils with Déhle bodies
a. Sphingomyelinase d. Lymphocytes with Alder—Reilly bodies
b. Hexosaminidase A
c. B-Glucocerebrosidase See answers at the back of this book.
_ d. B-Galactosidase
REFERENCES with Gaucher disease. Gaucher Clin 20. Rosenbloom, BE, et al: Gaucher disease
irene mr cole Gaucher disease: New Persp 5:9, 1997. and cancer incidence: a study from the
: ? h an ore poe wane Il. Chaudhuri, TK, and Paul, S: Protein- Gaucher Registry. Blood. 105(12):4569,
Satna 83-748 2001 misfolding diseases and chaperone-based 2005.
pee Nee MC rgSa Veena eee therapeutic approaches. FEBS J 21. Pastores, GM, et al: Therapeutic goals in
7 Se hie ot eee nie Food 273(7):1331, 2006. the treatment of Gaucher disease. Semin
P eee u eee with storage Cis- 12. Bussink, AP, et al: The biology of the Hematol 41(4 Suppl 5):4, 2004.
ee a Sate SAG. gta Gaucher cell: The cradle of human chiti- 22. Weinreb, NJ, et al; International Collabo-
3 AS oe ae esa aes nases. Int Rev Cytol 252:71, 2006. rative Gaucher Group (ICGG): Gaucher
; ee eee Naa era families ILS). Premkumar, ret al:X-ray structure of disease type 1: revised recommendations
Fam Commnnniy Health 30(1):50 3007. human acid-beta-glucosidase covalently on evaluations and monitoring for adult
ABrady. RO: Gaucherand Bibi bd gaeiades bound to conduritol-B-epoxide. Implica- patients. Semin Hematol 41(4 Suppl 5):
; aes are ee ae = tions for Gaucher disease. J Biol Chem 15, 2004.
Ere ea. ee a rha aoe 280(25):23815, 2005. 23. Machado de Paula, MT, et al (eds):
92(443):19 ae) PP 14. Liou B, et al: Analyses of variant acid Gaucher patients treated at HEMORIO.
5 oe ™M ene Seas SARC Oe beta-glucosidases: Effects of Gaucher Gaucher Clin Persp 6:12, 1998.
i ss of A ne iineeli Ate dane. disease mutations. J Biol Chem 23. Pastores, GM, et al: Therapeutic goals in
ree ] eee ne 29(2-3):449 281(7):4242, 2006. the treatment of Gaucher disease. Semin
2006 eeg 15. Mignot, C, et al; French Type 2 Gaucher Hematol 41(4 Suppl 5):4, 2004.
6. Grabowski, GA: Recent clinical progress Disease Study Group. Type 2 Gaucher 24. Starzyk, K, et al: The long-term interna-
a COD eat OS einoun bone disease: 15 new cases and review of the tional safety experience of imiglucerase
17(4):519, 2005 literature. Brain Dev 28(1):39, 2006. therapy for Gaucher disease. Mol Genet
7 Erik a A ; ee T, 3 : 16. Kaye, EM (ed): Type 3 Gaucher disease. Metab 90(2):157, 2007.
ee EE ey ee Gaucher Clin Persp 5:12, 1997. 25. Charrow, J, et al: Enzyme replacement
of enzyme infusion therapy of Norrbot- 17
Ten ee Gauche mows ces 2 Aoki, M, et al: Improvement of neuro- therapy and monitoring for children with
Paediatr 95(3):312, 2006. logical symptoms by enzyme replace- type | Gaucher disease: Consensus recom-
Beetle ce al Bvianow er tres ment therapy for Gaucher disease mendations. J Pediatr 144(1):112, 2004.
ra Non eee ee 8 es OR type Ub. Eur J Pediatr 160(1):63, 2001. 26. Brooks, DA, et al: Significance of
OF Cen icescc hsb Gg:Mseab Dis 18. McGovern, MM, and Desnick, R: immune response to enzyme-replacement
28(4):585, 2005 eae of the eas therapy for patients with a lysosomal
ey GE ee ok ae macrophage system. In Greer et al: storage disorder. Trends Mol Med
‘ Rene ene Rien Wintrobe’s Clinical Hematology, ed 11. 9(10):450, 2003.
ee ae re ae cae Lippincott, Williams & Wilkins, 27. Pick, L: Niemann-Pick’s disease and
pe ite y Philadelphia, 2003. other forms of so called xanthomatosis.
replacement
Gre Med Restherapy in Gaucher
Onin 53/6) 1045 patients.
2006 19 . Savage,
RA: Specific and not-so-specific Am J Med Sci 185:601, 1933.
et es ae a os ane ; histiocytes in bone marrow. Lab Med
: 28. Das, S, et al: Niemann-Pick disease.
LG; all, S): 15:467, 1984. J Indian Med Assoc 92(3):87, 1994.
Hematologic complications associated
Chapter 23 Lipid (Lysosomal) Storage Diseases and Histiocytosis 54]
Zo Kumar, V, et al: Robbins & Cotran leukemia. In Harley, RD. Harley’s Pedi- bone marrow transplantation in infancy.
Pathologic Basis of Disease, ed. 7. WB atric Ophthalmology, ed 5. Lippincott, Int JPaediatr Dent 16(3):207, 2006.
Saunders, Philadelphia, 2004. Williams & Wilkins, Philadelphia, 2005. 40,a Mahalingam, K, et al: Diagnosis of
30. Sevin, M, et al: The adult form of Bi): Sandhoff, K, et al: The GM, gangliosi- mucopolysaccharidoses: How to avoid
Niemann-Pick disease type C. Brain doses. In Scriver. CR, et al (eds): The false positives and false negatives.
130(Pt 1):120, 2007. Metabolic and Molecular Bases of Indian J Pediatr 71(1):29, 2004.
ail. Muro, S, et al: Lysosomal enzyme Inherited Disease 4 volume set, ed 8. 4]. Hoogerbrugge, PM, and Vossen, JMJJ:
delivery by ICAM-1-targeted nanocarri- McGraw-Hill, New York, 2000. Bone marrow transplantation in the treat-
ers bypassing glycosylation- and . Nakaya-Onishi, M, et al: Observations ment of lysosomal storage disease. In
clathrin-dependent endocytosis. Mol on time course changes of the cherry red Fernandes, J, et al (eds): Inborn Meta-
Ther 13(1):135, 2006. spot in a patient with Tay-Sachs disease. bolic Diseases. Diagnosis and Treatment,
oy; Dodge, JC, et al: Gene transfer of human Br JOphthalmol 84(11):1320, 2000. ed. 4. Springer, Berlin, 2006.
acid sphingomyelinase corrects 37. Emre, S, et al: Biochemical and molecu- 42. McPherson, RA, and Pincus, MR (eds):
neuropathology and motor deficits in a lar analysis of mucopolysaccharidoses in Henry’s Clinical Diagnosis and Manage-
mouse model of Niemann-Pick type A Turkey. Turk J Pediatr 44(1):13, 2002. ment by Laboratory Methods, ed. 21.
disease. Proc Natl Acad Sci USA . Laradi, S, et al: Mucopolysaccharidosis WB Saunders, Philadelphia, 2006.
6;102(49):17822, 2005. I: Alpha-L-iduronidase mutations in
3 Charrow, J: Ashkenazi Jewish genetic three Tunisian families. J Inherit Metab
disorders. Fam Cancer 3(3-4):201, 2004. Dis 28(6):1019, 2005.
34. Robb, RM: Ocular abnormalities in Dy: Hingston, EJ, et al: Hurler’s syndrome:
childhood metabolic disease and Dental findings in a case treated with
it I
PART 4
HEMOSTASIS AND INTRODUCTION TO THROMBOSIS
Chapter —
Introduction
to Hemostasis
Denise M. Harmening, PhD, MT(ASCP), CLS(NCA)
Claudia E. Escobar, MT(ASCP)SH
David L. McGlasson, MS, CLS(NCA)
Thrombin-Mediated Platelet
Aggregation
Thrombin Formation: Role
of Extrinsic Pathway
Thrombin Formation: Role
of Common Pathway
Thrombin-Mediated
Anticoagulant Activity
Fibrinogen Conversion and
Fibrin Stabilization via
Thrombin
Thrombin-Mediated Tissue
Repair
Blood Coagulation: A
Cell-Based Model of
Hemostasis
Fibrin-Lysing (Fibrinolytic)
System
Action of Plasmin
Kinin System
Protease Inhibitors
Complement System
Laboratory Evaluation of
Hemostasis
Case Study
Platelets and Hemostatic Mechanisms effect on cellular function, in particular, endothelial cell
function. Mechanical or pathologic disruption of intact
Overview endothelium within the vessel wall initiates localized hemo-
static and thrombotic responses. If the vascular injury
Hemostasis is the complex process by which the body spon-
exceeds the capacity of the platelets mediated by thrombin to
taneously stops bleeding and maintains blood in the fluid state
form a hemostatic platelet plug, then the clinical signs and
within the vascular compartment. The major role of the hemo-
symptoms of hemorrhage occur. Stagnation of blood flow as
static system is to maintain a complete balance of the body’s
tendency toward clotting and bleeding (Fig. 24-1). Normal
hemostasis is both rapid and localized. Hemostasis is
achieved by the highly integrated and regulated interaction of:
1. Blood vessels
2. Platelets
3. Coagulation proteins
4. Fibrinolysis
5. Serine protease inhibitors
Minor ancillary systems involved are also listed in
Table 24—1. Through a complex series of molecular interac-
tions involving cells and biochemicals, a balance between
procoagulant and fibrinolytic activity is achieved. A fine line
separates clot formation in circulating blood from bleeding.
It has long been recognized that an imbalance, either
acquired or inherited, in any one of the many factors that Hemostasis
contribute to hemostasis often leads to hemorrhage or throm-
bosis. Significant changes in blood flow have a profound Figure 24-1 @ Hemostasis: A system in balance.
Chapter 24 Introduction to Hemostasis 545
*In general, the larger the vessel, the more hemostatic system involvement is required to seal the breach.
546 Chapter 24 = Introduction to Hemostasis
the middle layer (media), and the inner layer (intima) is outlined (1) von Willebrand factor (vWF), (2) collagen fibers, and
in Table 244 The endothelial surface of the blood vessel is usu- (3) platelet membrane glycoprotein Ib (GPIb; the receptor
ally inert to platelets and coagulation factors. It is termed a non- for vWF).°
thrombogenic surface because the physical and biochemical The adherent platelet undergoes activation events,
characteristics of the endothelium render the cell surface throm- including shape change, platelet aggregation, and secretion of
boresistant. This 1s achieved through: ADP from the platelet-dense bodies. Secretion of platelet
ADP acts in the recruitment of additional platelets to form the
. Synthesis and secretion of a vasodilator, prostacyclin (PGI,)
primary hemostatic «plug. Activated platelets provide an
. Secretion of tissue plasminogen activator (t-PA)
important phospholipid surface that enhances activation of
. Inactivation and clearance of thrombin
the coagulation proteins in both the intrinsic and extrinsic
. Activity
RWNe of the cofactor thrombomodulin in the thrombin-
system. In addition, platelet secretion of thromboxane A,
dependent activation of protein C
(TXA,), a prostaglandin; and serotonin, further promotes
5. The degradation of proaggregating substances such as
vasoconstriction. Endothelial release of tissue factor (for-
adenosine diphosphate (ADP) and vasoactive amines?
merly known as tissue thromboplastin) during vascular injury,
However, when the endothelial lining is disrupted, the represents one of the body’s major procoagulant abilities.’*
vascular system acts to prevent bleeding by promoting rapid The initiation of the extrinsic pathway of coagulation
vasoconstriction of the injured vessel as well as adjacent requires tissue factor, factor VII/VIla, calcium, and a nega-
vessels. This process diverts blood flow around the damaged tively charged phospholipid membrane. The factor VIIa—
vessel and enhances contact activation of platelets and coag- tissue factor complex is now known to initiate the activation
ulation factors (Table 24—5S). Platelet adhesion occurs almost of both factor X and factor IX. Release of t-PA by the dam-
immediately to the exposed collagen fibers of the subendothe- aged endothelial cell provides the mechanism for clot disso-
lium. The principal mechanism of platelet adhesion involves lution, necessary to re-establish vascular patency.? PDGF,
secreted by platelets, assists in proliferation and repair of
damaged endothelium. The vascular response involved in the
hemostatic mechanisms usually lasts less than 1 minute.
Some of the major substances contained within the endothe-
lium, subendothelium, or both, that play primary roles in
hemostasis are listed in Table 24—6. Other substances known
to bind to endothelial cells that promote coagulation are
Vessel ayer" ) Composinon and Eiancucn listed in Table 24-7.
Substance(s)
Collagen Binds to Satter Toone GPIb/Ila: activates plasma coagulation factors via
intrinsic pathway
Heparin sulfate A glycosaminoglycan that has anticoagulant activity and contributes to the
activation of antithrombin
von Willebrand factor (VWF) Protein synthesized in endothelial cells and megakaryocytes; primarily binds to
platelet membrane receptor GPIb to promote adhesion; also functions as a carrier
for factor VII, providing factor stability in circulation; may also bind to platelet
GPIIb/IIIa in conditions of high shear stress to promote adhesion
PGI, A prostacyclin synthesized by endothelial cells that physiologically inhibits platelet
aggregation and limits thrombus formation beyond the damaged vessel
Tissue factor (TF) A lipoprotein released following vascular trauma that initiates coagulation by
activating factor VII with ionized calcium (extrinsic pathway) and contributes to
the activation of factors X and IX (intrinsic pathway)
Tissue factor pathway inhibitors (TFPIs) glycoproteins found on endothelial surfaces that serves as an anticoagulant by
. inhibiting factors VIla/TF and Xa
Thrombomodulin A large protein found on endothelial surfaces that serves as a cofactor in protein C
activation when bound to thrombin
Tissue plasminogen activator (t-PA) Serine protease secreted by endothelial cells that regulates fibrinolysis; when
bound to fibrin, t-PA activates plasminogen to form plasmin
Plasminogen activator inhibitors (PAIs) Proteins found in endothelial cells that regulate fibrinolysis by neutralizing the
activity of plasmin, plasminogen, and t-PA; associated with risk of thrombosis
Figure 24-2 = Normal platelets: Wright-stained blood smear 1. Providing a negatively charged phospholipid surface for
(peripheral blood). factor X and prothrombin activation
548 Chapter 24 Introduction to Hemostasis
PERIPHERAL ZONE
BASIC CONCEPTS The peripheral zone is a complex region
of the platelet consisting of the glycocalyx (an amorphous
MT Microtubules exterior coat), platelet membrane, numerous deeply penetrat-
G Other granules ing surface-connecting channels known as the Open Canalic-
DB Dense bodies
DTS Dense tubular system
ular System (OCS), and a submembranous area of specialized
M Mitochondria microfilaments (see Fig. 24~3).
Gly Glycogen lakes (particles) A number of glycoproteins anchored in the platelet
Lys Lysosomes
OCS Open canalicular system
membrane are present in the glycocalyx and mediate the crit-
Mf Microfilaments ical events of platelet adhesion and aggregation. Specific
EC Exterior coat (glycocalyx) binding of platelet membrane receptors to adhesive macro-
GA Golgi apparatus
molecules such as fibronectin and vitronectin in plasma or
collagen in vascular subendothelium, further facilitates the
Figure 24-3 @ Discoid platelets; (top) summary diagram of the spreading of platelets on the damaged vessel wall and leads to
platelet organelles; (bottom) transmission electron micrograph platelet activation and the formation of large platelet aggre-
(TEM) of cross-sectioned platelets illustrating basic ultrastructure. gates.'° Platelet membrane glycoproteins serve as receptors
Chapter 24 Introduction to Hemostasis 549
ADVANCED CONCEPTS Microtubules disappear from the Dense granules store ADP, ATP, ionic calcium, serotonin, and
center of the platelet after secretion, and reappear in other phosphates.
peripheral areas such as pseudopods. Microtubules appear to Lysosomes appear similar to the azurophilic granules
monitor the internal contraction of platelets, preventing platelet found in granulocytes and contain microbicidal enzymes,
secretion in response to only minimal stimulation and thereby neutral proteases, and acid hydrolases. Proteases may con-
regulating the degree of platelet response to external stimuli. tribute to disruption of the subendothelial structure following
vascular injury. Glycogen granules are also found within the
ORGANELLE ZONE organelle zone and function in platelet metabolism.
BASIC CONCEPTS The organelle region is responsible for The contents of both a and dense granules are released
the metabolic activities of the platelet. Like many other cells, during secretion into the OCS. Secretion promotes the recruit-
platelets possess mitochondria and various cytoplasmic gran- ment of additional platelets to the platelet aggregate at the site
ules. Unlike many cells, platelets are anucleated and do of vascular injury. Platelet secretion is an energy-dependent
not possess either a Golgi body or rough endoplasmic reaction that relies on the metabolic function of mitochondria.
reticulum (RER). The estimated 10 to 60 mitochondria present per platelet require
Generally, the most numerous organelles are the platelet glycogen as their source of energy for metabolism.° In the rest-
granules, which are heterogeneous in size, electron density, ing platelet, ATP (energy) production is generated by glycolysis
and chemical content. Platelets contain three morphologically and the oxidative Krebs cycle. In the activated state, about half
distinct types of storage granules: dense granules, a granules, the ATP production in platelets occurs through the glycolytic
and lysosomes. The a granules are most numerous (20 to pathway. The important platelet-secreted proteins and their
200 per platelet) and store a number of different substances. known role in hemostasis are summarized in Table 24—10.
The content of the a granules along with their major function The dense tubular system (DTS) is another important
in hemostasis are listed in Table 24—10.° structure present in the cytoplasm of the organelle zone of
Dense granules or bodies, are smaller and fewer in num- platelets. Similar to the sarcotubules in skeletal muscle, the
ber (2 to 10 per platelet) and appear as dense opaque granules DTS is derived from the smooth endoplasmic reticulum
in transmission electron microscope (TEM) preparations. (ER) of immature megakaryocytes. The DTS is the site of
Granules
Alpha eianuies
Platelet-Specific Proteins Inhibits heparin; chemotactic; promotes smooth muscle growth for vessel
Beta-thromboglobulin (8-TG) repair
Platelet factor 4 (PF4) Inhibits heparin
Platelet-derived growth factor (PDGF) Promotes smooth muscle growth; involved in atherosclerosis and lipid
metabolism
Thrombospondin Promotes platelet-to-platelet interaction; mediates cell-to-cell interaction
Factor V Once activated complexes with Xa to convert prothrombin to thrombin
Plasma Proteins
Fibrinogen Fibrin formation
von Willebrand factor (vWF) Promotes platelet adhesion
Factor V Cofactor in fibrin formation
Factor VUI Cofactor in fibrin formation
Fibronectin Cellular adhesion molecule; promotes platelet spreading
Albumin Uncertain
High-molecular-weight kininogen (HMWk) Activation of the intrinsic pathway via contact
a.-Antiplasmin Inhibits plasmin
Plasminogen Precursor to plasmin; functions in fibrinolysis
Dense Granules
ADP (nonmetabolic) Promotes platelet aggregation
ATP (nonmetabolic) Uncertain
Calcium Primary and secondary messenger regulates platelet activation/ aggregation
Serotonin Promotes vasoconstriction
Lysosomes
Neutral proteases* Uncertain
Acid hydrolase* Uncertain
Bacteriocidal enzymes* Uncertain
*The specific function of these proteins is uncertain: heemay serve to pices plas activation in response to thrombin.
Chapter 24 Introduction to Hemostasis 551
1. vWF, a plasma protein that links the platelet to subendothe- Figure 24-6 @ Pictorial representation of platelet adhesion to
lial binding sites subendothelium through von Willebrand's factor (vWF) bridge.
Chapter 24 Introduction to Hemostasis NnNnWwW
Figure 24-8 @ TEM showing disk-to-sphere transformation of an activated platelet. Note progression from (1) disk shape to (2) pseudopod
formation to (3) degranulated ballooned sphere.
nn‘nS Chapter 24 Introduction to Hemostasis
from the a granule have been well characterized to date and are
Figure 24-10 @ A typical biphasic response of in vitro platelet currently used as ‘markers of platelet activation.” These include
aggregation to ADP, as recorded via an aggregometer. beta-thromboglobulin (B-TG), platelet factor 4 (PF4), throm-
bospondin, and PDGF, which confirm the degranulation of
platelet a granules.'’ Because B-TG, PF4, thrombospondin, and
containing citrated whole blood in a 1:1 saline dilution. The PDGF are proteins virtually absent from normal plasma and
electrodes produce a small electric current running through the found in small concentrations within platelet a granules, these
whole blood suspension. As platelet aggregation occurs, proteins specifically mark platelet activation.' A number of
the platelets collect on the electrodes, reducing or impeding clinical conditions such as arteriosclerosis, cerebrovascular dis-
the magnitude of the current. The change is directly propor- ease, cardiopulmonary bypass, shock, venous thrombosis, and
tional to the degree of platelet aggregation. The peak ampli- DIC are associated with increased plasma levels of these mark-
tude is measured in ohms. The whole blood aggregation ers, thus signifying platelet activation.
pattern resembles a platelet-rich plasma (PRP) aggregation
tracing but is measured in ohms. However, you rarely see STABILIZATION OF THE HEMOSTATIC PLUG The last
secondary aggregation demonstrated and the peak amplitude stage involved in arresting bleeding after vessel damage is the
is much lower than in a PRP tracing. One distinct advantage formation of a stable platelet plug. This stabilization is
of whole-blood aggregation is that it virtually eliminates achieved through the activation of the coagulation cascade
the need for PRP preparation, reducing the blood volume and formation and deposition of fibrin (the end product of
required for testing, and the platelet count does not have to be coagulation) on the platelet aggregates. Exposure of collagen
standardized. The operator pipettes an aliquot of properly within the subendothelium of the damaged vessel (via the
mixed whole blood into the cuvette, adds an equal volume of intrinsic pathway) and the release of tissue factor (via the
physiologic saline pre-warmed to 37°C, and a stir bar. After extrinsic pathway) directly initiate fibrin formation. Thus,
placing the cuvette into the reaction well, the electrodes are fibrin interweaves through and over the initial platelet
placed into the mixture and the aggregating agent is added. plug, compressing it into place at the site of the vessel injury.
The aggregation pattern is then measured.'* Recent studies What originally starts as a small gelatinous mass gradually
have also shown that whole blood aggregation may be more
sensitive, and reflect the true nature of platelet aggregation in
vivo than PRP methods, and may be more sensitive to aspirin
elfecise a0
10-20 sec.
resulting in the cross-linking of fibrin strands and, thus, clot Platelet Release Thrombin
stabilization.
Fibrinolysis, the complex series of enzymatic reac-
tions that promote clot dissolution, is simultaneously acti- |
as 1-3 min.
3. PLUG FORMATION
STIMULATED NUCLEUS
MEMBRANE
PHOSPHOLIPID
Phospholipase Az
ARACHIDONIC ACID
Cyclo- oxygenase
PROSTAGLANDIN
ENDOPEROXIDES
Thromboxane
Synthetase
iyTHROMBOXANE A:
~y (TXAg),
NORMAL
Figure 24-15 @ Synthesis of prostaglandins in platelets and endothelial cells during platelet plug formation.
that mediate platelet activation and aggregation, but only This system is mediated by many coagulation proteins
one, TXA, synthetase, is inhibited by aspirin.*? Thus, aspirin (coagulation factors), normally present in the blood in an
has a modest effect on platelet function in vivo, causing inactive state. The coagulation factors and their most
moderate prolongation of the bleeding time, moderate inhi- commonly used designations are listed in Table 24-13. Low
bition of platelet aggregation, increase in the PFA-100® clo- levels of the coagulation factors in secondary hemostasis may
sure time [epinephrine/collagen cartridge (Dade-Behring)]** be associated with bleeding that is generally delayed when
and reduction of the aspirin-resistance units using the compared to that observed in defects in the primary hemosta-
arachidonic-acid cartridge with the VerifyNow™™ (Accu- tic mechanism. Symptoms of defective secondary hemostasis
metrics) analyzer. Despite its limited effect on platelet may include soft tissue bleeding, hematomas, retroperitoneal
function, aspirin provides a clinically significant antithrom- bleeding or hemarthrosis. Good examples of these problems
botic effect. are the hemophilias, that are associated with factor VII and
IX deficiencies (see Chap. 26). Secondary hemostasis is the
Secondary Hemostasis: Fibrin-Forming phrase used to encompass the coagulation factors’ role in the
hemostatic mechanism.
(Coagulation) System
Most of the coagulation factors are designated by Roman
The fibrin-forming (coagulation) system is that system numerals. The numerical system adopted assigns the number
through which coagulation factors interact to eventually form to the factors according to the sequence of discovery and not
a fibrin clot. The purpose of fibrin clot formation (secondary to the point of interaction in the cascade. Some factors are
hemostasis) is to reinforce the platelet plug (primary hemosta- routinely referred to by their common names, such as fibrino-
sis). Secondary hemostasis may be started by the release of gen and prothrombin, whereas others are more commonly
tissue factor from epithelial or endothelial cells that are referred to by Roman numeral (such as factor XI, plasma
exposed to the circulatory system at the site of a vascular thromboplastin antecedent factor).
injury. Defects in secondary hemostasis decrease fibrin pro- Activation of a factor is designated by the addition of
duction and reduce the stability of the formed clot. a small “a” next to the Roman numeral in the coagulation
558 — Chapter 24 Introduction to Hemostasis
*Factors V and VIII are not present in stored plasma because of their labile nature, but factors I and XIII are present. PK = prekallikrein;
HMWK = high-molecular-weight kininogen; BaSO, = barium sulfate.
factors may also be selectively removed from plasma by Blood Coagulation: The “Cascade” Theory
adsorption on barium sulfate (BaSQO,).
Drugs that act as antagonists to vitamin K (such as war- The process of blood coagulation involves a series of bio-
farin [Coumadin], commonly used for oral anticoagulant ther- chemical reactions that transforms circulating substances into
apy) inhibit this vitamin K-—dependent reaction, which is an insoluble gel through conversion of soluble fibrinogen to
required for functionally active coagulation factors of the pro- fibrin. This process requires plasma proteins (coagulation
thrombin group.'® Factors II, VII, [X, and X, proteins C and factors) as well as phospholipids and calcium.
S, and protein Z, are still synthesized by the liver, but are non- Blood coagulation leading to fibrin formation can be
functional because they lack the specific receptors for calcium separated into two pathways, extrinsic (Fig. 24-14) and
and cannot enter into the formation of an enzyme-substrate intrinsic (Fig. 24-15), both of which share specific coagula-
complex.” Therefore, patients who are nutritionally vitamin tion factors with the common pathway (Fig. 24-16).*° Both
K-deficient also exhibit decreased production of functional extrinsic and intrinsic pathways require initiation, which
prothrombin proteins. These dysfunctional factors are known leads to subsequent activation of various coagulation factors
as “Proteins Induced by Vitamin K Absence or Antagonists” in a cascading, waterfall, or domino effect. Useful demonstra-
(PIVKAs). tions can be derived from the waterfall or domino concept.
Acquired deficiencies of the vitamin K—dependent coagu- According to the cascade theory, each coagulation factor is
lation factors are relatively common because the body does not converted to its active form by the preceding factor in a series
contain appreciable stores of vitamin K. Clinical situations in of biochemical chain reactions. Ca** participates in some of
which a vitamin K deficiency can develop include: patients the reactions as a cofactor. Each reaction is promoted by the
who have just had surgery and are receiving parenteral feeding; preceding reaction, and if there is a deficiency of any one of
patients who are receiving high doses of intravenous antibi- the factors, the consequences listed in Table 24-16 result.
otics; and patients suffering from liver disease. Table 24-16 Eventually, both the extrinsic and intrinsic systems lead
lists the consequences of a factor deficiency. to the common pathway with generation of the enzyme
The fibrinogen group (see Table 24—15) consists gener- thrombin, which converts fibrinogen to fibrin (Figs. 24-17
ally of high-molecular-weight proteins that include factors I and 24-18). The term extrinsic is used because this pathway
(fibrinogen), V (labile factor), VIII:C (antihemophilic factor), is initiated when tissue factor, a substance not found in blood,
and XIII (fibrin-stabilizing factor). During coagulation, gen- enters the vascular system (see Fig. 24-14). The tissue factor
erated thrombin acts on all the factors in the fibrinogen group. includes a phospholipid component that is the source of
Thrombin enhances the activity of factors V and VIII:C required phospholipid in the extrinsic system (Fig. 24-19).
by converting these proteins into active cofactors, that are Phospholipid provides a surface for interaction of various fac-
involved in the assembly of macromolecular complexes on tors. The phospholipids required in the intrinsic pathway are
the surface of activated platelets. Thrombin converts fibrino- provided by the platelet membrane. In the intrinsic pathway,
gen to fibrin and also activates factor XIII.
Factors V and VIII:C are the least stable factors,
because their activity is relatively labile to degradation and
denaturation. Therefore, testing for factor V and VIII:C
should be rapid, unless appropriate storage measures are
taken. In addition to their presence in plasma, these factors ¢ Coagulation cannot proceed at a normal rate
¢ Initiation of the next subsequent reaction is delayed
are also found within platelets. Some factors within the
¢ The time required for the clot to form is prolonged
fibrinogen group have been reported to increase with ° Bleeding from the injured vessel continues for a longer
inflammation, pregnancy, and with the use of oral contra- time (or there may be a physiologic tendency toward
ceptives. Other physical properties of the coagulation thrombosis present if the patient is deficient in factor XII)
factors are summarized in Table 24-17.
“UOSOUTUTY JYSIOM-IV[NOOTOWI-YSIYy = YMINH ‘Uo [eyeid = Yd
“vurseyd ysory [eULIOU Ul JUASoId O7e S1O}ORJ [Vx
poqgiospe
MWH
O'S-O'e0'6-0°9
JUdSIIg
MH
J0
vuiseyd
vurseyd
ATUO SISULQUT
pure
O9IT-OSI OST
UI
SE
vurseyd pogiospe
ur quasaid ‘ssaoo0id uonrpnseoo Aemyyed uoulw09
Sulinp podonsop AWANIY S Ca0il OTE ‘SISULNXO ‘OISULQUT
vuiseid
poqiospe pure windos UI JUASOIg OVO 08 ATWO OISULnUT
1981S a1qris
vurseyd
poqiospe pur windos ul JUdSaIg VS 091 A]UO SISULQUT
0
JUISIIg
Aemyjed uouluwi09
WNIIS
a1qras
JUISIIg
“SISULI]XO “OISULNUT
WINIOS
Ons 6S
Ul
vuuseyd
IIAAMA/TILA
10-100
poqiospe ut juasaid ‘ssaooid uon
0'c-S$0'0
-R[NSvod SuLinp podoNsop APWANIY oTge7 OSE AJUO wWiaisds SISULNUT
UINIIS UI JUISIIg a1qras QP Ajuo wiaysAs SISULN XY
vurse[d paqiospe
Aemyjed uourtu05
apiqey
ur quasaid ‘ssoooid uone[nseos
Sulinp podonsop AWANOY OEE “OISULIYXO ‘OISULUT
Gles 0
powinsuod
ssooo1d
0
SuLInp
Aemyjed uourlu0s
SI-0l
OL “‘OISULXO ‘OISULQU
vuiseid paqiospe
Introduction to Hemostasis
IJVI
OALA
(GD) 481M Kemyed
(Y)
T-SIPH
4
SOSLIOIVABY 1YIO a8es0}5 AGEs ("Tp/sut)
AB[NID[OJA| SUIRIO[D
OV
UL
LCUISe[
UOHRIJUIIUOD
Chapter 24
560
Chapter 24 Introduction to Hemostasis 56]
all the factors necessary for clot formation are intrinsic to the the VIa- tissue factor complex can activate factor [IX to [Xa
vascular compartment because they are all found within the in the intrinsic pathway. In the laboratory, the prothrombin
circulating blood (see Fig. 24-15). time (PT) test is used to monitor the extrinsic pathway (for a
review of the procedure, see Chap. 33).
Extrinsic Pathway (Factor VII)
In the extrinsic pathway, factor VII is activated to factor VIIa Intrinsic Pathway (Factors XIl, XI, IX,
in the presence of Ca** (factor IV) and the tissue factor and VIII)
(factor III), which is released from the injured vessel wall
(see Fig. 24-14). Only factor VIa, Ca**, and factor III (tissue BASIC CONCEPTS
factor) are needed to activate factor X to Xa. Following exposure to negatively charged foreign substances
Figures 24-17 and 24-18 show that the extrinsic path- such as collagen, subendothelium, or phospholipids, activation
way provides a means for very quickly producing small of factor XII involving “contact factors” and factor XI initiate
amounts of thrombin, leading to fibrin formation. In addition, clotting through the intrinsic pathway (see Fig. 24-15).
Extrinsic Pathway
P nat OrA A} wing.
‘ Pen, 25
ppeioes
athwa
A Vila
TF
complex
Activates
Figure 24-14 m The extrinsic pathway and the role of factor Vila in activation of factor X and IX. (From American Bioproducts Company.
Modified with permission.)
562 Chapter 24 Introduction to Hemostasis
Intrinsic Pathway
AE: aaHMWK
~~ Kallikrein
ee Xil
Tapa dligem 4 Xila
pathway Teen alice)
pP athwa y
F XII
|
Contact with Collagen
F Xila
HMWK F IX
Prekallikrein
FX! ——— > F Xia —> | CA
Common Pathway
F lXa Ca'PF 3] Activates (X———— Xa) etc.
Villa
Intrinsic System
(Factors XIl, XI, IX, VIII)
Figure 24-15 @ The intrinsic pathway and its role in activation of factor X. HMWK = high-molecular-weight kininogen; PF3 = platelet
factor 3. (From American Bioproducts Company. Modified with permission.)
Once generated, factor XIla, in the presence of Fitzger- (Table 24-18). Factor VII complex consists of two main por-
ald factor (HMWK) and Fletcher factor (prekallikrein), con- tions, factor VIII:C (the procoagulant protein) and vWF (the
verts factor XI to XIa. Factor XIa is capable of activating carrier protein).
factor XI without HMWK, but the activation takes place It should be noted that factor VIII requires enhancement
much more slowly.”’ by the generated enzyme thrombin to amplify its activity.?’ In
The next reaction in the intrinsic pathway is the activa- the laboratory, the activated partial thromboplastin time
tion of factor [IX to factor [Xa by factor XIa, in the presence (APTT) test is used to evaluate the intrinsic pathway. During
of Ca**. Activated factor [X (IXa) participates, along with the laboratory testing, the intrinsic pathway is initiated in vitro by
essential cofactor VIII:C, in the presence of ionized calcium activation on negatively charged surfaces, such as glass or
and PF3, a source of phospholipids, to activate factor X, kaolin; or chemically by ellagic acid (for a review of this pro-
which leads to the generation of thrombin and formation of cedure, see Chap. 33).
fibrin. The complex consisting of factor [Xa-factor VIIla- The factor VIII complex, which consists of several com-
phospholipid-Ca** has been called the tenase complex ponents, comprises the largest protein involved in the coagu-
because it activates factor X (Fig. 24-20). '” lation cascade. The major portion of this protein complex is
The macromolecular complex of factors [Xa, VIIIa, considered to be the carrier protein called von Willebrand
X, PF3, and Ca’* assembles on the surface of the activated factor (vWF), although this is not the portion active in the
platelet (providing the phospholipid) during the intrinsic coagulation cascade. A smaller subunit or protein that is
pathway of blood coagulation.*’ This surface provides a associated with factor VIII is responsible for the clotting or
protective environment that facilitates the enzymatic reac- procoagulant activity of factor VII (VIII:C). The C in the
tions of the coagulation cascade without interference from expression VIII:C stands for “coagulant.” It is factor VIII:C
the physiologic anticoagulants normally present in plasma. that is functionally active in the coagulation cascade.
In regard to the intrinsic pathway, it is also important to The vWF portion of the complex carries the VIII:C pro-
be familiar with the properties of the factor VIII complex coagulant portion. The vWF portion may exhibit a stabilizing
Chapter 24 Introduction to Hemostasis 563
INTRINSIC SYSTEM
Contact Collagen
‘a -e oeaa
XII alte Xlla MWK
HMWK PK
X| —— Xla
|Catt 2
IX ——-> IXa
t
AO) SSN
Lab Tests
Figure 24-17 @ Blood coagulation: The “cascade” theory of coagulation. The extrinsic system, the intrinsic system, and the common
pathway and the appropriate laboratory tests for evaluation of each. HMWK = high-molecular-weight kininogen; PF3 = platelet factor 3;
PK = prekallikrein; PT = prothrombin time; APTT = activated partial thromboplastin time.
formation, following vascular injury. Thus, thrombin has a cen- Thrombin Formation: Role of Extrinsic
tral role in the bioregulation of hemostasis in both normal and Pathway
pathologic conditions. The following discussion provides an
overview of thrombin-mediated mechanisms in hemostasis. Thrombin’s main duty is to cleave fibrinopeptides A and B
The various roles of thrombin in hemostasis are summarized in from the alpha and beta chains of the fibrinogen molecule,
Table 24—21. thus triggering fibrin polymerization. Also thrombin amplifies
the coagulation mechanism by activating cofactors V, VIII
Thrombin-Mediated Platelet Aggregation and factor XI.'’ Formation of thrombin occurs by way of the
extrinsic pathway and tissue factor. Tissue factor, a membrane
Thrombin is considered to be a potent platelet-aggregating glycoprotein, is released from cells and tissues and binds to
agent. The binding of thrombin to specific platelet membrane factor VII in circulation, thus activating factor VII to factor
receptors initiates cellular events leading to platelet secretion Vila. The TF—VIla complex then activates either factor LX or
and aggregation. Thrombin promotes secretion of serotonin, a X. The prominent pathway is thought to be activation of
vasoconstrictor; and TXA,, a platelet-aggregating agent. As factor IX to promote coagulation and fibrin formation.
a result of thrombin-induced secretion, vessels constrict,
limiting blood flow, and platelets aggregate. The hemostatic Thrombin Formation: Role of Common
plug grows in size, eventually being enmeshed within fibrin. Pathway
Activated platelets provide surface phospholipids for the
assembly of coagulation factors and further thrombin Factors X, II (prothrombin), I (fibrinogen), and cofactor V
generation. are critical to the formation of a thrombus in circulation.
Chapter 24 Introduction to Hemostasis 565
Coagulation Cascade
Intrinsic Pathway Extrinsic Pathway
@e gpgHMWK
oe << & PK Tissue Factor eo.
Ss : Kallikrein
Cat platelets a
Vg
LO 7 in-vitro 7 vil
v@ pathway in-vivo _Mila
7athway
Fibrinogen
Laey.\
Ee Xxill
mleldim(oyatelint=\g
Fibrin Polymers
Fibrin Clot
QD
ae |
Fibrinolytic Pathway
»
Figure 24-18 @ An overview of coagulation cascade, the intrinsic and extrinsic pathways, and the interaction between the two. The fibrinolytic
pathway and its action on fibrinogen and fibrin. HMWK = high-molecular-weight kininogen; PK = prekallikrein; FoA = fibrinopeptide A. (From
Diagnostica-Stago, Inc., with permission.)
Activated factor [Xa (via factor VIIa) factor X, Ca**, and thrombin from prothrombin at a faster rate. Increased throm-
thrombin-activated VIIla assemble on membrane platelet bin concentrations serve to amplify the activation of cofactors
phospholipid and catalyze the activation of factor X. Newly V and VIII, which, in turn, leads to enhanced thrombin forma-
generated factor Xa, membrane-bound thrombin-activated tion from its precursor prothrombin.
factor Va, Ca?*, and factor II (prothrombin) assemble on the
platelet membrane, forming the prothrombinase complex. Thrombin-Mediated Anticoagulant Activity
This complex catalyzes the conversion of prothrombin to
thrombin. Thus, this thrombin-modulated pathway provides a Antithrombin (AT), formerly known as AT III, is the main
positive feedback mechanism to amplify the generation of physiologic inhibitor of thrombin, factors Xa, [Xa, XIa, and
566 Chapter 24 Introduction to Hemostasis
©
"Prothrombinase @
TF Tissue Factor Complex" %$$6666666066666
|
Gla = gamma-carboxy-glutamic acid
Catt Calcium ions
a activated factor Factor
PL Phospholipid source Thrombin (Ila)
Figure 24-19 m Platelet membrane phospholipid provides a surface for the interaction of coagulation factors and the formation of “tenase
complex” and “prothrombinase complex.”
Figure 24-20 @ Activation of factor X at the beginning of the APTT = activated partial thromboplastin time.
common pathway and the “tenase” complex.
Chapter 24 Introduction to Hemostasis 567
THROMBIN ACTION interesting to note that the reactions that lead to thrombin
on formation from its precursor prothrombin are regulated in
FIBRINOGEN
Thrombin Cleaves part by thrombin-mediated mechanisms (see Chap. 28 for a
+4 detailed discussion of anticoagulant therapy).
alpha chain be
sc chains mine ae _ [eae
gamma chains Cia
> Fibrinogen Conversion and Fibrin
thrombin
Stabilization via Thrombin
= EEE a
ome =al Thrombin proteolytically cleaves first fibrinopeptide A, then
sr) o
fibrin
fibrin monomer
peptides
fibrinopeptide B from the @ and B chains of fibrinogen,
A,B respectively, forming a soluble fibrin monomer. Fibrin
spontaneous polymerization monomers polymerize in an end-to-end and side-to-side
manner held together by weak hydrogen bonds. Factor XIII
is activated to factor XIIa by thrombin (in the presence of
hydrogen bonds Ca**) and catalyzes the cross-linking of glutamine and lysine,
weak fibrin polymer thus polymerizing soluble fibrin monomers to an insoluble
fibrin meshwork.
(Thrombin)
Inactive
AT Complexes
Figure 24-23 @ The inhibitor pathway of coagulation. AT = antithrombin; PC = protein C; APC = activated protein C; PS = protein S;
C4b-BP = C4b-bound protein; TM = thrombomodulin.
XIa. In addition, factor Va cleaves factor VIII from von The fibrinolytic system is mediated mainly by the
Willebrand’s factor (vWF) activating it to VIIa. Recent studies enzyme plasmin, which acts primarily on fibrin to produce
have shown that activated platelets can synthesize TF.’***° lysis of the clot. Plasmin is generated from the inactive zymo-
Phase 3, propagation, occurs when large amounts of gen called plasminogen. Plasminogen is activated to plasmin
thrombin are generated on the platelet surface. The factor XIa by t-PA and other substances (see Chap. 27).
generated from the amplification phase binds to the activated A number of plasmin inhibitors exist to keep fibrinolysis
platelets and subsequently activates factor X to Xa. The acti- from getting out of control. In addition to plasmin, plasmino-
vated platelet surface protects FXla from an inhibitor by a gen and plasminogen activators, inhibitors of plasmin are a
specific protease. FXIa can efficiently activate FLX to F[Xa on part of the fibrinolytic system.*” Some inhibitors of plasmin
the platelet surface, inducing an explosive thrombin forma- are listed in Table 24-22.
tion by activating X to Xa, which moves directly into a pro- It is important to realize that some of the same substances
tected complex with factor Va. This results in a burst of that initiate or enhance clot formation also initiate clot degrada-
thrombin generation as the Xa/V complex activates prothrom- tion. For example, in tissue, both tissue thromboplastin (initiator
bin to thrombin (Fig. 24-24). of extrinsic pathway of fibrin formation) and t-PA (which acti-
The extrinsic or tissue factor (TF) pathway works on the vates plasminogen) are released with endothelial damage. t-PA
initiating cell during phase 1. The prothrombin time (PT) is produced by vascular endothelial cells and selectively binds
assays the factors in the extrinsic/initiation pathway. The to fibrin as it activates fibrin-bound plasminogen. Because cir-
intrinsic pathway works on the platelets during the amplifica- culating plasminogen is not activated by t-PA, this biologic sub-
tion and propagation stage. The APTT assays the factors in stance is efficient in dissolving a clot without causing systemic
the intrinsic/platelet surface pathway. fibrinolysis and serves as an ideal therapeutic fibrinolytic agent.
Biologic t-PA has been successfully produced by recom-
binant deoxyribonucleic acid (DNA) technology and is cur-
Fibrin-Lysing (Fibrinolytic) System rently available. Inhibitors of t-PA also exist and contribute to
The fibrin-forming and fibrin-lysing systems are intimately controlling its actions.*? It is also important to note that
related. Activation of coagulation also activates fibrin lysis. thrombin generates both fibrin and plasmin formation.
Fibrinolysis, the physiologic process of removing unwanted
fibrin deposits, represents a gradual progressive enzymatic
cleavage of fibrin to soluble fragments. These fragments are
then removed from the circulation by the fixed macrophages
of the mononuclear phagocytic system (MPS). This action of
the fibrinolytic system re-establishes blood flow in vessels ¢ «,-Antiplasmin (a rapid inhibitor of plasmin)
occluded by a thrombus and facilitates the healing process * a,-Macroglobulin (a slower inhibitor of plasmin)
following injury. For a detailed description of the fibrinolytic ¢ Others (see discussion in text of protease inhibitors)
system refer to Chapter 27.
Free vWF
1. Initiation (Small amount
of thrombin)
Tissue-factor bearing
Villa
cell (fibroblast)
IX
Platelet
2. Amplification
lla
(Large amounts
of thrombin)
Activated platelet
Figure 24-24 m Cell-based model of hemostasis. Phase 1 = initiation; phase 2 = amplification; phase 3 = propagation.
570. Chapter 24 Introduction to Hemostasis
(M.W. 340,000) -
Fibrinopeptide
plasmin cleaves
AandB Kinin System
The kinin system, important in inflammation, vascular perme-
ability, and chemotaxis, is activated by both the coagulation
and fibrinolytic systems (see Chap. 27).
Prekallikrein (PK) and high-molecular-weight kininogen
(HK) are additionally needed to enhance or amplify the contact
thrombin clottable factors involved in the intrinsic system (Fig. 24-26). Specifi-
cally, factor XI]a in the presence of HK converts prekallikrein
plasmin cleaves
to kallikrein. Kallikrein feeds back to accelerate the conversion
AlYva
ddd of factor XII to XIIa, speeding up intrinsic system processes.
Y, Intrinsic
YA XI >Xla— coaguiation |
System
: (C, cleaved into} |
HMW (Fitzgerald) C;, and C5,)
Kallikrein > ;
Kininogen
| Plasminogen
Kininogens
LMW HMW
Kininogen Kininogen
(Fitzgerald)
“a v
: a Bradykinin Kinins formed 27
Se Kallidin
Figure 24-26 @ Interrelationship of coagulation, fibrinolytic, kinin, and complement systems. HMW = high molecular weight; LMW = low
molecular weight.
The activation of factor XII acts as the common link clotting factors, play an important role as mediators of both
between many aspects of the hemostatic mechanism, includ- immune and allergic reactions. The reactions in which com-
ing the fibrinolytic system, the kinin system, and the comple- plement participates take place in the blood or in other body
ment system (see Fig. 24-25 and Chap. 27). fluids. The most important biologic role of complement is the
production of cell membrane lysis of antibody-coated target
cells. Two independent pathways of activation of the comple-
Protease Inhibitors ment cascade may occur along with a common cytolytic path-
Because the fibrinolytic system is activated when coagula- way. These are designated the classic and alternate pathways
of complement activation (see Chap. 13 for a review of the
tion is activated, extra fibrin is degraded and eliminated
along with some of the coagulation factors. However, complement system).
enzymes such as plasmin and kallikrein still circulate until Plasmin activates complement by cleaving C3 into C3a
and C3b. Cl esterase inhibitor inactivates complement and
they are eliminated by:
also has a role in hemostasis, as described earlier.
1. Liver hepatocytes (which have an affinity for activated en-
zymes),
2. MPS cells (which pick up particulate matter), or
3. Serine protease inhibitors present in plasma.”
Serine protease inhibitors attach to various enzymes and
inactivate them. Some important serine protease inhibitors are ¢ Antithrombin III
listed in Table 24—24. ¢ a,-Macroglobulin
¢ a,-Antiplasmin
* a,-Antitrypsin
Complement System ¢ Cl esterase inhibitor
¢ Protein C inhibitor
The complement system is composed of approximately 22 ¢ Protein S inhibitor
serum proteins that, working together with antibodies and
572. Chapter 24 Introduction to Hemostasis
Both the coagulation system and the fibrinolytic system The bleeding time has been utilized as a screening tool
are interrelated with the complement system.** The interrela- to determine the risk of bleeding in many clinical settings;
tionship of the coagulation, complement, and fibrinolytic however, there is now a large body of literature that
systems is discussed in Chapter 27. has shown no correlation with risk of bleeding in these
instances.
Recent development of an automated platelet function
Laboratory Evaluation of Hemostasis analyzer (PFA-100) has made the bleeding time almost
of fibrinogen to fibrin. Cross-linked stabilized fibrin, which 1. Screening tests for vascular or platelet dysfunction (such as
develops later through mediation of factor XIIa, does not bleeding time, PFA-100, platelet aggregation using platelet-
have an impact on the PT or APTT. Special testing to assess rich plasma or whole blood, and PF3 assay)
factor XIII activity must be done (see Chap. 33). 2. Tests for coagulation (such as factor assays)
PT and APTT testing has been reported in a variety of 3. Special tests (e.g., for fibrinolytic disorders, tests for deter-
ways, such as patient seconds and control seconds. Past mination of FDPs, D-dimers, plasminogen, t-PA, or ELISA
reporting, using ratios for PT testing and percentage, have assays for detection of fibrin monomers)
largely been abolished. The International Normalized Ratio
The reader may refer to subsequent chapters for a detailed
(INR) is the method of choice for PT reporting, because it
discussion of vascular and platelet-related disorders (see
adjusts for source-related thromboplastin sensitivity differ-
Chap. 25), plasma clotting factor defects (see Chap. 26), inter-
ences and testing methodology through use of a mathematical
action of systems involved in hemostasis (see Chap. 27), throm-
exponent, the International Sensitivity Index (ISI).*? The ISI
bosis and anticoagulant therapy (see Chap. 28), and laboratory
is unique to each batch of thromboplastin/instrumentation
methods (see Chap. 33).
combination and is furnished by the manufacturer.
Numerous articles state the method for reporting
INR values. INR standardizes PT reporting worldwide by
adjusting all reported values to a World Health Organiza-
tion international reference thromboplastin standard. Case Study
Therefore, all PT results reported by INR methodology are
A 4-year-old boy was brought to the emergency department
theoretically comparable. Using the INR values facilitates
by his parents. The boy had fallen off of the monkey bars in a
optimal oral anticoagulant therapy in patients at risk for
park and hit his thigh against one of the bars during the fall.
thrombosis, especially those on warfarin who travel exten-
His thigh became swollen and painful. There was a history of
sively, and require frequent monitoring. Thromboplastins bleeding in the male family members. They had been labeled
with a low ISI (less than 1.2) correlate better to recombi- as “bleeders” in their family history. Coagulation studies were
nant thromboplastin, which replaced human-brain thrombo- performed that showed a normal PT, prolonged APTT, and
plastin for standardizing INR values by the World Health normal platelet count and bleeding time.
Organization.*° Because the PT was normal, all extrinsic and common path-
The thrombin time is a measure of the ability of thrombin way deficiencies were ruled out. The abnormal APTT led the
to convert fibrinogen to fibrin and is particularly useful laboratory technologist to believe the child had a problem with
in the evaluation of circulating anticoagulants (pathologic a factor deficiency in the intrinsic pathway (VIII, IX, XI, and
XI). Factor XII deficiency was ruled out, because it does not
inhibitors). The thrombin time is prolonged in the following
present with bleeding and patients with this disorder are more
conditions:
likely to develop thromboses. Factor assays were performed
1. Hypofibrinogenemia and dysfibrinogenemia for VIII, LX, and XI, to rule out any deficiencies. Results of the
2. Treatment with heparin factor IX and XI assays were normal. However, factor VIII
3. Circulating FDPs assays revealed abnormal results. VWF antigenic assays were
also performed to rule out von Willebrand’s disease.
4. Pathologic circulating inhibitors
The results of all the assays confirmed a diagnosis of hemo-
Table 24-26 can be used as a general guide toward cate- philia A resulting from deficiency of factor VIII. Hemophilia
gorizing bleeding disorders into the groups previously listed, A is an X-linked, recessive disorder passed from the mother,
using the suggested screening tests. who is the carrier, to her male children. Because of inheritance
Additional laboratory testing is designed to narrow patterns, male children have a 25% chance of being affected,
depending on what X gene is inherited. Female children will
down the abnormality to one of these specific areas. As a
be carriers if they inherit the affected gene. More information
result, laboratory testing can be divided into the following
on hemophilia A and other factor deficiencies can be found in
categories: Chapter 26.
Test Vascular Disorder Quantitative Platelet Disorder Qualitative Platelet Disorder Factor Deficiency
Platelet N AbN N
N N N
N N N
AbN AbN AbN
PFA-100 AbN AbN AbN
*Dependent on the factor deficiency: see Table 23-20 for specific information.
N = normal: AbN = abnormal: PT = Prothrombin time: APTT = activated partial thromboplastin time: IBT = Ivy bleeding time.
574 Chapter 24 Introduction to Hemostasis
OD reectonn
1 . Which of the following is true concerning the organelle _ ®¢, Factors VII, X, V, IL, I
zone? d. Factors XII, XI, IV, PF3, VIII
a. Responsible for metabolic activities of the platelet . What events take place in the extrinsic system?
b. Contains dense granules, a granules, and glycocalyx a. Activation of factor X to Va
c. Contains the dense tubular system which is the site of b. Acceleration of intrinsic pathway by enhancement of
prostaglandin synthesis, calcium release, and platelet the activity of factors XII and XI
relaxation c. Activation of factor XII to XIa to initiate clotting
d. Contains the OCS to deliver stored products to the d. Activation of factor VI to VIla in the presence of
platelet surface Ca** and factor I
. What events are involved in the normal formation of a
to
. Which of the following factors are unique to the intrinsic
platelet plug? system?
a. Adhesion, activation, fibrinolysis, secondary hemostasis a. Factors XII, XI, IX, VIII, X, V, I, I, Fletcher, Fitzgerald
b. Aggregation, coagulation, release reaction, lupus anti- b. Factors XII, XI, LX, VIII
coagulant c. Factors VII, X, V, I, I
c. Release action, adhesion, lupus anticoagulant, sec- d. Factors III, VII, X, V, IL, I
ondary hemostasis
d. Activation, adhesion, aggregation, release reaction ee What factor is not found in the common pathway?
a. Factor X
3. What product is responsible for stabilization of the b. Factor V
hemestatic plug? ; PE3
a. TXA, d. Factor XII
b. PF3
c. Fibrin . Which of the following is a function of thrombin?
d. GPIb a. Conversion of fibrinogen to fibrin
b. Activation of factor XIII to stabilize fibrinolysis
. Which of the following are classified as contact group c. Conversion of factor VIT to XIla
proteins? d. Enhancement of factor V, VIII, and XI activity
a. Factors I, VU, IX, X
b. Factors XII, XI, PK, HMWK 135 Which of the following is a function of plasmin?
c. Factors I, V, VII, XI a. Cleavage of T-PA
d. Factors I, II, V, X b. Destruction of fibrin
c. Conversion of XII to Xla
. Which of the following are classified as prothrombin
d. Inhibition of XIa
group proteins?
a. Factors II, VU, IX, X 14. What is the purpose of the PT test in monitoring
b. Factors XII, XI, PK, HMWK hemostasis?
c. Factors I, V, VUI, XU a. Measures factors of the extrinsic pathway
d. Factors I, If, V, X b. Detects platelet decrease or dysfunction
c. Detects presence of aspirin
. Which of the following are classified as fibrinogen group
d. Monitors heparin therapy
proteins?
a. Factors II, VU, Ix, X i). What is the purpose of the APTT test in monitoring
b. Factors XII, XI, PK, HMWK hemostasis?
c. Factors I, V, VII, XI a. Monitoring heparin anticoagulation
d. Factors I, If, V, X b. Detects deficiency of factors for both intrinsic and
extrinsic pathways
. Which of the following factors are dependent on vitamin
c. Measures circulating FDPs
K for synthesis?
d. Detects platelet dysfunction
a. Factors IL, VII, IX, X
b. Factors XII, XI, PK, HMWK
See answers at the back of this book.
c. Factors I, V, VIII, XIII
d. Factors I, Il, V, X
. Which of the following factors are unique to the extrin-
sic system?
a. Factors XII, XI, X, IV, VIII, V, Il, I
b. Factors Ill, VIT
Chapter 24 Introduction to Hemostasis 575
REFERENCES Nn . Nasdala, I, et al: A transmembrane tight thrombin and protein C. J Biol Chem
junction protein selectively expressed on 280(9): 7956, 2005
. Colman, RW, et al: Hemostasis and endothelial cells and platelets. J Biol 29. Dahlback, B, and Villoutreix, BO: Regu-
Thrombosis: Basic Principles and Clini- Chem 277:16294, 2002. lation of blood coagulation by the pro-
cal Practice, ed 4. Lippincott-Raven, . Nesbitt, WS, et al: Intracellular calcium tein C anticoagulant pathway: Novel
Philadelphia, 2000. communication regulates platelet aggre- insights into structure-function relation-
bo . Brandt, JT: Overview of hemostasis. In gation and thrombus growth. J Cell Biol ships and molecular recognition.
McGlatchey, KD (ed): Clinical Labora- 160:1151, 2003. Arterioscler Thromb Vasc Biol
tory Medicine, ed 2. Lippincott Williams . Kolde, H-J: Haemostasis, Physiology, 25(7):1311, 2005.
and Wilkins, Baltimore, 2003, Pathology, Diagnostics, ed 2. Pen- 30. Hoffman, M, and Monroe, DM: A cell-
pp 987-1009. tapharm, Basel, Switzerland, 2004. based model of hemostasis. Thromb
oS) . Kottke-Marchant, K, and Corcoran, G: . Fritsma, G: Evaluation of hemostasis. In Haemost 85(6): 958, 2001.
The laboratory diagnosis of platelets Rodak, B et al (eds): Hematology: Clini- . Monroe, DM, et al: Platelets and throm-
disorders. Arch Pathol Lab Med cal Principles and Applications. WB bin generation. Arterioscler. Thromb
126(20):133, 2002. Saunders, Philadelphia, 2002, pp 71-753. Vasc Biol 22:1381, 2002.
. Liu, MC, and Kessler, CM: A systematic . Dyskiewica-Korpanty, AM, and Sarode, 32. Maugeri, N, et al: Human polymorphonu-
approach to the bleeding patient. In R: Abstract: Aggregation and platelet clear leukocytes produce and express
Kitchens, CS et al (eds): Consultative function analyzer (PFA-100) in detecting functional tissue factor upon stimulation.
Hemostasis and Thrombosis. WB Saun- aspirin effect on platelet function. J Am J Thromb. Haemost 4:1323, 2006.
ders, Philadelphia, 2002, pp 181-196. Soc Hematol 100:11, 2002. . Engelmann, B, et al: Intravascular tissue
5 Lichtman, M, et al: William’s Hematology, 20. Dyskiewica-Korpanty, AM, et al: factor pathway: A model for rapid initia-
ed 7. McGraw-Hill, Columbus, OH, Approach to the assessment of platelet tion of coagulation within the blood
2006. function: Comparison between vessel. J Thromb. Haemost 89:3, 2003.
6 Hillman, R, et al: Hematology in Clinical optical-based platelet-rich plasma and 34. Butenas, S, et al: Tissue factor activity in
Practice, ed 4. McGraw-Hill, Columbus, impedance-based whole blood platelet whole blood. Blood. 2005: 105: 2764-
OH, 2005S. aggregation methods. Clin Appl 2770.
—. Panes, O, et al: Human platelets synthe-
Thrombos/Hemostas 11(1):25, 2005. 315, Mackman, N: Role of tissue factor in
size and express functional tissue factor. 2 ren. Ruggeri, ZM, et al: Activation- hemostasis, thrombosis, and vascular
Blood 109 (12):5242, 2007. independent platelet adhesion and development. Arterioscler Thromb Vasc
oo. Weyrich, A, et al: Platelet tissue factor
aggregation under elevated shear stress. Biol 24:1015, 2004:
comes of age. Blood 109(12):5069, Blood 108:1903, 2006. 36. Day, SM, et al: Macrovascular thrombo-
2007. Ppl Undas, A, et al: Antithrombotic proper- sis is driven by tissue factor derived
.Zillman, A, et al: Platelet-associated primarily from the blood vessel wall.
ties of aspirin and resistance to aspirin:
tissue factor contributes to the collagen- Beyond strictly antiplatelet actions. Blood 105:102, 2005.
triggered activation of blood coagula- Blood 109: 2285, 2007. . Roemisch, J, et al: Blood Coagulat
tion. Biochem Biophys Res Commun . Konrad, CJ, et al: Duration of effects of Fibrinolysis 13(8):657, 2002.
281(2):603, 2001. aspirin on platelet function in healthy 38. Mosnier, LO, and Bouma, BN: Regula-
. Wintrobe, MM: In Greer JP (ed):
volunteers: an analysis using the PFA- tion of fibrinolysis by thrombin activat-
Wintrobe’s Clinical Hematology, ed 11. 100. J Clin Anesth 18(1):12, 2006. able fibrinolysis inhibitor, an unstable
Lippincott Williams & Wilkins, 24. Ley, EI, et al: Aspirin and clopidogrel carboxypeptidase B that unites the path-
Philadelphia, 2004. drug response in patients undergoing ways of coagulation and fibrinolysis.
. Dopheide, SM, et al: Shear-dependent
percutaneous coronary intervention: The Arterioscler Thromb Vasc Biol
tether formation during platelet translo- role of dual drug resistance. J AM Coll 26(11):2445, 2006.
cation on von Willebrand factor. Blood Cardiol 47(1):27, 2006. 3H. Jensen, R: Oral anticoagulation and the
99:159, 2002. . Magy, N: Protein Z: A new factor of INR. Clin Hemost Rev 13(5), 1999.
. Shattil, SJ, and Newman, PJ: Integrins:
thrombophilia? Rev Med Int 26(11):891, 40. Tripodi, A, et al: International collabora-
dynamic scaffolds for adhesion and 2005. tive study for the calibration of a
signalling in platelets. Blood 104:1606,
. Tapper, H, and Herwald, H: Modulation of proposed reference preparation for
2004.
hemostatic mechanisms in bacterial infec- thromboplastin human recombinant,
. Maxwell, MJ, et al: Identification of a
tious diseases. Blood 96(7):2329, 2000. plain; on behalf of the subcommittee on
two-stage platelet aggregation process
. Wolberg, AS, et al: Impact of procoagu- control of anticoagulation. Thrombos
mediating shear-dependent thrombus
lant concentration on rate, peak and total Hemostas 79(2):439, 1998.
formation. Blood 109:566, 2007.
thrombin generation in a model system.
. Ahmad, SS, et al: The assembly of the
J Thrombos Haemostas 2(3), 402, 2004.
factor X-activating complex on activared
. Xu, H, et al: Thrombomodulin changes
human platelets. J Thrombos Haemostas
the molecular surface and interaction and
1:48, 2003.
the rate of complex formation between
Chapter —
Disorders of Primary
Hemostasis
Quantitative and Qualitative Platelet
Disorders and Vascular Disorders
Darla K. Liles, MD
Charles L. Knupp, MD
Introduction OBJECTIVES
Laboratory Evaluation At the end of this chapter the learner should be able to:
of Disorders of Primary
Hemostasis 1. Describe the laboratory tests that may be utilized in the evaluation of quantitative and
qualitative platelet disorders.
Quantitative Platelet
Disorders: . 2. Describe the pathophysiologic processes that cause thrombocytopenia. List the
Thrombocytopenia thrombocytopenic disorders caused by each process.
Deficient Platelet Production
ee
Abnorinal. Distribution of 3. Define 1 -medi
efine immune-mediated :
thrombocytopenia
Platelets 4 . List conditions that are associated with autoimmune and alloimmune thrombocytopenia.
Increased Destruction of ! bea ese
Secs SSUUCHO ORO P). Describe how the diagnosis of idiopathic thrombocytopenic purpura (ITP) is made.
6. Compare ITP and thrombotic thrombocytopenic purpura (TTP).
Quantitative Platelet
3
Disorders: Thrombocytosis . Compare thrombotic thrombocytopenic purpura (TTP) and hemolytic uremic
Primary Thrombocytosis syndrome (HUS).
Reactive Thrombocytosis
8. List the characteristics of the inherited platelet membrane defects and
Qualitative Platelet Disorders compare Bernard-Soulier syndrome to Glanzmann’s thrombasthenia.
Congenital Disorders of
Platelet Function 9. Describe the types of von Willebrand disease and how they differ in the
von Willebrand Disease management of bleeding.
Acquired Qualitative Platelet 1(. Differentiate von Willebrand disease from Bernard—Soulier syndrome and
Disorders hemophilia A.
Vascular Disorders 11. Describe the pathophysiology responsible for storage pool and platelet release
Primary Purpura
defects and how it relates to laboratory studies used for diagnosis of these disorders.
Secondary Purpura
12. Differentiate between reactive and primary thrombocytosis and describe the
Vascular and Connective
hemostatic problems expected.
Tissue Disorders
Case Study 1
Case Study 2
Case Study 3
Case Study 4
Sve
578 Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders
Because von Willebrand disease mimics the bleeding platelet surface and release defects can be identified with this
diathesis of platelet disorders, assays to exclude this diagno- test. Drugs and disorders such as hepatic or renal disease can
sis are an integral part of the evaluation. To determine the interfere with platelet function and make interpretation of this
presence or absence of von Willebrand disease, factor VIII test difficult. Lumi-aggregation or °'Cr release assays may be
coagulant activity, von Willebrand’s antigen (VWF:Ag), von useful to specifically measure platelet release.
Willebrand’s activity by ristocetin-induced platelet aggrega- Bone matrow aspiration and biopsy are useful in deter-
tion, and von Willebrand multimers must be assayed mining the etiology of quantitative platelet disorders. The
together. Further discussion of these tests is provided later in bone marrow specimen can be used to assess adequacy of
this chapter in the section on von Willebrand disease. megakaryocytes, overall cellularity, and to identify infiltrative
Platelet antibody testing determines the amount of processes such as myelodysplastic syndromes, malignancy, or
immunoglobulin G (IgG) bound on the platelet surface fibrosis.
by various methodologies. Increased amounts of platelet-
associated IgG are often found in immune-mediated throm-
bocytopenias, but this finding is not specific enough to Quantitative Platelet Disorders:
establish a diagnosis of an immune origin. Flow cytometry Thrombocytopenia
can be used to measure platelet surface glycoproteins and
Platelets must be present in adequate numbers to maintain
platelet-bound IgG.
normal hemostasis. Platelet production remains relatively
Platelet aggregation assesses platelet function by mea-
constant for an individual over time. The body senses the
suring the response of the platelet to various stimuli such as
total platelet mass, related both to number and mean volume
epinephrine, adenosine diphosphate (ADP), collagen, throm-
of platelets, and regulates this tightly. Thrombopoietin (TPO),
bin, and ristocetin. The procedure for performing platelet
a growth factor produced by the liver and to a lesser degree
aggregation is described in Chapter 33. Abnormalities of the
by the spleen, is responsible for this regulation. When the
platelet mass is normal, TPO is cleared from plasma by TPO
receptors on megakaryocytes. With thrombocytopenia, the
overall clearance is low and the plasma concentration of TPO
increases to boost megakaryocyte and platelet production.
ay peaBroce” The average platelet count ranges from 150 to 400 10°/L of
of Primary Hemostasis whole blood. Thrombocytopenia is defined as a platelet count
below the lower limit of normal, although clinical signs and
Platelet count
Peripheral blood smear symptoms of thrombocytopenia typically are not manifested
Ivy bleeding time or PFA-100 until the platelet count falls below 100 x 10°/L and usually
Von Willebrand studies not until the platelet count falls below 50 X 10°/L. Overt
FVUI:C spontaneous hemorrhage is not usually seen until the platelet
vWE antigen
count falls to less than 20 X 10°/L. Platelet counts less than
vWF activity
Platelet antibody testing 10,000 may result in life-threatening hemorrhage and may
Flow cytometry require emergency platelet transfusions. Quantitative platelet
Platelet glycoprotein analysis disorders are the most commonly encountered group of
Platelet-associated IgG platelet abnormalities and can be simply divided into two
Platelet aggregation studies
categories, thrombocytopenia and thrombocytosis. Thrombo-
Lumiaggregometry
*1Cr release cytopenia results from three distinct mechanisms: deficient
_Bone marrow aspiration aud biopsy platelet production, abnormal platelet distribution (splenic
sequestration), or increased destruction (Table 25-4). Qualita-
“EVIL C= acter VIE: = yon Willebrand factor;
VWF =
PFA= platelet function analyzer. tive platelet defects may coexist with quantitative platelet
defects to increase bleeding risk.
580 Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders
with more chronic form, which is treated like the more common
Table 25-5 Congenital Disorders. adult form of ITP, except that splenectomy is often avoided
_
| Associated with— because of the infectious complications of splenectomy in
ROUSE: - Decreased Platelet young children.
ee - Production | a
Adult ITP
eDisonden: mssociated Abnormalities In adults, ITP commonly presents in the 20- to 50-year-old
group, as a chronic disease process with a greater predilection
Alport’s stuns Giant aieicn
Thrombocytopenia for women. Occasionally patients will have an acute immune
Deafness thrombocytopenia after a viral illness or exposure to drugs,
Nephritis but this is uncommon. In most cases of adult immune throm-
Chédiak-Higashi Partial oculocutaneous bocytopenia, there is usually not a recent history of drug
syndrome albinism
exposure or infectious illness that can be related to the onset
Increased susceptibility to
pyogenic infections
of thrombocytopenia. Platelet counts are typically less than
Storage pool defect of dense 30 X 10°/L in patients who present with bleeding manifesta-
granules tions. Clinically, patients present with mucosal bleeding
Hermansky-Pudlak Platelet deficiency of typical of a primary hemostatic defect, such as menorrhagia,
syndrome nonmetabolic ADP epistaxis, easy bruisability, or petechiae (Figs. 25-1 and
Oculocutaneous albinism
May-Hegglin anomaly Thrombocytopenia 25-2). Some patients are diagnosed while still asymptomatic
Giant platelets based on a low platelet count on a routine complete blood
Dohle bodies count obtained for other reasons. These patients usually have
TAR syndrome Multiple skeletal and cardiac a platelet count greater than SO < 10°/L. Adult ITP does not
abnormalities usually remit spontaneously.
Storage pool defect
Wiskott-Aldrich Disorders of dense granules
syndrome Recurrent pyogenic infections Common Clinical Findings
Eczema The bone marrow is characterized by increased or normal
Thrombocytopenia numbers of megakaryocytes (Fig. 25—3). Platelet life span is
shortened and circulating platelets are morphologically large
on the smear, reflecting the early release from the marrow in
Increased Destruction of Platelets response to the peripheral destruction. There are also changes
in the splenic microcirculation that cause a reduction of
PRIMARY IMMUNE-MEDIATED THROMBOCYTOPENIAS platelet transit time in the spleen and increased destruction of
This group of thrombocytopenias all have an immune-mediated the antibody-coated platelets.’ The bleeding time in ITP may
mechanism by which there is increased platelet destruction that not be as prolonged as the bleeding time in other disorders
can be primary (idiopathic) or secondary to an underlying associated with the same degree of thrombocytopenia, sug-
disease (see Table 25-4). gesting that the younger platelets in ITP may be more effec-
tive at maintaining hemostasis.
IDIOPATHIC THROMBOCYTOPENIC PURPURA Idiopathic Because ITP is a diagnosis of exclusion, other causes of
or immune thrombocytopenic purpura (ITP) is one of the thrombocytopenia must be considered and eliminated first.
most common disorders causing severe isolated thrombocy- Drugs that may cause thrombocytopenia must be stopped, and
topenia and is caused by an autoantibody to the patient’s
platelets. There is not a specific test that readily confirms the
diagnosis of ITP, so it is typically a diagnosis of exclusion.
ITP can present in children and adults; however, there are
important differences between the two groups when compar-
ing the long-term prognosis.°
Childhood ITP
Young children may present with an immune thrombocytopenia
that typically develops acutely with a |- to 2-week duration,
usually with bruising or petechiae. Serious bleeding is uncom-
mon. Most children present with an initial platelet counts of less
than 20 X 10°/L. Bone marrow aspiration and biopsy is often
performed to exclude the diagnosis of acute leukaemia. This dis-
order is usually self limited. Spontaneous remissions, with or
without therapy, occur in the majority of these patients. How-
ever, immunoglobulin G and corticosteroids are often used to Figure 25-1 @ Oral cavity of a patient with idiopathic thrombocy-
decrease the period of thrombocytopenia. A minority of children topenic purpura (ITP).
582 Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders
used to help control bleeding until the platelet count can be To confirm the presence of a platelet-specific antibody, a panel
corrected. These agents are safe to use either before or after with Pl‘'-positive cells should be run. The patient’s platelets
splenectomy and are most effective at sites of high fibrinolytic should also be phenotyped but the patient’s own blood type
activity (i.e., urinary tract, nose, and mouth). Platelet transfu- will not be evaluable until after recovery. Confirmation that
sions are usually not effective because transfused platelets are the patient is PI‘! negative provides additional support for the
usually rapidly destroyed; however, in some patients a platelet diagnosis. In some cases of PTP, isosensitization to HLA anti-
response may occur and bleeding may improve.'? gens found on platelets occurs, as well as the appearance of
platelet-specific antigens other than PI‘', making serologic
POSTTRANSFUSION PURPURA (PTP) In this disorder, sud- typing difficult.
den onset of thrombocytopenia occurs approximately | week Treatment with platelet transfusions is usually not
after transfusion of blood or blood products containing effective as the alloantibodies destroy even PIl*!-negative
platelets (Fig. 25-4). The majority of cases are a result of an platelets. Plasmapheresis without exchange and intravenous
alloantibody directed against the platelet antigen PI4', also IgG infusions have each been effective means of treating the
referred to as HPA-1a.'* The PI“! antigen is found in approx- hemorrhagic complications associated with PTP. In a num-
imately 97% of the normal population; the 3% of people who ber of cases, patients have had repeated episodes of PTP
lack the PI*' (HPA-1a) antigen on their platelets are consid- following re-exposure to PI‘'-positive blood, but some do
ered at risk for developing PTP. It is believed that posttrans- not. Pl‘'-negative blood is indicated for all subsequent
fusion purpura (PTP) results from an anamnestic immune transfusions when possible because patients are considered
response from prior exposure to the antigen. Most reported at risk for recurrence of PTP with subsequent transfusions.
cases have been in middle-aged women who have had chil-
dren. It is believed that primary immunization occurs during ISOIMMUNE NEONATAL THROMBOCYTOPENIA Similar
pregnancy, when PI*'-positive fetal platelets sensitize a to the pathogenesis of erythroblastosis fetalis, isoimmune
Pl*'-negative mother (see the following discussion of isoim- neonatal thrombocytopenia results from immunization of the
mune neonatal thrombocytopenia). Other mechanisms for the mother by fetal platelet antigens and placental transfer of
development of PTP have been suggested.'* Why these alloan- maternal antibody.'® Isoimmune neonatal thrombocytopenia
tibodies destroy the patients PI*!-negative platelets is not clear. is most often caused by maternal alloantibodies to the PI*'
Recycling of antigen from destroyed platelets onto the (HPA-la) antigen: however, as in posttransfusion purpura,
patient’s platelets may explain this phenomenon. Rare reports isoimmune neonatal thrombocytopenia has been reported
of cases of PTP have occurred in association with other only rarely with other platelet antigens such as Pl**, Bak’,
platelet antigens, including Pl**, Bak*, Bak”, Pen*, and Br*.'° Bak”, Br*, and Br®.'> It is an uncommon disorder, generally
Complement fixation, release of °'Cr, or '*C-serotonin affecting the first-born child. Based on gene frequency of the
have been some of the reliable laboratory tests used to detect PL“! (HPA- 1a) antigen in fathers, there is a high probability
and measure anti-Pl*! (HPA-1a) antibodies in PTP. Currently, that a Pl*'-negative mother will have a Pl*!-positive child.
direct and indirect laboratory tests have been developed Once isoimmune neonatal thrombocytopenia has developed,
to increase specificity and sensitivity in the detection of there appears to be an increased risk of the next child being
platelet antibodies. These tests employ some of the following affected, because most fathers are homozygous for PI*'.
techniques: enzyme-linked immunosorbent assay (ELISA), A large percentage of PI*'-negative mothers who give birth
Western blot followed by ELISA or radioimmunoassay to an affected child are phenotype-positive for the HLA-B8
(RIA), platelet suspension immunofluorescence, and immuno- antigen. It has been suggested that the HLA-B8 antigen serves
precipitation of radiolabeled glycoprotein Hla (GPIIIa). to protect from immunization, which accounts for the rela-
tively low incidence of isoimmune neonatal thrombocytope-
nia, despite the frequency of the PI*! antigen and the chance
for maternal sensitization. The relationship of ABO incompat-
ibility to symptomatic isoimmune neonatal thrombocytopenia
is unclear.
Infants who develop isoimmune neonatal thrombocy-
topenia appear normal at birth but within hours develop scat-
tered petechiae and purpuric hemorrhages, with platelet
counts below 30 X 10°/L. Intracranial hemorrhage is the pri-
mary cause of mortality in these infants. Characteristically, in
this disorder the platelet count begins to decrease shortly
after birth with low levels reached several hours later.
Therapy is aimed at preventing intracranial hemorrhage
and keeping platelet counts at hemostatically safe levels. Cae-
sarean delivery is usually performed to prevent intracranial
hemorrhage from birth trauma when the disorder is suspected
prior to delivery. Corticosteroids or intravenous IgG may be
Figure 25-4 m Posttransfusion purpura (PTP). used prior to delivery. Postnatal treatment is not necessary if
584 Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders
the infant is asymptomatic and the platelet count remains after initial exposure to heparin; however, in individuals who
above 30 X 10°/L. When the infant manifests clinical signs of have been previously exposed to heparin it can develop within
bleeding and the platelet count falls below 10 < 10°/L, com- | to 3 days after re-exposure. Patients with myocardial infarc-
patible platelet transfusions utilizing maternal platelets or tion and cardiogenic shock or those who have undergone major
P]*!-negative donor platelets is the preferred treatment. vascular surgery may be particularly susceptible. Venous or
arterial thrombosis results in an incidence of morbidity and mor-
DRUG-INDUCED IMMUNE THROMBOCYTOPENIA Drugs tality of 20% of the patients who develop HITTS (see Chap. 28).
may also cause thrombocytopenia. Quinine has been recog- Pathologically, this syndrome develops secondary to anti-
nized as one of the most frequent causes of drug-induced body produced to platelet factor 4-heparin complex with
thrombocytopenia since the first report in 1928.'° The drug immune complex Fe receptor-induced activation of platelets.'”°°
appears to act as a hapten, eliciting an antibody response Laboratory confirmation of HITTS is difficult. Origi-
when complexed with a larger carrier molecule. Antibody nally the test available to confirm HITTS was platelet aggre-
binding with drug appears to be the initial step in the forma- gation test utilizing donor platelets mixed with patient serum
tion of the complex. Cellular binding or adsorption of the after the patient has been off heparin for 8 hours or longer.
antibody—drug complex to the platelet membrane via the Heparin is added to the patient samples at low and high con-
antibody Fab region results in platelet injury and splenic centrations, and platelet aggregation is observed in the
sequestration. Removal of these platelets by the spleen results heparin sample compared to a control sample without
in thrombocytopenia. heparin. In patients with HITTS the aggregation test should
The list of drugs that have been implicated to cause show aggregation of platelets at the low concentration of
immune drug purpura is rather extensive. However, many of heparin but not at the high concentration of heparin or the
these reports do not provide sufficient evidence to prove causa- sample without heparin. This test is approximately 40% sen-
tion. The drugs most frequently cited are quinine, quinidine, sitive but 90% specific for confirming the diagnosis of
salicylates, thiazides, and sulfa drugs.'’ When possible, any sus- HITTS. Platelet release assays increase the sensitivity of
pect drug should be stopped in a thrombocytopenic patient. detection. Aggregation and release assays are not readily
Drug-induced immune purpura appears to occur more available in many hospitals. Newer tests utilizing flow cytom-
frequently in the elderly population as a result of the etry or ELISA are even more sensitive for the diagnosis of
increased usage of medication; however, cases have been HITTS and can be performed in the presence of heparin.
reported in children and young adults. Purpura occurs approx- ELISA testing is usually more readily available for clinical
imately 7 days after initial use of the drug but may occur laboratories. However, this method may detect the presence
within 3 to 5 days owing to an anamnestic response on of antibody that has not caused the syndrome in patients with
re-exposure to the drug. It is estimated that 1 in 100,000 indi- thrombocytopenia resulting from other causes. Using a suffi-
viduals using prescription medication per year will require ciently high optical density cutoff has led to much greater sen-
hospitalization as a result of a drug-induced blood disorder. sitivity and has resulted in use of ELISA as the most common
The frequency for users of quinine and quinidine is approxi- laboratory method used to diagnose the disorder.?!
mately | in 1000. The disorder is generally self-limiting In patients with suspected HITTS, heparin must be dis-
because the platelet count returns to normal once the drug has continued. The platelet count should return to normal within
been removed from circulation. Re-administration of a drug 4 to 6 days. Alternative anticoagulation is recommended with
known to cause purpura should be avoided. or without thrombotic complications in any individual in
whom the diagnosis of HITTS is strongly suspected. In some
HEPARIN-INDUCED THROMBOCYTOPENIA instances, patients have been given warfarin without intra-
AND THROMBOSIS venous anticoagulation; however, recent reports have sug-
Heparin therapy is associated with the development of two dis- gested warfarin may increase the risk for skin necrosis and
tinct types of thrombocytopenia. One type develops early in venous limb gangrene in this syndrome.?? In patients who
treatment and is benign. The platelet count rarely falls below have life- or limb-threatening thrombosis, there are a limited
100 < 10°/L, and there are no resultant bleeding or thrombotic number of choices for intravenous anticoagulation that all
complications. The second type is associated with severe throm- have potential hemorrhagic risk.% In the past, Dextran has
bocytopenia and paradoxically thrombotic episodes instead of been administered; however, there is no laboratory measure
hemorrhagic complications (see Chap. 28). Initially, it may be other than clinical observation to measure its efficacy.
difficult to distinguish between these two types of thrombocy- Dextran coats circulating red blood cells, complicating type
topenia based on laboratory values alone. and crossmatch for red cell transfusion products. Some data
In the second type of thrombocytopenia, platelet counts as now suggest that Dextran may be detrimental; therefore it is
low as 20 * 10°/L occur in association with arterial and venous no longer used. Newer agents that are approved for treatment
thrombosis; this type has been termed heparin-induced throm- in this setting include the direct thrombin inhibitors lep-
bocytopenia and thrombosis syndrome (HITTS). The actual thirudin and argatroban.*> Lepthirudin is the recombinant
incidence of this syndrome is not well defined, but it probably form of the leech salivary gland protein hirudin. Argatroban
occurs in fewer than 1% of patients receiving heparin.'’ It has is a synthetic peptide direct thrombin inhibitor. Bivalirudin,
been reported more often with bovine heparin administration a synthetic derivative of hirudin, has been used off-label
than with porcine heparin. HITTS typically develops 4 to 7 days and has a somewhat shorter half-life than lepthirudin and
Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders 585
argatroban. Fondaparinux, which is the synthesized pen- The pathogenesis of HIV-related thrombocytopenia
tasaccharide portion of the heparin molecule, does not appears to be heterogeneous. An immune etiology is suggested
appear to cross-react with heparin in the manner that by studies which have shown a diminished in vivo platelet sur-
low-molecular-weight heparins can in promoting the clini- vival. Detection of platelet-associated IgG and circulating
cal syndrome. The bleeding that may occur with these treat- immune complexes that contain antiplatelet glycoprotein anti-
ments may not be easily corrected. Fortunately, the direct bodies provides support for immune destruction in HIV-related
thrombin inhibitors have short plasma half-lives so their thrombocytopenia. The pattern of IgG subclasses found in HIV-
effects resolve relatively quickly. Fibrinolytic therapy may infected patients, as well as the level of anti-[gG immune com-
play a treatment role to dissolve thrombus, especially in plexes on platelet surfaces in HIV disease, are significantly
individuals who develop neurovascular compromise related different from patients with “classic ITP” (Table 25-6).
to their thrombosis. Immunohistochemical markers show increased CD8+ T cells
in the spleens of patients with HIV-related immune thrombo-
ACQUIRED SECONDARY IMMUNE-MEDIATED cytopenic purpura. However, the finding that thrombocytope-
THROMBOCYTOPENIA nia does not frequently occur in infants of HIV-infected
LYMPHOPROLIFERATIVE DISORDERS/COLLAGEN mothers with thrombocytopenia suggests an immune etiology
VASCULAR DISORDERS Lymphoproliferative disorders may not be the principal cause. Viral infection of hematopoi-
such as Hodgkin’s disease and non-Hodgkin’s lymphoma, as etic cells, altered marrow microenvironment or dysfunction
well as other hematologic malignancies such as chronic lym- of the reticuloendothelial system contribute to ineffective
phocytic leukemia, have been reported with an ITP-like thrombopoiesis in HIV-related thrombocytopenia. Develop-
thrombocytopenia associated with decreased platelet survival. ment of marrow fibrosis or marrow involvement by AIDS-
In systemic lupus erythematosus (SLE), roughly 14% of the related lymphoma may also lead to thrombocytopenia. These
patients develop thrombocytopenia resembling ITP during the factors may be more important in producing HIV-related
course of the disease. The hematologic manifestations of thrombocytopenia.
SLE, which include immune-mediated thrombocytopenia and Treatment with antiretroviral therapy alone is often
thrombocytopenia secondary to bone marrow suppression, effective.** HIV-related thrombocytopenia which is more severe
may precede the other clinical manifestations of the disease. or is associated with bleeding is treated similarly to ITP. Intra-
Thrombocytopenia resulting from either etiology in SLE venous IgG and anti-D globulin have been used successfully.
responds well to corticosteroid therapy. Corticosteroids may be effective but have the potential to
increase the risk of infection in these immunosuppressed indi-
VIRAL INFECTIONS Viral infections may also transiently viduals. Splenectomy may also be effective. As there seems to
impair megakaryopoiesis without a reduction in marrow be increased morbidity with use of Rituximab, when added to
cellularity. Chronic viral infections such as human immun- standard chemotherapy for the treatment of HIV-related non-
odeficiency virus (HIV) or hepatitis may lead to marrow Hodgkin’s lymphoma, its use in HIV-related thrombocytopenia
hypocellularity and, subsequently, to thrombocytopenia in cannot be recommended.
affected individuals.
Thrombocytopenia as a result of acute viral, bacterial, or MICROANGIOPATHIC THROMBOCYTOPENIA
parasitic infections has been well documented. An immuno- THROMBOTIC THROMBOCYTOPENIC PURPURA Throm-
logic mechanism appears likely in the development of throm- botic thrombocytopenic purpura (TTP) is a rare and often
bocytopenia as a result of infection.*°’’ Viral infections such fatal syndrome associated with thrombocytopenia and
as mononucleosis, mumps, and rubeola may be complicated microangiopathic hemolytic anemia (Fig. 25-5). Despite being
by severe thrombocytopenia. In bacterial sepsis; thrombocy- recognized since 1924, the pathogenesis of this syndrome
topenia may be present with or without disseminated intravas-
cular coagulation (DIC). Malaria is frequently associated with
thrombocytopenia as a result of increased destruction and
splenic sequestration.
in these patients is thrombocytopenia. The incidence of throm- Age at onset Child Adult
bocytopenia appears to vary according to the stage of the dis- Previous infection Yes No
ease. There appears to be a correlation between CD4+ T cell Platelet count Usually Usually 30—50,000
depletion, viral load in plasma and the occurrence of thrombo- = 20,000
Bleeding episode Abrupt Slow
cytopenia. Many affected patients also have active hepatitis Duration of Transient Prolonged
infection. Most patients with HIV-related thrombocytopenia thrombocytopenia
do not have significant bleeding Hemorrhagic complications Spontaneous Yes No
may occur but are difficult to predict based solely on the remission
platelet count.
586 Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders
than Bernard—Soulier syndrome, another surface glycoprotein 2. Normal platelet count and normal platelet morphology,
abnormality. Glanzmann’s thrombasthenia appears to cluster in 3. Prolonged bleeding time,
ethnic populations where consanguinity is prevalent. Bleeding 4. Absent platelet aggregation to ADP, thrombin, collagen,
is most commonly from mucosal surfaces and includes easy and epinephrine, with normal platelet agglutination to
bruisability, epistaxis, spontaneous gingival bleeding, prolonged ristocetin (Fig. 25-7);
bleeding from minor cuts, and menorrhagia. Gastrointestinal 5. Flow cytometric assessment of GPIIb/IIIa surface protein,
hemorrhages are less common. Facial petechiae and subcon- revealing deficiency or absence;
Junctival hemorrhages may be seen in infants associate with 6. Normal lumi-aggregation and °'Cr release; and
crying. The formation of deep hematomas and _ recurrent 7. Abnormal clot retraction.
hemarthroses that are typical features in hemophilia are not
Treatment
present in Glanzmann’s thrombasthenia.
Prevention of bleeding with good dental care and avoidance
Diagnostic Criteria of antiplatelet drugs is important. Patients with Glanzmann’s
Criteria for diagnosing Glanzmann’s thrombasthenia include: thrombasthenia who present with severe bleeding episodes
require platelet transfusions to replace dysfunctional platelets.
1. An autosomal-recessive trait with clinical manifestations Supportive therapy should be used judiciously, because
expressed in homozygotes only; patients may develop alloantibodies to GPIb and GPIIla on
transmittance
Percent
30
8 10 12 14
Time (min)
Figure 25-7 m This graph depicts the lack of platelet aggregation to e pinephrine, 10 um (blue); ADP, 5 um
(black); collagen, 2 ug/mL (red);,
and arachidonic acid, 0.5 g/mL (green)—typical of a patient with Glan zmann’s thrombasthenia.
Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders 591
the transfused platelets or anti-HLA antibodies and become (which is the receptor for vWF) is the cause of the aggregation
refractory to platelet transfusions. Antifibrinolytic agents such defect. 25-The differences in platelet aggregation tests with
as EACA may help in controlling hemorrhage, especially various platelet agonists among von Willebrand’s disease,
from the nose and mouth where fibrinolytic activity is promi- Bernard—Soulier syndrome, and Glanzman’s thromboasthenia
nent. Topical measures such as thrombin or pressure packing are depicted in Table 25-10. Crossed immunoelectrophoresis
may be effective. DDAVP has been used but has not been or flow cytometry of platelet membrane glycoproteins should
helpful. Estrogen therapy in the form of birth control pills is demonstrate a decrease in GPIb, GPIX, and GPV to confirm a
especially useful in controlling menorrhagia. Recombinant diagnosis of Bernard—Soulier syndrome.
factor VIIa has been successfully used but thromboembolism Heterogeneity among the glycoprotein abnormalities in
has occurred in one reported case after discontinuation of the Bernard-Soulier indicates that there are multiple genetic
recombinant factor VIIa treatment. defects. Heterozygotes present with recognizable abnormalities
such as occasional large platelets on the peripheral smear, with
BERNARD-SOULIER SYNDROME Bernard—Soulier syn- a history of bleeding yet few clinical problems. Homozygotes
drome is a rare autosomal-recessive bleeding disorder caused present with abnormal platelet function and morphology,
by a deficiency of the platelet GPIb/IX complex. GPV, which thrombocytopenia, and hemorrhagic disease.
is associated with the GPIb/IX complex on the platelet sur-
face and is a thrombin substrate, has also been documented to Diagnostic Criteria
be deficient in Bernard—Soulier platelets. Gene mutations of Rather specific criteria for the diagnosis of this bleeding syn-
GPIb and GPIX, but not GPV, appear to be the cause of this drome have been established. These criteria are:
disorder. An acquired form of the disorder has been reported
1. An autosomal trait with clinical manifestations expressed
due to autoantibodies to these platelet glycoproteins. As dis-
in homozygotes (or double heterozygotes with combined
cussed in Chapter 24, GPIb/IX plays a major role in various
genetic abnormalities of GPIb, GPV, and GPIX)
hemostatic events. GPIb/IX is the platelet receptor involved in
2. Normal platelet count or moderate thrombocytopenia
the vWF-dependent contact adhesion of unactivated platelets
(despite a normal number of marrow megakaryocytes)
to exposed subendothelium at high shear rates and for the
3. A significant number of giant or large platelets present on
binding of platelets to fibrin. It also serves as a high-affinity
the peripheral blood smear and prolonged bleeding time in
thrombin-binding site and regulates platelet shape and reac-
excess of the degree of thrombocytopenia
tivity. Because the platelet membrane is attached to the
4. Absent platelet agglutination in response to human vWF
cytoskeleton via GPIb, the loss of normal membrane—
and ristocetin
cytoskeletal function may account for the abnormal platelet
5. Normal platelet aggregation in response to ADP, collagen,
morphology seen in Bernard—Soulier platelets.” Bleeding in
and epinephrine, with reduced aggregation in response to
Bernard—Soulier syndrome is due to a combination of hemo-
thrombin
static defects including thrombocytopenia, abnormality of
6. Normal factor VIII coagulant activity (FVII:C) and vWF
platelet-von Willebrand’s factor interaction important for
antigen
platelet adhesion, abnormality of platelet-thrombin interac-
tion, and abnormality of platelet coagulant activity.
Treatment
Affected individuals who present with active bleeding should
Clinical Manifestations and Laboratory Results
be treated with red blood cell transfusions to replace blood
Patients with Bernard—Soulier syndrome present with the typ-
loss and an antifibrinolytic agent such as EACA. Antifibri-
ical symptoms of a primary hemostatic disorder with varying
nolytic agents do not correct the defect but only allow the pri-
severity. Gingival bleeding, epistaxis, purpura, menorrhagia,
mary hemostatic plug to remain intact at the site of injury.
and gastrointestinal bleeding are the typical hemorrhagic
DDAVP has been reported to shorten the bleeding time in
manifestations. Symptoms occur early in life and have a ten-
some individuals*® but not in others. Estrogen therapy may be
dency to decrease with age.
helpful in controlling bleeding.*°*! In instances in which
Laboratory abnormalities in patients with Bernard—
bleeding is not controlled by these measures, platelet transfu-
Soulier syndrome include normal or moderately reduced
sions may be necessary; however, these should be used judi-
platelet counts with large irregularly shaped platelets noted on
ciously to avoid alloimmunization to GPIb. There has been
the peripheral smear, and a prolonged bleeding time. The
successful use of recombinant factor VIla to treat bleeding in
platelets seen on peripheral blood smear may be large enough
this disorder
to resemble lymphocytes. Normal numbers of megakary-
ocytes are found on bone marrow examination. Platelet
aggregation is normal with ADP, epinephrine, and collagen, PLATELET RELEASE (SECRETION) DEFECTS
and is reduced with thrombin. Ristocetin-induced platelet As described in Chapter 24, platelet glycoprotein receptors
aggregation is absent in Bernard—Soulier syndrome, similar to transmit signals intracellularly to cause release of platelet
von Willebrand’s disease. However, the deficient ristocetin- granule substances and platelet shape change. Phospholi-
induced aggregation defect is corrected by the addition of pases (A and C) are liberated from the internal surface of
normal plasma in von Willebrand’s disease, but not in the platelet membrane in response to platelet agonists
Bernard-Soulier syndrome, where the deficiency of GPIb/IX and cause an increase in intracellular calcium levels. This
592 Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders
vWD = +
BSS +
GT = -
produces centralization of granules and fusion of granule Laboratory features of storage pool deficiencies include
membrane with the open canalicular system to transport a prolonged or normal template bleeding time. Aspirin inges-
granule proteins to the environment to allow adhesion tion may cause a marked prolongation of the bleeding time.
and aggregation. The disorders of platelet release primarily Plasma coagulation screening tests and factor assays are
involve absence of platelet granules or defective enzymatic normal in these disorders. Platelet aggregation testing in
pathways. 8 storage pool deficiency demonstrates normal primary wave
aggregation but absent or reduced secondary wave aggrega-
STORAGE POOL DEFICIENCIES (GRANULE DEFECTS) tion in response to ADP and epinephrine. Collagen-induced
Storage pool deficiencies are classified according to an analy- aggregation is reduced at lower but not high collagen concen-
sis of the granule proteins and the morphological appearance trations. High concentrations of thrombin do not produce nor-
of the platelets. Most commonly, a decrease in platelet dense mal release, in contrast to platelet secretion defects. Platelet
granules is present, with decreased amounts of secretable aggregation testing is variable in a storage pool deficiency.
ADP, adenosine triphosphate (ATP), calcium, and serotonin ADP and epinephrine-induced aggregation are often normal.
(delta [5] storage pool deficiency). In other patients, alpha (a) Collagen and thrombin-induced aggregation are more fre-
granules and dense granules are decreased, and decreases in quently abnormal. Platelet aggregation patterns in a,5 storage
amounts of a-granule proteins such as platelet factor 4, beta pool deficiency are similar to ad storage pool deficiency as
(6)-thromboglobulin, and platelet-derived growth factor as the deficiency in 6 granules appears more severe than the
well as the dense granule proteins are noted (a6 storage pool deficiency of a granules in these individuals. Platelet counts
deficiency). Other patients have a decrease in @ granules only, are normal in the storage pool deficiencies. Morphology is
with normal dense granules and normal amounts of dense normal except in the gray platelet syndrome and Chediak—
granule constituents (ad storage pool deficiency). Deficiency Higashi syndrome.
of the « granules leads to an agranular appearance of platelets
on the Wright-stained peripheral blood smear and has been PRIMARY SECRETION DEFECTS (ENZYMATIC PATHWAY
termed gray platelet syndrome. The Quebec platelet disorder DEFECTS) These disorders resemble storage pool deficien-
is reported to have a deficiency of a granule proteins related cies but have no abnormalities of the platelet granules
to excessive proteolysis. Platelet factor 3 (PF3) activity is and have normal amounts of platelet granule contents. The
reduced in these disorders and appears related to the defects causes of release defects are deficiencies of enzymes and
in platelet aggregation. other “second messengers” that transmit the signal from
The inheritance pattern of these disorders has not been surface receptors to cause platelet release of granule con-
well defined but in some instances appears to be an autoso- tents. The bleeding tendency and laboratory findings in
mal dominant pattern. Patients with storage pool deficiency these disorders are similar to those found in the storage
have a mild to moderate mucosal bleeding tendency. Easy pool deficiencies.
bruising, epistaxis, menorrhagia, postpartum bleeding Cyclo-oxygenase deficiency results in deficient conver-
and bleeding after dental extractions and tonsillectomy are sion of membrane-associated arachidonic acid to thrombox-
often present. Gastrointestinal bleeding is uncommon, and ane A,. Thromboxane synthetase deficiency and other defects
hemarthrosis is not present. Storage pool deficiencies have of thromboxane A, generation and calcium mobilization have
been found in association with other congenital abnormali- been described.”
ties (see Table 25-5). The Hermansky—Pudlak syndrome Treatment of storage pool deficiencies and primary
describes patients with oculocutaneous albinism and dense secretion defects has primarily been with judicious use of
granule storage pool deficiency. Individuals with Chediak— platelet transfusions for prevention or treatment of bleeding.
Higashi syndrome also have oculocutaneous abnormalities, For procedures with low risk of bleeding or where bleeding is
including ocular albinism and characteristic silver-gray hair easily controlled by local measures, no additional treatment
with dense granule storage pool deficiency. Other congeni- may be needed. Prednisone has been reported to improve the
tal abnormalities, including Wiskott—Aldrich syndrome and bleeding time but not platelet aggregation defects, presum-
the syndrome of thrombocytopenia with absent radii (TAR ably owing to an effect to promote vascular integrity. Cryo-
syndrome), have been described to have an associated precipitate and DDAVP have improved the bleeding time in
platelet storage pool deficiency. patients with Hermansky-Pudlak syndrome. DDAVP has been
Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders 593
used in patients with gray platelet syndrome to shorten the THE FACTOR VIII/VWF COMPLEX IN PLASMA
prolonged bleeding time. However, efficacy in treating
bleeding has not been reported in those studies. Red blood Chromosome 12
X Chromosome
cell transfusion may also improve the platelet adhesion Endothelial cell
defect, possibly by supplying ADP. Avoidance of nons- Liver cell Megakaryocyte
teroidal anti-inflammatory agents or other drugs that induce
platelet dysfunction is important in the management of these
disorders. Factor VIII VWEF subunit
GP Ib GP Ilb-Illa
von Willebrand disease is an inherited deficiency of vWF that
may be confused with a primary platelet defect because of the
similarities in clinical presentations between von Willebrand
disease and platelet disorders. von Willebrand disease was orig-
inally termed parahemophilia, because it is a congenital bleed-
Plasma
ing disorder; however, it has a different inheritance pattern and
different bleeding pattern than hemophilia.
vWE is a large, multimeric glycoprotein coded for by a
gene located on chromosome 12 (Fig. 25-8). VWF is synthe-
sized by vascular endothelial cells and stored in Weibel-Palade
H
Palade bodies. A small amount of vWF is also synthesized in
Endothelium 4 arian aA
megakaryocytes and stored in the platelet « granules. Polymer-
ization of VWF occurs in endothelial cells with the production Subendothelium COLLAG
of high-molecular-weight multimers. vWF-cleaving protease VWF
(ADAMTS 13) reduces the size of the multimers at the time Figure 25-9 ™ Schematic representation of the interactions
vWE is secreted into the plasma. Circulating vWF forms a between vWF, platelets, and collagen of the subendothelium. vWF
complex with factor VIII, the protein that is deficient or defec- synthesized by endothelial cells is released in plasma and is stored
tive in hemophilia A. vWF acts as a carrier protein for factor in the a granules of platelets, and can be released after stimulation.
VWF mediates platelet adhesion through binding to collagen and to
VIII, serving to protect the factor VIII molecule from prote-
platelet glycoprotein Ib (GPlIb) in the presence of ristocetin, as well
olytic degradation and increase its concentration at the site of as the platelet GPIlb/Illa in the presence of physiologic agonists
tissue injury. In normal individuals, plasma levels of factor VIII (thrombin, collagen, ADP). vWF also binds to factor VIII (FVIII) and
closely correlate with plasma levels of vWF. heparin.
594 Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders
VWE. VWE promotes secondary hemostasis by functioning as binding of normal vWF to platelets caused by point mutations
a carrier for factor VIII. In von Willebrand disease, absence or in platelet glycoprotein Iba (GPIba).*°*’ The disorder also
dysfunction of vWF results in decreases in factor VII and appears tobe inherited in an autosomal dominant pattern. There
abnormal secondary hemostasis. is variable thrombocytopenia, possibly due to shortenedplatelet
VWF can be measured antigenically and functionally survival from this increased vWF binding to platelets.
using the antibiotic ristocetin. Ristocetin was removed from
use after clinical trials revealed it produced thrombocytope- LABORATORY EVALUATION
nia. Further evaluation revealed it produced agglutination of The routine laboratory evaluation of patients suspected of
platelets in the presence of vWF. This has led to its use as a having von Willebrand’s disease includes a bleeding time,
diagnostic agent in the evaluation of von Willebrand disease platelet count, factor VIII coagulant activity, quantitative
in vitro with the ristocetin-induced platelet aggregation measurements of plasma vWF antigen (vWF:Ag) by crossed
(RIPA) test. Factor VII is measured by a functional clotting immunoelectrophoresis or agarose gel electrophoresis, and
assay. Measurements of factor VII, vWF antigen, and vWF functional assays of plasma vWF by RIPA (vWF activity).**
activity are important in differentiating von Willebrand Subtypes of von Willebrand disease can be distinguished
disease from hemophilia A and to define the types of von according to the vWF:Ag, vWE activity, factor VII coagulant
Willebrand’s disease. activity, and multimeric pattern on gel electrophoresis. All
von Willebrand disease comprises both quantitative and types of von Willebrand disease will have reduced vWF
qualitative abnormalities of the large multimeric VWF glyco- activity but other laboratory findings will vary between the
protein. Affected individuals exhibit mucocutaneous bleeding types. Platelet-type von Willebrand disease also has reduced
typical of defects of primary hemostasis, in contrast to the plasma vWF levels, reduced low molecular weight vWF mul-
joint and deep muscle bleeding typical for hemophilia. The timers and enhanced RIPA, similar to type 2 von Willebrand
disorder has an autosomal pattern of inheritance but has vari- disease. Since the defect occurs on the platelet surface, VWF
able penetrance for expression in affected individuals. from affected individuals will bind normally to normal
Severe cases are characterized by recurrent, potentially life- platelets and vWF from normal individuals will bind abnor-
threatening bleeding, whereas mild cases may go undetected. mally to platelets of affected individuals.
Diagnosis of von Willebrand disease is often difficult
because of the variability of abnormal laboratory results TREATMENT
within affected individuals or family members. vWF and fac- For patients with type I von Willebrand disease, DDAVP is
tor VIH are both acute phase reactant proteins, and plasma considered the initial treatment of choice.” DDAVP stimu-
levels may be increased with stresses such as inflammation, lates the release of vWF from endothelial cell Weibel-Palade
surgery, and pregnancy or with estrogen therapy. Further- bodies. Baseline plasma vWF antigen and activity increase
more, the levels of VWF vary with ABO blood types, with the about three- to fourfold after DDAVP administration. vWF
lowest levels being seen in individuals of O blood type. stores within the endothelial cells are depleted after about 3 to
4 days of DDAVP treatment. Patients requiring longer treat-
CLASSIFICATION ment or those who do not achieve an adequate response to
von Willebrand disease can be classified into three general DDAVP may require the use of intermediate purity factor VIII
categories: products such as Humate P, which contain intact vWF, or
cryoprecipitate. In type 1 patients who receive cryoprecipi-
1. Type 1, an autosomal-dominant disorder accounting for
tate, there is evidence that the endothelial cells are also stim-
70% of all cases, which is characterized by a quantitative
ulated to increase de novo synthesis of VWF.
decrease in normal vWF and mild bleeding;
Patients with type 3 von Willebrand disease do not make
2. Type 2, which has variable inheritance with numerous sub-
vWE and do not respond to DDAVP. Therefore, they must
types, accounts for most of the remaining cases, and is
characterized by a qualitative abnormality in the structure be treated with intermediate purity factor VIII products or
cryoprecipitate.
of vWF; and
3. Type 3, a rare autosomal-recessive disorder characterized There are many subtypes of type 2 von Willebrand dis-
by absent levels of VWF multimers and a severe bleeding ease, some of which respond to DDAVP and some of which
do not. DDAVP has been reported to cause thrombocytopenia
diathesis very similar in presentation to hemophilia A.*
in the type 2b variant.** In many of the type 2 variants,
von Willebrand Normandy is an unusual type 2 variant response to therapy is difficult to quantitate and may require
of von Willebrand disease that is characterized by a defect in measurement of the bleeding time or PFA-100 with determi-
the vWF binding to factor VIII. This disorder is sometimes nation of correction of platelet function after administration
confused with mild hemophilia A because the factor VIII of appropriate therapy. Addition of DDAVP to cryoprecipitate
coagulant level is reduced but the vWF antigen and activity has been utilized as an effective combination in certain
levels are normal or elevated. Clinically, the bleeding diathe- patients.
Sis associated with von Willebrand Normandy is similar to Antifibrinolytic agents can be utilized as an adjunctive
other patients with type 2 von Willebrand’s disease.*5 therapy in all patients with von Willebrand disease. For
A platelet-type von Willebrand’s disease has also been dental procedures, local measures are used and replacement
described. It is a platelet disorder characterized by increased therapy may not be necessary.
Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders 595
Treatment of platelet-type von Willebrand disease with correlation of the metabolic changes with the exact mechanism
measures to increase plasma vWF may actually cause throm- of the functional defect has not yet been firmly established.
bocytopenia by inducing clearance of platelets from the circu- There may also be a defect in platelet activation since platelets
lation. Infusion of smaller amounts of cryoprecipitate than in some uremic individuals demonstrate reduced fibrinogen
used to treat the other forms of von Willebrand disease and binding, secretion and aggregation responses.
platelet transfusions if the thrombocytopenia is severe may be Mild thrombocytopenia with platelet counts as low as
useful. There has been a report of successful use of recombi- 100 X 10°/L, decreased adhesion, abnormal aggregation, and
nant factor VIIa in this disorder. increased bleeding times are abnormal laboratory findings
often seen in patients with uremia. Platelet aggregation studies
in uremic patients typically show no characteristic patterns.
Acquired Qualitative Platelet Disorders The anemia of renal failure may contribute to the hemo-
Platelet dysfunction can also be caused by many systemic ill- static defects observed in uremic patients. Hematocrit values
nesses and medications (Table 25-11). In these diseases, the of less than 25% may prolong bleeding times. Red cells may
platelet dysfunction is a secondary manifestation and not typi- play a role in hemostasis by improving factor VIII coagulant
cally one of the primary presenting features of these disorders. function and by providing ADP, a platelet-aggregating agent.
Acquired causes of platelet dysfunction are more commonly Erythropoietin (r-HuEPO) therapy has proved to be effective
encountered than congenital platelet disorders; however, the in improving hemostasis and decreasing the bleeding tenden-
results of platelet function tests can appear similar to those seen cies in uremic patients on dialysis. It has been theorized that
with congenital disorders. To differentiate between acquired t-HuEPO may also increase platelet number and aid aggrega-
and congenital causes of platelet dysfunction, a careful history tion in addition to raising the haemoglobin and hematocrit.
and physical examination as well as family history must be Hemodialysis or peritoneal dialysis is the treatment of
obtained. choice to correct the hemostatic defect in uremia. However,
platelet function abnormalities may remain. Administration of
UREMIA
cryoprecipitate to patients unresponsive to dialysis has been
Bleeding has long been recognized as a common complication reported to shorten the prolonged bleeding time and decrease
of uremia. Patients with acute and chronic renal failure gener- bleeding. The use of DDAVP in uremic patients has also
ally exhibit bleeding from mucous membranes. Petechiae, pur- prevented clinical bleeding in patients following surgical pro-
pura, epistaxis, ecchymosis, and gastrointestinal bleeding are cedures and has shortened the prolonged bleeding time.°°
common. Severe hemorrhage within serous cavities and Conjugated estrogens have been reported to shorten the
muscles may also occur. bleeding time in uremia and limit bleeding in patients who
A number of different laboratory findings and clinical develop gastrointestinal telangiectasias. The mechanism of
symptoms suggest that platelet dysfunction with abnormal action is not clear.
platelet—vessel wall interaction is the major cause of platelet
LIVER DISEASE
dysfunction and hemorrhage. Studies have shown an abnormal-
Chronic liver disease is often associated with a significant
ity in the interaction of vWF and platelet GPIIb/IIIa complex in
hemorrhagic diathesis as a result of multiple alterations in
uremic patients; however, platelet membrane glycoproteins Ib,
hemostasis, including platelet dysfunction. Mild to moderate
IIb, and IIa are quantitatively normal. Other platelet abnormal-
thrombocytopenia is seen in approximately one-third of
ities seen in uremia include abnormal prostaglandin synthesis,
patients with chronic liver disease as a result of splenic
decreased membrane procoagulant activity, decreased platelet
sequestration secondary to congestive splenomegaly associ-
serotonin release, abnormal B-thromboglobulin levels, elevated
ated with portal hypertension. Abnormal platelet function
intracellular calcium, and decreased thromboxane synthesis.
tests found in patients with chronic liver disease include
Increased levels of “uremic toxins,” such as guanidosuccinic
reduced platelet adhesion; abnormal platelet aggregation to
acid and phenols, are rather consistent findings in patients with
ADP, epinephrine, and thrombin; and abnormal PF3 availability.
uremia. The acquired platelet defects associated with uremia
An acquired storage pool deficiency has also been suggested.
are thought to be primarily mediated by these products. A
The exact mechanism responsible for the platelet defects seen
in chronic liver disease is not known.
Treatment of bleeding in a patient with chronic liver dis-
ease may require several modalities simultaneously to correct
the multiple abnormalities present. Transfusion with platelet
concentrates may ameliorate the bleeding and thrombocy-
Renal disease (uremia)
Liver disease topenia associated with chronic liver disease; however, the
Paraproteinemias expected rise in platelet count following transfusion may be
Myeloproliferative disorders blunted if splenic sequestration or increased platelet con-
Acquired von Willebrand disease sumption occurs. DDAVP may improve the qualitative
Cardiopulmonary bypass
platelet defect associated with this disorder. The numerous
Acquired storage pool deficiencies
Drug therapy coagulation factor deficiencies common in chronic liver dis-
ease typically require the administration of fresh frozen
596 Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders
plasma to correct the coagulation abnormalities. Use of con- manifestations include ecchymosis, epistaxis, and mucocuta-
jugated estrogens may decrease the overall bleeding tendency neous bleeding from the gastrointestinal and genitourinary
after an acute episode. tracts. Thrombosis occurs in both the arterial and venous cir-
culation and includes deep vein thrombosis, pulmonary
PARAPROTEINEMIAS embolism, stroke, myocardial infarction, and thrombosis of
Patients with lymphoproliferative malignancies may exhibit the hepatic, portal, splenic, and mesenteric veins. Patients
both hemorrhagic diatheses and hypercoagulability. These may present with bleeding and thrombosis simultaneously, or
hemostatic alterations are complex and multifactorial. Clini- alternate with bleeding and thrombotic episodes as the disease
cally significant bleeding is a manifestation associated with progresses. The precise mechanisms that cause the problems
multiple myeloma, Waldenstr6m’s macroglobulinemia, and are unknown.°! Intrinsic platelet function defects are noted
other related malignant paraprotein disorders. The pathogen- frequently. In many cases, thrombocytosis is a contributing
esis of hemorrhage and other recognized hemostatic abnor- factor, especially for thrombotic problems in patients with an
malities has been proposed to be caused by the interaction of MPD. In patients with polycythemia rubra vera increased
the paraprotein with platelets and coagulation factors. Clini- whole blood viscosity may play a role.
cal bleeding and platelet dysfunction are seen in approxi- Aggregation patterns in these disorders are usually not
mately 60% of patients with IgM myeloma or Waldenstrém’s characteristic. The most consistently noted laboratory abnor-
macroglobulinemia as compared to approximately 40% of the malities include abnormal release and aggregation in response
patients with IgA myeloma and 15% of patients with IgG to epinephrine, collagen, and ADP, but these findings vary
myeloma. from patient to patient with the same type of MPD. The bleed-
The hemorrhagic diathesis of paraproteinemias is charac- ing time is prolonged in many cases but does not correlate
terized by a prolonged bleeding time and platelet dysfunction. with the risk for bleeding or thrombosis in these patients.
The occurrence of bleeding correlates with the impairment of Other reported abnormalities include acquired storage pool
platelet function. Thrombocytopenia and inhibition of coagula- defects, abnormal prostaglandin and arachidonic acid metab-
tion factors may also contribute to the bleeding tendency. olism, and platelet hyperactivity.
Patients may present clinically with spontaneous epistaxis and Thrombosis is a major complication of PV owing to an
ecchymosis, or unexplained postoperative bleeding in the face increased red cell mass and whole blood viscosity and throm-
of a normal platelet count and coagulation profile. Patients bocytosis. Thrombocytosis is generally moderate, in the range
with malignant paraprotein disorders should not ingest aspirin of 500 X 10°/L to 1 x 10!°/L, and is seen in approximately
or aspirin-containing drugs because this may exacerbate the 50% to 60% of cases. Platelet function defects occur in
platelet defect. Uremic platelet dysfunction associated with approximately 50% to 60% of cases. Platelet function defects
renal failure often seen in the malignant paraproteinemias may are thought to be responsible for many of the hemostatic
also contribute to the hemorrhagic episodes. problems seen. Abnormal platelet aggregation in response
Platelet abnormalities include decreased aggregation in to epinephrine, ADP, and collagen has been reported in
response to various aggregating agents, altered shape change, PV. Platelet activation and intravascular coagulation with
abnormal release reactions, and a prolonged bleeding time. increased plasma levels of B-thromboglobulin, low platelet
Acquired von Willebrand’s syndrome causing reduced levels of serotonin levels, and abnormal levels of fibrinogen and pro-
vWF/factor VIII complexes, other circulating anticoagulants, thrombin have also been reported in cases of PV.
amyloid-associated coagulopathies, impaired fibrin formation Patients with idiopathic myelofibrosis may experience
and polymerization, hyperviscosity syndrome, nephrosis, and bleeding, such as ecchymosis and urogenital hemorrhage.
DIC represent additional abnormalities that predispose patients Thromboembolic complications, with the exception of
with paraproteinemia to bleeding. hepatic and portal vein thrombosis, occur rather infrequently.
Thrombocytopenia, unrelated to the paraprotein effect, It has been suggested that of all the MPDs, idiopathic
may result from marrow replacement, chemotherapy, or hyper- myelofibrosis has the greatest degree of platelet function
splenism and is acommon finding contributing to the incidence defects. Decreased platelet adhesion and prolonged bleeding
of significant bleeding. times are relatively common findings in patients with idio-
Therapy for patients with qualitative defects resulting pathic myelofibrosis. Storage pool deficiencies and impaired
from multiple myeloma and related disorders includes platelet aggregation have also been reported.
plasmapheresis to reduce the circulating paraprotein concen- ET presents with thrombocytosis and is associated with
trations and chemotherapy to inhibit paraprotein production. both bleeding and thrombosis, although bleeding episodes
Blood product support and other therapies may be required occur more often. Evidence suggests that thrombocytosis
when additional hemostatic abnormalities are present. alone does not account for the bleeding and thrombotic
episodes seen in ET, but the combination of thrombocytosis
MYELOPROLIFERATIVE DISORDERS and abnormal platelet function determine the hemostatic
The MPDs include polycythemia vera (PV), idiopathic defects in ET. Decreased platelet aggregation in response to
myelofibrosis, CML, and essential thrombocythemia (ET) epinephrine as well as decreased platelet retention are noted
(see Chaps. 17 and 18). This group of disorders has many in patients with ET. Impaired PF3 availability and platelet
clinical and hematologic features in common, but the hemo- hyperactivity have been reported in patients with thrombosis
static and thrombotic problems are variable. The hemorrhagic in ET. Several platelet defects have been described in ET, but
Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders 597
Waterhouse—Friderichsen syndrome or purpura fulminans. defect in the synthesis of collagen in the walls of small
Activated Protein C (Drotrecogin alpha) concentrate and blood vessels. Vitamin C is a cofactor for hydroxylation of
hyperbaric oxygen have been used to treat this disorder. proline and lysine in the production of collagen and keratin.
A platelet function defect may also be present, with a
ALLERGIC PURPURA decrease in the platelet adhesion reaction. Scurvy may pre-
Allergic purpura is the result of an allergic vasculitis. sent with leg swelling, pain, or discoloration. Subperiosteal
Schénlein-Henoch purpura is a vasculitis involving the bone bleeding is characteristic of scurvy and may be noted
skin, gastrointestinal tract, kidneys, heart, and central ner- radiologically. Gingival bleeding and hemarthrosis may also
vous system.°** This syndrome is considered an immune be present. Perifollicular hemorrhages with hyperkeratosis
complex disease, and is characterized by involvement of the hair follicles and “corkscrew” hairs are characteristic
of capillaries with a diffuse perivascular infiltration by neu- of this problem. Vitamin C administration orally cures the
trophils, lymphocytes, and macrophages. Some patients disorder.
have IgA deposition in the kidney and elevated serum IgA Cushing’s syndrome caused by corticosteroid excess
levels. This syndrome may be related to IgA nephropathy results in purpura (Fig. 25-11). The pathogenesis is identical
whereby similar IgA deposition in the kidney is noted. In to that seen with steroid or senile purpura, including atrophy
Schonlein—Henoch purpura, the onset is abrupt, with a mac- of the subcutaneous tissue and increased blood vessel
ular rash (usually symmetric) involving the lower extremi- fragility. Other cutaneous stigmata of Cushing’s syndrome
ties that becomes purpuric. In some individuals, the joint include a “moon” face, pigmented abdominal striae, a “buf-
and gastrointestinal symptoms predominate and the cuta- falo hump” on the lower neck and upper back, with wasting
neous expression is minimal. This disorder is seen most of the extremities and an obese abdomen.
commonly in children. Renal dysfunction is common and Diabetes mellitus may be associated with retinal hem-
typically reversible in children. Overt renal failure is most orrhages. This is a result of vascular proliferation caused
common in affected adults. Schonlein—Henoch purpura may by retinal hypoxia. These fragile vessels may leak fluid or
last several weeks and then recur (Fig. 25-10). hemorrhage because of their increased permeability. Laser
Drug hypersensitivity may also result in allergic pur- photocoagulation and optimal control of hyperglycemia are
pura. This must be differentiated from drug-induced thrombo- treatments used for this problem. No endogenous platelet or
cytopenias that result in purpura. The mechanism by which coagulation disorder is present in this situation.
these drugs cause purpura is not understood. Aspirin, Protein C deficiency may occur on a congenital or
atropine, iodides, penicillin, quinine, and sulfonamides have acquired basis and is a risk factor for thrombosis. An
been described to cause allergic hypersensitivity purpura. unusual syndrome that resembles purpura fulminans can be
present in neonates, resulting from inherited homozygous
METABOLIC PURPURA protein C deficiency.’ This disorder presents hours after
Metabolic purpura are caused by hormonal or biochemical birth and is usually lethal. Protein C deficiency results from
abnormalities. Scurvy is caused by a deficiency of vitamin oral anticoagulant therapy, because protein C is a vitamin
C. This was a common problem several centuries ago in K-dependent protein. Rapid depletion of protein C by war-
sailors who did not have access to vitamin C on long voy- farin appears to cause a unique thrombotic disorder termed
ages. In modern times, this disorder is found in refugees the warfarin skin necrosis syndrome. This unusual disorder
from war or famine, adults with inadequate or fad diets, and presents as painful, erythematous areas on the buttocks,
in alcoholics. Deficiency of vitamin C appears to cause a breasts, legs, or penis, which rapidly turn to hemorrhagic
Case Study3
2. Hemolytic-uremic syndrome, pre-eclampsia/eclampsia
syndromes, and disseminated intravascular coagulation
are other considerations for a pregnant female with
microangiopathic hemolytic anemia. Idiopathic throm- A 67-year-old white man with a history of cigarette smoking
bocytopenic purpura and gestational thrombocytopenia developed sudden onset of tingling and weakness of the left
do not cause microangiopathy. ADAMTS 13 testing arm that lasted 10 minutes before resolving, consistent with
often reveals the presence of autoantibodies to the a transient ischemic attack. He was examined soon after-
ADAMTS 13 metalloprotease and ADAMTS 13 levels ward in the hospital emergency department and no residual
are less than 5%, not in the other disorders. neurologic abnormalities were found. The spleen was not
palpable and no bleeding or bruising were present. Labora-
tory findings included a hematocrit of 43%, white blood
count of 11.2 X 10°/L, with a normal white blood cell dif-
ferential, and platelet count of 760 x 10°/L. Bone marrow
aspirate and biopsy revealed megakaryocytic hyperplasia
with clustering of megakaryocytes but no granulocytic or
erythrocytic hyperplasia, no fibrosis, and normal storage
iron. Bone marrow culture exhibited growth in the absence
of added growth factors, consistent with a diagnosis of a
continued
604 Chapter 25 Disorders of Primary Hemostasis: Quantitative and Qualitative Platelet Disorders and Vascular Disorders
Woo ont
c. Abnormal platelet aggregation in response to ADP, 7. What disorders are classically associated with
collagen, and epinephrine thrombocytosis?
d. Abnormality in platelet membrane GPIb/IX a. Myeloproliferative syndromes
4. Which of the following is not characteristic of b. Autoimmune disorders
Glanzmann’s thrombasthenia? c. Immunodeficiency syndrome (HIV)
a. Prolonged bleeding time d. Renal disease
b. Giant platelets with thrombocytopenia 8. Which of the following is not an inherited vascular defect?
c. Absent platelet aggregation in response to ADP, a. Ehlers—Danlos syndrome
thrombin, collagen, and epinephrine b. Marfan syndrome
d. Normal platelet aggregation to ristocetin and c. Amyloidosis
bovine vWF d. Giant hemangiomas
5. Which of the following statements is not true regarding 9. Which condition is classified as nonimmunologic throm-
acute idiopathic thrombocytopenia (ITP)? bocytopenia?
a. Thrombocytopenia occurs following a viral infection. a. DIC
b. Spontaneous remissions are common. bs TTk
c. It is found primarily in young children. c. Storage pool defects
d. Platelet counts are generally higher than in d. Post-transfusion purpura
chronic ITP. a
10. Which condition is classified as immunologic thrombo-
6. Thrombocytopenia, fever, renal disease, microangio- cytopenia?
pathic hemolytic anemia, and neurologic complications a. DIC
are hallmark characteristics of: barEe.
a. Hemolytic uremic syndrome (HUS) c. Storage pool defects
b. Disseminated intravascular coagulation (DIC) d. Bernard—Soulier syndrome
c. Thrombotic thrombocytopenic purpura (TTP)
d. Idiopathic thrombocytopenic purpura (ITP) See answers at the back of this book.
American Society of Hematology. Blood Que Francois, B, et al: Thrombocytopenia in Normandy by abolishing binding to fac-
89:1464, 1997. the sepsis syndrome. Role of hemo- tor VIII: Studies with recombinant von
. Cines, DB, and Bussel, JB: How I treat phagocytosis and macrophage colony Willebrand factor. Blood 79:563, 1992.
idiopathic thrombocytopenic purpura stimulating factor. Am J Med 103:114, 46. Russell, S, and Roth, G: Pseudo-von
(ITP). Blood 106: 2244, 2006. 1997, Willebrand disease: A mutation in the
10.CO Vesely, SK, et al: management of adult . Scaradavou, A: HIV-related thrombocy- platelet glycoprotein Iba gene associated
patients with persistent idiopathic throm- topenia. [review] Blood Rev 16(1):73, with a hyperactive surface receptor.
bocytopenic purpura following splenec- 2002. Blood 81:1787, 1993.
tomy: A systematic review. Ann Intern . George, J, et al: Thrombotic thrombocy- 47. Moriki, T, et al: Expression and func-
Med 140(2):112, 2004. topenic purpura—hemolytic uremic syn- tional characterization of an abnormal
__ . Koulova, L et al: Rituximab for the treat- drome: Diagnosis and management. platelet membrane glycoprotein Iba
ment of refractory idiopathic thrombocy- J Clin Apheresis 13:120, 1998. (Met 239— Val) reported in patients
topenic purpura (ITP) and thrombotic . Furlan, M, et al: von Willebrand with platelet-type von Willebrand dis-
thrombocytopenic purpura (TTP): report factor-cleaving protease in thrombotic ease. Blood 90:698, 1997.
of three cases Am J Hemat 78(1):49, thrombocytopenic purpura and the 48. Triplett, D: Laboratory diagnosis of von
2005. hemolytic-uremic syndrome. N Engl Willebrand disease. Mayo Clin Proc
iP. Carr, JM, et al: Efficacy of platelet trans- J Med 339:1578, 1998. 66:832, 1991.
fusions in immune thrombocytopenia. . Tsai, H, and Lian, E: Antibodies to von 49. Mannucci, P: Desmopressin (DDAVP) in
Am J Med 80:1051, 1986. Willebrand factor-cleaving protease in the treatment of bleeding disorders: The
. Kickler, T, et al: Studies on the patho- acute thrombotic thrombocytopenic first 20 years, Blood 90:2515, 1997.
physiology of posttransfusion purpura. purpura. N Engl J Med 339:1585, 1998. 50. Eberst, M, and Berkowitz L: Hemostasis
Blood 68:347, 1986. . George, JN: Thrombotic thrombocy- in renal disease: Pathophysiology and
. Christie, D, et al: Posttransfusion pur- topenic purpura. N Engl J Med management, Am J Med 96:168, 1994.
pura due to an alloantibody reactive with 354:1927, 2006. 5 i . Tefferi, A: Pathogenetic mechanisms in
glycoprotein Ia/Ila (anti- HPA-5b). Blood . Thrompson, C, et al: Thrombotic chronic myeloproliferative disorders:
THANE, NOD. microangiopathics in the 1980s: Clinical Polycythemia vera, essential thrombo-
. Bussell, J, et al: Fetal alloimmune features, response to treatment, and the cythemia, agnogenic myeloid metaplasia,
thrombocytopenia. N Engl J Med impact of the human immunodeficiency and chronic myelogenous leukemia.
SVL, WOT. virus epidemic. Blood 80:1890, 1992. Semin Hematol 36(I Suppl 2):3, 1999.
. Rosenthal, N: The blood picture in pur- . Torok, TR, et al: Increasing mortality 2), Barbui, T, and Finazzi G: Clinical para-
pura. J Lab Clin Med 13:303, 1928. from thrombotic thrombocytopenic pur- meters for determining when and when
. Kaufman, D, et al: Acute thrombocy- pura in the United States—Analysis of not to treat essential thrombocythemia.
topenic purpura in relation to the use of national mortality data, 1968-1991. Semin Hematol 36(1 Suppl 2):14, 1999.
drugs, Blood 82:2714, 1993. Am J Hematol 50:84, 1995. Dr Tefferi, A, and Nichols, W: Acquired von
. Warkentin, T: Clinical presentation of 315 Melnyk, A., et al: Adult hemolytic- Willebrand disease: Concise review of
heparin-induced thrombocytopenia. uremic syndrome: A review of 37 cases, occurrence, diagnosis, pathogenesis and
Semin Hematol 35(4 Suppl! 5): 9, 1998. Arch Intern Med 155:2077, 1995. treatment. Am J Med 103:536, 1997.
. Suh, J, et al: Antibodies from patients 36. McCrae, K, et al: Pregnancy-associated 54. Woodman, R, and Harker, L: Bleeding
with heparin-induced thrombocytopenia/ thrombocytopenia: Pathogenesis and complications associated with cardiopul-
thrombosis recognize different epitopes management. Blood 80:2697, 1992. monary bypass. Blood 76:1680, 1990.
on heparin: Platelet factor 4. Blood 3 Loos, W: HELLP syndrome: Clinical 55: Kestin, A, et al: The platelet function
91:916, 1998. aspects and coagulation abnormalities. defect of cardiopulmonary bypass. Blood
20. Amiral, J: Antigens involved in heparin- Biomed Progress 7:26, 1994. 82:107, 1993.
induced thrombocytopenia. Semin 38. George, J, et al: Glanzmann’s thrombas- 56. Childers, BJ, et al: Acute infectious pur-
Hematol 36(1 Suppl 1):7, 1999. thenia: The spectrum of clinical disease. pura fulminans: a 15 year retrospective
2 — . Zwicker, JL, et al: Thrombosis and elisa Blood 75: 1383, 1990. review of 28 consecutive cases.
optical density values in hospitalized . Fox, J: Identification of a membrane Ann Surg. 69(1): 86-90, 2003
patients with heparin-induced thrombo- skeleton in platelets. J Cell Biol DIE Hautekeete, M, et al: Purpura fulminans
cytopenia. J Thromb Haemost 2(12): 106:1525, 1988. in pneumococcal sepsis. Arch Intern
2133, 2004. . Cuthbert, R, et al: DDAVP shortens the Med 146:497, 1986.
. Walenga, J, et al: Laboratory tests for bleeding time in Bernard-Soulier syn- 58. Jennette, J, and Falk, R: Small vessel
heparin induced thrombocytopenia: A drome. Thromb Res 49:649, 1988. vasculitis. N Engl J Med 337:1512,
multicenter study. Semin Hematol . Mant, M: DDAVP in Bernard-Soulier IQS.
36(1 Suppl 1):22, 1999. syndrome. Thromb Res 52:77, 1988. 3). Manco-Johnson, MJ: Activated Protein C
Uke Warkentin, T, et al: The pathogenesis of . Rao, K, et al: Impaired cytoplasmic cal- concentrate reverses purpura fulminans
venous limb gangrene associated with cium mobilization in inherited platelet in severe genetic protein C deficiency.
heparin-induced thrombocytopenia. Ann secretion defects. Blood 74:664, 1989. J Pediatr Hematol Oncol 26(1):25, 2004.
Intern Med 127:804, 1997. . Ginsberg, D, and Bowie, E: Molecular 60. Ferreiro, J, et al: Benign hypergamma-
24. Warkentin, T: Limitations of conven- genetics of von Willebrand disease. globulinemic purpura of Waldenstré6m
tional treatment options for heparin- Blood 79:2507, 1992. associated with Sjégren’s syndrome.
induced thrombocytopenia. Semin 44. Sadler, J: A revised classification of von Am J Med 81:734, 1986.
Hematol 35(4 Suppl 5):17, 1998. Willebrand disease for the subcommittee 6 —_. Jameson, JJ, et al: Hormonal and
. Ortel, T, and Chong, B: New treatment on von Willebrand factor of the scientific antihormonal therapy for epistaxis in
options for heparin-induced thrombocy- and standardization committee of the hereditary hemorrhagic telangectasia.
topenia. Semin Hematol 35(4 Suppl International Society on Thrombosis and Laryngoscope 114(4):705, 2004.
5):26, 1998. Haemostasis. Thromb Haemost 71:520, 62. Grahame, R: Heritable disorders of con-
. Kelton, J, et al: Elevated platelet-associated 1994. nective tissue. Best Practice and
IgG in the thrombocytopenia of sep- . Elodee, S, et al: The mutation Arg53— Research in Clinical Rheumatology
ticemia. N Engl J Med 300:760, 1979. Trp causes von Willebrand disease 14(2):345, 2000.
Chapter |
Disorders of Plasma
Clotting Factors
Sharon L. Schwartz, MS, MT(ASCP)SH, CLS(NCA)
Introduction OBJECTIVES
The Plasma Clotting Factors, At the end of this chapter, the reader should be able to:
Associated Disorders, and
Laboratory Evaluation 1. List various defects that impair the coagulation system.
Factor | (Fibrinogen) . List the clotting factors and their synonymous names.
Factor Il (Prothrombin)
Factor V (Proaccelerin; . List the vitamin K-dependent factors.
Labile Factor) . Describe the genetic mutations known to affect factor II and factor V, and the conse-
Factor VII (Proconvertin; quences thereof.
Stable Factor; Plasma
Thromboplastin . Describe the differences between hemophilia A and von Willebrand disease.
Component) . Name the respective factor deficiency responsible for causing hemophilias A, B, and C.
Factor VIII (Antihemophilic
Factor) and von . Describe various etiologies and treatment modalities for various factor deficiencies.
Willebrand Factor . Describe laboratory methods used to identify factor deficiencies.
Factor IX (Christmas Factor;
Plasma Thromboplastin . Describe circulating anticoagulants/inhibitors.
cn
ONG
Nec
1)
COn
NOR
Component [PTC]) 10. Describe laboratory methods used to identify circulating anticoagulants/inhibitors.
Factor X (Stuart-Prower
Factor)
Factor XI (Plasma
Thromboplastin
Antecedent [PTA])
Factor XII (Hageman Factor)
Factor XIII (Fibrin-Stabilizing
Factor)
Prekallikrein (Fletcher Factor)
High-Molecular-Weight
Kininogen (Fitzgerald
Factor; Williams—
Fitzgerald—Flaujeac Factor)
Circulating
Anticoagulants/Acquired
Inhibitors
Specific Inhibitors
Nonspecific Inhibitors:
Lupus Anticoagulants
Case Study 1
Case Study 2
Case Study 3
Case Study 4
Case Study 5
607
608 Chapter 26 Disorders of Plasma Clotting Factors
Case Study 6
Case Study 7
Case Study 8
Case Study 9
Case Study 10
INTRODUCTION have the amino acid serine in their active sites. The activation
of the zymogens that participate in the coagulation pathways
This chapter covers disorders of secondary hemostasis: the leading to the formation of an insoluble fibrin clot were dis-
plasma clotting factors, their defects, and how these defects cussed in Chapter 24.
directly affect hemostasis. Information is provided in detail on Plasma clotting factor defects can impair hemostasis.
each of the clotting factors, the effects of their deficiencies, and This can occur as a result of:
associated disorders (Tables 26-1 and 26-2). Recommended
1. Decreased synthesis of one or more factors
laboratory tests and their results are discussed. In addition, dis-
2. Production of abnormal molecules that interfere with the
cussions include information on specific and nonspecific
coagulation pathways
inhibitors. The recommended therapy for each plasma clotting
3. Loss or consumption of the coagulation factors
factor defect is discussed. A summary of the materials pro-
4. Inactivation of one or more factors by inhibitors or
vided, and several case studies are presented at the end of the
antibodies. '
chapter.
The transformation of liquid blood into a semisolid clot Bleeding disorders related to clotting factor deficiencies
is a very complex process. The end result is the cessation of or dysfunction (secondary hemostasis) have a clinical presen-
bleeding, or hemostasis. Clotting factors circulate as inactive tation that differs from platelet-related (primary hemostasis)
zymogens, Or precursors of enzymes, known as serine pro- disorders. Most of the clotting factor deficiencies involve
teases: proteolytic enzymes that cleave peptide bonds that delayed and/or inadequate fibrin formation (except in cases of
von Willebrand nenI 30-40% 16-24 Variable ety rear Mucosal scans
and variants: variable studies, BT, aPTT, superficial wound
inheritance FVII:C, vWF:Ag, RCA, bleeding—variable
multimers depending on level of F
VIII:C, vWF
Ix Hemophilia B 30-40% 20 Abnormal aPTT; May be severe, moderate,
(Christmas disease) normal PT mild—spontaneous
_ hemorrhage, hemarthroses,
crippling, muscle
hemorrhage, post-
traumatic/postoperative
bleeding
xX Stuart-Prower 10% 65 Abnormal PT, aPTT Menorrhagia, ecchymoses,
deficiency CNS bleeding, excessive
bleeding postpartum
XI Hemophilia C 20-30% 65 Abnormal aPTT; Mild bleeding, bruising,
normal PT epistaxis, retinal
hemorrhage, menorrhagia
XII Hageman trait Unknown 60 Abnormal aPTT; Asymptomatic: thrombotic
normal PT tendency
XIll Factor XIII deficiency 1% 150 Normal PT, aPTT; clot Umbilical cord stump
lysis in 5 Molar urea bleeding, poor wound
solution healing, minor injuries
causing delayed bleeding,
fetal wastage, excessive
fibrinolysis, male sterility,
intracranial hemorrhage
PK Prekallikrein Unknown 35 Abnormal aPTT: shortening Asymptomatic
(Fletcher factor) with interval incubation
with activator
HMWK Fitzgerald factor Ua pown 156 _ Abnormal aPTT _ Asymptomatic
PT == aries time; aPTT= activated Gi ieee time; BT =Fert time; HMWK = =ee molecular-weight eee
Ie = thrombin time; CNS= central nervous system.
Source: From Pittiglio, DH, et al: Treating hemostatic disorders. A problem oriented approach. In Pittiglio DH: Hemostasis Overview. American Asso-
ciation of Blood Banks, Arlington, VA, 1984, p 28, with permission.
XI ABN
XI
Prekallikrein ABN*
HMWK ABN |
: Pa
BOG
far
dr
MAAN| l\Zz2AzZAZAZZZZZZzZ
ZZ
Ze
Ae
Wee
*The aPTT willihe after aa contact
a activation.
N = normal; ABN = abnormal; BT = bleeding time; PT = prothrombin time; aPTT = activated partial thromboplastin time;
(bh thrombin time; N/A = not applicable.
Source: From Pittiglio, DH, et al: Treating hemostatic disorders. A problem-oriented approach. In Pittiglio, DH: Hemostasis Overview. American
Association of Blood Banks, Arlington, VA, 1984, p 31, with permission.
610 Chapter 26 Disorders of Plasma Clotting Factors
factor XIII deficiency and most cases of von Willebrand dis- aggregate. In the final stages of coagulation, these soluble
ease); whereas platelet function is normal. Bleeding or hemor- fibrin monomers spontaneously polymerize to form soluble,
rhage can be more profuse because larger vessels are usually but unstable, fibrin strands. The production of an insoluble,
involved (during surgery, childbirth, or after trauma); the stable clot requires the formation of covalent cross-linkages
___location of the hemorrhage is within muscle, deep-tissue, or of the alpha and gamma chains between the soluble fibrin
joint cavities; or it can be delayed, (i.e., a stabilized injury strands, catalyzed by thrombin-activated factor XIII (see
begins oozing or actively bleeding again). Secondary hemo- Factor XIII). This insoluble, stable clot is resistant to enzy-
static disorders may be inherited as sex-linked or autosomal matic digestion or chemical degradation. Normal serum does
genetic traits; they may also be acquired due to dietary not contain detectable fibrinogen; it is consumed in the clot-
vitamin K deficiency, liver disease, hemorrhage, or a consump- ting process.
tive coagulopathy (e.g., disseminated intravascular coagulation
[DIC]); induction of anticoagulant therapy, inflammatory dis- AFIBRINOGENEMIA
orders, or treatment with various drugs. Afibrinogenemia refers to the absence of any chemically, anti-
Laboratory evaluation for clotting factor defects always genically, or functionally detectable fibrinogen in the plasma. It
begins with screening tests: prothrombin time (PT) and acti- is a rare (estimated prevalence 1:1,000,000), homozygous, con-
vated partial thromboplastin time (aPTT). Abnormal results genital disorder inherited as an autosomal recessive trait. Fib-
obtained from one or both screening tests must be followed- rinogen levels may be less than 5 mg/dL. There can be profuse
up with differential tests which include repeating the abnor- bleeding after slight trauma and delayed wound healing. Initial
mal screening tests on mixtures of patient plasma with pooled symptoms include bleeding from the umbilical cord stump and
normal plasma (known as mixing studies), to further charac- mucosal tissues (e.g., epistaxis, gastrointestinal bleeding),
terize the abnormal screening test results. The addition of which can be fatal. Other symptoms can include intracranial
pooled normal plasma to the patient’s plasma sample will nor- bleeding, menorrhagia, and hemarthrosis. Mild to moderate
malize, or “correct” the results of the abnormal screening test if thrombocytopenia can occur, but platelet counts are rarely less
there is a deficiency of one or more factors. The inability to cor- than 100,000/uL.*° Platelet adhesion and aggregation are
rect the screening test result, or a correction followed by pro- abnormal in patients with this disorder; fibrinogen is required
longation after a 2-hour incubation at 37°C, would suggest for normal primary platelet aggregation because it binds to stim-
the presence of an inhibitory substance. The inhibitor sub- ulated platelets at the glycoprotein IIb/Ila (GPIIb/IIIa) receptor,
stance can be the result of an immune response (antibody) forming a linkage with adjacent platelets and reinforcing
produced by the patient, or an anticoagulant such as heparin the platelet plug at the site of vascular injury (see Chap. 24). *
or other thrombin inhibitor which has been administered to Afibrinogenemia, therefore, produces a prolonged bleeding time
the patient or contaminated the specimen. (see Table 26-1).
The laboratory evaluation for this disorder should
The Plasma Clotting Factors, Associated include: PT, aPTT, fibrinogen assay (clottable activity and
antigen), and thrombin time (TT) (Table 26-3). The results of
Disorders, and Laboratory Evaluation these tests will show marked abnormalities: terminal prolon-
Factor | (Fibrinogen) gation of the PT, aPTT, and TT (no clot forms; no detectable
endpoint), and the absence of measurable fibrinogen. Mix-
Factor I, more commonly known as fibrinogen, is a large gly- ture of the patient’s plasma with pooled normal plasma will
coprotein (molecular weight 340 kD) produced solely by the correct these defects.
liver, and is critical in the final stages of coagulation. It is the Treatment for afibrinogenemia requires the administra-
most abundant of the plasma coagulation proteins; up to tion of cryoprecipitate; this preparation contains appreciable
5 grams/day are produced by the liver, with a normal plasma amounts of fibrinogen. Fresh frozen plasma (FFP) may be
concentration of approximately 200 to 400 mg/dL. A minimum used; however, volume overload is a major concern due to the
of approximately 100 mg/dL is required to maintain normal number of units required to sufficiently elevate the fibrinogen
hemostasis. The normal plasma half-life is 3 to 5 days.?* While level. Whole blood transfusions may be required if significant
the majority of fibrinogen is present in the plasma (75%), it can bleeding has occurred.
also be found in interstitial fluid, and is present in platelet alpha
granules by absorption from the plasma. The fibrinogen mole- HY POFIBRINOGENEMIA
cule consists of three pairs of different polypeptide chains, des- Hypofibrinogenemia may be inherited as a heterozygous,
ignated as A-alpha (Aq), B-beta (BB), and gamma (vy), joined autosomal recessive disorder. The level of fibrinogen is found
by disulfide bonds. The interaction of these chains, combined to be in the range of 20 to 100 mg/dL. Bleeding episodes are
with the activity of the enzyme thrombin, ultimately yields a usually mild and not spontaneous. Bleeding may occur after
network of insoluble strands called fibrin. Fibrin monomers are trauma or surgery. Acquired hypofibrinogenemia can be
produced by thrombin’s enzymatic cleavage of fibrinopeptides caused by liver disease or DIC.
A and B from the terminal ends of the Aw and Bf chains of The degree of prolongation of the PT, aPTT, and TT is
the fibrinogen molecule. These fragments contain negatively dependent on the level of fibrinogen present in the plasma.
charged amino acids; once removed, the intermolecular Treatment for this condition may include the use of cry-
repulsive force is reduced, allowing the fibrin monomers to oprecipitate and/or FFP.
Chapter 26 Disorders of Plasma Clotting Factors 611
ABN N N
ABN ABN N/ABN
ABN ABN ABN
Thrombin time ABN ABN ABN
Reptilase time ABN ABN
Fibrinogen (clottable) Undetectable ABN
Fibrinogen (antigen) Absent ABN
Platelet aggregation ABN N
N = normal; BT = bleeding time; ABN = abnormal; aPTT = activated partial thromboplastin time; PT = prothrombin time.
Source: From Girolami, A, et al: Rare and quantitative and qualitative: Abnormalities of coagulation. Changes Hematol 14:388, 1985,
with permission.
activated factor V (F Va), platelet phospholipids (PF3), and disease, or the presence of antibodies to prothrombin must be
calcium ions. This is known as the prothrombinase complex, differentiated from dysprothrombinemia and hypoprothrom-
which is assembled on the platelet surface and activates binemia in order to determine the proper course of therapy.
prothrombin (F II) to thrombin (F Ia). The generally accepted Treatment may include administration of FFP or PCC.
normal adult reference range is 50% to 150% (0.5 to 1.5 U/mL),
PROTHROMBIN G20210A MUTATION
and may vary by institution or published reference. Defi-
A single-point mutation in the prothrombin gene, located on
ciency of prothrombin delays the generation of thrombin, thus
chromosome 11, was discovered in 1996 that was associated
contributing to hemorrhagic symptoms. The prothrombin
with a mildly elevated prothrombin concentration and an
time (PT) is an in vitro analysis of the time required to con-
increased risk of venous thrombosis.'’ The mutation, known as
vert plasma prothrombin into endogenous thrombin, using an
prothrombin G20210A, consists of a single base change
exogenous source of phospholipid, tissue thromboplastin
resulting from a G (guanine) to A (adenine) transition at
(tissue factor), and calcium ions; thereby cleaving fibrinogen
nucleotide 20210, in the 3’-untranslated region of the pro-
to a fibrin endpoint. This assay is affected by defects or defi-
thrombin gene.'* It is the second most common cause of inher-
ciencies in any of the components of the prothrombinase
ited thrombophilia (predisposition to thrombosis). Patients
complex, as well as the concentrations of fibrinogen, pro-
who are heterozygous for this mutation carry a two- to fivefold
thrombin, and factor VII (F VII).
increased risk of venous thromboembolism as compared with
normal individuals. This genetic defect is not commonly
HYPOPROTHROMBINEMIA
found in patients with arterial thromboembolic disease, but it
It has been suggested that the mode of inheritance of
has been identified as a risk factor for myocardial infarction in
hypoprothrombinemia is autosomal recessive.'> Patients who
young women! and cerebrovascular ischemic disease in
are either doubly heterozygous or homozygous with this rare
young patients.”? Prothrombin G20210A is predominantly
condition may have hemorrhagic symptoms with prothrom-
(although not exclusively) restricted to the Caucasian popula-
bin levels from 2% to 25% of normal activity. Deficiency
tion. In the presence of other acquired risk factors such as
may also be acquired by dietary vitamin K deficiency or oral
pregnancy, oral contraceptives, malignancy, antiphospholipid
anticoagulant (warfarin, Coumadin®) therapy. However, the
antibody (lupus anticoagulant), recent surgery or trauma, the
type and severity of symptoms may vary with the level of
incidence of thrombosis is increased.*!”?
functional prothrombin available. Epistaxis, menorrhagia,
The elevation of F II activity, which may only be slightly
postpartum hemorrhage, hemorrhage following surgery or
greater than levels in patients without the mutation (approxi-
trauma, and broad-spectrum antibiotic use (which destroy the
mately 115% to 130%; 1.15 to 1.30 U/mL), may still be
normal flora of the gastrointestinal tract responsible for vita-
within normal limits, making this an insensitive method of
min K synthesis) are exhibited with prothrombin levels rang-
detection. Therefore, the mutation can be detected only by
ing from less than 2% to less than 50%. Ingestion of aspirin
direct analysis of genomic DNA, since the polymerase chain
increases the bleeding tendency by affecting platelet function.
reaction (PCR) technology is only one (initial) part of analyz-
Prothrombin levels approaching 50% activity generally do
ing such single nucleotide polymorphisms.
not cause hemorrhagic symptoms. Variable prolongation of
both the PT and aPTT, and a normal TT, are observed in indi-
viduals with hypoprothrombinemia. These screening assays Factor V (Proaccelerin; Labile Factor)
are not specific for hypoprothrombinemia. Definitive diagno-
sis is dependent on specific assays for functional activity or Factor V (F V) ( molecular weight 350 kD) is a single-chain
antigenic concentration of prothrombin, or both. In hypopro- glycoprotein, synthesized by the liver and also present in the
thrombinemia, both the functional activity and antigenic con- alpha granules of platelets, which is secreted during platelet
centration of prothrombin are decreased. Mixing studies with activation.**** It has a short half-life in vivo, and is also
pooled normal plasma will show full correction of the PT or referred to as the “labile factor” because of rapid deterioration
aPTT (see Tables 26—1 and 26-2). of its activity in plasma at room temperature. F V, also known
Treatment of hypoprothrombinemia may include the as proaccelerin, is the catalyst in the conversion of prothrom-
administration of fresh frozen plasma or prothrombin com- bin (F II) to thrombin (F Ia), functioning as a cofactor within
plex concentrate (PCC), which contains factors II, VII, IX, the prothrombinase complex. It is converted to its active
and X. Vitamin K supplementation will restore only low form, F Va, by the actions of thrombin (F Ia) or F Xa, in the
factor levels associated with dietary deficiency or oral antico- presence of calcium ions and phospholipids. The normal adult
agulant therapy. reference range is 50% to 150% (0.5 to 1.5 U/mL), and may
vary by institution or published reference.
DYSPROTHROMBINEMIA In 1947, Owren discovered F V deficiency in a 21-year-
The mode of inheritance in dysprothrombinemia is the same old woman with a lifelong bleeding history.*>?° Studies demon-
as in hypoprothrombinemia.'® A structural defect causes strated that this clotting factor was not vitamin K dependent.
impaired functional activity, although the antigenic concen- Congenital F V deficiency, also known as parahemophilia, is
tration is normal. Bleeding manifestations may occur that are inherited as an autosomal recessive trait.” It is an extremely
similar to those described for hypoprothrombinemia. Vitamin rare condition that occurs with a probability of 1 individual per
K deficiency, induction and therapeutic warfarin therapy, liver 1 million population (see Table 26-1).
Chapter 26 Disorders of Plasma Clotting Factors 613
Ecchymoses, epistaxis, menorrhagia, and gingival, gas- 90% of the cases positive for APCR; approximately 20% to
trointestinal tract, umbilical, and central nervous system 60% of patients with recurrent venous thrombosis were found
bleeding are associated with deficiencies of F V. Hemorrhagic to be positive for the mutation.'® The clottable activity of F V
manifestations are usually noted in individuals with less than is unaffected by this mutation. Detection of the R506Q defect
10% F V activity. Hemarthrosis seldom occurs even in can be accomplished only by genomic DNA analysis. In
severely deficient patients. Combined deficiencies of factors V addition, APCR has been found to be present in patients with-
and VIII have been reported in several families. The com- out the factor V Leiden mutation; other mutations have been
bined factor V and VIII activities and antigen levels are characterized, however, they are still undergoing research.
decreased, ranging from 5% to 30% of normal.” This syn- There are also cases of acquired APCR associated with malig-
drome has been found to be the result of a gene mutation nancies, lupus anticoagulants, and third trimester pregnancy.
which produces a protein that affects the intracellular trans- This illustrates the importance of detecting APCR in patients
port of these factors from the endoplasmic reticulum to the who may be at an increased risk of thromboembolic disease.
Golgi apparatus. Its defective transport leads to reduced Screening for the presence of APCR is easily performed by
secretion of these coagulation factors.’® clottable assays utilizing modified aPTT or Russell’s viper
Acquired F V deficiency is associated with a variety of venom time methods. By either method, the plasma clotting
disorders such as liver disease, carcinoma, tuberculosis, and time with the addition of activated protein C (APC) is com-
DIC.*° It can also result from the presence of F V-specific pared to the clotting time of the plasma without added APC.
antibodies acquired after childbirth, due to autoimmune dis- Normal patients produce prolonged clotting times in the pres-
eases, Or exposure to certain types of fibrin glue used in sur- ence of APC, because of its ability to inactivate F Va and F
gical procedures.*! Villa; the clotting time without APC will therefore be signif-
In the laboratory evaluation, the PT and aPTT are icantly shortened. Patients with APCR will produce shortened
prolonged, and both are corrected on mixing with pooled nor- clotting times for both tests, indicating the inability of APC
mal plasma. The TT is normal. Approximately one-third of to exert its effect on F Va or F VUIa, hence the description
patients may present with an abnormal bleeding time (BT), “resistance to APC” (refer to Chap. 28 for a further discussion
possibly related to platelet alpha granule-associated F V defi- of the technique.)
ciency.*” Prior to analysis, the centrifuged plasma must be
platelet-poor (less than 10,000/uL) to avoid contamination of
the plasma F V level by platelets’ endogenous F V. Plasma Factor VII (Proconvertin; Stable Factor;
factor activity by clotting factor assay is decreased or absent. Plasma Thromboplastin Component)
Correction with the addition of pooled normal plasma will not
occur in the presence of an inhibitor. A predominant plasma protein of the extrinsic coagulation
F V-deficient patients are often treated with fresh plasma pathway is F VII (molecular weight 50 kD), a heat- and
or FFP. Cryoprecipitate does not contain adequate amounts of storage-stable component for which it is also known as “‘sta-
F V to be used therapeutically. ble factor.” It is also known as proconvertin, for its pivotal
role in coagulation. It is produced by the liver requiring vita-
FACTOR V LEIDEN MUTATION (R506Q) min K for its synthesis (vitamin K—dependent), and circulates
Under normal conditions, F Va (and, as will be discussed, as an inactive zymogen (F VII), as well as in low levels of its
F VIIa) must be inactivated to prevent ongoing thrombin gen- activated form, F VIla.*° The normal adult reference range is
eration. This is accomplished by the naturally occurring antico- generally 50% to 150% (0.5 to 1.5 U/mL).
agulant, protein C; and its cofactor, protein S. Protein C is The division of the coagulation cascade into the intrinsic
activated by thrombin (which is bound to thrombomodulin on and extrinsic pathways was created as a useful tool for labo-
the endothelial cell surface; see Chap. 28). Activated protein C ratory diagnoses. It is now recognized that this division does
(APC) is responsible for inactivation of F Va and F VIIa . not exist in vivo. Initiation of coagulation in vivo begins with
In 1993, a new and very common inherited throm- the extrinsic pathway, involving components of the vascula-
bophilic syndrome was described: activated protein C resis- ture and cellular elements of the blood. A major component is
tance (APCR). A molecular defect (single point mutation) was tissue factor (TF), also known as tissue thromboplastin, which
identified involving the transition of a guanine (G) to adenine functions as a cofactor. TF, a transmembrane lipoprotein nor-
(A) at nucleotide 1691 of the F V gene. This results in the sub- mally found exclusively extravascular, is synthesized by
stitution of arginine (R) with glutamine (Q) at amino acid monocyte/macrophages and endothelial cells. Its surface
number 506; this is one of the cleavage sites where APC expression can be induced by inflammatory cytokines; this
must bind to the factor V molecule.** This mutation, named seems to be one of the most important pathophysiological
factor V Leiden (R506Q), obliterates this site and leads to mechanisms of DIC in sepsis. TF is also present in many
impaired degradation (inactivation) of F Va by APC, resulting other tissues, such as brain, lung, and placenta. In the event of
in continued thrombin generation, promoting thrombosis vessel injury, TF and F VII/F Vila form a complex (TF/F
(Fig. 26-1). Factor V Leiden is present in 3 to 7% of Vila) in the presence of calcium ions. This complex, known
Caucasians of Northern European descent,” but it is at least as an extrinsic tenase complex, activates F X to F Xa. The
10 times more prevalent than other known genetic throm- TF/F Vila complex is also capable of activating F IX, bypass-
bophilic disorders. This mutation is responsible for more than ing the need for contact activation of factors XI and XII.
614 Chapter 26 Disorders of Plasma Clotting Factors
APC
© i
i = inactivated
a = activated
Thrombin
Figure 26-1 ™ Activated protein C (APC) resistance of factor V Leiden. (Top) Normal inactivation of factors V and VIII:C by APC. (Bottom)
The binding site for APC on the factor V Leiden molecule is altered, thereby permitting activated factor V (factor Va Leiden) to continue
thrombin generation and subsequent fibrin formation.
Positive feedback mechanisms exist that further activate more Factor VII deficiency can be acquired with liver disease,
F VII. Another name for F VII is “plasma thromboplastin warfarin therapy, broad-spectrum antibiotic use, or dietary
component (PTC)” for its involvement in this mechanism. vitamin K deficiency. Treatment can include FFP, PCC, and/or
Homozygous congenital F VII deficiency is a rare, vitamin K supplementation. In addition, a genetically engi-
autosomal-recessive trait.*°°’ Clinically, these patients can neered preparation structurally similar to human plasma-derived
present with deep muscle hematomas, hemarthroses, epistaxis, F VIIa: recombinant activated factor VII (rF VIIa), can be used
and menorrhagia. Patients having less than 1% F VII activity in congenitally deficient patients with a hemorrhagic diathesis.
can have severe hemorrhagic manifestations. Heterozygotes rFVIla was also approved by the FDA in 1999 as a treatment
are usually asymptomatic, having factor activities between option in hemophiliacs with inhibitors (see Hemophilia A; see
40% and 60% of normal. also Circulating Anticoagulants/Acquired Inhibitors).
In the laboratory, the PT is prolonged, with a normal
aPTT (F VII is not measured by the aPTT test system). The
PT can be fully corrected by mixing with pooled normal Factor VIII (Antihemophilic Factor) and von
plasma. Testing with a Russell’s viper venom reagent, Willebrand Factor
known as the Stypven time, will be normal because it
Factor VIII (F VIII) is a large glycoprotein ( molecular weight
directly activates F X (Table 26-4). Documentation of F VII
330 kD), essential in the middle phase of coagulation.'* It is
deficiency requires the performance of a clotting factor
not completely known where the procoagulant F VIII is
activity assay.
synthesized, although studies have tended to support the liver
as the source.**“? However, severe hepatic failure does not
cause F VIII deficiency. Some evidence suggests that a cell
type present in several organs may be the major source, such
as fibroblasts, lymphocytes, monocytes or macrophages, and
vascular endothelial cells.*°
Results Normal Values F VIII is secreted into the plasma, where it circulates as a
macromolecular complex with von Willebrand factor (vWF),
>14 sec 10.0-14.0 sec
<36 sec designated as FVIII:vWF (Table 26-5).41 vWF is synthesized
Factor VII assay <50% within megakaryocytes and endothelial cells as a high-molecu-
Stypven time << SEG lar-weight (HMW) monomeric glycoprotein (molecular weight
Other factor activities >50% 250 kD), which dimerizes and polymerizes into even higher
PT = prothrombin time; aPTT = activated partial thromboplastin molecular weight molecules known as multimers. These multi-
time; sec = seconds. mers are secreted and circulate in the plasma as a heterogeneous
mixture, with molecular weights ranging from 600 to 20,000
Chapter 26 Disorders of Plasma Clotting Factors 615
A 1 Minute
0% sdb
10%
20%
30%
B, and
common of the hemophilic syndromes (hemophilias A,
ve bleedin g disorde r
C). Hemophilia A is a sex-linked recessi
es. Approx imatel y | in
that has been documented for centuri
5.000 to 10,000 affected males are born annually. The fifth-
d
century Talmud described a bleeding episode that occurre
after circumcision. Modern rabbinic comma nd forbids the cir-
cumcision of any child in whom the diagnosis of hemophilia
has been made.°! The disorder was found in the Royal House
of Stuart in Europe and Russia. Queen Victoria, a carrier, was
the source of hemophilia in four subsequent generations.*””*
The defect in hemophilia A is not an absence of the
F VIII complex but rather a molecular defect or absence of its
coagulant portion, F VIII:C,** caused by either a point muta-
tion involving a single nucleotide, deletion of all or part of the
gene, or a mutation affecting gene regulation. The vWF anti-
genic component (vWF: Ag) of the F VIII complex is found to
be normal in patients with hemophilia A. The gene for F
VIII:C resides on the X chromosome. Males inheriting an
affected X chromosome will manifest the disease, whereas
females with one affected X chromosome will be silent
(obligatory) carriers of the trait (asymptomatic). They do not
exhibit clinical bleeding because one allele is capable of pro-
ducing sufficient, functional F VIII:C to maintain hemostasis.
The ratio of F VIII:C to the vWF antigen (F VIII:C/vWF:Ag)
should be approximately 1:2 in the carrier.’ In approximately
one-third of newly diagnosed cases of hemophilia A, there
may be no previous family history of bleeding. This suggests
that a spontaneous mutation may have occurred, or that
there could be several generations of female silent carriers of
thetrait:2
1 2 3
Patients with a F VIII:C activity of less than 1% (less
than 0.01 U/mL) are classified as severe hemophiliacs. They
have a profound hemorrhagic disease, evidenced by intracra-
Figure 26-4 = vWF multimers. Samples of (7) normal plasma,
nial and intramuscular bleeding, hematuria, and spontaneous
(2) von Willebrand’s plasma, and (3) cryoprecipitate, underwent
electrophoresis in sodium dodecyl! sulfate (SDS)-agarose gel. hemorrhage, requiring daily factor replacement therapy.
A Western blotting technique was performed. The gel on nitrocellu- Frequent, spontaneous hemarthroses are the primary symp-
lose paper was incubated first with a rabbit anti-human vWF: tom, involving the knees, elbows, ankles, shoulders, hips, and
Ag antibody and then with goat anti-rabbit IgG, after which it was wrists, that causes crippling deformities. Moderately severe
stained.
hemophilia demonstrates levels of F VII:C activity between
2% and 5% (0.02 to 0.05 U/mL). These patients have less fre-
after a citrated whole blood sample is added. This subjects the quent spontaneous bleeds; however, profuse bleeding still
platelets to the shear forces that may be encountered in vivo, as occurs with circumcision, surgery, trauma, or even minor
they are drawn up through the capillary tube. When they injuries. Patients with activities greater than 5% to 30%
encounter the coated membrane aperture, they are stimulated to (greater than 0.05 to 0.30 U/mL) are classified as having mild
adhere and aggregate, ultimately occluding the aperture. The hemophilia. These patients may go undiagnosed until a bleed-
time required for this occlusion to occur is known as the ing episode occurs, such as with surgery or trauma. Approxi-
“closure time” (CT). Normal values for the CT are established mately 10% to 15% of patients with hemophilia A develop
by each laboratory. Patients with vWD or other platelet dysfunc- antibodies, or inhibitors, to F VIII:C°° that are usually time
tion disorders have abnormal CTs. This test parallels results of and temperature dependent in vitro, and are immunoglobulin
the BT test, but is superior to the BT in standardization, preci- (IgG) in nature. They are capable of destroying F VIII:C
sion, and causing less patient distress. Its limitations, however, at 37°C, neutralizing the coagulant present in therapeutic
are also related to platelet count, as well as hematocrit, erythro- infusions, and complicating treatment.°’ Administration of
cyte sedimentation rate, and specimen age. F VIII:C concentrates may lead to a rise in antibody titers in
patients who have developed antibodies (an anamnestic
HEMOPHILIA A response) to F VIIN:C (see Circulating Anticoagulants/
Hemophilia A (classic hemophilia) is a hereditary coagulopa- Inhibitors).>!
thy, second in overall frequency of the inherited bleeding dis- There are many forms of treatment available for hemo-
orders after von Willebrand disease. It is, however, the most philia A. Concentrates of human plasma F VIII:C were widely
Chapter 26 Disorders of Plasma Clotting Factors 619
used prior to 1984. Since then, many patients have been the aPTT method, known as the “Bethesda Assay,” and
treated with highly purified, heat- or solvent-detergent quantifying the inhibitor titer in Bethesda units (see Spe-
processed F VIII concentrates, to inactivate hepatitis B, C, cific Inhibitors).°° Patients with inhibitors may show dilu-
and human immunodeficiency viruses (HIV). Although tional increases in factor activity during factor assays.
cryoprecipitate is a rich source of F VIII:C, it is not a product These inhibitors must be differentiated from a nonspecific
of choice due to the high incidence of parenteral viral trans- inhibitor (see Circulating Anticoagulants/Inhibitors) by
mission, because antiviral processing is not possible. New additional testing and clinical presentation.
technologies that produce synthetic DNA-recombinant F VIII
(rF VII:C) have significantly lowered the incidence of viral VON WILLEBRAND DISEASE
transmission. Patients in whom antibodies to human F VIII:C In 1926, Dr. Erich von Willebrand evaluated a large family
have developed, may often respond to treatment with porcine from the Aland Islands for a severe bleeding disorder. Both
F VIII; however, they may form antibodies to this product as sexes were affected, and presented with prolonged bleeding
well. Factor IX concentrate, activated PCC, or Factor Eight times despite normal platelet counts and clot retraction tests.°!
Inhibitor Bypassing Activity™ (FEIBA™), an anti-inhibitor Dr. von Willebrand concluded that this was a previously
coagulant complex agent, has been used in severe cases. undescribed bleeding disorder.
These products contain factors II, IX, X, activated F VII, and vWD differs from classic hemophilia A in three cardinal
a small amount of F VIII:C antigen, which can restore manifestations: (1) autosomal inheritance rather than sex-linked,
hemostasis by providing factors beyond F VIIE:C in the coag- (2) consistently prolonged BTs, and (3) mucocutaneous bleed-
ulation mechanism to form thrombin. As discussed under the ing rather than hemarthroses and deep muscle hemorrhage. It
topic of F VII, rFVIa was approved by the FDA as a treat- was not until the 1970s that vWF and F VIII:C were found to
ment option in hemophiliacs who have inhibitors to control be different proteins produced by different cells under different
life-threatening hemorrhage. Thirty percent (0.30 U/mL) genetic control.
FP VUI:C activity is the minimum level required to maintain von Willebrand’s disease is the most common inherited
normal hemostasis. bleeding disorder, affecting both males and females equally. It
The laboratory findings for patients with hemophilia A results from mutations within the vWE gene, which causes
would be: prolonged aPTT, normal PT, and a normal BT either quantitative or qualitative defects of plasma vWF.
(Table 26-7). Mixing studies with pooled normal plasma These differences arise due to the characteristics of the muta-
corrects the prolongation of the aPTT. The deficiency 1s tion(s) and the autosomal inheritance pattern, which may be
further characterized by low to absent levels of F VIII:C, either dominant or recessive. The gene for vWF resides on
normal levels of vWF:Ag and RCA, and normal platelet chromosome 12; therefore, the disorder may occur in either a
function assays. The presence of an inhibitor can be estab- heterozygous or homozygous mode. These variations in
lished when mixing studies with pooled normal plasma do genotype produce various phenotypes of the disease. vWD
not correct the prolonged aPTT. The inhibitor activity is can be divided into three main types, designated as types 1, 2,
determined using a modified one-stage factor assay utilizing and 3. Type 2 vWD is further subdivided into four subtypes,
See FVIC WE
Inheritance Sex-linked recessive Autosomal, dominant
Clinical presentation Hemarthroses; muscle, soft tissue bleeding Mucosal bleeding: gingival, gastrointestinal,
menorrhagia
Bleeding tendency Moderate to severe Mild to moderate
Laboratory Tests
Bleeding time N N/ABN (variable)
PFA-100° N ABN
Platelet count N N
N N/ABN (variable)
N N
A BN N/ABN (variable)
A BN N/ABN (variable)
N ABN
Caray 2k
N ABN
WE Multimer ‘Pattern N ma Pattern with decreased total concentr, ation
owe= er. ABN= ANS FF VIL C = factor VIII aera activity; PT = Ain Rewari time; aPTT= activated pane tirombontestin ti
time;
Ag = von Willebrand factor antigen; RCA= ristocetin cofactor assay; PFA-100°= platelet function analyzer; RIPA= ristocetin-induced
vWF:
platelet agglutination.
620 Chapter 26 Disorders of Plasma Clotting Factors
designated as 2A, 2B, 2M, and 2N. There is an additional dis- prophylactically with a synthetic analog of vasopressin (the
ease category related to the platelet VWF receptor, known as antidiuretic hormone): desmopressin, also known as DDAVP
platelet-type/pseudo-vWD, which has a similar presentation (1-deamino-8-b-arginine-vasopressin), before dental work,
as vWD, but does not involve a mutation of the vWF gene. surgery, and after bleeding episodes, thereby avoiding expo-
There are also acquired forms of vWD. Each is discussed sure to blood products. This medication causes the vWF and
separately. F VIII:C activity to transiently increase, approximately two-
Since vWF circulates as a complex with F VIII:C, to-fivefold, by stimulating the release from endothelial stores.
patients having reduced or absent levels of VWF:Ag may also This medication can also be useful in patients with mild
have reduced or absent levels of F VIII:C. However, levels of hemophilia A. Some patients do not respond to DDAVP; a
F VUI:C are usually slightly greater than the level of trial course may be administered in advance of elective pro-
vWF:Ag, presumably because there is a normal gene present cedures to determine the degree of response by the patient.
for F VHI:C, and the potential binding sites are saturated Patients who fail to respond to DDAVP must receive cryopre-
when levels of vWF:Ag are reduced.*! The ABO blood type cipitate infusions for prophylaxis or treatment.
has also been found to have a significant effect on the amount
of vWF:Ag produced; individuals who are type A, B, or AB TYPE 2 vWD The type 2 vWD subtypes (15% to 20% of
have much greater mean plasma vWF:Ag concentrations than cases) exhibit qualitative defects of the VWF molecule, with
type O individuals, affecting the severity of symptoms." unequal reductions in the components of the FVIII/vWF
Patients with vWD present with mucocutaneous bleeding: complex, and abnormalities in the multimeric distribution.
frequent instances of epistaxis, ecchymosis, easy bruisability, They are associated with missense mutations of the vWF
gastrointestinal bleeding, menorrhagia, and hemorrhage gene, and inherited as autosomal dominant traits unless
following surgery, childbirth, or dental extractions. These stated otherwise.
manifestations are caused by the inability of platelets to adhere
to subendothelial collagen following injury to the blood Type 2A
vessel.°° vWE and F VII:C are acute-phase reactants, known Type 2A vWD is the most common of the type 2 disorders. It
to increase during stress, inflammation, pregnancy, with oral exhibits a dysfunctional defect of vWE, suggested by a
contraceptive use, estrogen replacement, or after surgery. This disproportionately lower vWF activity than antigenic concen-
variability creates difficulties in the evaluation and diagnosis of tration. There is an absence of the intermediate and HMW
patients suspected of having vWD. Therefore, it may be neces- multimers in the plasma and platelets. Bleeding symptoms are
sary to study these patients on multiple occasions to determine moderately severe; BTs, CTs, RIPA, and vWF activity are
if vWD is the cause of their bleeding symptoms. markedly abnormal; platelet count, PT, and LD-RIPA are nor-
Due to the various types of vWD that have been described, mal. Levels of F VIII:C and vWF:Ag may be normal or
the clinical presentation may vary. Patients who have severe decreased. The aPTT may be affected, dependent on F VIII:C
forms of vWD generally present with a normal PT, a prolonged levels. Treatment involves the use of vWF concentrates or
aPTT that corrects on mixing with pooled normal plasma, nor- commercial F VII concentrates with a near- normal comple-
mal platelet counts, and an abnormal BT test (see Table 26-7). ment of multimers.*! DDAVP is ineffective as a treatment
Laboratory assessment should include assays for F VIII:C modality.
activity, VWF:Ag, RCA, RIPA, LD-RIPA, and vWF multi-
meric analysis. Multimeric analysis will further confirm the Type 2B
specific variant type; identification is essential for proper Type 2B vWD exhibits a qualitative defect of the vWF in
treatment (Table 26-8). which there is an increased affinity for platelet GP Ib. In this
disorder, the plasma shows an absence of only the HMW mul-
VON WILLEBRAND DISEASE TYPES AND SUBTYPES timers, whereas the platelet-associated multimers are nor-
TYPE 1 vWD Type 1, or classic vWD, is the most common mal.°’ The HMW multimers are most important for normal
type (70% to 80% of cases), inherited as a heterozygous auto- platelet function; each subunit contains numerous binding
somal dominant trait. It is associated with partial deletions of sites for the GP Ib receptor on resting platelets and the
the vWF gene that produces a partial quantitative defect, GP IIb/IIIa receptor on activated platelets. This high number
causing only mild symptoms. Patients have reduced levels of of platelet-binding sites correlates with the highly adhesive
a normally functioning vWF, with parallel decreases in properties of this fraction, thereby facilitating aggregation.
vWF:Ag concentration. This may produce either normal A differential laboratory finding in this variant is an abnormal
or abnormal BTs, or CTs according to PFA-100® analysis. LD-RIPA response, i.e., enhanced aggregation is seen when
F VUEC activity can be normal or decreased; this may cause testing with very low concentrations of ristocetin (e.g., 0.2 to
the aPTT to be normal or increased dependent on the F VII:C 0.6 mg/mL). This increased affinity for the GP Ib receptor is
level. The PT and platelet counts are normal. RIPA may be not an intrinsic abnormality of the platelets. The production of
normal or decreased, with a normally absent response to low- the abnormal vWF protein spontaneously binds to normal
dose RIPA, as well. The multimeric pattern is normal but platelets, increasing their clearance from the circulation and
decreased, i.e., the distribution of the various molecular thereby removing the HMW multimers.*! This may be seen
weight multimers on agarose gel electrophoresis is complete in vitro as spontaneous self-aggregation: an aggregation
but equally reduced. Patients with type 1 vWD can be treated response without the addition of any agonist. This subtype
621
Disorders of Plasma Clotting Factors
WoL “ao4unog
‘uorsstuniad yim “772 d “pg6] “erydjapepiyg “‘stapunes gM ‘L] po ‘WuowWaseuryy puke slsouseIg [koIUl]D (pe) q ¢ Aruopy uy “stskjourqy pue uoNe[nseos poo[g -Tf [TA
ysrom-Iepnoajowl-ysiy= MIWH ‘uoneunn[sse jojaiejd poonput-unad0}stl asop-MO[= VdII-CT ‘uoneunnysse jojaietd paonput-unssoysit= WT :AWANIe
10}
ee uNa00}S1I= vou ee sesgee = dud ‘uesnue ge J PURITAN UOA = BVA SAVATIORAiea DIA d = OIA « eIos = ONI eee = oad ‘yewouge= Nav ‘yeuou = N
~ gonesaid3e
pasvarisep
qI—d)
Nav Aesse SUIPUIg 4A\A
sulpulg
N
0}
JAA
Chapter 26
JO [VULION
SuIpulq JAVA
—:1A A
pasevaisep
JAC
Ndv Vdld—dT
N
N
Vdla
A[POyVJAL Ndv OAC PRIN Ndv JON
N
SOUT] NU
pue
ANH
OIPSULIOJUI
juasqy
N
SAMA Jo danjonays
IIIWI [NYA
OHd ION Odd OAC PATRI JAd 10 N Odd PHPIN JHC vod
OAC PAR SV:AMA
OAC Pye Od ION Jd Odd 10'N DAGON Odd 10 N JAd
Ad ION JAC 10 N Odd JO.N JAC JON JHd JON Odd ON OIA A
Odd Pre
JuNOd
ba
49]91"[
eee
OA PLA Oa pitt Jo N
Pe
N
N
N
N
dw) SUIPse,
Nav
Be ae Na@v/N
NEW
ie Nav Nav
ody,
“az
€ addy, ake 4a)91eId NZ adky, adKy, Vz addy ifaddy, ‘sBupurg
A10}VIOGR'T
/AM4-Opnoesg
622 Chapter 26 Disorders of Plasma Clotting Factors
must be differentiated from another form of vWD with simi- variant requires the use of VWF concentrates or cryoprecipi-
lar findings (see platelet-type/pseudo-vWD). There is no evi- tate. DDAVP is ineffective owing to the vWF defect.
dence of an increased incidence of thromboembolic disease.*!
A mild transient or persistent thrombocytopenia may be seen Type 3 vWD
in some patients. Bleeding symptoms are moderate; BTs and Type 3 vWD is the most severe form (1% to 3% of cases),
CTs are increased. The levels of vWF activity, antigen, and F inherited as a homozygous or double heterozygous autosomal
VIII:C activity may be either normal or decreased; the aPTT recessive trait. Total deletion of the vWF gene causes a com-
will be increased relative to the F VIII:C level. The PT is plete deficiency of vWF in the plasma, plateiets, and endothe-
normal. The RIPA will show a normal response. Definitive lial cells. VWF antigen, activity, and multimers are nearly
diagnosis requires the presence of an increased LD-RIPA undetectable. F VII:C activity is usually 2% to 5% (0.02 to
response, an absence of HMW multimers in the plasma, and 0.05 U/mL), due to the lack of its carrier protein.”” BTs, CTs,
a normal platelet-associated multimer distribution. Treatment RIPA, and aPTTs are severely abnormal. The PT, platelet
of this subtype requires the use of vWF concentrates. DDAVP counts, and LD-RIPA are normal. This form may initially be
administration is contraindicated; the increased release of the confused with hemophilia A by its clinical presentation (e.g.,
abnormal vWE molecules with increased binding to normal hemarthroses, intramuscular bleeding). Treatment of this
platelets causes accelerated clearance of normal platelets, form of vWD requires the replacement of vWF and F VIII:C
resulting in a worsening of the thrombocytopenia.*! Subtyp- with concentrates that contain a normal complement of mul-
ing is essential in determining the appropriate treatment. timers, and which has been treated to inactivate any viruses.
Development of anti-vWF alloantibodies is possible if
Type 2M patients are infused with plasma products, which reduces the
Type 2M (‘“multimer”) vWD is a very rare form in which the effectiveness of therapy.’* Allo-antibody formation has been
qualitative defect of vWF causes decreased or absent binding reported in 10% to 15% of type 3 (severe) vWD patients as a
of vWF to GP Ib, as in type 2A; however, the multimer distri- result of transfusion of vWF-containing products, similar to
bution is normal. There are two known mutations in the GP Ib patients with hemophilia A who produce anti-F VHI:C anti-
binding site that selectively impair ristocetin-induced binding bodies (see Specific Inhibitors).”*
of vWF to the platelet, but do not affect botrocetin-induced
binding.” Occasionally, larger than normal multimers may be Platelet-Type/Pseudo-vWD
seen (Vicenza variant).”? vWF activity is disproportionately Platelet-type/pseudo-vWD (Pt/p-vWD) is an abnormality
decreased as compared to levels of the antigen and F VIII:C, intrinsic to the platelet, although its initial laboratory findings
which may be normal or slightly decreased. The binding of are identical to those of type 2B vWD. It is classified as a
vWF to F VUI:C is normal.“ BTs and CTs are increased with pseudo-vWD because there is no genetic mutation in the VWF
normal platelet counts. The PT is normal, and the aPTT gene. It is inherited as an autosomal recessive trait. A molec-
will be normal if the levels of F VIII:C and vWF antigen are ular abnormality in the GP Ib receptor causes spontaneous
normal. The RIPA response is decreased with a normal binding of the HMW multimers, clearing them from the plasma
LD-RIPA. Treatment requires the use of vWF concentrates or and causing increased platelet turnover. A mild thrombocytope-
cryoprecipitate. DDAVP is ineffective owing to the vWF nia develops and larger-than-normal platelets are present on a
defect. peripheral smear.*' The presence of a positive LD-RIPA
response, as in type 2B, may cause misdiagnosis. Classification
Type 2N is crucial, because different treatments are required for these
Type 2N (“Normandy”) vWD is inherited as an autosomal two disorders. Differentiation can be assisted by performing a
recessive trait, and is defined by a markedly decreased affinity test known as the vWF binding assay. In this method, the
of vWF to F VIII:C. Genetic (missense) mutations alter the F patient’s plasma vWF is tagged with a vWF-specific mono-
VUl:C-binding region of vWF. The majority of these clonal antibody, and reacted with normal formalin-fixed
patients are either homozygous or compound heterozygous for platelets, at various concentrations of ristocetin. This assay
these mutations; another group of patients with this variant cannot rule out Pt/p-vWD; however, a patient with type 2B
appears to be heterozygous for one of the mutations that pro- vWD would show increased vWF binding at lower concentra-
duces the F VIII:C binding defect as well as co-inheriting an tions of ristocetin than normal vWF or vWF in Pt/p-vWD.7
allele associated with type 1 vWD.°’’!” The levels of VWF An abnormal vWF binding assay is diagnostic of type 2B
antigen and activity are normal or decreased, the multimer vWD. Diagnosis of Pt/p-vWD requires a normal vWF bind-
distribution is normal, and the level of F VIII:C is dispropor- ing assay result, in conjunction with other test results typical
tionately lower than levels of VWF, which may have caused of this type (as mentioned earlier). Platelet concentrates are
some patients to be misdiagnosed in the past as hemophiliacs. used to treat patients with this defect.
The disorder may be considered an autosomal form of hemo-
philia A. The platelet-dependent function of vWF is normal, ACQUIRED VON WILLEBRAND DISEASE SYNDROME A
therefore the RIPA and LD-RIPA will be normal. Platelet rare bleeding disorder, with symptoms identical to vWD but
counts, BTs, and CTs are normal. The F VIHI:C is structurally without a genetic defect, can develop in association with a
normal, but has a shortened half-life in the plasma due to variety of conditions or without any apparent cause. This dis-
its inability to bind to its carrier protein. Treatment of this order is known as acquired von Willebrand Disease (AvWD)
Chapter 26 Disorders of Plasma Clotting Factors 623
syndrome because it does not involve mutations of the vWF normal hemostasis, the age at presentation, a negative family
gene. It can occur at any age, but usually affects the elderly; history, and the presence of an underlying disorder can pro-
either sex can be equally affected. Hemorrhagic symptoms vide a presumptive diagnosis. Detection of vWF-specific
vary considerably among patients and there is a negative fam- antibodies provides a definitive diagnosis. In addition, analy-
ily history. While the incidence of AvWD syndrome is sis of the vWF propeptide (vWF:Ag II): a fragment cleaved
extremely rare, it is most often associated with monoclonal from the mature vWF antigen within the endothelial cell prior
gammopathies, such as multiple myeloma and Waldenstr6m to its release, can assist in distinguishing congenital vWD
macroglobulinemia; and with lymphoproliferative disorders, from AvWD syndrome. vWF:Ag II and mature vWF antigen
such as the lymphomas or chronic lymphocytic leukemia. occur in equimolar amounts in congenital WWD. However, in
Other disorders can include: Wilms tumor; congenital cardiac AvWD syndrome, due to immune complex clearance of the
defects or mitral valve prolapse; angiodysplasia; other solid mature vWF antigen, vWF:Ag II is in excess.”
tumors; autoimmune disorders; and collagen vascular diseases, Some cases of AVWD syndrome can be eradicated by
such as systemic lupus erythematosus (SLE).”? Moderate to treating the underlying disorder. This may involve surgery,
severe bleeding from mucosal surfaces (e.g., epistaxis, gastroin- chemotherapy, radiation, or immunosuppression. Treatment
testinal and gingival bleeding) are the primary symptoms, also must be tailored to the specific cause; the presence of an
occurring following invasive procedures or surgery.’° autoantibody will affect the survival of any vWF-containing
AvWD syndrome can arise from either a reduced rate product administered to the patient. High-dose intravenous
of synthesis of vWF or rapid clearance from the circulation. immunoglobulin (IVIG) therapy is effective in these patients.
Reduced synthesis can occur in hypothyroidism; laboratory The circulating autoantibody is neutralized by the high con-
results resemble those found in type | vWD. Treatment with tent of anti-idiotypic antibodies (i.e., antibodies that interact
thyroid hormone replacement causes a return to normal with the antigen-combining region of other antibodies) in the
hemostasis. Rapid clearance can be attributed to: IVIG preparation.” This allows a spontaneous rise in F VII:C
and vWE levels to occur which can last 2 to 3 weeks, as pro-
* Development of vWF-specific antibodies which form immune
phylaxis for surgical procedures or to maintain hemostasis.
complexes that are cleared by the reticuloendothelial system;
However, most cases are managed with supportive measures
* Selective adsorption of HMW multimers onto the surface of
such as controlling acute hemorrhage or preventing complica-
malignant cells due t6 aberrant expression of the GP Ib recep-
tions from hemorrhage. These measures may include the use
tor, or to activated platelets in essential thrombocythemia; or
of DDAVP, F VIII:C/vWE concentrates, high-dose [VIG ther-
¢ Proteolytic or mechanical degradation: e.g., induced by certain
apy, corticosteroids, plasma exchange, immunoadsorption, or
drugs, or associated with congenital cardiac defects, such as
recombinant activated F VII concentrates.*°*!
ventricular septal defects or mitral valve prolapse; these create
turbulent blood flow disturbances around the defect leading to
the increased activity of a catabolic enzyme of vWF.
Factor IX (Christmas Factor; Plasma
Surgical correction of the defect or cessation of drug Thromboplastin Component [PTC])
usage can resolve the coagulopathy.
AvWD syndrome may also occur without the presence Factor IX (F IX) is a single-chain glycoprotein (Mr approx-
of an underlying disorder. An antibody spontaneously devel- imately 60 kD) that is synthesized by the liver and is
ops that is directed against the vWF:F VIII complex, resulting vitamin K dependent. The gene coding for its production
in a concomitant reduction of F VHI:C, vWF:Ag, and RCA; also resides on the X chromosome. In the coagulation
or it binds vWF alone, resulting in normal F VHI:C activity sequence, F IX participates in the intrinsic pathway, where
without any measurable vWF activity.”” Autoantibodies that it is activated by F Xla in the presence of calcium ions to
interfere with vWF function or cause rapid clearance from the become a serine protease, F [Xa. A second mechanism of
circulation are usually of the IgG type, but occasionally can activation, which bypasses the intrinsic contact system
be of the IgM or IgA type.°' (F XII, high-molecular-weight kininogen, prekallikrein),
Laboratory findings for AVWD syndrome are those typi- occurs through TF and F VIla (see Factor VI).** F [Xa, in
cally seen in vWD: variable aPTT, decreased vWF activity the presence of F VIIIa, calcium ions, and platelet phospho-
discordant with the level of vWF antigen, variable levels of F lipids (PF3), forms the intrinsic tenase complex, that
VIII:C and vWF antigen, and prolonged BTs. Multimer activates F X.° Although F [Xa alone is capable of slowly
analysis most commonly shows a loss of the HMW multi- activating F X in the presence of phospholipid and calcium
mers, as seen in type 2A vWD. The PT, platelet count, and ions, neither F VIII alone nor F VUI plus thrombin activates
other factor activity levels are normal. These results are indis- F X in the absence of F [Xa.® Factor IX is as critical to
tinguishable from the inherited form. It may be possible to hemostasis as F VHI:C. The generally accepted normal adult
demonstrate inhibitory activity by performing an RCA assay reference range is 50% to 150% (0.5 to 1.5 U/mL).
on a mixture of patient plasma with pooled normal plasma,
which would remain abnormal; in addition, adding patient HEMOPHILIA B
plasma to pooled normal plasma may cause a reduction of Factor [X deficiency, also known as hemophilia B, is also a
assayed vWF:Ag. One or both of these assays may be posi- sex-linked bleeding disorder, as in hemophilia A. Females
tive. Clinical history of the patient reflecting previously with the deficiency are quite rare; they are obligate carriers
624 Chapter 26 Disorders of Plasma Clotting Factors
of the disorder. However, spontaneous mutations do occur. chain. This reaction, in the intrinsic pathway, is catalyzed by
The clinical symptoms of hemophilia A and B are identical; the intrinsic tenase complex (F [Xa, F VIIa, calcium ions,
it was originally believed that these were the same disorder. and platelet phospholipid). This same bond is cleaved by the
In 1947, it was observed that mixing plasmas from two extrinsic tenase complex (F VIla, TF, and calcium ions) from
unrelated hemophilic patients would normalize the clotting the extrinsic pathway.’° The normal adult reference range is
time in the mixed plasma.***°Hemophilia B became known 50% to 150% (0.5 to 1.5 U/mL).
as “Christmas disease” by Biggs and colleagues,*’ named Inherited F X deficiency is extremely rare. F X, also
after the family of the first patient they had seen with the known as Stuart-Prower factor, gets its name from two cases
disorder. Since then, F IX has also been known as Christ- found to have this abnormality. At the time of their diagnoses,
mas factor, or synonymously as plasma thromboplastin Mr. Stuart was the 36-year-old son of a consanguineous rela-
component (PTC). tionship between an aunt and nephew within a large, interre-
The incidence of inherited F [X deficiency in the normal lated family living in the Blue Ridge Mountain region of
population is approximately | per 100,000.** The level ofF IX North Carolina®’; and Miss Prower was a 22-year-old English
activity determines the severity of the disorder, with the most woman with a history of two significant hemorrhagic
severe deficiencies having less than one percent activity, and episodes following dental extraction and _ tonsillectomy,
milder deficiencies having between 5% to 25% activity. In who also had a brother that died of postoperative hemorrhage
severe hemophilia B, there is a total absence of F [X, demon- following tonsillectomy at the age of 5.*' Its transmission is
strated by the absence of activity by factor assay as well as autosomal recessive, with an occurrence of less than 1 per
antigenic assay. Some hemophilia B patients synthesize a 500,000; however, the heterozygous state can occur in
nonfunctional variant of the F LX molecule, whereas others do approximately | per 500.
not have identifiable F IX antigen in the plasma and, there- Deficiency of F X may occur at any age, but the most
fore, have a true absence of synthesis of the molecule.*’ severe hemorrhagic symptoms occur in the very young.
Measurement of F [X antigen and clottable activity levels Bleeding sites vary according to the severity of the deficiency.
greatly increases the accuracy of determining carrier status.”° Clinical symptoms range from easy bruising; epistaxis; hema-
Acquired deficiency states can be seen in patients with liver turia; gastrointestinal bleeding; or menorrhagia in mildly
disease, nutritional vitamin K deficiency, and in patients on affected patients, to hemarthrosis; central nervous system
oral anticoagulant therapy. hemorrhage; and severe postoperative hemorrhage in the
Treatment of patients with severe to moderate F IX most severely affected patients.
deficiency consists of infusions of FFP, PCC, or purified F Usually, the diagnosis of inherited F X deficiency can be
IX concentrates. Patients with mild deficiencies usually made by an appropriate family history and laboratory data.
receive prophylactic treatment in association with both However, differentiating between inherited and acquired defi-
minor and major surgery. The most serious complication ciencies should include the consideration of liver disease and
in patients with hemophilia B, as in hemophilia A, is the vitamin K deficiency. Acquired F X deficiencies usually coin-
formation of antibodies/inhibitors to F IX. These are cide with other vitamin K—dependent factor disorders.
extremely difficult to handle, especially in life-threatening F X deficiencies have been reported in patients with
situations, and they occur in approximately 10% of patients amyloidosis.’* Rare, variant forms of F X deficiencies exist
with F IX deficiency.?!” due to amino acid substitutions or deletions, which produce
The laboratory findings in F IX deficiency include: a different patterns of results with PT, aPTT, Stypven time and
normal PT, TT, and BT. There is a prolongation of the aPTT F X antigen levels.”
that corrects when mixed with pooled normal plasma. Laboratory testing results typically seen in this defi-
Patients with inhibitors would demonstrate non-correction ciency include a prolonged PT and aPTT, that corrects when
of the aPTT in mixing studies. Factor assays for F IX mixed with pooled normal plasma. The TT and BT are nor-
activity produce decreased to absent levels. Patients with mal. The Russell’s viper venom time (Stypven time test) is
inhibitors may show dilutional increases in factor activity prolonged, due to its dependence on normal levels of factors
during factor assays; these must be differentiated from II, V, X, fibrinogen, and phospholipid. Factor activity assay,
a non-specific inhibitor (see Circulating Anticoagulants/ typically using the PT methodology, is decreased or absent
Inhibitors, later in this chapter) by additional testing and (Table 26-9). Factor assays based on either the PT or aPTT
clinical presentation. method may be used to determine factor activity (F X is part
of the common pathway, which is measured by both tests);
Factor X (Stuart-Prower Factor) however, there are variant forms of F X deficiency in which
discrepancies in the results can occur, depending on which
Factor X (F X) (M, 58.8 kD) is a vitamin K—-dependent glyco- methodology is used.
protein. It is composed of a light chain and a heavy chain, Treatment consists of transfusions with FFP or PCC.
held together by a single disulfide bond.”*°* The heavy chain However, the need for such measures should be guided by the
contains the catalytic ___domain of the protein.” It is synthesized severity of the hemorrhagic episode. Levels of 10% are con-
by the liver and released into the plasma as a precursor to a sidered adequate for hemostasis. If the deficiency is acquired
serine protease. The conversion of F X to its proteolytic form, by poor diet or oral anticoagulant therapy vitamin K supple-
F Xa, involves the cleavage of a peptide bond in the heavy mentation may be therapeutic.
Chapter 26 Disorders of Plasma Clotting Factors 625
charged surfaces in-vitro (solid-phase activation) such as such as placenta, platelets, macrophages, and prostate; and
glass, celite, or kaolin; or chemical activation with ellagic contain the active catalytic site for transglutaminase activity.
acid, causes the auto-activation of F XII and its conversion to The B chains are synthesized in the liver and circulates as
a serine protease.''® This process initiates the intrinsic path- a free dimer or as a complex with the A chains. It is postu-
way of coagulation. In-vivo (fluid-phase) activation occurs by lated that the B chain contributes to the stabilization of
contact with the contents of cell membranes or negatively the A chain; however, the function of the B chain remains
charged subendothelial structures, such as collagen. In this unknown.
process, F XII undergoes a conformational change exposing Congenital F XIII deficiency is inherited as an autosomal
its active site, which then converts PK to kallikrein and sub- recessive trait, with a high frequency of consanguinity in
sequently activates F XI.''’ Additional F XIla is formed due families with this disorder.'*! Clinically, the homozygous
to cleavage by enzymes such as trypsin, plasmin, or deficiency presents with moderate to severe hemorrhagic
kallikrein. The proteolytic product is a two-chain molecule diatheses characterized by initial hemostasis followed by the
composed of a heavy chain and a light chain held by a disul- recurrence of bleeding 12 to 36 hours or more after the initial
fide bond.'°! The presence of small amounts of F XIla leads traumatic event. This results from the dissolution of the solu-
to the activation of its substrates: PK, F XI, and HMWK. ble, unstabilized fibrin clot. These soluble fibrin clots are
These in turn activate more F XII, which serves to amplify highly susceptible to degradation by plasmin. This disorder
the reactions. F XIla, F Xa, and kallikrein stimulate the con- can be diagnosed at birth because the most common clinical
version of plasminogen to plasmin, the enzyme critical to symptom is bleeding from the umbilical cord stump. There is
fibrinolysis. also poor wound healing and abnormal scar formation. An
Deficiency of F XII, also known as Hageman trait, acquired partial deficiency has been reported with several dis-
after the first patient identified with this deficiency in 1955, eases, including leukemias, DIC, and severe liver disease.
is a very rare disorder (1:1 million) inherited in an autoso- The development of [gG autoantibodies to F XIII have been
mal recessive fashion. Homozygotes are severely deficient reported, which have resulted in severe bleeding diatheses.
(less than 1% activity; 0.01 U/mL), while heterozygotes Treatments had once consisted of transfusion with FFP or cry-
may have 40% to 60% activity (0.40 to 0.60 U/mL). oprecipitate, which could cause circulatory overload and
Normal F XII activity levels fall in the range of 70% to virus transmission; there now are viral-inactivated F XIII con-
140% (0.70 to 1.40 U/mL). This disorder is asymptomatic; centrates available.
it is not associated with clinical bleeding or surgical hemor- All routine laboratory screening tests for hemostasis will
rhagic risk. Paradoxically, there may be an increased risk of indicate normal results; PT, aPTT, fibrinogen, BT, and platelet
thrombosis due to inadequate activation of the fibrinolytic count. The screening test for F XIII deficiency is based on the
pathway; it is notable that Mr. Hageman’s death was due to solubility of a recalcified plasma clot in a 5 molar (M) urea or
a pulmonary embolism. The deficiency is usually an inci- 1! M monochloroacetic acid solutions. If the plasma is defi-
dental discovery during presurgical screening. Laboratory cient in F XIII, the clot will dissolve within 24 hours. This is
analysis indicates a normal PT and a prolonged aPTT: a qualitative assay and does not reflect the level of F XIII
greater than 200 seconds in homozygotes that corrects on present, although activity of approximately 1% of normal is
mixing with pooled normal plasma. Specific factor analysis sufficient to prevent both an abnormal test result and symp-
yields decreased or absent F XII activity, which is required toms in-vivo.'** Actual F XIII activity can be assayed by a
for definitive diagnosis. chromogenic enzymatic method. The normal reference range
is 60% to 150% (0.6 to 1.5 U/mL).
Factor XIll (Fibrin-Stabilizing Factor)
Prekallikrein (Fletcher Factor)
In the final stages of the plasma coagulation process, the gen-
eration of thrombin, polymerization of fibrin, and activation Prekallikrein (PK) (M, of approximately 100 kD), is a single-
of factor XIII (F XIIa) is responsible for the formation of a chain protein synthesized by the liver.'?? Conversion to its
stable fibrin clot. F XIII is a proenzyme for a plasma transg- active form, kallikrein, is catalyzed by F XIla and HMWK.
lutaminase; in the presence of fibrin, thrombin and calcium Approximately 75% circulates bound to HMWK,'4 and 25%
ions convert F XIII to the enzymatic F XIIIa."'*"!? It is the circulates as free PK. Kallikrein, a serine protease, is impor-
only enzyme in the coagulation system that is not a serine tant in the activation of three other biologic systems: the fib-
protease. F XIIa functions as a catalyst, forming covalent rinolytic, complement, and kinin systems. Kallikrein cleaves
bonds between various protein substrates such as fibrin HMWKkK into bradykinin: an important mediator in the inflam-
monomers, alpha-2-antiplasmin, fibronectin, and collagen.!?° matory response; it leads to the conversion of plasminogen to
The action of this cross-linking of various plasma and extra- plasmin: a potent enzyme that degrades fibrin, fibrinogen, and
cellular matrix proteins contributes to hemostasis, wound factors V and VIII:C; and, through plasmin, it also activates
healing, and the maintenance of pregnancy. the complement components C3 and CS. Neutrophils and
F XII (M, 320 kD) circulates with fibrinogen. It has a monocytes are also attracted to the site of injury by the pres-
very long half-life of approximately one week. Extracellular ence of kallikrein.
or plasma F XIII consists of two subunits: two A chains and Deficiency of PK, known as Fletcher trait, is not associ-
two B chains. The A chains exist in various tissues and cells ated with clinical bleeding, even during surgery or after
Chapter 26 Disorders of Plasma Clotting Factors 627
Specific Inhibitors
High-Molecular-Weight Kininogen
(Fitzgerald Factor; Williams—Fitzgerald- The specific inhibitors are characterized as antibodies. They
Flaujeac Factor). either directly inhibit (neutralize) the factor’s activity or cause
its increased clearance by forming immune complexes. They
High-molecular-weight kininogen (HMWK) (M, 200 kD) is a may occur secondary to transfusion, replacement therapy, or
single-chain glycoprotein produced by the liver. HMWK is both. They can also arise spontaneously, in a person without
known as the “‘contact activation cofactor” because it is required any underlying disorders. Inhibitors could potentially arise
for contact activation.'’? Studies indicate that plasma HMWK against any factor.
exists as a pro-cofactor that can be activated through cleavage F VIII:C inhibitors, the most frequently encountered, as
by kallikrein.'°° The kinin system is important in chemotaxis, well as rare instances of F IX inhibitors, can develop in
vascular permeability, and inflammation. Kallikrein cleaves patients with severe hemophilia as a result of transfusion and
HMWK into a two-chain molecule held by a disulfide bond. exposure to the respective deficient factor. These would be
This reaction releases bradykinin, a small peptide with actions classified as alloantibodies: antibodies produced against
related to pain and inflammation. The binding of HMWK to “nonself’ antigens. Their presence, however, does not
endothelial cells is necessary for the binding of F XI (with increase the frequency of bleeding episodes.'*? Hemarthroses,
which it is in complex) and its activation to F XIa, as well as for muscle and soft tissue hemorrhage continue to be the clinical
the activation of F [IX to F [Xa by F XIa."”! symptoms. F VIII:C inhibitors are predominately IgG anti-
Deficiency of HMWK, known as Fitzgerald trait or bodies with a specificity to the coagulant’s antigenic portion
Williams trait, has been described as an autosomal recessive of the F VIH/vWE complex, but do not interfere with the
disorder, and like PK, shows no predilection to race. HMWK function of vWF; there is no prolongation of the BT. An
deficiency is rare and not associated with any bleeding diathe- inhibitor should be suspected in any hemophiliac if transfused
sis. It presents with a markedly prolonged aPTT that corrects factor replacement products appear to have reduced effective-
in mixing studies with pooled normal plasma. Mild to moder- ness, hemostasis is difficult to achieve, or both.
ate acquired deficiencies can occur with liver disease or DIC. More frequently, however, inhibitors to F VIII:C may oc-
While there is no associated clinical bleeding disorder, cur as an autoantibody: an antibody produced against “self”
patients with HMWK deficiency have been observed with antigens; most often seen in patients with rheumatoid arthritis,
deep vein thrombosis and pulmonary embolus.'** systemic lupus erythematosus (SLE) and other autoimmune
Replacement therapy is not indicated with F XII, PK, or diseases; drug reactions, and lymphoproliferative diseases or
HMWK deficiencies. Patients requiring major surgery have multiple myeloma. They also occasionally occur in women
not had any incidence of bleeding. This is presumably be- during pregnancy or postpartum, and in the elderly with no
cause the extrinsic pathway of coagulation, through F VII and apparent underlying disease. This disorder is known as
tissue factor, remains intact, and F XI can be activated by acquired hemophilia. Its etiology is unknown and is apparently
thrombin generated from the extrinsic pathway.'**'** There- unrelated to any previous exposure to blood or blood
fore, F XI can be activated without the need for HMWK, PK, products.'* The clinical symptoms of this disorder may be
or F XI. severe and life-threatening, including large hematomas, gross
628 Chapter 26 Disorders of Plasma Clotting Factors
hematuria, retropharyngeal or retroperitoneal hematomas, or a low titer inhibitor (less than 5 BU) with no rise in titer after
cerebral hemorrhage.'* Hemarthroses are less common. The exposure to additional F VIII:C (i.e., “low responders”) can
bleeding cannot be controlled with factor replacement, and usually be treated with high concentrations of F VIII:C, in an
could further exacerbate the situation. Acquired hemophilia effort to overwhelm the antibody by saturating all its binding
should be suspected in anyone with no prior history who pre- sites. Other options include: porcine F VIII:C, recombinant
sents with massive bruising or hematoma.'*° The mortality rate F VIII concentrates, F IX concentrates, [VIG, and rFVIla.'*”
associated with acquired hemophilia is approximately 20%. Patients with high titer inhibitors (greater than 5 BU) with
A normal PT and prolonged aPTT, with non-correction a marked increase in titer after exposure to F VIII:C (ie.,
in a mixing study with pooled normal plasma, is a classical “high responders) present a treatment dilemma: options can
laboratory finding. However, some patients with mild to mod- include porcine F VIII:C concentrates; steroids, alone or in
erate inhibitors may still show correction on the initial mixing combination; immunosuppressive therapy; cytotoxic agents;
study. These antibodies also demonstrate a characteristic PCCs; high-dose IVIG, extracorporeal immunoadsorption or
known as “‘time-and-temperature dependency”: a 2-hour incu- plasmapheresis for patients with very high inhibitor titers.'°°
bation at 37°C, of the patient plasma/pooled normal plasma rFVIla is the newest therapeutic option available, used pri-
mixture from the initial mixing study, will show a further pro- marily to bring hemostasis under control while other modali-
longation of the aPTT when repeated after the incubation ties focus on suppressing the immune response. Inhibitors can
period. An “incubation control,” consisting of a new mixture persist for weeks or months; some titers may decline to near
prepared from separate, individually incubated patient and zero, only to recur as an anamnestic (memory) response in
pooled normal plasma samples, will not demonstrate the fur- some patients. Spontaneous remissions have been reported in
ther prolongation, indicating the effect of the inhibitor on the approximately 38% of cases, most often occurring in women
additionally provided F VIII:C from the pooled normal postpartum or in drug-induced reactions.!**'°°
plasma during the 2-hour period. This step is extremely Spontaneously acquired inhibitors to factors II, VII,
important in order to properly characterize the disorder, and and X are very rare. F II inhibitors are described in associa-
should be routinely performed for all abnormal screening tion with the LAC (see Nonspecific Inhibitors, following).
aPTTs with an order for follow-up testing. There have been several reports of F XI and F XII inhibitors;
Inhibitors to specific clotting factors are demonstrated by several cases were reported as F XI deficiency with acquired
performing factor assays for all suspected factors indicated by inhibitors posttransfusion and other cases occurring in
the results of the PT and/or aPTT. In the case of a F VIII:C patients with SLE.'**:'%° Acquired F V inhibitor is also rare;
inhibitor, only this factor will be reduced or absent. It is pos- approximately 100 cases are reported in the literature, with
sible for an inhibitor effect (dilutional increase in activity) to only one reported case in a F V-deficient patient; the remain-
occur in other factor assays, since the inhibitor in the patient’s ing cases occurred in elderly patients who previously had
plasma can react against the F VIII:C that is present in the normal levels.'*°
single factor deficient plasma used in the assay; e.g., F IX Antibodies to vWF can occur in patients congenitally
deficient plasma used in assays for F IX activity contains a deficient of vWF (severe vWD-type 3) as alloantibodies, or
normal amount of F VIII:C activity with which it can react. as autoantibodies in normal patients or in those with underly-
The inhibitor titer can be quantitated in a method ing disorders, causing AVWD syndrome (see acquired von
known as the Bethesda Inhibitor Assay. In this procedure, Willebrand disease syndrome, earlier in this chapter).
the patient’s plasma and various dilutions are incubated in
mixture with an equal volume of pooled normal plasma for
2 hours at 37°C; pooled normal plasma diluted with an Nonspecific Inhibitors: Lupus
equal volume of buffer serves as the control. The FVIII:C Anticoagulants
activity of the patient plasma mixture, each dilution, and
the control, is subsequently determined by factor assay. The Nonspecific inhibitors are not directed toward specific factors
percentage of residual F VIII:C in each mixture is then and are not usually associated with a hemorrhagic risk.'*°!*!
calculated, by comparison to the F VIII:C activity of the They are frequently discovered as an unexpectedly prolonged
control. A residual F VIII:C activity of 50%, or the amount aPTT on a routine preoperative evaluation. These results
of inhibitor that will inactivate 50% of the F VIII:C origi- would give the impression that the patient may have a factor
nally present, is defined as one Bethesda unit (BU) of deficiency. However, mixing studies with pooled normal
inhibitor. The inhibitor titer is determined by the dilution/ plasma does not produce a correction of the aPTT. This phe-
mixture with a residual FVIII:C activity of, or closest to nomenon was first recognized in patients with SLE,'? and
50%: e.g., 50% residual F VII:C at a 1:20 dilution is inter- consequently became known as the lupus anticoagulant
preted as a titer of 20 BU. This method can be applied to (LAC). This designation is a misnomer, however, because
any of the clotting factors. these inhibitors often occur in patients without SLE, who are
Treatment of patients with specific inhibitors is first otherwise healthy and have no underlying medical conditions.
aimed at stabilization of hemostasis; the overall goal is to Therefore, it would be more appropriate that they be termed
eradicate the antibody. The choice of therapeutic modality is lupus-like anticoagulants. LACs are immunoglobulins, usually
dependent on the titer (low versus high) and the patient’s IgG and/or IgM, that interfere with one or more phospholipid-
response on exposure to additional F VIII:C.'*° Patients with dependent coagulation tests, e.g., PT and aPTT. This laboratory
Chapter 26 Disorders of Plasma Clotting Factors 629
phenomenon is a direct reaction against the negatively APLS'**; and thromboses associated with aCL syndrome are
charged (anionic) phospholipids present in the reagents used far more common than with LAC syndrome alone.'*
for coagulation screening assays. LACs do not inactivate the Venous thrombosis presents as deep vein thrombosis (DVT)
clotting factors in vitro: they inhibit the formation of the pro- with or without pulmonary embolism (PE); there may also be
thrombinase complex (F Xa, F Va, calcium ions, F II, and thromboses in unusual sites. Arterial thromboses present as
phospholipids) by binding to the phospholipids, causing a stroke, transient ischemic attacks, or myocardial infarction.'*°
prolongation of the clotting time.'**'°°'4? This in vitro aPL antibodies are actually directed against negatively
effect of “anticoagulation” is paradoxically associated with a charged phospholipid-protein complexes; they require the
hypercoagulable (thrombotic) state in-vivo. presence of specific proteins to facilitate phospholipid binding,
It was also noted that there was a high incidence of bio- such as prothrombin, or a natural anticoagulant protein: beta-
logical false-positive results on serologic tests for syphilis 2-glycoprotein-1 (B,GP-1), also known as apolipoprotein H.
(VDRL) in SLE patients. This method utilizes a phospholipid There have been some cases of patients with LAC that have
known as cardiolipin (diphosphatidylglycerol) extracted from had clinically significant bleeding, which in nearly all cases
bovine heart tissue: it is an anionic phospholipid found in the could be attributed to an abnormality other than the LAC.'*° In
inner mitochondrial membranes of cardiac and skeletal mus- some cases, the PT is prolonged and may or may not correct
cle cells, and some bacteria. These results were found to be with mixing studies. This could be attributed to an acquired
significantly associated with LACs and the risk of thrombo- hypoprothrombinemia, caused by an anti-prothrombin anti-
sis. It was determined that the sera of these patients contained body that binds prothrombin but does not neutralize its coagu-
IgG, IgM, and/or IgA anticardiolipin (aCL) antibodies. aCL lant activity in-vitro; the prothrombin activity and antigen
antibodies and LACs both complex with negatively charged are decreased to the same extent, due to immune complex
phospholipids bound to protein. These antiphospholipid anti- formation and rapid clearance.'**:'°° This has been named the
bodies (aPL) each comprise two related but clinically distinct hypoprothrombinemia-LAC syndrome. If the antibody was of
syndromes: the anticardiolipin antibody (aCL) syndrome and a neutralizing type, the prothrombin antigenic concentration
the LAC syndrome. Both are frequently associated with would have been normal. Serum anti-prothrombin antibodies
thrombosis, fetal loss, or thrombocytopenia, with or without can be detected by ELISA assays. Other causes of bleeding
autoimmune disorders.'*° These syndromes belong to a fam- can be attributed to thrombocytopenia, alone or in combina-
ily of disorders known as the Antiphospholipid antibody syn- tion with a moderate prothrombin deficiency.'*'*!
dromes (APLS). They are one of the most common causes of Antibodies to 8,GP-1 are frequently detected in patients
acquired coagulation defects associated with venous and/or with clinical symptoms of APLS. Approximately 20% of
arterial thrombosis.'** The APLS can occur spontaneously, patients testing negative for aCL antibodies will test positive
without any underlying autoimmune disorder, known as for anti-B,GP-1.'°* The presence of anti-8,GP-1 tends to sup-
primary APLS. A serious and often fatal manifestation of port a diagnosis of APLS in patients with symptoms even
APLS, known as catastrophic antiphospholipid syndrome though aCL or LAC testing may be negative.'** If patients test
(CAPS), is characterized by the development of multiple positive for aCL antibodies, but their clinical presentation is
organ thromboses (infarctions) over a very short period of not consistent with APLS, a negative result for anti-8,GP-1
time (days to weeks). The mortality rate is approximately could rule out the presence of APLS. aCL antibodies associ-
50%. APLS occurring in association with SLE and other ated with infections do not tend to be 8,GP-1-dependent.
autoimmune diseases, acquired immune deficiency syndrome They can often be observed during the convalescent phase of
(AIDS), infections of bacterial, viral, or protozoal origin; acute bacterial or viral infections and in individuals with
drugs such as procainamide or chlorpromazine, inflammation, syphilis. These infection-induced antibodies are usually tran-
malignancies, and lymphoproliferative disorders is known as sient and are not associated with an increased risk of clinical
secondary APLS.'*° Other antiphospholipid antibodies can be complications. Therefore, testing for anti-8,GP-1 can be more
detected using phosphatidylinositol, phosphatidic acid, phos- specific for APLS than aCL antibody testing.'* In general, all
phatidylserine, or phosphatidylethanolamine. patients that test positive for aCL antibodies should be
The LAC syndrome is much more frequently associated retested after 6 to 8 weeks to rule out transient antibodies that
with venous than with arterial thrombosis. The presence of the are usually of no clinical significance.'* Definitive diagnosis
LAC is more specific for APLS than the aCL antibody, how- of APLS requires a positive result for LAC and/or aCL anti-
ever, as the titer of aCL antibody increases its specificity for bodies that persist for at least 8 to 10 weeks and accompany
APLS also increases, especially if it is of the IgG isotype.'* the presence of arterial or venous thrombosis or complica-
Approximately 90% of patients with LACs have also been tions of pregnancy.
found to have at least one isotype of aCL antibody pre- The detection of aCL antibodies requires the perfor-
sent!4°-!47- however, there are clinical situations in which aCL mance of serological tests via ELISA methodology, using
antibody is present without the LAC.'* There is also a high microtiter plates coated with purified cardiolipin antigen,
incidence of thrombotic disease in patients with elevated aCL because they do not prolong clot-based assays. The ability to
antibodies; as the level of antibody increases so does the inci- detect LACs, however, is dependent on the sensitivity of the
dence of thrombosis, fetal loss, and thrombocytopenia.'** screening reagents and the preparation of the plasma prior to
Generally, the presence of aCL antibodies is considered to analysis. Platelets, that are rich in phospholipids, must be
be a more sensitive indicator than LAC in the detection of depleted from the test plasma by centrifugation or filtration to
630 Chapter 26 Disorders of Plasma Clotting Factors
less than 10,000 platelets/uL, to avoid neutralization of the phospholipid for prothrombinase production, and a shorter
inhibitor, especially if the sample is to be frozen for further clotting time is produced. A ratio, calculated from the two
testing at a later time (freezing and thawing will lyse the clotting times and their respective mean normal clotting
platelets). Hemolysis also poses an interference due to cell times, is used to determine the presence of an LAC. If the
lysis, which would also include platelets, as well as red and ratio is greater than 1.2, an LAC is present (see Table 26-10):
white blood cells; those specimens must be recollected.
Patient plasmas containing LACs usually demonstrate a pro- Example:
longed aPTT without correction on mixing study, and a nor- RVVT screening: 50 seconds (NL = less than
mal PT, mostly because PT reagents have a much higher 40 sec; mean =
phospholipid content which masks the effect. However, it has B8Isec)
been observed that up to 30% of LACs can initially correct dRVVT 31 seconds (NL = less than
and then exhibit the time-and-temperature-dependent effect confirmatory: 35 sec; mean =
on the incubated mixing study.'°*'°° In addition, aPTT 28 sec)
reagents are manufactured with variable sensitivities to the Screening ratio: 50/33 = 1.52
LAC; some institutions do not desire a sensitivity to LACs, as Confirmatory ratio: 31/28 = 1.11
determined by the medical director, since these usually do not dRVVT ratio: 1.52/1.11 = 1.37 (NL = less than
represent a surgical risk. The LAC may also affect aPTT-based 1.20)
factor assays; a prolongation of the clotting time in factor
assays translates to a decreased factor activity. The patient The commercially available dRVVT reagents contain an
could possibly be misdiagnosed with a factor deficiency/ anti-heparin agent to prevent interferences by this frequent
inhibitor. For this reason, factor assays must be performed on contaminant and therapeutic agent. In addition, this test can
at least two to three serial dilutions of the patient’s plasma to be performed on patients with a normal aPTT because these
demonstrate an increase in factor activity with the dilutional reagents can vary in their sensitivity to LACs; the dRVVT is
decrease of the inhibitor’s concentration. The presence of mul- a more sensitive test than the aPTT.
tiple factor assays with this “inhibitor effect,” non-correction The PNP/high phospholipid aPTT method is a modified
on mixing studies with pooled normal plasma, and no clinical aPTT in a two-step process: the phospholipid is an extract of
evidence of bleeding are most suggestive of a nonspecific human platelets. The first step is an aPTT with added buffer,
inhibitor. producing a prolonged aPTT. The second step is an aPTT with
Because LACs are such a heterogeneous group of anti- the added platelet extract. This increases the phospholipid
bodies, using only one type of assay is insufficient to diagnose content and will shorten the aPTT if an aPL antibody is pre-
the condition; i.e., no one assay is specific. It is possible that sent. The difference between the two clotting times should be
one type of assay is positive while others are negative. Testing at least 3 to 5 seconds, the actual value determined in studies
for, and confirmation of the LAC should include the use of test by the performing laboratory. In addition, the patient’s aPTT
systems that have different types, concentrations, and configu- must be abnormal before this test can be performed, in order
rations of phospholipids, to demonstrate the specificity and to demonstrate the effect of the added phospholipid. False-
neutralization or bypassing of the inhibitor. These tests can positive tests may occur in heparinized patients due to platelet
include the diluted Russell’s viper venom time (dRVVT), factor-4, a potent anti-heparin substance contained in platelets.
platelet-phospholipid neutralization/high phospholipid aPTT Patients with factor deficiencies of the intrinsic and common
procedure (PNP), and the hexagonal phospholipid aPTT pathways will show equal prolongations in both steps.
(Staclot-LA®, Diagnostica Stago, Parsippany, NJ). These are Methodologies using altered phospholipid configurations
the most sensitive assays currently in use. Other less sensitive (i.e., hexagonal phospholipids) have also been used as confir-
assays have included the kaolin clotting time, and diluted matory techniques. By changing the epitope (antigenic deter-
thromboplastin inhibition time (Tables 26-10 and 26-11). minant) to which the antibody may be directed, increases the
The dRVVT method utilizes a two-step test system. odds of detection (see Table 26-11). Altered configurations
Russell’s viper venom, an extract from the venom of Vipera display different antigenic determinants than the other lamel-
russellii, directly activates F X, so only the common pathway lar, or flat, molecular structures. The Staclot LA® method is
is focused upon. The first step or “screening” reagent contains similar to the PNP principle of a two-step aPTT, however, it
the diluted venom and a low concentration of buffered utilizes an LAC-sensitive aPTT reagent and incorporates
cephalin (phosphatidylethanolamine) as the cofactor for pro- pooled normal plasma as part of the assay system, simultane-
thrombinase production; this increases the sensitivity to ously eliminating the effects of a factor deficiency. A shorten-
inhibitors of the prothrombinase complex, and a prolonged ing of more than 8 seconds with the addition of the hexagonal
clotting time is produced. A mixing study with pooled normal phospholipid versus the buffer is suggestive of a LAC. This
plasma and this reagent will correct in the presence of a factor assay also contains anti-heparin agents.
deficiency of the common pathway, e.g., during warfarin ther- The continuing association between aPL antibodies and
apy. The next step, if the screening dRVVT is abnormal, con- thrombotic episodes has supported advances in the diagnosis
sists of a “confirmatory” dRVVT: the same reagent containing of this syndrome. The most recent guidelines for LAC testing
a higher concentration of the same phospholipid is used. This were developed by the International Society on Thrombosis
will neutralize or bypass the inhibitor, providing additional and Hemostasis (ISTH), Scientific and Standardization
631
Disorders of Plasma Clotting Factors
(LOM)
aouasaid ul uonoval [enpisar
LOM uovoys peseq-LLd? 0] dATIISUS
AVIANISUOS AIA LOM ou
Ayueoyiusts ‘uoTjOvAl UT posn
QSVOIOUI [[LM ‘prdipoydsoyd suMo[D
vurseyd 100d
TT sjojareyd pidijoydsoyd
aantsod asyey aantsod os[e aantsod as[ey Jo 90U9SeId vurseyd s juoned poppe ON urjory
-jajaye|d Jo os aattsod as[ey
uonovel poseq (LAAMP)
sasesop -Kemujed UOWwWoD _ SUIUDIIOS,,
Aressooou oq AvU
‘poyjour stu} Aq days Aroyeusyuod onnodeisy} ‘(A1OJULIYUOS) uOT}e.QUIDUOS our
juasaid jou vy] Z days 0}
Jodi,
sazt[e.noU MOT UWLOUSA,
soyeorpul Jowqryur Agualayap
‘pammnbar qudSeaI Ul ILL pue LLd® 08 ‘aamtsod jt (oururepouRyya
vurseyd X 10 A XIOA
‘TI 20308] YIM Apnjs SUIXTUI popnypour juase qm poreduros SuraysAs yuasRol -[Apneydsoyd) S [[essny
jewtiou pojood ‘TI Foyoey YIM DATIISUAS ISO ureyda) peywytd
aantsod ase} aanisod as[ey ‘aaniisod as[ey ulredoy-nuy 7 JO | Wed
YIM UONIALI0D
ANUITIIYIC, quRNSsvOOnUY uliedoy AVIAQISUS S9.1je3,J pidyjoydsoyg
JUIUTLOD
199,
SaoqIqryuy Jo dINjeN
ayweds 10338] 1810 1310
Chapter 26
a1njen
jo LYO
pidyjoydsoyd J $9.1NjBI uniedayy
410
10JIBY
dywoads
ureydag Hed JOC
C ISO DATISUAS ase faanisod =
JUIseoI
-[Apneydsoyd) poreduros
UM
S
AJIAISUAS
AQUAIDYIG
SLOVIQIyUyT
oney
PuedeAT
syUIUIWOD
SuraysAs
1S9], poinyid
Ul
Apnjs
juase
ystH,
Disorders of Plasma Clotting Factors
uOoUIWIODd
juejnseoonuy
dUdIIJ1NIUI
JO
Surxrur
[[essny
popnyjour
uone.uadU0S -Aemyyed
uleday-nuy
10}9e} AQUAIDYAap
quaseol
‘posmnba.
poseq
Jodi,ou]
soqeurUnyo
SUONRUTULIO}Op
uoTnoval
sozipennou
(ourWwe[ouRyjo
sasesop
-1yuo,),,
onnodeioy}
YIM
ueuny joyoqeyd OS/) DAISUIS
UI ase] danisod JVI IOJOVJ =
{19k.1)X9 DATIISUDS JSOU Sasevd JOJORJ
YPM FOUGTYULTA
as[ey
(LAAUP)
LLd® Gia Lid? oq
d[ICI
A, Auatoyop
Avul
dattsod
UIST ul
euiouge asyry aantsod
uOneZ
WOUdA, ,MATOyeUL -1[e.NON
WIa}sAs osuvl
DANISUIS-DYT jeuosexoy paloog [euiou poloog [euoU IIQISsog
asTRJ =
(dNd)
LLd® uoneinsyuoo vurseyd vuiseyd UM
oanisod
aInpsd01g
JUdSvII soAaoiduit popraoid
ul popraoid
ul 40}9eJINA
UL
juase
popraoid AVLATISUS Juaseal
Ly Juaseol
Ly Ur QTY STOW
[euosexoy
oV
alee
popnyour SOJCUTULT[S
uueday-nuy
-[Apneydsoud
SJUOSROI
yooqyo
JO
ouTueouryya
-pidrjoydsoyg
TI9[Pehs
sozipe.nou
sosesop
onnodeioy}
IOORJ
QUESTIONS
1. What material did the laboratory professional use when
performing this mixing study?
2. What treatment is indicated for this patient? Case Study 5
ANSWERS A 29-year-old woman was seen by her obstetrician for pre-
natal care. She had three previous pregnancies, two resulting
1. The mixing study in this case would have been per- in spontaneous first-trimester miscarriages and the third in
formed with pooled normal plasma or adsorbed plasma. fetal demise at 28 weeks’ gestation. Laboratory values were
Aged serum would not have corrected the aPTT. as follows:
2. This patient requires infusion of factor VIII concentrate.
Test Result Ref. Values
QUESTIONS
Piel 13.0 sec 10.0-14.0 sec
F VII 30% 50-150% 1. What complex is inhibited by LAC, causing a prolonga-
tion of the clotting time?
continued continued
Chapter 26 Disorders of Plasma Clotting Factors 635
ANSWERS
Case Study 8
1. The PT measures the extrinsic pathway, in which PK is A 60-year-old woman was admitted to the emergency
not involved. Therefore, only the aPTT will reflect an department with hemorrhage into the right arm and right
abnormal clotting time. breast. No previous history of bleeding or medication was
2. No treatment is indicated: there is no associated risk of indicated. Laboratory data upon admission were as follows:
bleeding. continued
636 Chapter 26 Disorders of Plasma Clotting Factors
Case Study 9
Case Study 10
A 37-year-old man was seen by his physician for a routine
physical examination. A full blood workup was drawn and A 66-year-old man with a diagnosis of amyloidosis was
sent to a reference laboratory for analysis. Results were as admitted to the hospital with severe gastrointestinal bleed-
follows: ing. Admission laboratory data included:
continued continued
Chapter 26 Disorders of Plasma Clotting Factors 637
Oi ceticans
L Which of the following can lead to impairment of the a The factor deficiency that produces a normal aPTT and
coagulation system? an abnormal PT is:
a. Decreased factor synthesis a. Afibrinogenemia
b. Interference by abnormal molecules b. Prekallikrein
c. Loss, consumption, or inactivation of factors c. Factor VII
d. All of the above d. Factor X
. Which of the following shows decreased activity of F
i) 6. A patient with amyloidosis can have a deficiency of
VII:C? which factor?
a. Hemophilia A a. Factor V
b. Hemophilia B b. Factor X
c. Parahemophilia c. Factor VIU:C
d. Hemophilia C d. Factor I
. Which coagulation disorder has decreased activity of F 7. The reptilase time will be normal in the presence of:
VUI:C, vWF:Ag, and vWFR:Co, and a prolonged bleed- a. Heparin
ing time test? b. Dysfibrinogenemia
a. Thrombocytopenia c. Afibrinogenemia
b. von Willebrand disease d. Hypofibrinogenemia
c. Hemophilia A 8. Which of the following is not considered a test for iden-
d. Acquired hemophilia tification of a lupus anticoagulant?
. The lysis of a clot in a 5 M urea solution is an indication a. Platelet neutralization procedure (PNP)
of which disorder? b. Anticardiolipin antibodies
a. Dysfibrinogenemia c. Diluted Russell’s viper venom time (dRVVT)
b. F XIII deficiency d. Hexagonal phospholipid aPTT
c. Antiphospholipid antibody syndrome
d. Factor V Leiden
638 Chapter 26 Disorders of Plasma Clotting Factors
9. All of the following statements are true regarding the 10. Activated protein C resistance:
detection of a lupus anticoagulant (LAC) except: a. Is the result of a single point mutation of the F V gene
a. Platelet-poor plasma samples are required. in 90% of cases;
b. The aPTT is frequently prolonged and is usually the b. Leads to increased thrombin generation as a result of
first indication of a LAC. impaired F V inactivation;
c. A mixture of patient plasma with pooled normal c. Is found in 20% to 60% of patients with venous
plasma will correct the prolonged aPTT. thrombosis;
d. The diluted Russell’s Viper Venom Time may be used d. All of the above
to confirm the presence of LA.
See answers at the back of this book.
Acknowledgment
The author gratefully acknowledges the contributions of
Judith Brody, MD.
Chapter 26 Disorders of Plasma Clotting Factors 639
REFERENCES . Chong, LL, et al: A case of “super” 34, Dahlback, B, et al: Familial throm-
warfarin poisoning. Scand J Haematol bophilia due to a previously unrecog-
— . Rock, G: Defects of plasma clotting fac-
36:314, 1986. nized mechanism characterized by poor
tors. In: Pittiglio, DH, and Sacher, RA
. Jensen R: Screening and molecular diag- anticoagulant response to activated pro-
(eds): Clinical Hematology and Funda-
nosis in hemostasis. Clin Hemost Rev tein C: Prediction of a cofactor to acti-
mentals of Hemostasis, ed 1. FA Davis,
13:12, 1999. vated protein C. Proc Natl Acad Sci
Philadelphia, 1987.
18. Poort, SR, et al: A common genetic vari- USA 90:1004, 1993.
i). Hantgan, RR, et al: Fibrinogen structure
ation in the 3~-untranslated region of the boNn . Morrissey, JH, et al: Quantitation of acti-
and physiology. In Colman, RW, et al
prothrombin gene is associated with ele- vated factor VII levels in plasma using
(eds): Hemostasis and Thrombosis:
vated plasma prothrombin levels and an tissue factor mutant selectively deficient
Basic Principles and Clinical Practice,
increase in venous thrombosis. Blood in promoting factor VII activation. Blood
ed 3. JB Lippincott, Philadelphia,
88:3698, 1996. 81:734, 1993.
1994,
. Rosendaal, FR, et al: A common pro- 36. Hall, CA, et al: A clinical and family
. Collen, D, et al: Metabolism and distrib-
thrombin variant (20210 G to A) study of hereditary proconvertin
ution of fibrinogen. I. Fibrinogen
increases the risk of myocardial infarc- (factor VII) deficiency. Am J Med
turnover in physiological conditions in
tion in young women. Blood 90:1747, 37:172, 1964.
humans. Br JHaematol 22:681, 1972.
1997. . Dische, FE, and Benfield, V: Congenital
. Bommer, W, et al: Kongenitale
. DeStefano, V, et al: Prothrombin factor VII deficiency. Haematological
Afibrinogename. Teil I. Ann Paedlatr
G20210A mutant genotype is a risk fac- and genetic aspects. Acta Haematol
200:46, 1963.
tor for cerebrovascular ischemic disease AEST, NOSE).
. Gralnick, HR, and Connaghan, W:
in young patients. Blood 91:3562, 1998. . Park, F , Ohashi, K, and Kay, MA:
Hereditary abnormalities of fibrinogen.
. DeStefano, V, et al: Prevalence of mild Therapeutic levels of human factor VIII
In Williams WJ, et al (eds): Hematology,
hyperhomocysteinaemia and association and IX using HIV- |- based lentiviral
ed 5. McGraw-Hill, New York,
with thrombophilic genotypes (factor V vectors in mouse liver. Blood 96
1994.
Leiden and prothrombin G20210A) in (3):1173-1176, 2000.
ON. Henschen, A, et al: Genetically abnormal
Italian patients with venous thromboem- 3 Rick, ME , Walsh, CE, and Key, NS :
fibrinogens—some current characteriza-
bolic disease. Br JHaematol 106:564, Congenital Bleeding Disorders. Hema-
tion strategies. In Haverkate F, et al
1999. tology : 559-574, 2003.
(eds): Fibrinogen Structure. Functional
i)to . DeStefano, V, et al: The risk of recurrent AQ. Bloom, AL: The biosynthesis of factor
Aspects, Metabolism. Walter de Gruyter,
deep venous thrombosis among het- VUI. Clin Haematol 8:53, 1979.
Berlin, 1983. 4
erozygous carriers of both factor V . Tuddenham, EGD, et al: Response to
—~—. McDonagh, J, et al: Dysfibrinogenemia
Leiden and the G20210A prothrombin infusion of proelectrolyte fractionated
and other disorders of fibrinogen struc-
mutation. N Engl J Med 341:801, 1999. human factor VIII concentrates in
ture and function. In Colman, RW, et al
. Tracy, PB, et al: Radioimmunoassay of human haemophilia A and von
(eds): Hemostasis and Thrombosis:
factor V in human plasma and platelets. Willebrand disease. Br JHaematol
Basic Principles and Clinical Practice,
Blood 60:59, 1982. SZ aw), Oey.
ed 3. JB Lippincott, Philadelphia, 1994.
24. Chiu, HC, et al: Biosynthesis of coagula- . Montgomery, RR, and Coller, BS: von
oo. Reptilase is a registered trademark of
tion factor V by megakaryocytes. J Clin Willebrand disease. In Colman, RW,
Pentapharm, Ltd. Invest 75:339, 1985. et al (eds): Hemostasis and Thrombo-
\O. Latallo, ZS, and Teisseyre, E: Evaluation
. Owren, CA: The coagulation of blood: sis: Basic Principles and Clinical Prac-
of Reptilase-R and thrombin clotting Investigation on a new clotting factor. tice. ed 3. JB Lippincott, Philadelphia,
time in the presence of fibrinogen degra- Acta Med Scand (Suppl) 194:1, 1947. 1994.
dation products and heparin. Scand J 26. Owren, CA, and Cooper, T: Parahe- 43, Wagner, EE, et al: Immunolocalization
Haematol 13(Suppl):261, 1971. mophilia. Arch Intern Med 95:194, 1955. of von Willebrand protein in Weibel-
10. Donati, MB, et al: Fibrinogen degrada- 27 Colman, RW: Factor V. Prog Hemost Palade Bodies of human endothelial
tion in vivo: Effect on the reptilase time Thromb 3:109, 1976. cells. J Cell Biol 95:355, 1982.
and on the thrombin time. Scand J 28. Jandl, JH: Blood: Textbook of Hematol- 44, Enbal, A, and Loscalzo, J: Glycocalicin
Haematol 13(Suppl):259, 1971. ogy. Little Brown, Boston, 1987. binding to vWf adsorbed onto
— . Francis, CW, and Marder, VJ: Physio-
Zo) Kaufmann, RJ, et al: Factor VII and collagen -coated or polystyrene surfaces.
logic regulation and pathologic disorders hemophilia A. In Colman, RW, et al Thromb Res 56:347, 1989.
of fibrinolysis. In Colman RW, et al (eds): Hemostasis and Thrombosis: 45. Howard, NA, and Firkin, BG:
(eds): Hemostasis and Thrombosis: Basic Principles and Clinical Practice, Ristocetin—a new tool in the investiga-
Basic Principles and Clinical Practice, ed 4. JB Lippincott, Philadelphia, 2000. tion of platelets. Thromb Diath
ed 3. JB Lippincott, Philadelphia, 1994. . Nesheim, ME, et al: Isolation and study Haemorrh 26:362, 1971.
. Brick, W: Dysfibrinogenemia. April 13, of acquired inhibitor of human coagula- 46.©) Brinkhous, KM, et al: Botrocetin
2006. Online. Available: http://www. tion factor V. J Clin Invest 77:405, 1986. (venom agglutinin): Reaction with a
emedicine.com/med/topic599.htm. . Grund, S: Factor V deficiency. May 4, broad spectrum of multimeric forms of
(May 26, 2006). 2004. Online. Available: http://www. factor VII macromolecular complex.
. Hirsh, J, et al: Approach to the throm- drugs.com/enc/factor_v_deficiency.html. Proc Natl Acad Sci USA 80:1463,
bophilic patient. In Colman, RW, et al (May 26, 2006). 1983.
(eds): Hemostasis and Thrombosis: WwWto. Peyvandi, F, and Mannucci, PM: Rare 47. Coller, BS, and Gralnick, HR: Studies
Basic Principles and Clinical Practice, coagulation disorders. Thromb Haemost on the mechanism of ristocetin-induced
ed 3. JB Lippincott, Philadelphia, 1994. SIAL, WSL) platelet agglutination: Effects of
. Davie, EW, et al: The coagulation cas- 318), Press, RD, and DeLoughery, TG: structural modification of ristocetin
cade: Initiation, maintenance and regula- Thrombotic risk assessment; molecular and vancomycin. J Clin Invest
tion. Biochemistry 30:10363, 1991. and conventional testing for a common, 60(2):302, 1977.
. Shapiro, SS, and McCord, IS: Prothrombin. treatable genetic disease. Am Assoc 48. National Committee for Clinical Labora-
In Spact, TH (ed): Progress in Clin Chem Clin Lab News 126:10, tory Standards. Assays of von Willebrand
Hemostasis and Thrombosis, vol. IV. 2000. factor antigen and ristocetin cofactor
Grune & Stratton, New York, 1978.
640 Chapter 26 Disorders of Plasma Clotting Factors
activity: Approved guideline H51-A. 65 . Lombardi, R, et al: Alterations of factor 78. Phillips, MD, and Santhouse, A: von
Wayne, Pa: NCCLS; 2002. VIII:vWf in clinical conditions associ- Willebrand disease: Recent advances in
49. Hoyer, LW, and Shainoff, JR: Factor ated with an increase in its plasma con- pathophysiology and treatment. Am J
VIII-related protein circulates in normal centration. Br JHaematol 49:61, 1981. Med Sci 1998;316:77-86.
human plasma as high molecular weight 66. Scholtes, MC, et al: The factor VIII ratio 7). Kazatchkine, MD, and Kaveri, SV:
multimers. Blood 55:1056, 1980. in normal and pathological pregnancies. Immunomodulation of autoimmune and
. Nilsson, IL, et al: The Duke and Ivy Eur J Obstet Gynecol Reprod Biol inflammatory diseases with intravenous
methods for determination of bleeding 16:89, 1983. immune globulin. N Engl J Med 2001
time. Thromb Diath Haemorrh 10:223, 67. Ruggeri, ZM, et al.: Multimeric compo- Sept 6; 345(10): 747-55.
1963. sition of Factor VII/vWf administration 80. Nitu-Whalley, IC, and Lee, CA:
SIE Miale, JB: Hemostasis and blood coagu- of DDAVP: Implications for pathophysi- Acquired von Willebrand syndrome—
lation: Hemophilia (factor VIII defi- ology and of vWd subtypes. Blood report of 10 cases and review of the lit-
ciency). In Miale, JB (ed): Laboratory SEND, SIs. erature. Haemophilia 1999; 5: 318-326.
Medicine: Hematology, ed 6. CV Mosby, 68. Geil, JD: von Willebrand Disease. May 8 —_. Friederich, PW, et al.: Successful treat-
St. Louis, 1982. 19, 2006. Online. Available: http://www. ment with recombinant factor VIla of
. McGlasson, DL: Defects of plasma clot- emedicine.com/PED/topic2419.htm. therapy- resistant severe bleeding in a
ting factors. In Harmening, DM (ed): (June 5, 2006). patient with acquired von Willebrand
Clinical Hematology and Fundamentals 69. Hillery, CA, et al. Type 2M von disease. Am J Hematol 66:292, 2001.
of Hemostasis. ed 2. FA Davis, Philadel- Willebrand disease: F606I and 1662F 82. @sterud, B, and Rapaport, SI: Activation
phia, 1992. mutations in the glycoprotein Ib binding of factor IX by the reaction product of
. Hoyer, LW, and Rick, ME: Implications ___domain selectively impair ristocetin- but tissue factor and factor VII: Additional
of immunological methods for measur- not botrocetin-mediated binding of von pathway for initiating blood coagulation.
ing antihemophilic factor (factor VIII). Willebrand factor to platelets. Blood Proc Natl Acad Sci USA 74:5260, 1977.
Ann NY Acad Sci 240:97, 1975. 1998;91:1572-81. . Hultin, M, and Nemerson, Y: Activation
54. Ratnoff, OD, and Bennett, B: The genet- 70. Endres-Brooks, J, and Golden, T: von of factor X by factors [Xa and VII. A
ics of hereditary disorders of blood coag- Willebrand disease: Monograph and specific assay for factor [Xa in the pres-
ulation. Science 179:1291, 1973. Case Report. The Blood Center of ence of thrombin-activated factor VII.
55: Miller, JL: Blood coagulation and fibri- Southeastern Wisconsin, November Blood 52:928, 1978.
nolysis. In Henry, JB (ed): Clinical Diag- 1994. 84. Pavlovsky, A: Contribution to the patho-
nosis and Management, ed 17. WB TAN. Peerlinck, K, et al: A patient with von genesis of hemophilia. Blood.
Saunders, Philadelphia, 1984. Willebrand’s disease characterized by a 1947;2:185-191.
56. Weiss, AE: Circulating inhibitors in compound heterozygosity for a substitu- 85. Biggs, R, and Macfarlane, RG: The reac-
hemophilia A and B: Epidemiology and tion of Arg 854 by Gln in the putative tion of haemophilic plasma to thrombo-
methods of detection. In Brinkhous, KM, factor VI-binding ___domain of von Wille- plastin. J Clin Pathol 4:445, 1951.
and Henker, HC (eds): Handbook of brand factor (VWF ) on one allele and 86. Aggeler, PM, et al: Plasma thromboplas-
Hemophilia. Excerpta Medica, Amster- very low levels of mRNA from the sec- tin component (PTC) deficiency: A new
dam, 1975. ond vWF allele. Br J Haematol 80:358, disease resembling hemophilia. Proc Soc
ihe Shapiro, SS: Hemorrhagic disorders 1992 Exp Biol Med 79:692, 1952.
associated with circulating inhibitors. In . Kroner, PA, et al: The defective interac- 87. Biggs, R, et al: Christmas disease: A
Ratnoff, OD, and Forbes, CD (eds): Dis- tion between von Willebrand factor and condition previously mistaken for
orders of Hemostasis. Grune & Stratton, factor VIII in a patient with type | von haemophilia. Br Med J 2:1378, 1952.
Orlando, 1984. Willebrand disease is caused by substitu- 88. Biggs, R: The inheritance of defects in
58. Larson, L: Disorders of secondary hemo- tion of Arg 19 and His 54 in mature von blood coagulation. In Biggs, R, and
stasis. In McKenzie, SB: Clinical Labo- Willebrand factor. Blood 87:1013, 1996. Rizza, CR (eds): Human Blood Coagu-
ratory Hematology. Pearson Education, WS: Shelton, IBB, et al: Gene deletions cor- lation, Haemostasis and Thrombosis,
Upper Saddle River, 2004. relate with the development of alloanti- ed 3. Blackwell Scientific, Oxford,
. Baxter Healthcare CorporationTM bodies in vWd. J Clin Invest 79:1459, 1984.
FEIBATM-VH Anti-Inhibitor Coagulant 1987. 89. Meyer, D, et al: Cross-reacting material
Complex. April 2005. Online. Available: 74. Pollack, ES: von Willebrand disease. in genetic variants of hemophilia B. J
www.hemophiliagalaxy.com/pdfs/ August 18, 2004. Online. Available: Clin Pathol 25:443, 1972.
therapies/Feiba.pdf. (May 30, 2006). http://www.emedicine.com/med/ 90. Orstavik, KH, et al: Detection of carriers
60. Kasper, CK, et al: A more uniform mea- topic2392.htm. (June 26, 2006). of hemophilia B. Br J Haematol 42:293,
surement of factor VIII inhibitors. US. Scott, JP, and Montgomery, RR: The L979;
Thromb Diath Haemorrh 34:869, 1975. rapid differentiation of type IIB von 9 — . Nilsson, IM, et al: Hemophilia prophy-
6 jek. Gralnick, HR: von Willebrand disease. Willebrand’s disease from platelet-type laxis in Sweden. Acta Paediatr Scand
In Williams WJ, et al (eds): Hematology, (pseudo-) von Willebrand’s disease by 65:129, 1976.
ed 5. McGraw-Hill, New York, 1994. the “neutral” monoclonal antibody bind- , Brinkhous, KM, et al: Prevalence of
62. Bennett, B, et al: Studies on the nature of ing assay. Am J Clin Pathol 96:723, inhibitors in hemophilias A and B.
antihemophilic factor (factor VII): Fur- 1991. Thromb Diath Haemorrh 51(Suppl):315,
ther evidence relating the AHF-like anti- 76. Nikiforova, MN, and Retzinger, GS: 1972.
gens in normal and hemophiliac plasma. Acquired von Willebrand disease. OB DiScipio, RG, et al: A comparison of
WElinvinvesto2 219 Tal 73% September/October 2003. Online. Avail- human prothrombin factor LX (Christ-
63. Coller, BS: Von Willebrand’s disease. In able: http://www.pathology.uc.edu/ mas factor), factor X (Stuart factor),
Ratnoff, OD, and Farbes, CD (eds): Dis- LABLINES/V9I5.pdf. (June 28, 2006). and protein S. Biochemistry 16:698,
orders of Hemostasis. Grune & Stratton, Jae Lazarchick, J, et al.: Acquired von OTe
Orlando, 1984. Willebrand syndrome due to an 94. DiScipio, RG, et al: Activation of human
64. Holmberg, L, and Nelson, IM: von inhibitor specific for von Willebrand factor X (Stuart factor) by a protease
Willebrand’s disease. Clin Hematol Coag factor antigens. Am J Hematol, from Russell’s viper venom. Biochem-
Disord 14:461, 1985. 1986;21:305-314. istry 16:5253, 1977.
Chapter 26 Disorders of Plasma Clotting Factors 641
05: Titani, K, et al: Bovine factor X, (Stuart Principles and Clinical Practice, ed 3. 124. Mandle, R, Jr, et al: Identification of
factor): Amino acid sequence of heavy JB Lippincott, Philadelphia, 1994. prekallikrein and high-molecular-
chain. Proc Natl Acad Sci USA 72:3082, 109. Roberts, HR, and Hofman, M: Hemo- weight kininogen as a complex in
Sia philia and related conditions—inherited human plasma. Proc Natl Acad Sci
96. Ichinose, A, and Davie, EW: The blood deficiencies of prothrombin (factor II), USA 73:4179, 1976.
coagulation factors: Their cDNAs, factor V, and factors VII to XII. In . Hattersley, PG, and Hayse, D: Fletcher
genes, and expression. In Colman, RW, Beutler E, et al (eds): Williams Hema- factor deficiency: A report of three
et al (eds): Hemostasis and Thrombosis: tology, ed 5. McGraw-Hill, New York, unrelated cases. Br JHaematol 18:411,
Basic Principles and Clinical Practice, 199m. 1970.
ed 3. JB Lippincott, Philadelphia, 110. Colman, RW: Surface mediated defense 126. Sollo, DG, and Saleem, A: Prekallikrein
1994. reactions: The plasma contact activation (Fletcher factor) deficiency. Ann Clin
DM Giangrande, PLF: Historical review: six system. J Clin Invest 73:1249, 1984. ab Scilla 279985:
characters in search of an author: the itil. Meiers, JCM, et al: High levels of 27. Goodnough, LT, et al: Thrombosis or
history of the nomenclature of coagula- coagulation factor XI as a risk factor myocardial infarction in congenital
tion factors. Br J. Haematol 121:703, for venous thrombosis. N Engl J Med clotting factor abnormalities and
2003. 342:696, 2000. chronic thrombocytopenia: A report of
98. Griep, PR, et al: Factor X deficiency in iN, Naito, K, and Fujikawa, K: Activation 21 patients and a review of 50 previ-
amyloidosis: A critical review. Am J of human blood coagulation factor XI ously reported cases. Medicine 62:248,
Hematol 11:443, 1981. independent of factor XI by thrombin in 1983.
Oo; Roberts, HR, and Leflowitz, JB: plasma results in additional formation 128. Saito, H, et al: Defective activation of
Inherited disorders of prothrombin con- of thrombin that protects fibrin clots clotting, fibrinolytic, and permeabil-
version. In Colman, RW, et al (eds): from fibrinolysis. Blood 86:3035, 1995. ity-enhancing systems in human
Hemostasis and Thrombosis: Basic iL}. von dem Borne PAK, et al: Feedback Fletcher trait plasma. Circ Res 34:641,
Principles and Clinical Practice, ed 3. JB activation of factor XI by thrombin in 1974.
Lippincott, Philadelphia, 1994. plasma results in additional formation 1293 Schiffman, S, and Lee, P: Preparation,
100. Thompson, RE, et al: Association of of thrombin that protects fibrin clots characterization, and activation of a
factor XI and high-molecular-weight from fibrinolysis. Blood 86:3035, 1995. highly purified factor XI: Evidence that
kininogen in human plasma. J Clin 114. von dem Borne PAK, et al: Thrombin- a hitherto unrecognized plasma activity
Invest 60:1376, 1977. mediated activation of factor XI results participates in the interaction of factor
10 — . Kurachi, K, and Davie, EW: Activation in a thrombin-activatable fibrinolysis XI and XII. Br J Haematol 27:101,
of human factor XI (plasma thrombo- inhibitor-dependent inhibition of fibri- 1974.
plastin antecedent) by XIla (activated nolysis. J Clin Invest 99:2323, 1997. . Colman, RW, et al: Regulation of the
Hageman factor). Biochemistry IIS. Fujikawa, K, and Davie, EW: Human coagulant activity and surface binding
IE srsisul, DWT. factor XII (Hageman factor). Methods of high molecular weight kininogen.
102. Bouma, BN, and Griffin, JH: Human Enzymol 80:198, 1981. Clin Res 32:551a, 1984.
blood coagulation factor XI: Purifica- 116. Cochrane, CG, et al: Activation of . Berrettini, M, et al: Assembly and
tion, properties, and mechanism of acti- Hageman factor in solid and fluid expression of an intrinsic factor IX acti-
vation by activated factor XI. J Biol phases: A critical role of kallikrein. vator complex on the surface of cul-
Chem 252:6432, 1977. J Exp Med 138:1564, 1973. tured human endothelial cells. J Biol
103. Fair, BD, et al: Detection by fluores- 117. Loscalzo, J: Pathogenesis of thrombo- Chem 267:19833, 1992.
cence of structural changes accompany- sis. In Beutler E, et al. (eds): Williams IS. Cheung, PP, et al: Total kininogen defi-
ing the activation of Hageman factor Hematology, ed 5. McGraw-Hill, ciency (Williams trait) is due to an Arg
(factor XII) (39773). Proc Soc Exp Biol New York, 1995. stop mutation in exon 5 of the human
Med 155:199, 1977. 118. Lorand, L, and Kornishi, K: Activation kininogen gene. Blood 78:391a, 1991.
104. Fujikawa, K, et al: Amino acid of fibrin stabilizing factor of plasma by 3. Gailani, D, and Broze, GJ: Factor XI
sequence of human factor XI, a blood thrombin. Arch Biochem Biophys activation in a revised model of blood
coagulation factor with four tandem 105:58, 1964. coagulation. Science 253(5022):909,
repeats that are highly homologous with iLiUy, Naski, MC, et al: Characterization of 1991.
plasma prekallikrein. Biochemistry the kinetic pathway for fibrin promotion 134. Naito, K, and Fujikawa, K: Activation
25:2417, 1986. of alpha-thrombin-catalyzed activation of human blood coagulation factor XI
105. Baglia, PA, et al: Functional domains in of plasma factor XIII. Biochemistry independent of factor XII. Factor XI is
the heavy chain region of factor XI: A 30:934, 1991. activated by thrombin and factor Xa in
high-molecular-weight kininogen- 120. Folk, JE, and Finlayson, JS: The the presence of negatively charged sur-
binding site and substrate binding site epsilon gamma-glutamyl lysine faces. J Biol Chem, 1991;
for factor IX. Blood 74:244, 1989. crosslink and the catalytic role of trans- 266(12):7353-8.
106. Roberts, HR, and Hoffman, M: Hemo- glutaminases. Adv Protein Chem 31:3, SI, Lusher, JM: Acquired inhibitors to fac-
philia and related conditions—inherited 1977. tor VIII in non-hemophilic patients. In
deficiencies of prothrombin (factor II). il. McDonagh, J: Structure and function of Kessler, C, et al. (eds): Acquired Hemo-
factor V, and factors VI to XII. In Factor XII. In Colman, RW, et al: philia, ed 2. Excerpta Medica, Prince-
Beutler, E, et al (eds): Williams Hemostasis and Thrombosis: Basic ton, NJ, 1995.
Hematology, ed 5. McGraw-Hill, New Principles and Clinical Practice, ed 3. 136. Feinstein, DI: Immune coagulation
Yorkesl99a: JB Lippincott, Philadelphia, 1994. disorders. In Colman, RW, et al (eds):
107. Seligsohn, U: High gene frequency of 2. Miletich, JP: Factor XII: Laboratory Hemostasis and Thrombosis: Basic
factor XI (PTA) deficiency in analysis. In Beutler E, et al (eds): Principles and Clinical Practice, ed 3.
Ashkenazi Jews. Blood 51:1223, 1978. Williams Hematology, ed 5. JB Lippincott, Philadelphia, 1994.
108. DeLa Cadena, RA, et al: Contact acti- McGraw-Hill, New York, 1995. . Hedner, U, and Glazer, S: Management
vation pathway: Inflammation and 3. Bagdasarian, A, et al: Immunochemical of hemophilia patients with inhibitors.
coagulation. InColman, RW, et al (eds): studies of plasma kallikrein. J Clin Hematol Oncol Clin North Am 6:1035,
Hemostasis and Thrombosis: Basic Invest 54:1444, 1974. 1992.
642 Chapter 26 Disorders of Plasma Clotting Factors
138 . Reece, EA, et al: Spontaneous factor XI 145. Bick, RL. Antiphospholipid thrombosis 2. Triplett, DA: Coagulation abnormali-
Inhibitors: Seven additional cases and syndromes. Clin App! Thromb Hemost ties. In McClatchey, KD (ed): Clinical
review of the literature. Arch Intern 7(4):241, 2001. Laboratory Medicine, ed 2. Lippincott
Med 144:525, 1984. 146. Harris, EN, et al: Anticardiolipin anti- Williams & Wilkins, Philadelphia,2002,
139. Poon, MC, et al: A unique precipitating bodies: Detection by radioimmunoassay pp. 1033-1049.
autoantibody against plasma thrombo- and association with thrombosis in sys- 3. Harris, EN, et al: Diagnosis of the
plastin antecedent associated with temic lupus erythematosus. Lancet antiphospholipid syndrome: A proposal
multiple apparent clotting factor defi- ZOE OSS: for use of laboratory tests. Lupus 7
ciencies in patients with systemic lupus 147. Alving, BM: The antiphospholipid (Suppl 2):S144-S148, 1998.
erythematosus. Blood 63:1309, 1984. syndrome: clinical presentation, diag- 154. Adcock, DM, et al: Coagulation Hand-
140. McGlasson, DL, et al: Platelet neutral- nosis and patient management. In book. Austin Texas: Esoterix Coagula-
ization procedure (PNP): Atypical Kitchens, CS, et al, eds. Consultative tion; 2002.
results seen with a factor V deficiency Hemostasis and Thrombosis. I55: Brandt, JT, et al: Laboratory identifica-
with and without the presence of an Philadelphia, Pa: W.B. Saunders; tion of lupus anticoagulants: Results of
inhibitor. Clin Lab Sci 3:119, 1990. 2002:181-196. the second international workshop for
141. Triplett, DA, and Brandt, JT: Lupus 148. Harris, EN, et al: Thrombosis, recurrent identification of lupus anticoagulants.
anticoagulants: Misnomer, paradox, fetal loss and thrombocytopenia: Pre- Thromb Haemostas 74:1597, 1995.
riddle. Epiphenomenon. Hematol Pathol dictive value of the anticardiolipin anti- 156. Lazarchick, J, and Kizer, J: The labora-
2:121, 1988. body test. Arch Intern Med 146:2153, tory diagnosis of lupus anticoagulants.
142. Conley, CL, and Hartmann, RC: A 1986. Arch Pathol Lab Med 113:177, 1989.
hemorrhagic disorder caused by circu- 149. Loeliger, A: Prothrombin as a co-factor Sy7/. Lug, R: Cost-effective and rapid solu-
lating anticoagulant in patients with dis- of the circulating anticoagulant in sys- tion to the diagnosis of lupus anticoagu-
seminated lupus erythematosus. J Clin temic lupus erythematosus. Thromb lants. Clin Hemost Rev 10:3, 1996.
Invest 31:621, 1952. Diath Haemorrh 3:237, 1959.
143. Shapiro, SS, and Thiagarajan, P: Lupus 150. Bajaj, SP, et al.: A mechanism for the
anticoagulants. In Spaet T (ed): hypoprothrombinemia of the acquired
Progress in Hemostasis and Thrombo- hypoprothrombinemia-lupus anticoagu-
sis, vol 6. Grune & Stratton, New York, lant syndrome. Blood. 61:684-692, 1983.
1982. Nn . Bonnin, JA, et al: Coagulation defects
144. Jensen, R: Antiphospholipid antibody in a case of systemic lupus erythemato-
syndrome diagnosis. Clin Hemost Rev sus with thrombocytopenia.
10:3; 1996. Br JHaematol 2:168, 1956.
Chapter |
Interaction
of the Fibrinolytic,
Coagulation, and Kinin
Systems; Disseminated
Intravascular
Coagulation; and
Kelated Pathology
John Lazarchick, MD
Melanie Oswald, MHS, MT(ASCP)SH
Fibrinopeptides
6. Thrombin-activatable fibrinolysis inhibitor that cleaves A and B
plasminogen/plasmin binding sites on fibrin
7. Fibrin/ fibrinogen, which serve as substrate for the active Figure 27-1 m Summary of the interaction of the coagulation, fibri-
nolytic, and kinin systems. High-molecular-weight kininogen (HMWK)
enzyme plasmin (Table 27-1).
and prekallekrein catalyze the activation of factor XII to factor Xlla.
Factor Xlla then promotes the conversion of prekallekrein to kallekrein.
The latter liberates the kinins from HMWK, thus completing the posi-
Plasminogen tive feedback loops of the contact phase of coagulation. These compo-
nents stimulate endothelial cells to release plasminogen activator.
Native plasminogen is a single-chain plasma zymogen of Thrombin generated through the extrinsic and intrinsic coagulation
approximately 90 kD that circulates in two molecular forms, cascades then converts fibrinogen to fibrin, induces platelet activation,
differing only in their carbohydrate content.’ It is synthesized activates protein C, and stimulates both tissue plasminogen activator
by the liver and has a half-life of 2 days. The plasma content (tPA) and urokinase-like plasminogen activator (uPA) from the en-
dothelium, which then can convert plasminogen to plasmin. Thrombin
is approximately 20 mg/dL. Each form of this molecule has
exerts a negative feedback by simultaneously stimulating plasminogen
an amino acid terminal glutamic acid (Glu-plasminogen) and activator inhibitor 1 (PAI-1) release from the endothelial cells. PAI-1 will
is capable of undergoing limited proteolytic cleavage of this serve to bind tPA and uPA to dampen plasmin generation. Plasmin,
region to an incomplete molecule with lysine as the new ter- once formed, will initiate clot lysis by proteolytically cleaving fibrin and
minal amino acid (Lys-plasminogen). This latter form is more fibrinogen into fibrin/fibrinogen degradation products (FDPs). In addi-
tion, plasmin will inactivate a number of coagulation factors including
readily converted to active plasmin by plasminogen activators factors V and VIII and convert activated factor XII into Xlla fragments.
than the Glu-plasminogen form and is probably of greater Excess plasmin in circulation is prevented by the presence of
physiologic significance. a-antiplasmin which forms a complex with plasmin, thus inactivating it.
Chapter 27 Interaction of the Fibrinolytic, Coagulation, and Kinin Systems; DIC; and Related Pathology 645
by the kidney in addition to the vascular endothelium, as pre- The earliest recognized component is fragment X,
viously mentioned, and is excreted in the urine.'* It can also be which is still capable of clotting. A recent finding has been
identified in vitro using kidney cell cultures and is a potent the identification of a small peptide fragment from the B-B
direct activator of plasminogen. Its major drawbacks are its chain of fibrinogen, which is released simultaneously with
expense and its relatively lower affinity for fibrin as compared the formation of the X fragment. Measurement by radioim-
with t-PA. A consequence of the latter property 1s that the plas- munoassay of the B-B 15-42 related peptide may prove of
min generated will not only digest fibrin but also circulating value in the documentation of early fibrinolytic states.*!*
fibrinogen and, therefore, the development of severe hypofib- The X fragment undergoes further plasmin attack to yield
rinogenemia is not uncommon with its use. The other exoge- unclottable Y and D fragments. The Y fragment is further
nous activator, streptokinase, is a product of beta (8)-hemolytic digested to yield an additional D fragment and a single
streptococci. It is not a serine protease and has no intrinsic pro- E fragment. It is now realized that the proteolytic cleavage
teolytic activity but is capable of forming a 1:1 stoichiometric of cross-linked fibrin (i.e., fibrin transaminated through the
complex with plasminogen. This interaction results in a confor- action of factor XIa and calcium) results in other interme-
mational change of the plasminogen molecule and exposure of diate degradation products (e.g., D2E without the genera-
its active serine site.'° The streptokinase—plasminogen complex tion of fragment D or E). This proteolytic product is
can then undergo autocatalysis to yield other activators namely, referred to as the D-dimer.
streptokinase-Glu-plasmin and streptokinase-Lys-plasmin. Any These breakdown products have specific inhibitory
of these forms will readily convert free plasminogen to plas- effects on the coagulation system and thereby suppress fur-
min. Because streptokinase is a bacterial protein, a major limi- ther clot formation. Fragment X is capable of clotting slowly
tation with its use in thrombolytic therapy is the induction of an and exerts an anticoagulant effect by competing with fib-
immune response, with resulting antibody development and an rinogen for thrombin. It also forms slowly polymerizing
inhibition of its activity. complexes with fibrin monomer and inhibits the polymer-
ization step. Fragment D forms abnormal complexes with
fibrin monomers as it polymerizes. Fragment E is not known
Plasminogen Activator Inhibitor-1 to have any specific anticoagulant effect. In high concentra-
tions (more than 100 pg/mL), the degradation products are
Plasminogen activator inhibitor-| is a member of the
capable of inhibiting platelet aggregation and release. Plas-
family of protease inhibitors that includes antithrombin-HI,
min also exerts a direct limiting effect on the coagulation
a,-macroglobulin, a,-antitrypsin, and a,-antiplasmin, and
process by being able to proteolytically cleave and render
that are collectively referred to as serpins (serine protease
inactive factors V and VII, in addition to proteolysing fac-
inhibitors). PAI-1 is a53-kD glycoprotein also synthesized by
tor XII and platelet glycoprotein Ib, the von Willebrand’s
vascular endothelium and released primarily in an inactive,
factor receptor.
latent state.'® It is an acute-phase reactant and can be induced
by a variety of stimuli, including interleukin-1 (IL-1), endo-
toxin, and thrombin. PAI-1 is the primary inhibitor of both Plasmin Inhibitors
tPA and uPA. Thus, regulation of fibrinolysis is dependent on
Although plasmin formation characteristically takes place
the interaction of t-PA and uPA with PAI-1. Under basal con-
in the area of fibrin deposition with little free plasmin cir-
ditions, most of the t-PA released is bound to PAI-1.
culating, this enzyme if unchecked by the presence of spe-
Excess levels of this inhibitor have been associated with
cific inhibitors, would result in circulating fibrinogen being
thrombotic disease. A common diallelic polymorphism has
digested and the blood being rendered unclottable. The pri-
been described for PAI-1 in which the prevalence of the 4G
mary physiologic inhibitor of plasmin in vivo is a,-plasmin
allele is significantly higher in patients with myocardial
inhibitor.* It rapidly binds to the lysine binding site on
infarction who are younger than 45 years old.'’? However,
plasmin in a 1:1 molar ratio in an irreversible manner. Mea-
other studies have failed to find excess PAI-1 levels to be
surement of these plasmin—a,-antiplasmin inhibitor com-
associated with an increased risk of thrombosis.'®
plexes has been suggested as an indicator for activation of
the fibrinolytic system. Plasmin adsorbed onto fibrin during
Plasmin the fibrinolytic process appears to be protected from this
inhibitor because it binds to fibrin through the same lysine
The pivotal serine protease generated through these complex binding site. Because this binding site on plasmin is
biochemical processes is plasmin. This protein has a molecu- occupied, the inhibitor cannot bind, and clot lysis can pro-
lar weight of 77 to 85 kD, depending on whether Lys-plasmin ceed. The overall effect is to ensure that plasmin activity is
or Glu-plasmin is formed, and has a transient plasma half-life limited to the area of fibrin deposition and to prevent free
measured in seconds.'? Plasmin has the ability to proteolyti- plasmin from circulating. Other protease inhibitors in plasma
cally degrade both fibrin in clots and native fibrinogen in the include a,-macroglobulin, C1 inactivator, and q,antitrypsin.
circulation into a series of well-characterized end products Of these, only a,-macroglobulin has a role in plasmin inhi-
collectively known as fibrin/fibrinogen degradation products bition during normal hemostasis, but it participates only
(FDPs). This process results in an asymmetric, progressive when q,-antiplasmin inhibitor binding sites for plasmin are
breakdown of fibrin and fibrinogen.”° saturated.
Chapter 27 Interaction of the Fibrinolytic, Coagulation, and Kinin Systems; DIC; and Related Pathology 647
Thrombomodulin elevated TAFI levels are felt to increase the risk for recurrent
venous thromboembolism.” Acquired abnormalities of the
Thrombomodulin is an endothelial cell membrane glycopro- fibrinolytic system are much more common and are discussed
tein that is a receptor for thrombin and forms a 1:1 stoichio- in the next section on disseminated intravascular coagulation
metric complex with thrombin.** Thrombin, once bound to and related disorders.
thrombomodulin, no longer has proteolytic activity to convert In summary, an integrated system of serine proteases is
fibrinogen to fibrin but retains esterolytic function. The com- brought into play once the coagulation process is initiated in
plex serves to convert protein C to activated protein C after the response to disruption of blood vessel integrity (Fig. 27-2).
zymogen binds to endothelial cell protein C receptor (EPCR). The response is balanced so that the same reaction that initi-
The rate of this reaction is accelerated by the presence of free ated thrombin formation and fibrin deposition also initiates a
protein S complexed to the protein C. Activated protein C series of reactions to lyse the clot. Factor XIla, with other
(aPC) then dampens the coagulation process by proteolytically components of the contact phase of coagulation, converts plas-
cleaving factor Va and VIIa. The thrombin—-thombomodulin minogen to plasmin; thrombin stimulates endothelial cells to
complex also serves to convert procarboxypeptidase B to its release t-PA with subsequent plasmin generation. Excess t-PA
activated form, thrombin-activatable fibrinolysis inhibitor activity is controlled by the presence of plasminogen activator
(TAFI). The number of thrombomodulin sites on the vascular inhibitor. Because of the high affinity of plasmin for fibrin,
endothelium is down-regulated by proinflammatory cytokines most of these fibrinolytic processes are taking place at the site
including IL-6 and tissue necrosis factor (TNF) and by other of fibrin deposition within the damaged blood vessel. The
inflammatory modulators released from activated neutrophils presence of plasmin inhibitors further ensures that the prote-
during the inflammatory process. olytic process is limited to this area.
Congenital Abnormalities
Congenital abnormalities of the fibrinolytic system causing
either a hemorrhagic or thrombotic diathesis are rare.** Only
three cases of an abnormal plasminogen have been reported in Lysyl residue —_Lysyl residue
which the patients had a history of recurrent thrombotic GB) TAFI Plasmin
episodes. In essentially all other cases reported, patients have ><
I
been asymptomatic even though a hypercoagulable state v
would have been expected.”’ Low levels of t-PA activity have FDPs FDPs
been documented in two families and were associated with a D-dimers D-dimers
similar thrombotic tendency. Whether deficiencies of tPA or Figure 27-2 @ Control of fibrinolysis. Plasminogen activators tissue
uPA cause a hypercoagulable state is controversial. A recently plasminogen activator (t-PA) and, to a lesser degree, urokinase (u-PA)
described platelet disorder, Quebec platelet disorder, leading are synthesized in the endothelium. Both t-PA and u-PA are rapidly
to a bleeding diathesis associated with moderate to severe inhibited by activated plasminogen activator inhibitor-1 (PAI-1), also
synthesized in the endothelium. The interaction of t-PA with PAI-|
thrombocytopenia, has been found to be due to excess uPA
regulates fibrinolysis, and under basal conditions most t-PA released
content in the a granules of platelets.** As mentioned previ- is bound to PAI-1. Plasminogen-tPA binds to fibrin in clots generat-
ously, allelic polymorphism has now been documented for ing plasmin at the LYSYL (lysine) residue site and clot lysis occurs
PAI-1, with the presence of the 4G allele being associated resulting in the formation of fibrin degradation products (FDPs)
with premature coronary artery disease.'’ Deficiencies of including D-dimers. The plasmin inhibitor «,-antiplasmin binds to
free circulating plasmin at the same lysine binding site, thus limiting
a,-antiplasmin inhibitor have been reported in four families plasmin activity to the area of fibrin deposition. Excess levels of
to date and, in contrast, are associated with a severe hemor- thrombin activate the plasma protein thrombin activatable fibrinoly-
rhagic tendency. Deficiencies of TAFI have not been reported sis inhibitor (TAFI), which binds with fibrin at the plasminogen bind-
to be associated with a hemorrhagic diathesis, however, ing site, thus preventing plasmin formation and fibrinolysis.
648 Chapter 27 Interaction of the Fibrinolytic, Coagulation, and Kinin Systems; DIC; and Related Pathology
Subendothelium
Endothelium
Protein C
MMalceyonleyial
ae 5 Extrinsic Plasminogen
Activated 6) patnway
platelets
ile
Plasmin
Platelet © Thrombin (small amounts)
Platelet
Thrombin
(large amounts)
Fibrin degradation products
D-dimer
Activated
platelets
@
Intrinsic pathway
Figure 27-5 @ Integrated system of hemostasis. 7. Disruption of endothelial continuity releases tissue factor (TF). 2. In the presence of ion-
ized calcium, TF forms a complex with factor VII or Vila leading to the conversion of factor X to Xa and rapid generation of small amounts of
thrombin and fibrin formation via the extrinsic pathway. In addition, the TF-factor Vila-Xa complex is rapidly inhibited by the presence of
tissue factor pathway inhibitor (TFPI). 3. The Vila-TF complex also activates factor IX to IXa, initiating the intrinsic pathway and the formation
of larger amounts of thrombin on the surface of activated platelets. 4. Enhanced expression of TF by monocytes also occurs in response to
endotoxin or cytokines secondary to sepsis. 5. Thrombin stimulates platelet activation and the release of platelet agonists. 6. Activated
platelets undergo shape change, resulting in the exposure of phospholipids and cofactors V and VIII on the surface of the platelet membrane.
7. Secondarily, and simultaneously, thrombin complexes with thrombomodulin (TM) on the endothelial surface. 8. Protein C, once bound to
this complex, is rapidly converted to its activated state. 9. Activated protein C indirectly causes release of tissue plasminogen activator in cells
from the subendothelium, in addition to direct stimulation and release of this glycoprotein by thrombin. 70. Plasmin-induced proteolysis of
the fibrin clot results in the formation of fibrin degradation products.
Although activation of the intrinsic system can result in may be more complicated, with endotoxin inducing release of
DIC, this is an uncommon mechanism. The exact sequence of a number of cytokines, including tumor necrosis factor-alpha
intermediary events by which certain stimuli initiate coagula- (TNF-a) and IL-1, and the primary activation of coagulation
tion is well understood, but with other stimuli this process occurring through the extrinsic (tissue factor-dependent) path-
remains uncertain. All pathologic stimuli that result in activa- way rather than the intrinsic pathway.*° TNF-a is capable of
tion of the intrinsic system probably do so indirectly by means inducing tissue factor activity in monocytes and on both the
of first inducing endothelial cell damage with subsequent expo- luminal and subendothelial surfaces of endothelial cells. Tissue
sure of the subendothelium. Platelet adherence and aggregation factor can bind and activate factor VII; the complex of TF-VIla
and factor XII activation can then occur. This is the proposed can then activate factor X, with subsequent thrombin forma-
mechanism of DIC associated with anoxia and immune com- tion. Binding of granulocytes to the endothelial receptors
plex formation, however, activation through the extrinsic sys- (selectins) induces the release of granulocyte cathepsins and
tem may also play a role. New insight regarding sepsis now elastases, which then lead to organ damage. The participation
suggests that the mechanism of DIC induced by endotoxins, the of the intrinsic system of coagulation in the DIC process then
lipopolysaccharide constituents of gram-negative organisms, involves activation of the kinin system through the interaction
650 Chapter 27 Interaction of the Fibrinolytic, Coagulation, and Kinin Systems; DIC; and Related Pathology
Clinical Presentation
a shearing effect as they traverse this area, with resultant Specific tests for direct evidence of thrombin activity
fragmentation and the development of a microangiopathic relate to the action of thrombin on fibrinogen. Other than cer-
hemolytic anemia (Fig. 27—5) or the lumen may be completely tain snake venoms, thrombin is the only enzyme that releases
occluded by the thrombus negating the formation of fragmented the specific peptides fibrinopeptide A (FPA) and B from the
RBCs in the circulation (Fig. 27-6). fibrinogen molecule. Both ELISA and radioimmunoassays
Conversion of prothrombin to thrombin is the result of are available commercially to measure each fibrinopeptide.**
cleavage from the parent molecule of a small, inactive peptide The major drawback to measuring these peptides, paradoxi-
referred to as prothrombin fragment F1.2. Measurement of cally, is the extreme sensitivity of the assay such that elevated
this activation peptide has thus far been utilized primarily for
diagnosis of a hypercoagulable state; however, it may have
equal utility as a molecular marker for DIC.
Figure 27-6 ® DIC (skin biopsy). Note both partial (small arrow)
Figure 27-5 m DIC (peripheral blood). Note presence of schisto- and complete (/arge arrow) occlusion of blood vessels by RBC/
cytes (arrows) and nucleated red cell (top border). fibrin clot.
652 Chapter 27 Interaction of the Fibrinolytic, Coagulation, and Kinin Systems; DIC; and Related Pathology
le
27-5
tab Profile of DIC
Conditions Suggested Clinical
Associated Triggering Clinical Laboratory Sequential
Synonyms with DIC Mechanisms Manifestations Findings Therapy
*Sequential therapy is used only after clotting is stopped (3% of patients may require this therapy).
PT 5 prothrombin time; PTT 5 partial thromboplastin time; AT-HI 5 antithrombin HI.
Related Disorders urologic procedures. The fibrinolytic state seen with metasta-
tic prostatic carcinoma is another example of this mechanism.
Primary fibrinolysis is an unusual situation in which plasmin The basis for the hemorrhagic state seen after cardiopul-
is formed in the absence of activation of the coagulation cas- monary bypass surgery is complex, with platelet dysfunction
cade. The clinical presentation in this disorder is similar to and hemodilution of coagulant proteins as the primary
that in DIC, with diffuse hemorrhage occurring as a result of defects; activation of the plasminogen—plasmin system with
increased plasma fibrinolytic activity. Several mechanisms increased fibrinolytic activity is also well documented.
can initiate this process. The presence of proteolytic enzymes The failure of the hepatic clearance mechanism to remove
in plasma that are capable of either directly or indirectly con- plasminogen activator accounts for the increased fibrinolytic
verting plasminogen into plasmin can occur in certain disease activity seen in a variety of hepatic disorders, particularly cir-
states. The genitourinary system is enriched in urokinases, rhosis. Under normal circumstances the hepatic reticuloen-
which can enter the systemic circulation following various dothelial system removes not only activated clotting proteins,
Chapter 27 Interaction of the Fibrinolytic, Coagulation, and Kinin Systems; DIC; and Related Pathology 655
but also plasminogen activator from the systemic circulation. Thrombotic thrombocytopenic purpura is the syndrome
When this function is impaired because of hepatic disease or in in which fibrin and platelet thrombi are formed diffusely
patients who have portocaval shunting procedures, the removal throughout the microvasculature, in contrast to the localized
of plasminogen activator is less than adequate and hyperplas- thrombus formation seen in DIC.*° The clinical picture con-
minemia occurs, with resultant hemorrhage. Increased fibri- sists of a pentad of findings:
nolytic activity can also be seen in patients undergoing liver
lhever
transplantation, especially during the reperfusion phase follow-
2. Microangiopathic hemolytic anemia
ing reanastomosis of vessels.
3. Thrombocytopenia
The occurrence of DIC with secondary fibrinolysis is well
4. Azotemia
documented in patients with acute promyelocytic leukemia,
5. Vacillating neurologic deficits.
although as previously discussed, this disorder clearly causes
primary fibrinolysis through surface membrane annexin II bind- Despite fibrin and platelet deposition, this disorder is not typ-
ing of t-PA and plasminogen. It is now recognized, however, ically associated with excessive activation of the coagulation
that the coagulopathy these patients develop may also result system. The pathologic mechanism in this disorder is
from a primary fibrinolysis. The mechanism(s) is unsettled but either a hereditary/familial deficiency of von Willebrand
may involve direct activation by the leukemic cells with release cleaving enzyme (ADAMTS-13) in endothelial cells or the
of a urokinase-type or tissue-type plasminogen activator. development of an acquired IgG inhibitory antibody to the
The coagulation abnormalities seen in these fibrinolytic enzyme that results in the secretion and circulation of ultra-
disorders are similar to those in DIC, with prolonged PT, large high-molecular-weight von Willebrand factor multimers
APTT, and thrombin times. These defects result from the that spontaneously bind to circulating platelets and form
hypofibrinogenemic state induced by the proteolytic cleavage occlusive platelet thrombi in the vasculature.’ An abnormal-
of fibrinogen by excess plasmin, in addition to the catabolic ity of the fibrinolytic system can also be present in some
effect of this enzyme on factors V and VII. FDP concentra- patients with diminished or absent fibrinolytic activity, partic-
tions are increased and, as previously noted, will further ularly of t-PA, in plasma and blood vessels affected with
interfere with coagulation by acting as antithrombins and microthrombi. It is likely these are secondary changes. Ther-
inhibitors of fibrin polymerization. With the excess plasmin apy has not been standardized, but antiplatelet drugs (e.g.,
activity, the euglobulih lysis time is typically shortened. aspirin or dipyridamole), plasmapheresis, and exchange trans-
Because thrombin is not generated during this pathologic fusion have been used either singularly or in combination
process those laboratory finding that are the direct result of with variable success (see Chap. 25). The other causes of fib-
thrombin activity will be absent. Several laboratory tests can rinolytic activation are summarized in Table 27-7.
serve to readily distinguish primary fibrinolysis from DIC. In
primary fibrinolysis, the platelet count is typically normal;
fibrinopeptides A and B levels are not elevated, and circulat-
ing fibrin-monomer complexes and elevated D-dimer levels
are absent in contrast to the results in DIC (Table 27-6).
Primary
Laboratory Test Fibrinolysis
Prothrombin time
Activated partial
thromboplastin time
Thrombin time
Fibrinogen
Platelet count
Fibrinopeptide A
SFMC
TAT
FDPs
D-Dimers
1. Chronic liver disease Abnormal fibrinolytic Often fulminant hemorrhage ite Hypofibrinogenemia
inhibitor (a@,-macroglobulin) with massive hemoptysis, (due to lysis)
hematachezia, melena, or
epistaxis
Abnormal hepatic clearance May also demonstrate . Elevated FDP (X, Y, D,
of plasminogen activators petechiae, purpura, spider and E) a. Defective fibrin
telangiectasia, ecchymoses monomer/polymerization
b. Platelet dysfunction
. Proteolysis of factors V,
VU, [X, XI
. Platelet defects
a. Thrombocytopenia
b. Platelet dysfunction
(EDPAPRE3)
. Coagulation protein defects:
a. Decreased synthesis of
factors II, VII, IX, and X
b. Decreased synthesis of
Fletcher factor
c. Decreased or dysfunc-
tional synthesis of AT-HI
2. Cardiopulmonary Unclear; possibly direct Hemorrhage; hematuria, . Hyperfibrinolysis results in
bypass (CPB) activation of fibrinolysis petechiae/purpura, a. Elevated FDP
by the oxygenation system and oozing from b. Hypofibrinogenemia
of pump-induced acceler- intravenous site in c. Low levels of cofactors V
ated flow rates may after conjunction with and VIII
endothelial plasminogen increased chest tube loss . Functional platelet defect
a. CPB-induced
b. Drug-induced
. Thrombocytopenia
. Hyperheparinemia-heparin
rebound(?)
. DIC(?)
3. Malignancy Poorly understood; in Thrombosis/hemorrhage . Thrombocytopenia
several instances tumor . Platelet function defects
extracts possess the . Elevated FDP
ability to activate directly . Decreased AT-III
or indirectly the fibrinolytic — . DIC(?)
MN
ABWN
REFERENCES . Miller, JL: Normal fibrinolysis. In 2. Owen, WG, and Esmon, CT: Functional
Henry, JB (ed): Clinical Diagnosis and properties of an endothelial cell cofactor
. Kaplan, AP, et al.: Interaction of the clot- Management by Laboratory Methods, for thrombin-catalyzed activation of pro-
ting, kinin forming, complement and fib-
ed 17. WB Saunders, Philadelphia, tein C. J Biol Chem 256:5532, 1981.
rinolytic pathways. NY Acad Sci 389:25,
1984, p 769. . Prowse, CV, and Cash, JD: Physiologic
1982. . Mandle, RJ, and Kaplan, AP: Hageman and pharmacologic enhancement of fibri-
. Sundsmo, JS, and Fan, DS: Relation-
factor-dependent fibrinolysis: Generation nolysis. Semin Thromb Hemost 10:51,
ships among the complement, kinin,
of fibrinolytic activity by the interaction 1984.
coagulation and fibrinoltic systems.
of human activated factor XI and plas- . Rickli, EE: The activation mechanism of
Springer Semin Immunopathol 6:231,
minogen. Blood 54:850, 1979. human plasminogen. Thromb Diath
1983.
9. Bachman, F, and Kruithof, IEKO: Tissue Haemorrh 34:386, 1975.
. Rosenberg, RD, and Aird, WC: Vascular-
plasminogen activator: Chemical and . Brogden, RN, et al: Streptokinase: A
bed-specific hemostasis and hypercoagu-
physiological aspects. Semin Thromb review of its clinical pharmacology,
lable states. N Eng J Med 340:994,
Hemost 10:6, 1984. mechanism of action and therapeutic
1999.
. Hosaka, Y, et al: Thrombomodulin in uses. Drugs 5:357, 1973.
. Wu, KK, and Thiagarajan, P: Role of
human plasma contributes to inhibit . Edelberg, JM, et al: Vascular regulation
endothelium in thrombosis and hemosta-
fibrinolysis through acceleration of of plasminogen activator inhibitor-1
sis. Annu Rev Med 47:315, 1996.
thrombin-dependent activation of plasma activity. Semin Thromb Hemost 20:319,
. Michiels, C: Endothelial cell functions.
procarboxypeptidase. Thromb Haemost 1994.
J Cell Physiol 196:430, 2003.
DEST, USS. . Vaughn D: Update on plasminogen acti-
6 . Castellino, FJ: Recent advances in the
. Owen, WG: The control of hemostasis. vator inhibitor-1. Can J Cardiol.
chemistry of the fibrinolytic systems.
Arch Pathol Lab Med 106:209, 1982. 14(Suppl D):14, 1998.
Chem Rev 81:431, 1981.
Chapter 27 Interaction of the Fibrinolytic, Coagulation, and Kinin Systems; DIC; and Related Pathology 659
18. Juhan-Vague, I et al.: Fibrinolytic factors 2 Demarmeis Biasiutti, F, et al: Is plas- 3 Menell, J, et al: Annexin II and bleeding
and the risk of myocardial infarction or minogen deficiency a risk factor? A in acute promyelocytic leukaemia.
sudden death in patients with angina study on 23 thrombophilic patients and N Engl J Med 340:994, 1999.
pectoris. ECAT Study Group. European their family members. Thromb Haemost 36. Dempfle, CE: Coagulopathy of sepsis.
Concerted Action on Thrombosis and 80:167, 1998. Throm Haemost 91:213, 2004.
Disabilities. Circulation 94:2057, 1996. 28. Kahr, WH, et al.: Platelets from patients . Fareed, J, et al: Impact of automation on
. Gonzalez-Gronow, M, et al: Purification with Quebec platelet disorder contain the quantitation of low molecular weight
and some properties of the glu-and lys- and secrete abnormal amounts of markers of hemostatic defects. Semin
human plasmin heavy chains. J Biol urokinase-type plasminogen activator. Thromb Hemost 9:355, 1983.
@hemZa2-2Wsn 977: Blood 104:159, 2004. . Hirsh, J: Blood tests for the diagnosis of
. Marder, VJ, et al: High molecular weight 29} Eichinger, S, et al: Thrombin-activatable venous and arterial thrombosis. Blood
derivatives of human fibrinogen pro- fibrinolysis inhibitor and the risk for DAIL. awl.
duced by plasmin. I. Physicochemical recurrent venous thromboembolism. 339). Levi, M, and deJonge, E: Current man-
and immunologic characterization. J Biol Blood 103:3773, 2004. agement of disseminated intravascular
Chem 244:2111, 1969. . Carey, MJ, and Rodgers, GM: Dissemi- coagulation. Hosp Pract 35:59, 2000.
Oe Kudryk, B, et al: Measurement in human nate intravascular coagulation: Clinical 40. Sakuragawa, N, et al: Clinical evaluation
blood of fibrinogen/fibrin fragments con- and laboratory aspects. Am J Hematol of low molecular weight heparin
taining the B-beta 15—42 sequence. SOS), WON). ((FR-860) on disseminated intravascular
Thromb Res 25:277, 1982. . Bick, RL: Disseminated intravascular coagulation—a multicenter co-operative
. Ockelford, A, and Carter, J: DIC: Appli- coagulation: Pathophysiological mecha- double-blind trial in comparison with
cation and utility of diagnostic tests. nisms and manifestations. Semin heparin. Thromb Res 72:475, 1993.
Semin Thromb Hemost 8:198, 1982. Thromb Hemost 24:3, 1998. 4]. Vincent, JL, et al: Effects of drotrecogin
. Aoki, N, and Harpel, PC: Inhibitors of . Bick, RL: Disseminated intravascular alfa (activated) in organ dysfunction in
the fibrinolytic enzyme system. Semin coagulation: Objective criteria for clini- the Prowess Trial. Crit Care Med 31:834,
Thromb Hemost 10:24, 1984. cal and laboratory diagnosis and assess- 2003.
. Xu, H, et al: Thrombomodulin changes ment of therapeutic response. Clin Appl 42. Tallman, MS, et al: New insights into the
the molecular surface of interaction and Thromb Hemost 1:3, 1995. pathogenesis of coagulation dysfunction
the rate of complex formation between . Levi, M, and Ten Cate, H: Disseminated in acute promyelocytic leukemia. Leuk
thrombin and protein C. J Biol Chem intravascular coagulation. N Engl J Med Lymph 11:27, 1993.
280:7956, 2005. 341:586, 1999. . Nabhan, C, and Kwaan, HC: Current
. Cesarman-Maus, G, and Hajjar, KA: . Gordon, SG, et al: Cancer procoagualnt: concepts in the diagnosis and manage-
Molecular mechanisms of fibrinolysis. a factor X activator, tumor marker and ment of thrombotic thrombocytopenic
Br J Haemotol 129:307, 2005. growth factor from malignant tissue. purpura. Hematol Oncol Clinics North
. Kwaan, HC: Disorders of fibrinolysis. Blood Coagul Fibrinolysis 8:73, Am 17:177, 2003.
Med Clin N Am 56:163, 1972. NOOTe
Chapter
Introduction
to Thrombosis
and Anticoagulant
Therapy
Aamir Ehsan, MD
Jennifer L. Herrick, MD
Introduction OBJECTIVES
Regulation of Coagulation At the end of this chapter, the learner should be able to:
andFibrinolysis
The Role of Endothelium iD Name natural anticoagulants and inhibitors present in plasma.
Platelets . Describe the role of endothelium in thrombogenesis.
Procoagulant Factors and
Generation of Thrombin . Describe the mechanism of thrombin/thrombomodulin.
Natural Inhibitors of . Compare and contrast the protein C and protein S systems.
Coagulation Factors
(Plasma Components) . List the inherited causes of thrombophilia in order of frequency of occurrence.
Fibrinolytic System . Name the risk factors and acquired conditions with hypercoagulable states.
Inherited Thrombophilia
. List the laboratory tests to evaluate patients with hypercoagulable states.
Activated Protein C
Resistance . Describe the issues to consider in laboratory testing in patients with thrombosis.
Protein C Deficiency
. List laboratory tests for evaluating a lupus anticoagulant and the antiphospholipid
Sey
fee
Na)
SSS)
SS
a
Protein S Deficiency
syndrome.
Antithrombin Deficiency
Prothrombin Nucleotide . Describe the mechanism of heparin-induced thrombocytopenia.
G20210A Mutation
. Name laboratory tests for evaluating heparin-induced thrombocytopenia.
Hyperhomocysteinemia
Tissue Factor Pathway . Explain the mechanism of action and define the conditions in which heparins, oral
Inhibitor Deficiency anticoagulants, direct thrombin inhibitors and thrombolytic agents are used.
Factor XII Deficiency
. List the most common laboratory tests to monitor oral anticoagulant therapy.
Heparin Cofactor Il
Deficiency . List laboratory tests to monitor heparin therapy.
Dysfibrinogenemia
Elevated Plasma Factor VIII
Coagulant Activity
Lipoprotein a and
Thrombosis
Other Coagulant Factors
Associated with
Thrombosis
660
Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy 66]
Acquired Thrombotic
Disorders
Lupus Anticoagulant/
Antiphospholipid
Syndrome
Heparin-Induced
Thrombocytopenia
Other Acquired Conditions
Associated with
Thrombosis
Thrombosis with Pregnancy
and Use of Oral
Contraceptives
Thrombosis and Nephrotic
Syndrome
Thrombosis and
Medications
Thrombosis in Cancers and
Other Conditions
Thrombosis and Major
Trauma
forced into sequentially narrower spaces (the surface area to plasminogen activator inhibitor (TPAI) to block degradation
volume ratio increases) to allow the exchange of oxygen from of a newly formed clot. Interestingly, endothelial cells can be
the red blood cell to the tissues. The peak of this activity induced by various cytokines and endotoxins to produce tis-
is reached in the capillary bed where, at a vessel diameter sue factor (also known as thromboplastin), which initiates
as small as 4 um, the surface area to volume ratio approaches coagulation through the activation of factor VII. This is a
1000 mM*/uL*’ More graphically, it is the same as spreading mechanism that may explain the procoagulant state seen in
1 mL of liquid uniformly over the surface of an average game septic and neoplastic conditions.
table, allowing no cell or molecule to pass through a capillary
without contacting the endothelial cells lining the vessel. As a
result, opportunities for ligand-receptor interactions are pro- Platelets
vided and, if needed, endothelial cell activation resulting in a Platelets are activated in response to trauma to the vessel wall.
slowing of velocity within these areas that allows for an Platelet activation has three steps: (1) adhesion, (2) secretion,
increased chance of unnecessary coagulation. In addition, as in and (3) aggregation (see Chap. 24). Adhesion to the underly-
rivers, both large- and medium-diameter vessels have slower ing collagen, which anchors the activity to the needed area
blood flow near the vessel wall, creating a need for endothe- and stimulates a change in platelet shape, defines the first step
lial cells to have anticoagulant properties along the surface to of platelet activation. Functional aspects of platelet shape
prevent pathologic clot formation. However, if the lining of change lie in the rearrangement of the phospholipid platelet
the vessel is breached, agonists contained in the damaged membrane. This not only provides a surface for the surface-
endothelial cell stimulate immediate hemostatic mechanisms dependent component of coagulation (contact factors, which
that localize clot formation to the area of trauma to prevent regionalizes the coagulation enzymes to enhance enzyme—
excessive blood loss and thus the cell contains procoagulant substrate encounters) but also exposes binding sites for
properties when needed. These prothrombotic and anticoagu- platelet-to-platelet binding (aggregation). The catalytic ability
lative roles are discussed subsequently. of the platelet surface may in part explain why thrombin gen-
eration is greatly accelerated in the presence of platelets. The
ANTICOAGULANT ROLE
second step is the secretion of agonists that function in
In the normal state, intact endothelium physically prevents recruiting additional platelets and promote further adhesion
adhesion of platelets to underlying collagen, preventing an and aggregation. The platelet release reaction (secretion)
initiating event of platelet activation. Endothelial cells dis- contributes to thrombogenesis through the release of the
courage platelet aggregation by the continual release of alpha (a) granule contents in response to various stimuli (e.g.,
prostacyclin (PGI,) and nitric oxide (both potent aggrega- thrombin). These granules are visible via light microscopy
tion inhibitors). A product of arachidonic acid metabolism, and contain the platelet-specific proteins, platelet factor
the PGI, molecule is the most important natural inhibitor of 4 (PF4), beta (8)-thromboglobulin, platelet-derived growth
platelet function. This is the mechanism by which aspirin factor, and a variety of other proteins, including fibrinogen,
and nonsteroidal anti-inflammatory medications (NSAIDs) fibronectin, factor V, factor VII, and vWF. These con-
exert their platelet inhibitory effects. The endothelial cell tribute to platelet aggregation, thrombin generation, as
affects secondary hemostasis by binding the endogenous well as local platelet adhesion. The other type of storage
anticoagulants, thrombomodulin and heparin-like mole- granule, called dense bodies, are visible only via electron
cules, to its surface, thereby inhibiting the coagulation cas- microscopy and contain ADP, ATP, calcium, histamine,
cade (described in more detail later). Lastly, endothelial serotonin, and epinephrine. The third step in platelet activa-
cells synthesize tissue plasminogen activator (t-PA), which tion is aggregation, which is linked through fibrinogen
by promoting fibrinolytic activity is instrumental in the and creates the end product of primary hemostasis: the
degradation of any wayward fibrin deposits on endothelial platelet plug (the main physical barrier). These three steps
surfaces. of platelet activation are the basis of the platelet function
assays. How platelets respond enables assessment of the
PROTHROMBOTIC ROLE etiology of platelet disorders.
When endothelial cells are torn, platelets adhere to the newly
exposed subendothelial collagen, become activated, and
secrete substances to recruit and activate additional platelets. Procoagulant Factors and Generation
The cells synthesize von Willebrand factor (VWE), which is of Thrombin
the bridging molecule essential for this platelet adhesion to
collagen. vWF is packaged in cytoplasmic secretory granules As stated earlier, endothelial cells express binding sites for acti-
called Weibel-Palade bodies. It also serves as a carrier mole- vated factors [Xa and Xa. They also provide vWF, which
cule for factor VIII. This coupling of vWF to factor VIII bridges platelets to collagen and also facilitates efficient clotting
serves two functions: (1) protection of factor VIII from degra- by binding and stabilizing factor VII, localizing this powerful
dation and (2) localization of factor VIII to the area of trauma. potentiator of clot formation to the required area of hemostasis.
The endothelial cell also expresses binding sites for activated The resultant environment of activated platelets and clotting
factors [Xa and Xa on its surface, further localizing coagula- factors generates thrombin (factor Ia) through both the extrin-
tion activity to the site of injury while producing tissue sic and intrinsic pathways of the coagulation cascade. Thrombin
664 Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy
then is principally responsible for the resultant generation of Natural Inhibitors of Coagulation Factors
fibrin monomers which become the insoluble fibrin strands (Plasma Components)
needed for blood clot formation. Thrombin is the last and
most important enzyme of the clotting cascade. The active Normal plasma contains a sophisticated system of serine pro-
site of the enzyme is buried deep within a groove that is pro- tease inhibitors capable of inhibiting many of the activated
tected by surrounding structures. Selective access accounts proteases generated during coagulation that slow the genera-
for the specificity of the enzyme. In addition, thrombin has tion of thrombin (Fig. 28-1). Three types of natural anticoag-
two positively charged regions that are sites for macromole- ulants are present in the plasma:
cule ligand binding (exosite I and exosite II). Exosite I is the
|. Serine-protease inhibitors, AT and heparin cofactor II
binding site for the thrombin receptor, fibrinogen, factor V,
(HC-II).
protein C, and thrombomodulin. Owing to its versatility, NO . Protein C (PC), which upon activation becomes APC and
thrombin is one of the most fascinating enzymes in the coag-
degrades factor Va and VIIa in the presence of its cofactor
ulation system. Among the various activities of thrombin are
Protein S (PS).
the following (Table 28-1):
3. Tissue factor pathway inhibitor (TFPI).
1. It proteolytically cleaves fibrinogen to produce two mole-
cules of fibrinopeptide A and two molecules of fibrinopep-
ANTITHROMBIN (ANTITHROMBIN-IID)
tide B from the Aa and BB fibrinogen chains, respectively.
According to the current international nomenclature, AT-III
This results in the conversion of fibrinogen to a fibrin
is renamed antithrombin (AT). AT combines with heparin as
monomer. The released fibrin monomers spontaneously
a cofactor to become the most powerful anticoagulant in the
polymerize to form fibrin.
circulation. On serum protein electrophoresis, it is an
2. It converts factor XIII (in plasma and platelets) to an active
a>-glycoprotein composed of a single chain of 432 amino
transglutaminase, which cross-links fibrin with covalent
acids with a molecular mass of 58 kD and an in vivo half-
amidé bonds that render the fibrin insoluble in order to
life of approximately 2 to 3 days. AT is synthesized by the
become a physically binding structure.
liver and belongs to the serine protease inhibitor (serpin)
3. It activates the procoagulant factors V, VIII, and XI, to par-
superfamily. Normal plasma levels are approximately
ticipate in amplifying its own generation.
150 ug/mL. AT is a major inhibitor of thrombin (factor Ila)
4. It also binds platelets at low concentrations and initiates
and factor Xa. Other coagulation factors such as XIla, XIa,
shape change, aggregation, and secretion promoting pri-
and [Xa are also inhibited but to a lesser extent.
mary hemostasis.
Normally, AT is a relatively weak inhibitor of the serine
5. It participates in self-regulation by binding to thrombo-
proteases, but it is activated in vivo by heparin released from
modulin which then activates PC and PS to inhibit exces-
granules within mast cells and heparan sulfate, a glu-
sive thrombin formation.
cosaminoglycan which lines the endothelial layer. Heparin,
The complexity of the thrombin molecule is demonstrated which is highly negatively charged, interacts with a short pen-
by its participation in self-regulation and although it is tasaccharide ___domain in the AT molecule containing a high
generally considered a powerful procoagulant, has the density of basic amino acids, particularly lysine. As a result of
capacity to become an anticoagulant. This property will be the conformational change in the AT molecule precipitated by
discussed in more detail in the following section. Interest- the binding of heparin, an arginine residue is made readily
ingly, in addition to these functions in coagulation, throm- available to the active site of a serine protease. A locked com-
bin has been shown to play a role in inflammation and cell plex that blocks enzymatic activity is formed between AT and
growth.* the factor, tying up the availability of the protease. As the
complex forms, the heparin molecule disassociates and is
ready to react with another AT molecule. Therefore, heparin
administered even in small doses converts AT from a slow,
relatively ineffective inhibitor to a fast, effective one. The active form in human platelets and that platelet HC-II could
conformational changes due to this pentasaccharide sequence play a role in the regulation of thrombin generated on the
accelerates by several hundred-fold the inhibition of factor Xa.° platelet surface. A number of clinical studies including a large
This effect has been exploited in the development of scale effort with a decade of follow-up have failed to demon-
the relatively new anticoagulant medication, fondaparinux, strate evidence of a correlation between HC-II deficiency and
which is a synthetic form of this pentasaccharide sequence!” arterial or venous thrombosis.'?
and a direct Xa inhibitor. However, to induce inhibition of
thrombin, longer heparin chains are required. Longer chain THROMBIN/THROMBOMODULIN INTERACTION
heparins such as those found in unfractionated heparin, Thrombomodulin is a transmembrane glycoprotein synthe-
inhibit through approximation (binding and linking the target sized mainly by endothelial cells with a crucial role in the reg-
serine protease to antithrombin via a second heparin binding ulation of coagulation. When thrombin is generated at a
site), as well as the short-chain pentasaccharide-induced con- remote ___location and is released into the circulation, it comes
formational change. into contact with and quantitatively binds to the thrombomod-
ulin on the surface of the endothelium.'*'° When bound, it
HEPARIN COFACTOR I (HC-ID changes its enzymatic specificity, no longer recognizing fib-
HC-II is a minor inhibitor of thrombin and was first identified rinogen as substrate and, paradoxically, acts on protein C
by Birginshaw in 1974"! and isolated by Tollefsen in 1981.!* (PC), generating activated protein C (APC). APC, in the pres-
The primary amino acid structure of HC-II is quite distinct ence of its cofactor, protein S (PS), forms the APC proteinase
from that of AT. The specificity of HC-II is narrowly restricted complex. This complex has powerful anticoagulant effects by
to thrombin and does not inhibit other coagulation factors. cleaving factors Va and VIIla and promoting fibrinolysis by
HC-I also possesses an affinity for heparin that is significantly inactivating plasminogen activator inhibitor-1 (PAI-1). The
less than that of AT; therefore, a higher concentration of APC-proteinase complex constitutes, on a molar basis, the
heparin is necessary to inhibit thrombin by this cofactor. How- most active inhibitor of plasma coagulation identified, and
ever, other mucopolysaccharides, such as dermatan sulfate, are this factor Va-protein C pathway plays a significant role in the
able to dramatically accelerate the action of this protease regulation of thrombus formation (Fig. 28-2). Therefore, as
inhibitor. Indeed, it appears likely that the observed anticoag- previously mentioned, thrombin indirectly possesses antithrom-
ulant effects of mucopolysaccharides other than heparin are botic activities, limiting the extent of its own generation.
primarily a result of interactions with HC-II. HC-II probably A soluble form of thrombomodulin is present in plasma
plays a minimal role when heparin is used clinically as an with physiologic levels of 3 to 50 ng/mL.'° Increased soluble
anticoagulant. Tollefsen has suggested that HC-II may func- TM levels are seen in conditions with vascular damage (i.e.,
tion as a second-line inhibitor of thrombin after its activation inflammation and sepsis).'’ The relationship between soluble
by dermatan sulfate present on the surface of the vessel wall. TM levels and coronary artery disease are controversial and
It has also been suggested that HC-II is present in a functional remain under debate.'*
Activation of APC
Endothelium
Va to Vi
Villa to Vili
Figure 28-2 # Thrombin binds to thrombomodulin on the surface of endothelial cell. Thrombin/thrombomodulin complex acts on protein C,
resulting in the formation of activated protein C (APC). APC cleaves factors Va and Villa in the presence of its cofactor, protein S. APC also inac-
tivates plasminogen activator inhibitor-1 (PAI-1). Inhibition of APC occurs by PCI-1 and PCI-2. TR = thrombin; TM = thrombomodulin;
PC = protein C; PS = protein S; PCI = protein C inhibitor.
666 Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy
THE PROTEIN C AND S SYSTEM formation. This activity is enhanced in the presence of heparin.
PC is a vitamin K-dependent zymogen with anticoagulant A second inhibitor of PC, named protein C inhibitor-2 (PCI-2),
properties that, once activated by the thrombomodulin-altered was found to be q,-antitrypsin by amino acid analysis and is
thrombin molecule, proteolytically degrades factors VIIa and believed to be heparin independent.°!
Va.'° PC is a glycoprotein with heavy and light chains linked
by a disulfide bond with a molecular weight of 62 kD. The TISSUE FACTOR PATHWAY INHIBITOR
anticoagulant role of PC is also described under thrombin/
TFPI circulates and is associated with a lipoprotein that
thrombomodulin interaction, previously, and is depicted in
inhibits plasma coagulation in a dual manner: (1) it serves to
Figure 28-2.
bind the activated form of either factor X or factor IX, thereby
Thrombin/thrombomodulin activation of PC is modu-
inhibiting their enzymatic activity and (2) the TFPI-factor
lated by activated factor V (Va) concentrations. Factor Va
Xa/or [Xa complex then binds to the membrane-bound factor
concentrations above a certain level enhance the activation VIla-tissue factor complex, competitively inhibiting further
rate of PC, whereas lower concentrations inhibit PC activa-
activation of factor X or factor IX through the extrinsic path-
tion. This provides a necessary feedback mechanism allowing way.”* These actions are depicted in Figure 28-3. The role of
factor Va activation to occur (favors coagulation) until the
TFPI in clinical thrombosis has been studied in animal mod-
level becomes high enough to necessitate control of the els. The absence of circulating TFPI activity has been shown
process (to avoid hypercoagulation) through PC activation.
to result in the reversal of aspirin inhibited intravascular
PC function is significantly enhanced in the presence of its
thrombus formation. TFPI plasma levels appear to be acutely
cofactor, PS, and phospholipids. In the absence of adequate
consumed in acute myocardial infarction from intracoronary
quantities of PS, PC function is inadequate to control the
thrombosis. Most intriguing is recent data regarding arterial
generation of thrombin. A role in inflammation has been
wall gene transfection with that coding TFP with promising
established by the favorable clinical response to infused
therapeutic implications in the prevention of intravascular
recombinant APC in patients with sepsis.°°
thrombus formation. Certainly TFPI is an important compo-
In the presence of calcium, protein S (PS) binds to the
nent of dysregulated coagulation and may represent a pivotal
phospholipid surface of endothelial cells and activated
protein in novel therapeutic interventions.”
platelets and associates with activated protein C. This local-
izes the PS-APC complex at the needed site of clot formation.
PS is a vitamin K—dependent protein that is produced in the Fibrinolytic System
liver, as well as, in megakaryocytes and endothelial cells. PS
is a single chain with a molecular mass of 69 to 84 kD. It is The fibrinolytic system in vascular hemostasis is critical for
unique among the vitamin K—dependent coagulation proteins controlling the amount of clot formation and for resolving
in that it is not the zymogen of a serine protease. Plasma PS clots no longer needed. The plasminogen inhibitors contribute
circulates in two forms: one is bound with a 1:1 ratio to the to clot stability by blocking premature dismantling of the
C4b-binding protein (C4bBP) from the complement system, thrombus. Among the components of the fibrinolytic system
the other is free. The free PS form represents about 40% of are plasminogen/plasmin and the activator system: urokinase-
total PS in normal individuals and is the only functional form plasminogen activator (u-PA) and tissue-type plasminogen
of PS. Platelets also contain PS; therefore, the activation of activator (t-PA), which convert plasminogen into the func-
platelets provides not only a surface for procoagulant reactions, tional form, plasmin. The major inhibitors are a,-antiplasmin,
but also a cofactor for eventual control of thrombin generation. and plasminogen activator inhibitors | and 2 (PAI-1, PAI-2).
Decreased PS synthesis 1s seen in many conditions, the most Physiologic fibrinolysis results in the proteolytic degradation
common being oral anticoagulant therapy; however, vitamin K of polymerized fibrin. The central reaction in this system is
deficiency, liver disease, chemotherapy, and L-asparaginase the conversion of a proenzyme, plasminogen, to the prote-
therapy are also known etiologies. Consumption of all factors olytic enzyme plasmin. Plasminogen is a 90 kD glycoprotein
including protein S can occur with acute thrombotic events or synthesized in the liver with a circulating half-life of 2 to
disseminated intravascular coagulation (DIC). The carrier mol- 3 days. Plasminogen circulates both in the free state or bound
ecule C4b-BP acts as an acute phase reactant with fluctuating to histidine-rich glycoprotein (HRGP), «,-antiplasmin, or fib-
concentrations associated with inflammatory states. An increase rinogen itself. The physiologic activation of plasminogen is
in C4b-BP correspondingly increases the percentage of bound achieved through the extrinsic pathway initiated by t-PA
PS antigen and a relative decrease in free PS antigen and there- release from the endothelial cells after stimulation, and by the
fore PS activity. Conditions that are associated with an increase intrinsic pathway through a factor XIIa-dependent activator
in C4b-BP concentrations are pregnancy, oral contraceptive and urokinase.
use, diabetes mellitus, and systemic lupus erythematosus. Free A specific group of proteins control fibrinolysis. They
PS antigen and activity are also often decreased in nephrotic are serine protease inhibitors and are members of the serpin
syndrome. superfamily. They are structurally homologous to many
Two PC inhibitors have been described. The first is inhibitors of the serine proteases of the coagulation system
protein C inhibitor-1 (PCI-1), also known as plasminogen (Table 28-2). Plasmin activity is regulated and inhibited by
activator inhibitor-3 (PAI-3). PCI-1 inhibits the thrombin— a number of plasma proteins such as a,-antiplasmin,
thrombomodulin complex which indirectly inhibits APC Q-macroglobulin, o,-antitrypsin, AT (especially in the
Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy 667
Active enzyme
Factor VilaV_|
Active complex
Inactivated complex
Figure 28-5 ™ Shown here is the interaction of tissue factor pathway inhibitor (TFPI) with factors VII, IX, and X. (Courtesy of John D. Olson,
MD; PhD, Department of Pathology, University of Texas Health Science Center, San Antonio, TX.)
presence of heparin), and Cl esterase inhibitor. Physiologi- Plasminogen activators such as uPA and tPA, are inhibited
cally, «,-antiplasmin, the main plasmin inhibitor, is a 60 kD by a number of specific proteins. PAI-1 and PAI-2 are found in
glycoprotein and circulates in the plasma at a concentration of plasma and urine (Table 28-3). A third, tentatively termed
1 uM. Its properties include: PAI-3, was subsequently shown to be the previously described
nonspecific serine protease heparin-binding APC _ inhibitor,
1. Drect inhibition of plasmin
PCI-1. These inhibitors are nonspecific in nature and deacti-
2. Interference with absorption of plasminogen to fibrin
vate enzymes extrinsic to the coagulation system. The most
3. Susceptibility to factor XIII-catalyzed cross-linking of
important inhibitor in the plasminogen activator system is
antiplasmin to fibrin which incorporates the enzyme into
PAI-1, the primary fast-acting serpin inhibitor of t-PA. The
the structure of the clot providing stability by inhibiting
physiologic plasma concentration of PAI-1 varies widely
intrathrombotic plasmin activity.
from 0 to 60 ng/mL, with an average range of 5 to 20 ng/mL
The combined effects of these three characteristics render and many patients with myocardial infarction have elevated
a,-antiplasmin much more specific and effective in inhibition levels. Because the role of this enzyme is to dampen the
of fibrinolysis than any of the other major inhibitors. degradation of a clot, there has been interest in defining a role
in myocardial infarction and stroke, especially in atypical
patients (younger than 45 years of age). Although a case
of theDeerine Grotensa |
Inhibitors _se
PAI-2
Pay, abibitory, Effects
Origin Plasma Placenta tic
ee Ahiplenin Plasmin
Platelets Leukocytes Plasma
a>-Macroglobulin Thrombin, plasmin, kallikrein
Endothelial Macrophages
a,-Antitrypsin Thrombin, trypsin,
cell
chymotrypsin, factor XIa,
Granulocytes
elastase, activated protein C
MW 54,000 47,000 50,000
AT Thrombin; factors Xa, XIIa,
(daltons)
IXa, and Xa; plasmin;
Inhibitory TRA TPA UK
kallikrein effect
C1 inhibitor Kallikrein, plasmin, factors
UK UK APC
XIJa and XIa, C1 esterase
Concentration O-1.3nM 2 uM -
Antichymotrypsin Chymotrypsin
_ Grd trimester)
HC-II Thrombin
Protein C inhibitor Activated protein C UK = Uiadee APC = = activated protein C; MW = molecular
PAI-1 and PAI-2 Plasminogen activators weight; nM = nanomoles; uM = micromoles.
668 Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy
control study reported a correlation of younger patients with The exogenous addition of activated protein C (APC) to
myocardial infarction and a PAI-1 genetic polymorphism, a test tube of normal plasma creates an anticoagulant effect
subsequent prospective studies did not support this finding. that can be measured as a prolongation of the activated partial
More recently, the noncoagulant role PAI-1 plays in adipocyte thromboplastin time (aPTT). The absence of this anticoagu-
formation has received attention. Some researchers have lant response reflects the decreased ability of APC to inacti-
induced weight loss in mice despite a high-calorie diet by vate factor Va and results in a thrombophilic state called APC
inhibiting PAI-1.** Additional studies suggest a role between resistance (APC-R). The concept of APC-R was first
the upregulation of PAI-1 and ethanol-induced liver injury.” described by Dahlback and associates in 1993.*7 In the major-
Another enzyme under investigation is thrombin acti- ity of cases, APC-R is the result of a genetic defect. The pre-
vatable fibrinolysis inhibitor (TAFI), also known as procar- dominant etiology of APC-R is a defect in the factor V gene
boxypeptidase B. This clot-stabilizing 55 kD glycoprotein involving the point mutation at codon 506 of exon 10 which
is synthesized in the liver and platelet alpha granules and substitutes a glutamine residue for the wild-type arginine
circulates in plasma bound to plasminogen. TAFI cleaves amino acid and is termed “factor V Leiden” (or FV:Q°”°).”8
C-terminal lysine groups essentially removing the binding This codon is the site where APC cleaves and inactivates
sites for plasminogen and tPA to fibrin strands. The exact factor Va. The activity of factor V and its role in coagulation
role in hypercoagulable states and inflammation is not clear are not affected. The mutated factor Va is resistant to cleavage
at the present time.*° by APC and results in prolonged procoagulant activity; there-
The fibrinolytic system is as complex as the coagulation fore, it increases the risk of pathologic thrombus formation.
system. The cause-and-effect relationship between the fre- Mutations at the arginine 306 residue in factor V, the second
quency of deficiencies in the physiologic fibrinolytic system and APC site, have been reported as well but are much less
the occurrence of thromboembolism is not fully understood. common.” Other possible mechanisms of APC-R_ include
However, it seems the most common abnormality is the pres- inhibitors (autoantibodies) to APC, functional PS deficiency,
ence of excess PAI-1 leading indirectly to a decreased functional and mutated forms of factor VIII; however, no genetic abnor-
availability of t-PA, which results in a hypercoagulable state. malities in the factor VIII gene have been identified to cause
APC-R in humans.
Among Caucasians, APC-R is the most common risk
Inherited Thrombophilia factor associated with inherited venous thrombosis.. It can be
detected in 20% to 60% of patients with recurrent venous
Inherited thrombophilia is a group of congenital hematologic
thrombosis and shows autosomal-dominant inheritance. The
disorders that includes a variety of hypercoagulable states
heterozygous state is relatively common in Western countries
that usually present clinically as venous and/or arterial
and occurs in approximately 6%*° of the general popula-
thrombosis. Hypercoagulability is usually defined as an alter-
tion.*'** The prevalence in Hispanic, African American,
ation of the blood coagulation mechanism that predisposes to
Asian, and Native American populations appears to be low.
thrombosis.
The thrombotic risk for patients carrying heterozygous factor
The homozygous deficiency of natural inhibitors of
V mutation is increased five to ten times, while the risk for
coagulation (e.g., deficiencies of PC and PS, factor V Leiden
homozygous patients is 50 to 100 times.
mutation) substantially increases the risk of thrombosis. For
Factor V Leiden—associated thrombotic risk is dependent
example, virtually all neonates with homozygous deficiency
on the underlying clinical setting. Although oral contracep-
of PC and/or PS deficiency suffer from purpura fulminans in
tives and pregnancy significantly increase the risk for venous
the perinatal period. In contrast, the heterozygous condition
thrombosis in women with the mutation, the increased throm-
for PC deficiency and factor V Leiden mutation is widely
botic risk in patients with preexistent cancer or recent surgery
variable in the expression of thrombosis. There is little ques-
cannot be demonstrated.*° There is no convincing data that
tion that these conditions impart a risk; however, it is notable
factor V Leiden mutation confers an increased risk for arter-
that the majority of people who carry heterozygous mutations
ial thrombosis. Data for factor V Leiden as an independent
for many of the genes associated with thrombosis never man-
risk factor for myocardial infarction are controversial.
ifest a thrombotic disease. For thrombosis to manifest clini-
Most of the screening tests for APC resistance are func-
cally, more than one and presumably several, risk factors need
tional aPTT—based assays. They can be used as a screening
to be present simultaneously to overcome the natural
method to identify patients in need of the confirmatory factor
processes in the blood that are normally protective against
V Leiden molecular test. The functional APC resistance assay
thrombosis.
is performed by measuring an aPTT both in the absence and
then the presence of a standardized amount of APC, with a
Activated Protein C Resistance calculation of the resultant ratio (the APC ratio, Fig. 28-4). In
a normal person, the addition of APC to plasma induces a pro-
When thrombin is generated at a remote ___location and is longed aPTT because APC cleaves and inactivates factors Va
released into the circulation, it passes to the capillary bed and VIIIa. In patients with APC resistance, the aPTT is rela-
where it will bind quantitatively to thrombomodulin on the tively shorter than in normal plasma. Factor Va is not cleaved
surface of the endothelium. This mechanism was described and therefore is not inactivated. The test results can be inter-
earlier and is depicted in Figure 28-2. preted by comparing the ratio to the normal range, or by
Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy 669
Hidsthabieststecliia
Note: The sensitivity of the test is increased by comparing the ratio to the normal
range or by normalizing it to the APC resistance obtained using normal pooled
plasma.
Figure 28-4 @ Data on the functional assay for activated protein C are shown here. This is an APTT-based assay and the APC-R ratio can be
calculated by measuring the patient clot time with and without activated protein C (APC) in the presence of CaCl,. In a normal person, the
ratio is more than 0.9. In case 1, the final ratio is 1.2 and thus the test is negative for activated protein C resistance (APC-R). In case 2, the
ratio is less than 0.9 and the test for APC-R is positive. In case 2, therefore, the patient should be tested for factor V Leiden mutation by poly-
merase chain reaction (PCR). APC-V = activated protein C-factor V ratio.
normalizing it to the APC resistance ratio obtained using sample DNA is analyzed, allowing specific genotypes to be
normal pooled plasma. By diluting the patient’s plasma with identified (normal, heterozygous, or homozygous).
an excess of factor V—deficient plasma, the sensitivity and The genetic test is usually performed to confirm the
specificity of the aPTT-based APC-R assay can be increased factor V Leiden mutation and has several advantages over
and allow the analysis of plasma from patients who are taking plasma-based functional assays. The genetic test is not
oral anticoagulants or have other factor deficiencies without affected if the patient is receiving anticoagulants or has
interference. Factor V—deficient plasma is made of normal plasma inhibitors (such as lupus anticoagulant). It does not
plasma with all the moieties needed for coagulation except fac- have a threshold range as does the functional plasma-based
tor V (which the patient’s plasma must contribute to the mix- assays, and it can reliably differentiate between heterozygous
ture) and thus normalizes the concentrations of other plasma and homozygous states.
proteins involved in the formation and regulation of thrombin. One of the approaches in diagnosing patients with factor
The limitation of this aPTT-based functional assay (and, in fact, V Leiden is to perform an aPTT-based functional assay for
all aPTT-based assays) is that the results are unreliable when the APC-R. Patients with low APC-R ratios (results may vary
test is performed on patients treated with heparins or in the pres- among different laboratories) should be genotyped for the
ence of an underlying inhibitor such as a lupus anticoagulant factor V mutation. For example, in our coagulation labora-
that can artificially prolong the baseline aPTT. The test; how- tory, a normalized ratio (obtained using normal pooled
ever, may be performed during an acute thrombotic episode. plasma) of less than 0.9 is considered abnormal. Even in lab-
The genetic test for FV:Q°*° mutation is often referred to oratories in which there is an excellent concordance between
as the factor V Leiden assay. In this test, the genomic DNA is APC-R assays and the results of factor V Leiden mutation
isolated from blood mononuclear cells. Using allelic discrim- assays, Some patients with a low APC-R ratio and negative
ination methodology by polymerase chain reaction (PCR), the genetic results can still be identified. The significance of this
670 Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy
finding is uncertain and may be related to mutations other measured via a chromogenic assay. Using the clotting and
than FV:Q°°. amidolytic assays in tandem, useful information regarding the
nature of molecular defect in type I] PC deficiency may be
Protein C Deficiency detected. The cases have been described with normal PC anti-
gen level but reduction in PC anticoagulant activity and
PC is vitamin K—dependent glycoprotein which is synthesized normal amidolytic activity via chromogenic assay. For this
by the liver. Hereditary deficiency of PC shows an autosomal reason, clot-based PC assays are preferred (Table 28-4). High
dominant inheritance pattern. The prevalence for inherited PC levels of factor VIII (greater than 250%) may falsely decrease
deficiency among patients with venous thrombosis linked to PC levels in a clot-based assay.
inherited causes is estimated to be 6% to 10%. The frequency Acquired PC deficiency can be seen in a number of con-
of the heterozygous state varies between 1/300 and 1/1000, and ditions (Table 28-5), including liver disease, DIC, warfarin
the frequency of the homozygous state is estimated to be therapy, severe infection/septic shock, adult respiratory distress
1/50,000.** Homozygotes have a marked tendency for recurrent syndrome, postoperative states, acute thrombotic episode and
venous thrombosis and PE, neonatal purpura fulminans, and secondary to chemotherapy (e.g., L-asparaginase, methotrex-
warfarin-induced skin necrosis. Interestingly, heterozygotes ate, cyclophosphamide, and 5-fluorouracil). It is important to
commonly do not manifest thrombosis unless concomitant note that due to the vitamin K dependency of this factor, mea-
additional risk factors exist. surement cannot be taken while on oral anticoagulation and
Warfarin-induced skin necrosis has been associated with should be drawn only after an appropriate waiting period sub-
patients who have PC deficiency. It occurs during the first few sequent to the cessation of warfarin therapy (approximately
days of warfarin therapy. Warfarin ingestion results in a 4 weeks).
decrease in vitamin K—dependent factors (II, VU, IX, X, PC,
and PS). Because PC has a short half-life (six hours) as com-
pared to other vitamin K—dependent factors, the decrease in
PC level occurs before the decrease in factor II and X levels.
This causes a temporary imbalance between procoagulant and
anticoagulant factors, resulting in a transient hypercoagulable
state that can lead to thrombosis and subsequent skin necrosis.
Anticoagulant Amidolytic
Infants with inherited homozygous PC deficiency may Subtypes Antigen Activity Activity
develop thrombosis of smali capillaries including that of cere-
bral vessels and laboratory evidence of disseminated intravas- I Low Low
Ila Normal Low
cular coagulation (DIC). The condition is usually fatal and is
IIb Normal
called neonatal purpura fulminans. It is important not to mis-
interpret the physiologically low level of PC in a normal new-
born, which may be near zero in a sick preterm infant. The
level usually rises rapidly after birth.
The functional PC activity assay (mixing patient’s
plasma with protein C—deficient plasma and measuring the
aPTT) is used as the screening assay for PC deficiency.
The antigenic assay is used to determine the mechanism of
the deficiency (decreased production or abnormal protein) Inhibitors Causes of Acquired Defects
and is rarely necessary in diagnosis. Based on immunologic AT DIC
and functional assays, two subtypes of heterozygous PC defi- Liver disease
ciency have been described. Type I deficiency is the most com- Nephrotic therapy
mon, which shows a reduction in the activity due to a Oral contraceptives
L-Asparaginase
concomitant decrease in the amount of antigen (to approxi-
Protein C Oral anticoagulant treatment
mately 50% of normal). Type II deficiency is less frequent, and DIC
like type I, shows reduced functional activity but can be differ- Vitamin K deficiency
entiated by the measurement of normal antigen levels. This After plasma exchange
type is a result of defective molecular mechanisms that allow Postoperative state
expression of a protein that has lost functional capacity.***> L-Asparaginase
Liver disease
The procedures used for antigenic assays are enzyme- Protein S Oral anticoagulant treatment
linked immunosorbent assay (ELISA), electroimmunodiffu- Pregnancy
sion (Laurell rocket electrophoresis), and radioimmunoassay. Oral contraceptives
In the simpler functional assay, PC is activated in the presence Vitamin K deficiency
of the specific activator extracted from snake venom. The Liver disease
Diabetes type I
resulting APC inhibits the factors V and VIII, and thus pro-
Acute inflammation
longs the APTT. The enzyme’s anticoagulant activity can be Newborn infants
measured in a clotting assay or its amidolytic activity can be
Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy 671
Protein S Deficiency acts as a cofactor for APC, functional assays may show spu-
rious low PS levels in patients with APC resistance .
PS is a vitamin K—dependent nonenzymatic glycoprotein syn-
thesized in the liver, endothelial cells, megakaryocytes, testes,
kidney, and brain. It exists in an active free form that func-
Antithrombin Deficiency
tions as a cofactor for APC, and an inactive form that is bound As discussed earlier, because antithrombin serves to regulate the
to complement binding protein (C4bBP). The free form most powerful procoagulant, thrombin, a deficiency results in a
accounts for 40% of total PS and has a half-life of approxi- prothrombotic state. Homozygous states are largely incompati-
mately 45 hours.* ble with life except for a rare functional (type II) variant, and the
PS deficiency is inherited as an autosomal dominant majority of affected individuals are heterozygous with AT levels
disorder. The prevalence of PS deficiency in the general popu- 40% to 70% of normal. It is an autosomal dominant disorder
lation is 1 in 33,000. The incidence of PS deficiency in patients with a prevalence in the general population of approximately
with venous thrombosis is 6% to 10%. In contrast to PC defi- 1/2000 to 1/5000. One percent of all venous thrombosis patients
ciency, even heterozygotes have a strong tendency to develop and 5% of patients younger than 70 years of age with venous
DVT, PE, cerebral and mesenteric thrombosis, superficial thrombosis were found to have an AT deficiency.°°**
thrombophlebitis, and arterial thrombosis and may also develop The initial clinical manifestation of AT deficiency is com-
wartarin-induced skin necrosis. Patients with homozygous PS monly between the age of 10 and 50 years (peak 15 to 35), may
deficiency are severely affected and may develop neonatal occur spontaneously in half of the patients and are characterized
purpura fulminans just after birth, similar to that seen with PC by thrombosis of the deep veins of the lower extremities, mesen-
deficiency. teric veins, and PE. In the remaining patients, the thrombosis
Both immunologic and functional methods are available may occur in the presence of additional risk factors such as
to measure PS. Reliable measurements of total antigen are pregnancy, the use of oral contraceptive pills, surgery, or trauma.
performed using radioimmunoassay, ELISA, or electroim- The risk of arterial thrombosis is not well established but has
munodiffusion (Laurell rocket electrophoresis). Functional been reported as 2%.°°
assays are performed for the quantitative measurement of the Acquired antithrombin deficiency is common, arising
free functional PS level based on the ability of PS to serve as from conditions such as decreased synthesis (liver disease or
a cofactor for the anticoagulant effect of APC inhibition of L-asparaginase treatment), consumption (thrombosis, dissem-
factor Va. This inhibition is reflected by the prolongation of inated intravascular coagulation [DIC], surgery) or loss (pro-
the clotting time of a system that is enriched with factor Va, a teinuria in renal disease), and continuousheparin use (see
physiologic substrate for APC. Table 28—5).*°
Based on the immunologic and functional assays described Mild decreases can be seen with pregnancy, oral contra-
earlier, three subtypes of PS deficiency have been noted ceptive use, and colitis. Before a diagnosis of antithrombin
(Table 28-6). The most common, type I deficiency, is associated deficiency is rendered in any patient with these conditions, the
with decreased activity due to a low antigenic level of total and test should be repeated once the condition is no longer pre-
free PS levels. In type Ila deficiency, total PS is normal; how- sent. Confirmation of a hereditary antithrombin deficiency
ever, the free PS antigen levels are decreased and the functional may require documentation of a deficiency in a blood relative.
PS activity is low. In type IIb deficiency, total and free antigen Various methods have been described for the measurement
levels are normal, but activity level of PS is low. of AT. Immunologic assays such as radial immunodiffusion are
Acquired PS deficiency can be seen in liver disease, available that measure the antigen levels but cannot be used to
DIC, pregnancy, oral contraceptive use, vitamin K deficiency, detect dysfunctional molecules. To detect the qualitative abnor-
during an acute thrombotic event, and in therapy with mality of the AT molecules, the functional assay is preferred. A
warfarin and L-asparaginase (see Table 28-5). C4bBP is an known amount of thrombin is added to the patient’s plasma con-
acute-phase reactant and its increase will proportionately bind taining AT in the presence of heparin. The aliquot is removed
free PS; therefore, the amount of free PS may decrease in and added to a specific synthetic chromogenic or fluorescent
inflammatory conditions such as systemic lupus erythemato- thrombin substrate and the released compound is measured
sus (SLE), inflammatory bowel disease, pregnancy, and spectrophotometrically or via fluorometer, respectively.
acquired immunodeficiency syndrome (AIDS). Because PS Based on immunologic and functional assays, two types
of AT deficiency can be distinguished phenotypically. In the
common type I deficiency there is a proportionate decrease in
both the functional and antigen levels because the deficiency
is due to a decreased amount of a functionally sound molecule
(quantitative defect). Approximately 80 distinct mutations
have been described in patients with type I deficiency, and
Subtypes Total PS Free PS PS Activity most are a result of genetic mutations producing silent
alleles.*'** Type IL deficiency shows a more pronounced
I Low
Ila Normal
decrease in the functional level with a relatively normal anti-
IIb Normal gen level (qualitative or dysfunctional defect) because the
molecule is created but does not function properly. Type II is
672 Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy
associated with point mutations and can be further subdivided At present, the PCR test for G20210A mutation is avail-
into three subtypes: those characterized by abnormalities able in clinical laboratories. In this test, DNA is isolated from
primarily affecting (1) the serine protease inhibition site for blood mononuclear cells. Using allelic discrimination method-
thrombin/Xa, (2) the heparin-binding site, and (3) those with ology by PCR the sample DNA is analyzed, allowing specific
effects on both protease inhibition and heparin binding. The genotypes to be identified (normal, heterozygous, or homozy-
classification of hereditary AT deficiencies is summarized in gous). Among the Dutch population, the prothrombin gene
Table 28-7. mutation is the second most common genetic defect (the most
Based on functional assays, the prevalence of thrombo- frequent being the factor V Leiden mutation). Because of the
sis appears to be different in heterozygous patients with high prevalence of APC resistance (FV:Q**) and of the pro-
type II defects. Individuals with heparin-binding site defects thrombin gene mutation (G20210A), combinations of genetic
have infrequent thrombotic episodes while, in contrast, those defects are relatively common. Patients with both mutations
with thrombin-binding site defects sustain venous thrombosis (double heterozygotes) may have a high risk of thrombosis.
as often as type I patients. However, more studies are needed to improve our understand-
ing of the ethnicity-specific variability of the factor V Leiden
and prothrombin gene mutations.
Prothrombin Nucleotide G20210A
Mutation Hyperhomocysteinemia
Prothrombin (coagulation factor II) is a vitamin K—dependent
Hyperhomocysteinemia (an elevated plasma level of homo-
factor that is converted to thrombin by factor Xa in the pres- cysteine) is a risk factor for the development of early ather-
ence of factor Va, Ca**, and phospholipids. The combination
osclerotic vascular disease and venous thrombosis. Some
of factor Va, Xa, Ca**, and phospholipids is called the pro-
investigators include hyperhomocysteinemia under acquired
thrombinase complex. It is a macromolecular enzyme com-
disorders, whereas recent studies suggest that these should
plex in which prothrombin is a substrate and thrombin the
be included among the inherited causes of thrombophilia.
product. Hereditary disorders of prothrombin synthesis may
Homocysteine is an amino acid derived from metabolic
result in quantitative defects (decreased production) and qual-
conversion of methionine. It is usually metabolized within the
itative defects (dysfunctional prothrombin molecule). Clini-
cells to methionine (via the remethylation pathway) and to
cally, both of these defects may result in a bleeding diathesis.
cysteine (via the transulfuration pathway). Remethylation to
A 21-kb gene present on chromosome 11 codes for the
methionine involves two pathways. In the first remethylation
prothrombin molecule. This gene consists of 14 exons and
pathway, homocysteine accepts a methyl group from betaine.
13 introns. A mutation in the prothrombin gene has been
In the second remethylation pathway, homocysteine accepts a
described recently,** revealing the presence of a single gua-
methyl group from 5-methyltetrahydrofolate, with vitamin
nine (G) to adenine (A) mutation at the nucleotide position
B,, acting as a cofactor. This pathway is catalyzed by methio-
20210 to be associated with an elevated prothrombin level
nine synthase. The transulfuration pathway is catalyzed
and an increased risk of DVT.
by cystathione B-synthetase with vitamin B, as a cofactor*
A heterozygous G20210A mutation is found in 2% of
(Fig. 28-5).
healthy controls, 6% to 7% of venous thrombosis patients, and
18% of selected thrombophilia patients with a positive family
history. The relative risk for venous thrombosis in heterozygotes
HOMOCYSTEINE
is approximately threefold.“ Carriers of this mutation have
higher plasma prothrombin levels than those with a normal
20210 genotype. A high level of prothrombin alone is not a
reliable marker of disease predisposition, whereas detection of eet
5,10-methylene
tetrahydrofolate
AT-Heparin
Subtypes AT Antigen Cofactor Progressive AT Figure 28-5 = Homocysteine can be metabolized to methionine
(via remethylation) and to cysteine (via transulfuration). The num-
I Low Low Low bers represent the enzymes involved in the reactions: (7) Betaine
Ila Normal Low Low homocysteine methyltransferase; (2) methionine synthase in the
IIb Normal Low Normal presence of cofactor vitamin B,.; (3) cystathione -synthetase in the
IIc Normal Normal Low presence of cofactor vitamin B,; (4) methylene tetrahydrofolate
reductase (MTHFR).
Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy 673
Hyperhomocysteinemia can be inherited or acquired. factor, after the patient in whom the deficiency was first identi-
Inherited forms have genetic defects that involve (1) the fied in Cleveland in 1955. Patients with factor XII deficiency
remethylation pathway, as a result of a deficiency of methylene have a prolongation of APTT but no bleeding diathesis. Instead,
tetrahydrofolate reductase (MTHFR) and (2) the transulfuration a number of cases with venous thromboembolism and myocar-
pathway, owing to the deficiency of cystathione B-synthetase. dial infarction have been described in factor XII-deficient
The mutation of MTHFR gene (alanine substitution to valine at patients. Mr. Hageman himself died of a PE. Because
amino acid 677) is more common. This mutation can vary from factor XIla is involved in activating plasminogen, the throm-
1.5% to 15%, depending on the population studied.*° The fre- bophilic tendency in factor XII deficiency patients has been
quent causes of acquired hyperhomocysteinemia are secondary attributed to reduced plasma fibrinolytic activity. The incidence
to deficiencies of folate, vitamin B,,, and/or vitamin B,, which of thrombosis in patients with heterozygous or homozygous
are cofactors in homocysteine metabolism. factor XII deficiency in the absence of other thrombotic risk
Possible mechanisms by which hyperhomocysteinemia factors is uncertain and, thus, an increased thrombotic risk asso-
acts as a thrombogenic and atherogenic risk factor include ciated with factor XII deficiency remains to be well defined.
vascular smooth muscle proliferation, inhibition of endothe-
lial cell growth and intimal thickening, activation of factor V,
and inhibition of PC activation. Severe hyperhomocysteine- Heparin Cofactor II Deficiency
mia is usually secondary to genetic defects and characterized
Heparin cofactor II is a single-chain glycoprotein present in
by mental retardation, premature atherosclerosis, venous
human plasma that inhibits only thrombin but does not inhibit
thromboembolism, and skeletal abnormalities. Mild to mod-
other coagulation factors. The affinity of heparin cofactor II
erate hyperhomocysteinemia can be secondary to genetic
for heparin is lower than that of AT and requires a higher con-
or acquired conditions and is an independent risk factor
centration of heparin to affect in vitro testing (more than the
for stroke, MI, and peripheral vascular disease. Levels of
therapeutic concentration, i.e., more than | U/mL of heparin).
homocysteine can be measured by high-pressure liquid chro-
Therefore, heparin cofactor II probably plays an insignificant
matography. A PCR-based genetic test is available to detect
role in the clinical setting.’ On the contrary, the activity of
the common Ala 677 Val mutation in the MTHFR gene.
heparin cofactor II can be increased several-fold by dermatan
«
sulfate, which has no effect on the activity of AT. The clinical
manifestations of hereditary heparin cofactor II deficiency
Tissue Factor Pathway Inhibitor Deficiency
range from an absence of symptoms to arterial and venous
Tissue factor pathway inhibitor (TFPI) may play a significant thromboses. More studies are needed to determine the signif-
role in preventing thrombus formation; however, deficiency icance of this deficiency.
of this inhibitor is yet to be associated with thromboembolic
disease. Evidence suggests that TFPI neutralizes two pro-
teases simultaneously, and the inhibitory actions of TFPI Dysfibrinogenemia
occur through three domains.*’ The first ___domain inhibits Dysfibrinogenemia is defined as a condition in which there is
factor VHa—tissue factor (TF) complex, whereas the second a structural or functional abnormality of the fibrinogen mole-
___domain inhibits factor Xa. The function of the third ___domain is cule, or both. If congenital, the inheritance is autosomal dom-
unclear. TFPI can be found in endothelial cells, plasma, and inant. The clinical presentation is variable and can include
platelets. The endothelial cell pool represents the majority of bleeding or clotting tendencies. Sixty percent of the patients
the TFPI. Most of the plasma pool is complexed with various are asymptomatic, a bleeding diathesis can be seen in 20% of
lipoproteins, whereas only 10% of the TFPI is present as a cases, and 20% of cases may present with recurrent arterial or
free form and is biologically active. The concentration of venous thromboembolism.*!
TFPI is increased in plasma in patients receiving heparin A number of functional and biochemical defects of the fib-
infusion. Recombinant TFPI is now available and has been rinogen molecule have been described. These include an abnor-
evaluated in a few studies, with results showing a possible role mality in binding of thrombin to fibrinogen, the release of
of TFPI in the interruption of thrombus formation. Whether or abnormal fibrinopeptides, defective fibrin polymerization and
not there is a congenital deficiency of this inhibitor leading to cross-linking, decreased activation of plasminogen, and resis-
thromboembolic conditions is not yet known.” tance to lysis by plasmin. These abnormalities are reflected in
Recent evidence suggests that autoantibodies to tissue fac- the routine coagulation assays as prolongation of PT, APTT,
tor pathway inhibitor (TFPI) and/or antiphospholipid antibodies fibrinogen, reptilase time, and thrombin time. The fibrinogen
(aPL) may contribute to upregulation of the tissue factor (TF) concentration via functional assay is low, but is normal when
pathway of blood coagulation and the development of throm- measured immunologically. An important point to consider is
botic complications in the antiphospholipid syndrome (aPS).”° that the abnormal fibrinogen may not be incorporated into the
clot, and the soluble molecules that remain in the serum can be
mistaken for fibrinogen degradation products in some assays,
Factor XII Deficiency
leading to the misdiagnosis of active fibrinolysis and thus DIC.
Factor XII is one of the contact factors that initiates the intrinsic Dysfibrinogenemia can be seen in acquired conditions
pathway of coagulation in vitro. It is also known as Hageman such as severe liver disease, paraneoplastic syndromes, and
674 — Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy
with paraproteinemias.° Acquired dysfibrinogenemia is vascular thrombosis and pregnancy morbidity.” The thrombosis
rarely symptomatic and thrombotic complications have not may be arterial or venous and tends to recur in the same site.
been described. aPL syndrome requires the combination of at least one clinical
and one laboratory criterion (Table 28-8). aPL syndrome is con-
sidered secondary when it occurs in association with another
Elevated Plasma Factor VIII Coagulant disorder and primary when unassociated with any identified
Activity underlying disorder. Secondary aPL syndrome is most fre-
quently associated with systemic lupus erythematosus (SLE) or
The elevated plasma factor VII coagulant level (elevated func-
other autoimmune diseases, but has occasionally been seen
tional and antigenic levels) is considered an independent risk
after exposure to certain pharmaceuticals (such as phenoth-
factor for thrombosis.**>’This abnormality could not be attrib-
iazines, quinidine, hydralazine, procainamide) and in patients
uted to inflammation, because fewer than 10% of the patients
with underlying malignancies. The overall prevalence of aPL
with factor VIII:C levels of greater than 150% had elevations
syndrome is difficult to determine because of variations in
in an acute phase reactant (such as C-reactive protein, fibrino-
laboratory techniques and diagnostic criteria. In spite of this,
gen, and erythrocyte sedimentation rate [ESR]).°*°°
the prevalence of aPL syndrome in SLE patients has been
Studies have demonstrated that patients with factor VII
examined by many researchers. The frequency of lupus
greater than 150% had an increased risk of thrombosis when
anticoagulant (LA) and anticardiolipin (aCL) in nearly
compared to patients with factor VIII less than 100%.°°' One
2000 SLE patients reported in the literature is 31% and
study has demonstrated that an elevated factor VIII level may
40%, respectively. Patients with SLE and aPL antibodies
also be a strong thrombotic risk factor in the black popula-
had a 30% to 40% risk of thrombosis.”
tion. The clinical utility of routinely measuring factor VIII
The term /upus anticoagulant refers specifically to the lab-
levels in patients with thrombosis remains to be determined.
oratory in vitro phenomenon of prolongation of phospholipid-
dependent coagulation assays such as the APTT in the
Lipoprotein a and Thrombosis absence of an attributable factor deficiency. Laboratory
evidence of aPL antibodies can be in the form of either
Lipoprotein a (Lpa) represents a low-density lipoprotein immunologically demonstrated aPL antibodies or, more fre-
(LDL)-like particle having a protein moiety apoprotein B-100 quently, LA, or both.
linked by a disulfide bridge to a glycoprotein called apolipopro- The presence of aPL antibodies, either demonstrated by
tein a (Apo a). Elevated levels of Lpa are under genetic control LA or detected immunologically, is not sufficient for the diag-
and have been recognized as an atherothrombogenic factor. But nosis of aPL syndrome. Additional clinical features must also
the underlying mechanisms for this pathogenicity are not well be present to diagnose aPL syndrome (see Table 28-8) because
understood. There is a structural homology of Apo a with plas- aPL antibodies alone may not carry a similar clinical signifi-
minogen, and laboratory studies have shown that Lpa inhibits cance. In particular, infection-associated aPL antibodies are
fibrinolysis. It effectively competes with plasminogen for bind- usually transient and are rarely associated with thrombotic
ing to fibrin or endothelial cells. It also binds t-PA and may also complications.”°’’ Most aPL antibodies detected in children
stimulate the release of PAI-1 from endothelial cells. Studies do are of this type.
show evidence of increased levels of Lpa in atherogenesis/
thrombogenesis°* and an association with coronary heart dis- HISTORY OF ANTIPHOSPHOLIPID SYNDROME
ease and stroke. However, its role in venous thromboembolism The term antiphospholipid antibody is a misnomer. A more
is not established.°’ accurate term would be antiphospholipid-protein antibodies
because the antibodies have specificities for certain proteins
when those proteins are bound to phospholipids or other sur-
Other Coagulant Factors Associated
faces such as microtiter plates. “Lupus anticoagulant” is also
with Thrombosis not an optimal term because the anticoagulant phenomenon
High levels of factor XI, IX, fibrinogen, IL-8, and TAFI and seen in vitro is not always associated with SLE and causes
low levels of plasma fibrinolytic activity have been consid- thrombosis in vivo. However, for historical reasons, the terms
ered to be associated with venous thrombus. Assays for some aPL antibody and LA are well embedded in the literature and
of these proteins are not widely available, and the clinical util- clinical use.
ity of these measurements is currently uncertain.°*” In 1963, Bowie and colleagues published a paper calling
attention to a syndrome they had observed in patients with
SLE.” The syndrome, still unnamed, consisted of recurrent
venous or arterial thrombosis accompanied by paradoxical
Acquired Thrombotic Disorders
prolongation of clotting times in phospholipid-dependent
Lupus Anticoagulant/Antiphospholipid clotting assays. This phenomenon of prolonged clotting times
Syndrome in the absence of specific factor inhibitors or deficiencies came
to be known as lupus anticoagulant. As in many SLE patients,
Antiphospholipid (aPL) syndrome is one of the acquired throm- some of these patients were observed to have a false-positive
botic disorders. The clinical features of aPL syndrome include venereal disease research laboratory test (VDRL). The VDRL
Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy 675
aPL syndrome is present if at least one of the clinical criteria and one of the laboratory criteria are present.
Clinical
is a serologic screening test for syphilis that uses a mixture of antibodies bind prothrombin.*°** These antibodies usually do
cardiolipin (an acidic phospholipid found in beef heart), not deplete plasma levels of 82GPI and prothrombin because
lecithin, and cholesterol. Given the coexistence of the LA they bind only when these antigens are attached to phospholipid
phenomenon and the antiphospholipid antibodies demon- surfaces. Occasionally, patients with SLE have reduced pro-
strated by a positive VDRL, it seemed likely that LA antibod- thrombin levels and a resulting hemorrhagic diathesis. Other
ies were nonspecific antibodies that reacted with both platelet phospholipid-binding proteins may also be targets of thrombo-
and reagent phospholipids. The interference of such antibod- genic aPL antibodies.
ies with phospholipid-dependent coagulation reactions could
prolong clotting times. How these antibodies promote coagu- MECHANISM OF THROMBOSIS
lation in vivo was not understood and is still unproven, but It would be reasonable to question whether aPL antibodies are
eventually the concept of an aPL syndrome proposed by causative in aPL syndrome or merely an epiphenomenon that is
Harris in 1987 took hold and the terms are still with us.” useful for diagnosis. It is likely that multiple disturbances of
hemostatic mechanisms are required to result in thrombophilia.
IMMUNOLOGY One could assume that, if they were part of the etiology, they
In 1983, Harris and colleagues developed a solid-phase radioim- would affect phospholipid-dependent reactions in vivo and in
munoassay (RIA) for aCL antibodies.*° Recall that cardiolipin is vitro. In vitro evidence of such effects have been investigated
one of the components of the VDRL. High titers of these anti- and include inhibition of activated protein C, inhibition of
bodies correlated well with the presence of LA, thrombosis, and antithrombin-dependent anticoagulant mechanisms, inhibition
thrombocytopenia. The aCL assay, now usually done via ELISA of fibrinolysis, induction of increased soluble tissue factor, inhi-
rather than via RIA, is the most frequently used immunologic bition of prostacyclin secretion, promotion of platelet activation,
assay for the identification of aPL antibodies.*' Over the years it interference with the anticoagulant properties of 82GPI, and
has been shown that aPL antibodies can be detected by using displacement of annexin V from trophoblast.*’ Annexin V is a
various phospholipids, including phosphatidylinositol, phospha- protein that has potent anticoagulant properties in vivo and that
tidic acid, and phosphatidylserine. In addition, investigators may modulate thrombosis on the surfaces of cells lining placen-
have shown that LAs and aCL antibodies in a given patient can tal and systemic vasculatures. Interference with the function of
often be separated. In 1990, three groups working independently annexin V may play a role in aPL syndrome in which there is
found that the majority of aCL antibodies bind phospholipid- recurrent pregnancy loss.”
bound ,-glycoprotein I (B2GPI).*°* B2GPI, also known as
apolipoprotein H, is a 50-kD plasma protein present at a concen- THE LABORATORY’S CONTRIBUTION
tration of approximately 200 mg/mL that appears to have anti- TO THE DIAGNOSIS OF aPL SYNDROME
coagulant properties, although its physiologic role is uncertain. Overall, the presence of LA is a better predictor of thrombosis
Some LAs have also been shown to bind in a B2GPI-dependent and pregnancy loss than the presence of high-titer anticardi-
manner.® In addition, a large proportion of LAs and some aCL olipin antibodies.’' However, some patients with aPL syndrome
676 Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy
if This Screening Assay Is Positive ... One ofThese Techniques Should Be Used asa y Contiemainry Test
Eon assays detect eSa eioniea Runestime. CG assays demonstrate whether or not the prolongation
is decreased by phospholipids capable of neutralizing the LA antibody.
Source: Horbach, DA, et al: Lupus anticoagulant is the strongest risk factor for both venous and arterial thrombosis
in patients with systemic lupus erythematosus. Thromb Haemostas 76:916, 1996.
do not have both LA and aCL antibodies. Therefore, the labora- can be in the form of excess phospholipids (dRVV con-
tory diagnosis of aPL syndrome should include both coagulation firm), hexagonal (II) phase phospholipids (Staclot test) and
assays for LA and immunologic assays for aPL antibodies. platelets or platelet vesicles (platelet neutralization test).
Some authors have suggested that a broad panel of aPL antibod- The excess phospholipid binds and neutralizes the LA,
ies should be included when investigating aPL syndrome as a leaving enough phospholipid to provide a surface on which
possible cause for recurrent fetal loss.”? In the following discus- coagulation reactions can proceed.”*” A confirmatory
sion, we concentrate on the hematology laboratory’s role in assay that corresponds to the initial type of assay and mix-
working up a suspected case of aPL syndrome. ing study that were positive should be used. Solid-phase
As in most diagnostic algorithms, initial assays with broad assays for aPL antibodies, such as the aCL assay, should
sensitivity are performed and, if positive, they are followed by not be considered confirmatory for LA activity.
confirmatory assays that are more specific (Table 28-9). The 4. In addition, the absence of clinical or laboratory evidence
Subcommittee on Lupus Anticoagulant/Antiphospholipid ofa specific inhibitor of any one coagulation factor may
Antibody of the Scientific and Standardization Committee of also be necessary, because specific factor inhibitors can
the International Society on Thrombosis and Haemostasis cause mixing studies to remain uncorrected. The clinical
has recommended criteria, paraphrased here, for the diagnosis presentation is very important and may be informative.
of LA’ (Table 28-10). Patients with factor inhibitors have a bleeding diathesis
rather than thrombosis.
1. Prolongation of at least one phospholipid-dependent clot-
ting assay. Examples of such assays using a lupus-sensitive These steps can be carried out in various ways, using various
reagent include the APTT, dilute PT (dPT), kaolin clotting discrete assays or by using integrated systems for the diagno-
time (KCT), and the dilute Russell’s viper venom time sis of LA that incorporate an initial assay, mixing study, and a
(dRVVT) and colloidal silica clotting time.°*°’ Because of
the heterogeneity of these antibodies, no single screening
test will detect all of them. A minimum of two “screening” ‘Table 28-10 Criteria for Lupus —
assays should be available in a laboratory offering LA test-
_ Anticoagulants —
ing. Having three screening tests available is optimal.
NO. Evidence of inhibition of clotting is demonstrated by mix-
A tested sample should show each of Ieee
ing studies. This involves mixing the patient’s plasma and
. Prolongation of at least one phospholipid-dependent clot-
pooled normal plasma and repeating the phospholipid- ting test
dependent clotting assay in which the prolongation of clot- . Evidence of inhibitory activity shown by the effect of
ting time was observed. If the prolongation disappears patient plasma on pooled normal plasma
(“corrects”) with mixing, this usually indicates that there is . Evidence that inhibitory activity is dependent on phospho-
lipid
a factor deficiency.
. Satisfactory exclusion of another coagulopathy that could
3. If the results of the mixing study suggest an LA, then evi- “give similar laboratory results
dence of phospholipid dependence is required for confir-
Source: Horbach, DA, et al: Tae mitecieeent: is the strongest
mation. Confirmatory assays demonstrate phospholipid
risk factor for both venous and arterial thrombosis in patients with
dependence by demonstrating a reversal of the LA effect systemic lupus erythematosus. Thromb Haemostas 76:916, 1996.55
when excess phospholipid is added to the test mixture. This
Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy 677
procedure that confirms phospholipid dependence into a single laboratory diagnosis of an LA as depicted in Figure 28—7.'°!
kit. Table 28-9 lists Screening and confirmatory assays that Many, but not all, patients with a LA will have a prolonged
have been used are listed in Table 28-9, and an approach to the APTT. Some APTT assays are more sensitive to LA than oth-
diagnosis of LAs is illustrated in Figure 28-6.” ers and, therefore, they vary in their value as initial tests.!0*'°°
In addition to the requirements for diagnosis already Remember that prolongation of the APTT also occurs with
mentioned, several other elements are crucial to arriving at heparin therapy, heparin contamination, insufficient volume
the correct diagnosis in cases suspicious for aPL syndrome, as of blood for the amount of anticoagulant in the sample tube
follows: (short draw), DIC, factor deficiencies, and specific coagula-
tion factor inhibitors. Clinical history elucidates the cause of
1. Care should be taken to use properly prepared platelet-poor
some prolonged APTTs, and depending on the clinical situa-
plasma in order to avoid false-negative results caused by
tion other assays such as the thrombin time and fibrinogen
the neutralization of LA by platelet membrane phospho-
level, may be employed to identify heparin contamination or
lipids.'°°
DIC. If the prolonged APTT is unexpected or not explained
2. Immunologic assays for antiphospholipid antibody should
by the causes mentioned earlier, the laboratory investigation
also be done.
for LA is warranted, especially in a patient with a history of
3. Testing should be repeated after a minimum of 6 weeks to
thrombosis or other findings indicating aPL syndrome.
demonstrate persistence of the LA. Transient aPL antibod-
If the APTT is prolonged and investigation of a possible
ies do not appear to be associated with the clinical compli-
LA is indicated, the next step is to perform an APTT using a
cations of aPL syndrome.
1:1 mix of the patient’s plasma and normal pooled plasma. If
4. Finally, the diagnosis of aPL syndrome should make sense
the APTT does not correct, then either antibody to a phospho-
based on the patient’s history and clinical findings.
lipid in the test system or antibody to a specific coagulation
Laboratory testing for LA is currently hindered by the lack of factor is responsible. Correction can be defined in different
standardized controls. Laboratories have no option other than ways, but probably the most useful approach is to consider an
to use patient plasmas that have proved positive for LA that are APTT corrected when it returns to within 5 seconds of the
available commercially. Trials of potential standardized con- APTT of the pooled normal plasma.'™ If the APTT does not
trols containing monoclonal anti-B2GPI and antiprothrombin correct in the 1:1 mix, then confirmatory testing is indicated,
antibodies are ongoing. as discussed earlier.
If the APTT does correct, then the abnormality is
A WALK THROUGH THE ALGORITHM attributable to coagulation factor deficiency or a time- and
In the hematology laboratory, detection of aPL antibodies temperature-dependent antibody. Specific coagulation fac-
usually begins with the APTT. In this discussion, we trace the tor inhibitors are usually time and temperature dependent.
It has also been observed that up to 30% of LAs are time
dependent.'*'° An incubated 1:1 mix allows for detection
PROLONGED APTT of a time- and temperature-dependent inhibitor. Interpreta-
tion of incubated mixing studies can be challenging. The
incubated mix should be used only if the immediate mix
Explore | No [Thrombin
time ].Y°S | Fibrinog corrects. The incubated mix helps to detect inhibitors, but
et MNES _ measurable? _ >100 cannot be used to determine their etiology. The use of
inspecmen} Lo unmixed incubation controls, as shown in Figure 28-7, is
suggested.'°' Assays for specific coagulation factors can
Explore detect factor deficiencies, and serial dilutions are helpful
hypofibrinogenemia_ in detecting antibodies to specific coagulation factors.
: Incubate
Incubate Patient
Perform APTT
and compare results
Figure 28-7 @ Incubated mixing study can be performed as shown in this figure. For details, refer to the accompanying text. (Courtesy of
John D. Olson, MD; PhD, Department of Pathology, University of Texas Health Science Center, San Antonio, TX.)
anticoagulant for the first few days.'** The early warfarin- oral contraceptives prescribed in the United States contain
induced reduction of functioning protein C in the presence of low-dose estrogen. However, even the use of low-dose estro-
increased thrombin generation seen in HIT, puts the patient at gen contraceptives confers a fourfold increased risk of throm-
very high risk for this and other thrombotic complications. bosis when compared with nonusers. The risk may become
LMWH is also not recommended for the treatment of significant if the woman also has an inherited risk factor for
HIT. Although it is less likely to induce HIT once a patient has thrombophilia. The exact mechanism of the prothrombotic
a heparin-induced antibody, exposure to LMWH carries a risk state is not well understood, but it is thought that these med-
of thrombosis.!?’ ications may cause an acquired APC-R-like state.'*”
Anticoagulants available for use in HIT patients include
heparinoids such as danaparoid and direct thrombin inhibitors
such as hirudin, argatroban, and bivalirudin, which are dis- Thrombosis and Nephrotic Syndrome
cussed subsequently. The adjunctive use of medications that Nephrotic syndrome is characterized by heavy proteinuria
limit platelet function is also under investigation. Examples (more than 3.5 g/day), hypoalbuminemia, severe edema, and
include glycoprotein IIb/IIIa (GPIb/IHa) inhibitors such as hyperlipidemia and thrombosis of both venous and arterial
GPI 562 and ADP receptor antagonists, clopidogrel and systems may be seen. The renal vein is the most common
ticlopidine.'*°
site involved and is present in approximately one-third of
The current recommendation for HIT patients is therapy patients.'*° The incidence of thrombosis varies and correlates
with an alternate anticoagulant (direct thrombin inhibitors) to be
with the severity of the disease. Depending on the complex
followed by a transition to warfarin. The warfarin should be interaction between hepatic protein biosynthesis and the sever-
started at low doses and given concurrently with direct throm- ity of renal disease, the following changes of coagulation
bin inhibitors for 5 days until a therapeutic INR is achieved for parameters can be seen: AT levels are often decreased; PC and
2 days.'* Care should be taken in monitoring INR during this S are increased; and factors V, VII, VIII, X, and XIII are
period, as direct thrombin inhibitors may prolong the INR. increased, whereas factors XI and XII are decreased. In addi-
Obtaining a chromogenic X assay may help in transitioning a tion, platelet hyperactivity can also be seen in these patients.
patient from a direct thrombin inhibitors (DTI) to warfarin.
Warfarin is typically continued for 3 to 6 months; however, the
optimal duration of anticoagulation needs further study. Thrombosis and Medications
L-Asparaginase, used as a part of induction chemotherapy for
acute lymphoblastic leukemia (ALL), may cause venous
Other Acquired Conditions Associated thrombosis (intracranial thrombosis is observed most fre-
with Thrombosis quently). The incidence of venous thrombosis has been esti-
Thrombosis with Pregnancy and Use mated to be 1.2%.'*' This drug causes a significant reduction
in hepatic synthesis of both procoagulant (factors V, VU, VIII,
of Oral Contraceptives
IX, X, XI, and fibrinogen) and anticoagulant proteins (AT, PC,
There is a sixfold increased risk of thromboembolism during and PS). The alteration in the hemostatic balance in a particu-
pregnancy. The puerperium (6-week period after delivery) is lar patient determines the risk of thrombosis or bleeding. There
associated with a higher risk of thrombosis than pregnancy. may be a prolongation of PT, APTT, and thrombin time. These
The presence of coexistent inherited thrombophilia during transient abnormalities resolve within | to 2 weeks after ces-
pregnancy represents a significant added risk. In a Swedish sation of the drug. Recent studies indicate that, in children
population, approximately 50% of women with venous with ALL, L-asparaginase is a risk factor for thrombosis, but
thrombosis had APC-R, with 65% to 75% of the thrombotic those who develop thrombi have additional risk factors. Some
events occurring during pregnancy.'*>'*° Thrombosis during consideration should be given to the evaluation of other risk
pregnancy is attributable to the following conditions: factors for thrombophilia prior to beginning the medication. If
other risk factors are present, prophylaxis may be indicated.
1. Venous stasis in the lower extremities, caused by the gravid An increased risk of thrombosis has also been described
uterus. in women with breast cancer who are receiving chemother-
2. Trauma to pelvic veins during delivery. apy.'” The basis of thrombogenicity among these patients is
3. The placenta is rich in tissue factor and also releases plas- not well understood, and possible causes include low grade
minogen activator-2, which reduces fibrinolytic activity. DIC, impairment of vitamin K metabolism, and altered
4. Increased levels of most coagulation proteins (fibrinogen hepatic protein synthesis.
can be increased to 600 mg/dL).
5. A significant decline in plasma levels of free and total PS.
6. Elevated D-dimer levels during the third trimester. Thrombosis in Cancers
and Other Conditions
Although PC and AT levels remain normal during pregnancy,
both are decreased in preeclampsia. Patients with cancer are at high risk of VTE when compared to
The risk of venous or arterial thrombosis secondary noncancerous patients. The occurrence of thrombosis in patients
to oral contraceptives is related to estrogen dose. Most with malignancy can be caused by cancer itself or occur
682 Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy
Factor V dee = se ae
1. Age at onset
Prothrombin gene mutation sheets
. Any underlying associated risk factors
PC deficiency** ap ae
. Number of events (initial or recurrent)
PS deficiency ++
. Site of thrombosis
AT deficiency aimets
. Any documented evidence of pulmonary embolism or DVT
Hyperhomocysteinemia af
. Whether recurrent thromboembolism occurred despite
Lipoprotein a =
therapeutic anticoagulation
Antiphospholipid syndrome 3Par
. Any history of recurrent fetal loss, MI, or stroke
Heparin-induced sear
. Any medications that can lead to thrombosis (oral contra-
thrombocytopenia
ceptives, chemotherapy)
. Family history of venous thromboembolism *Women who smoke and take oral contraceptives have an increased
risk of myocardial infarction.
**A slight increased risk of stroke has been reported.
4 Table 28-14
1 Suggested Evaluation — Specific laboratory tests of interest in patients with
-.__. Criteria for. Inherited thrombophilia have been addressed in other sections of this
_ Thrombophilia chapter. There are, however, some issues that deserve consid-
eration when approaching the laboratory evaluation of a
1. Thrombosis at any age especially in younger patients* patient who has suffered from thrombosis. They are summa-
2. Recurrent thrombosis rized in Table 28-17 and addressed briefly below.
3. Significant family history of thrombosis
4.Thrombosis atunusual sites (other that ee veins of legs)
*Studies have Onset that the yield in feeultnherlied risk Complete History and Physical
factors for thrombophilia in the aged occurs at the same rate and
would have the same value in guiding clinical management.
Examination
A complete personal, family, and drug history is extremely
important in evaluating patients with acute, recurrent, or
remote thrombosis. The patient should be carefully questioned
about all the possible risk factors for thrombosis. A significant
Table 28-15 Testing for Inherited | family history of thrombosis strongly suggests the possibility
: _ Thrombophilia- coe of inherited thrombophilia. If the patient is female, history of
oral contraceptive use and obstetric history is important. Pres-
Plasma Gaaeatgion Screening ence of constitutional symptoms, hemoptysis, melena, or hema-
Pal turia may suggest underlying occult malignancy. Recurrent
APTT
Thrombin and reptilase times thrombosis despite anticoagulation therapy may suggest under-
lying malignancy. Atherosclerotic vascular disease and renal
Anticoagulant System
APC resistance assay (APTT-based assay)
Protein C (functional and antigenic)
Protein S (functional and antigenic)
AT (functional and antigenic)
| Table 28-17 Issues in Laboratory |
- Testing in Patients —
Fibrinolytic System
Fibrinogen (functional and antigenic)
(an Thrombosis
Factor XII activity
¢ Ensure a complete history and physical examination 1s
Plasminogen (functional)
obtained.
Genetic Tests * Careful selection of tests during the acute event.
Factor V (FV:Q°°°) Leiden mutation * Consider conditions that can interfere with test results.
Prothrombin gene mutation (nucleotide G20210A) ¢ Test in the appropriate clinical setting.
MTHFR gene mutation ¢ Use functional assays when possible.
¢ In arterial thrombosis, consider the additional evaluation of
Additional Tests hyperhomocysteinemia and lipoprotein a.
Homocysteine level ¢ Repeat testing prior to diagnosis.
Lipoprotein a level ¢ Ensure that the yield in performing testing is high, making
Factor VIII activity evaluation worthwhile.
684 Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy
disease (nephrotic syndrome) can lead to arterial or venous TESTING DURING THE ACUTE EVENT
thromboembolism. Thrombotic events, whether on the arterial or venous side of the
Ethnic background is also important in evaluating patients circulation, are capable of consuming naturally occurring coag-
with inherited thrombophilia. The factor V Leiden mutation is ulation inhibitors (PC, PS, AT) that are of interest for testing. In
common in people of European descent and extremely rare in addition, a number of these components are either positive or
African American, Native American, or Asian populations. negative acute phase reactants. Anticoagulant therapy also
Physical examination should be specifically directed to exam- affects these concentrations variably. For these reasons, testing
ining vascular system, skin, extremities, heart, chest, and during the acute event and during anticoagulant therapy will
abdomen. produce both false-positive and false-negative results.
development of a thrombus, regardless of the clinical setting, strategies will also evolve that will be directed by one or more
requires the accumulation of more than one inherited or of the thrombophilic risk factors.
acquired risk factor. Venous thrombosis in almost any clinical
setting would merit a comprehensive evaluation. For some
time, it was thought that evaluation for inherited throm- Anticoagulant Therapy
bophilia should be limited to the “young” patient with throm-
Patients with venous thrombosis are most frequently treated
bosis. Studies have demonstrated that the yield in detecting
with unfractionated heparin acutely, followed by long-term
inherited risk factors for thrombophilia in the aged occurs at
oral anticoagulants.'*"'*° This is done to prevent propagation
the same rate and would have the same value in guiding clin-
of the thrombus, to reduce the risk of embolus, and to allow
ical management.
for natural resolution. Because of difficulties with appropriate
monitoring, the use of LMWH is becoming more popular;
Functional Assays however, these medications are still considerably more
expensive than unfractionated heparin. Patients in a clinical
Many of the molecules of interest in thrombophilia can be setting (i.e., perioperative) with a high risk of thrombosis are
assayed using functional or antigenic assays. Whenever qual- also treated with low doses of anticoagulants to prevent
ity functional assays are available, they should be the first thrombus formation.
assays performed. In general, if the assay is within the refer-
ence range. further testing is not indicated. In contrast, if the
functional assay is below the reference range, evaluation by Unfractionated Heparin Therapy
antigenic assay is needed to determine if a normal amount of
abnormally functioning molecule is being produced. Heparin is an unbranched polysaccharide that is heavily sul-
fated, making it anionic. Commercial preparations are made
by extracting these molecules from bovine lungs or porcine
In Arterial Thrombosis, Consider intestines. Therefore, the heparins that are extracted are vari-
the Additional Evaluation of able in size (4 to 35 kD, averaging 12 to 13 kD). The nature
and degree of sulfation and the size of the molecule influence
Hyperhomocysteinemia and Lipoprotein a
the biologic activity.
The usefulness of testing for thrombophilia in patients with The anticoagulant activity of heparin is to function as a
arterial disease, particularly MI and stroke, continues to be dis- cofactor. When heparin is added to purified activated coagu-
cussed in the literature. A consensus has not evolved regarding lation factors (in the absence of AT), no anticoagulation
the usefulness of laboratory testing in these settings. Risks in occurs. Heparin acts by accelerating the rate at which AT is
relation to hyperhomocysteinemia and Lpa have been well capable of binding to the activated serine protease, irre-
demonstrated. LA and HIT can cause either venous or arterial versibly inhibiting its activity. In the absence of heparin, neu-
thrombosis. The absence of exposure to heparin or related tralization of thrombin or factor Xa will occur in a time frame
compounds eliminates the need to consider HIT. of approximately 15 minutes. In the presence of heparin, the
same reaction occurs more rapidly than it is possible to make
the first measurement.
Repeat Testing Prior to Diagnosis
Virtually all of the assays involved in the evaluation of throm- Administration and Monitoring
bophilia are influenced by a variety of factors in both the prean-
alytic and analytic phases. Because of this, false-positive testing Unfractionated heparin must be administered parenterally,
is acommon problem. Therefore, positive tests should be con- most frequently intravenously but it can also be used subcu-
firmed before the patient is labeled as having a deficiency state. taneously.'*” Clearance of heparin is primarily cellular (retic-
uloendothelial cells) and renal filtration. At usual doses, the
clearance is by the reticuloendothelial cells and provides an
Performing Testing and Making Evaluation average half-life of approximately 60 to 90 minutes. At higher
doses, renal filtration becomes a factor when the reticuloen-
Worthwhile
dothelial system is saturated. This may be an explanation for
These assays are not indicated as a routine preoperative eval- the varying half-life of heparin with increasing dose.
uation if the aim is to detect the risk of thrombosis in the The dose of heparin is determined by the weight of the
absence of a known personal or family history of thrombosis. patient. It is given as a bolus dose followed by continuous IV
Also, there is no valid indication for these tests in an elderly infusion. Owing to the length of the half-life of the drug,
patient who has a risk factor for venous thromboembolism. monitoring should not occur until equilibrium is reached at
Patients who present with thrombosis and who are compre- about 6 hours after beginning or changing therapy. Once
hensively evaluated will have more than a 50% probability of within the therapeutic interval, daily monitoring is sufficient.
having a thrombophilic risk factor identified. The usefulness When used for therapeutic purposes, heparin has a very
of determining thrombophilic risk factors continues to evolve. narrow therapeutic interval. The goal, of course, is to prevent
It is very likely that, as clinical studies evolve, treatment thrombosis without causing hemorrhage. If the drug is given in
686 — Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy
inadequate doses and serum concentrations are below effective disadvantage is the wide variability resulting from the vari-
levels, the consequences can be thrombosis and embolism, ables described earlier.
causing significant morbidity and even mortality. When con- The heparin assay has the advantage of being repro-
centrations are elevated, the anticoagulant properties will cause ducible with more precise therapeutic intervals, making clin-
hemorrhage. It is the obligation of the laboratory to inform clin- ical management less problematic. Disadvantages include the
icians of the method that the laboratory recommends for mon- cost and a more technically demanding assay technique than
itoring heparin and the therapeutic interval. the APTT. Some clinicians are also concerned that a target
Heparin therapy can be monitored by one of two strate- concentration approach may not accurately reflect the state of
gies. The first involves monitoring the effect of heparin on the anticoagulation due to the numerous variables influencing in-
patient’s coagulation system, most frequently via the APTT. dividual patient response.
The second is the target concentration strategy in which the However, with the advent of better defined heparin prod-
concentration of heparin in the blood is determined. Both of ucts with more predictable patient responses, it is likely that
these strategies have been demonstrated to be effective in assays of heparin will move toward the target concentration
monitoring populations of patients receiving unfractionated strategy.
heparin. Unfortunately, when the two are compared directly,
there is very poor correlation between the concentration of
heparin and the resulting APTT. There is no dose-response Low-Molecular-Weight Heparin
relationship between the amount of heparin and the APTT.
Low-molecular-weight heparin (LMWH) is prepared from
The APTT has been used for the monitoring of heparin
unfractionated heparin by fractionation or depolymerization,
since the 1950s. The general “rule of thumb” is a therapeutic
which produces heparins of a more uniform molecular mass
interval of one-and-a-half to two-and-a-half times the upper
(ranging from | to 12 kD; averaging 5 kD) and function. They
limits of the reference range for the APTT. Problems in the
tend to have a greater impact on the inhibition of factor Xa than
monitoring of heparin are based on:
on other enzymes. A significant advantage of LMWH is that it
¢ Variability of heparin is administered subcutaneously and, in many patients, it does
¢ Patient response not require therapeutic monitoring. Dosing is generally once or
e Reagent twice daily and can be administered by patients, similar to the
¢ Instrument'** way diabetic patients can administer their own insulin.
It appears that LMWH will replace unfractionated heparin
It is necessary for each laboratory to determine the respon-
in many clinical circumstances where heparin is required. These
siveness of the instrument/reagent method for the APTT to
products are still not satisfactory for use in extracorporeal circu-
heparin in the patients’ specimens. This process needs to be
lation (cardiopulmonary bypass, extracorporeal membrane oxy-
done each time there is a change in the reagent or instrument
genation). In addition, despite the fact that many patients do not
used in the laboratory.
require monitoring, those of unusually low or high body weight,
The concentration of heparin in the blood can be reflected
patients with renal failure, pediatric patients, and patients with
by assays that take advantage of the neutralization of the activ-
other complicating medical conditions that may influence
ity of thrombin or factor Xa. These assays can use either a clot
heparin metabolism require careful ongoing evaluation. Moni-
endpoint or chromogenic substrate with spectrophotometric
toring of LMWH requires the target concentration strategy
endpoint. The assays provide the enzyme (thrombin or acti-
because the therapeutic concentrations do not influence the
vated factor X), the inhibitor (antithrombin), and a marker
APTT or other coagulation tests in a dose-dependent way. These
(fibrinogen in the plasma or chromogenic substrate). The rate
tests may, however, be prolonged during therapy and this should
or amount of conversion of the substrate is inversely propor-
be noted in testing situations before assigning a diagnosis of a
tional to the concentration of heparin present in the specimen.
hemostatic disorder.
Another assay for quantifying the amount of heparin in
A final comment is warranted regarding some preanalytic
the blood uses protamine sulfate, an antagonist to heparin.
issues in monitoring heparin therapy. A natural inhibitor of
Protamine sulfate binds to heparin in a stoichiometric fashion,
heparin, called PF4, is released from platelets. The presence of
neutralizing its activity. Assays, based on the thrombin time,
platelets in the specimen or the activation of platelets during col-
can be constructed using known concentrations of protamine
lection can thus lead to a false lowering of heparin assayed in
sulfate, thereby judging the amount of heparin by the amount
the specimen. Therefore, care should be taken to collect a high-
of protamine sulfate required to neutralize heparin.
quality specimen, and its preparation should include assuring
All of these approaches to monitoring heparin therapy
that the specimen has a very low platelet count. In addition, if
have their advantages and disadvantages. The dominating
plasma from a heparinized sample sits on cells, the PF4 can neu-
methods being used at the present time are the APTT and the
tralize the heparin, resulting in a falsely decreased APTT.
chromogenic assay, which uses the inhibition of factor Xa.
The APTT has the advantage of being inexpensive (one-tenth
the cost of the Xa inhibitory assay), it can be rapidly and eas-
Oral Anticoagulation
ily performed by many different technologists in the labora-
tory, is available at all hours, and reflects the anticoagulant As described in the synthesis of coagulation factors in the pre-
activity of heparin in the patient’s specimen. Its significant vious chapter, vitamin K plays a key role in the postribosomal
Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy 687
modification of many of the serine proteases. Vitamin K is a When monitoring patients who are taking oral anticoag-
cofactor for a carboxylase that inserts an additional carboxyl ulant therapy, the INR is maintained between 2 and 3. The
group on the glutamic acid residues in the amino terminal end exceptions to this therapeutic interval include patients who
of coagulation factors II, VII, [X, and X in addition to PC and have a complicating lupus anticoagulant in whom a higher
PS. This process is a kinetic one in which vitamin K, through INR may be indicated and patients who have cardiac valves
two enzymatic steps, is recycled and can be reused for the car- in whom the INR is recommended to be 2.5 to 3.5.
boxylation of multiple molecules. The greatest difficulty in managing oral anticoagulation
A coumarin compound, ultimately named warfarin, in patients with thrombosis is the large number of medica-
capable of inhibiting the recycling of vitamin K was identified, tions and other factors that can increase or decrease the
isolated, synthesized, and first administered to patients in 1941. metabolism of warfarin. These drug interactions are important
Warfarin acts by interfering with the recycling of vitamin K considerations for clinicians who are treating patients.
after it has performed its carboxylation function. This makes Recently, it has been thought that warfarin dose-response
warfarin interesting in two respects: (1) Warfarin is not truly an may be regulated at the transcriptional level. The variants in
anticoagulant; it leads to the production of coagulation factors the gene encoding vitamin K epoxide reductase complex
that have reduced anticoagulant function (adding warfarin to a | (VKORC1) has been described that may explain differences
tube of blood does not inhibit coagulation). (2) Warfarin func- in dose requirements among patients of different ethnici-
tions by changing the action of vitamin K in the synthesis of ties.'°! Genetic polymorphisms (i.e., CYP2C9) have been
coagulation factors from a kinetic one to a stoichiometric demonstrated in patients with resistance to warfarin.
one; that is, one vitamin K molecule produces one change in
the glutamic acid molecule rather than a single vitamin K
molecule being able to modify several through a cycling
Alternative Anticoagulants
process. DIRECT THROMBIN INHIBITORS
Because warfarin functions by altering the synthesis of Because some patients cannot tolerate heparin, especially
coagulation factors, the synthetic rate of coagulation factors is those with HIT, additional anticoagulants are currently being
variable. In most patients it takes 5 to 7 days of warfarin ther- used in a limited way. Direct thrombin inhibitors (DTIs)
apy before a stable anticoagulant effect can be achieved. inhibit thrombin through the binding of exosite | and the
Anticoagulant therapy with warfarin has the same prob- active binding site of thrombin.'”
lem that was described earlier with heparin. The drug has a Hirudin, an anticoagulant produced in the salivary
narrow therapeutic interval and less than optimal tests to glands of leeches, is a direct thrombin inhibitor that binds
monitor the drug. Since 1935, when Armand Quick described irreversibly in a 1:1 stoichiometric relationship. It has a
the prothrombin time, and in 1941, when the first patient was plasma half-life of approximately | hour. It has no structural
treated with warfarin, the PT has been used to monitor ther- similarity to heparin and does not cross-react with heparin-
apy. As with the problem of heparin and the APTT, it was dependent antibodies. A hirudin derivative (lepirudin) and
discovered in the late 1970s that there was a wide degree of recombinant hirudin are available for treatment of HIT.
variability in the response of thromboplastin reagents and Hirudin therapy is commonly monitored using the APTT,
instruments that were used for the PT when evaluating with a target value of 1.5 to 2.0 times the median of the nor-
patients who were receiving warfarin therapy. It has been pos- mal range. However, variable responses of APTT reagents to
sible to overcome some of the difficulties with this variability hirudin can be problematic. Early data show promising results
by normalizing the responses of thromboplastin reagents for the Ecarin clotting time (a snake venom—derived assay);
against an international standard. This process is referred to however, clinical outcome data are still being gathered. Daily
as the international normalized ratio (INR). The INR is monitoring is recommended because hirudin is predomi-
described by the following formula: nantly cleared by the kidneys. Therefore, monitoring is espe-
Pi
ISI cially important in patients with impaired renal function.'°*'™
Antibodies to lepirudin develop in one-third of patients on
INR =(=—=
a
Bivalirudin is a reversible synthetic thrombin inhibitor ximelagatran shows oral bioavailability and has shown simi-
that can be used as an alternative drug in patients with HIT. It lar or enhanced efficacy when compared to warfarin with a
contains a C-terminus similar to hirudin that binds thrombin’s similar complication profile; however, questions regarding
exosite | and an amino terminus that binds thrombin’s active the risk of hepatotoxicity have currently delayed FDA
site. It is administered intravenously and has a half-life of approval. Another candidate for an oral thrombin inhibitor is
25 minutes. The current use is in coronary angioplasty for dabigatran, which is in clinical trials. Others in development
patients who cannot use heparins. DTIs can be monitored via may try to use tissue factor, factor VII, factor V, and factor VIII
anti-[la chromogenic assays. as potential targets. The use of recombinant activated protein
C is being developed.
FACTOR Xa INHIBITORS
Fondaparinux is a synthesized selective factor Xa inhibitor. A
pentasaccharide, it binds AT and increases its affinity for Antiplatelet Agents
factor Xa more than 300-fold. The molecule effects a confor- Used alone or in combination with other anticoagulants, a
mational change in AT and is released to inhibit another mol- number of antiplatelet function agents have been developed.
ecule. Because the inhibition is more specific for factor Xa, By far the most commonly used agent is aspirin. It functions
bleeding complications are expected to be decreased due to a by inhibiting prostaglandin synthesis. Its action has been well
small amount of thrombin generated by other uninhibited recognized for over 20 years, and its use (in low doses) for
coagulation factors. It is administered subcutaneously and has prophylaxis to prevent stroke and MI has been well character-
a half-life of 17 hours. A long acting variation, idraparinux, ized. Aspirin has been used in combination with heparin or
requires once a week dosing and is currently in clinical trials. LMWH in some patients who have arterial thrombotic events.
Oral factor Xa inhibitors are also in development and reliable Vascular surgeons and cardiologists are becoming
data are anxiously awaited. increasingly adept at using thrombolytic agents to lyse clots
formed in coronary arteries and other vessels. The site of
OTHER ANTICOAGULANTS
these thrombi can then be stented with devices that hold the
Danaparoid, a heparinoid, is a mixture of the glycosaminogly-
lumen of the artery open and allow recirculation beyond the
cans heparan sulfate, dermatan sulfate, and chondroitin sul-
previous occlusion. The difficulty that has occurred with these
fate. It is not available in the United States, but is used in
therapies has been the prevention of rethrombosis of the
Europe and Canada. Its anticoagulant effect is by the inactiva-
vessel. The most promising of the agents used in a postthrom-
tion of factor Xa. It also has some antithrombin (anti-IIa)
bolytic setting or with stents have been inhibitors of platelet
effects and interacts with heparin cofactor II. It can be admin-
function with or without heparin. Those used prevent platelet
istered subcutaneously or intravenously and has a half-life of
aggregation by binding to the GPIIb/IIla receptor on the sur-
18 hours. Because of a somewhat predictable distribution and
face of the platelet. In addition, new agents bind specifically
metabolism, dosing can be based on the patient’s weight.
to the ADP receptor on the surface of the platelet, thereby pre-
When used for the purpose of prophylaxis to prevent throm-
venting activation of the platelet. Three intravenous GP
bosis, it does not require monitoring. However, when needed
I[b/Ila inhibitors are in clinical use. Abciximab is a mono-
to therapeutically treat patients with thrombosis, the drug
clonal antibody directed against the receptor protein, and
requires monitoring using the Xa inhibitory assay. Thrombin-
tirofiban and eptifibatide are non-antibody receptor inhibitors.
based assays are not effective. Although recent publications
Of note, profound thrombocytopenia can develop within
suggest monitoring is not required during treatment unless the
hours of use of these drugs and should be suspected in the cor-
patient is very large (more than 90 kg) or very small (less than
rect clinical setting.
55 kg) or has some degree of renal failure, it seems prudent to
Oral GPIIb/IIIa inhibitors have been in development but
monitor danaparoid therapy. This is performed using the anti-Xa
have been associated with increased mortality and increased
activity assay (target range is 0.5 to 0.8 anti-Xa U/mL) and
bleeding risk as compared to placebo. The underlying mech-
requires the use of a standard curve prepared with danaparoid.
anism is not well understood.
H-PF4 antibodies may cross-react with danaparoid in vitro
Other antiplatelet agents are likely to be developed in the
but do not appear to have a negative effect on patient out-
near future. Their effectiveness is gradually increasing, as
come.'**'** However, new thromboembolic episodes occur
clinicians become more accustomed to their application. In
more frequently in patients treated with danaparoid than those
general, these antiplatelet agents have not required laboratory
using lepirudin.'* In addition, the rate of cross-reactivity of
monitoring.
danaparoid with heparin is approximately 3%. Another disad-
vantage is the absence of an antidote (protamine sulfate, the
antidote for unfractionated heparin and LMWH, has no effect
Thrombolytic Therapy
on danaparoid).
Anticoagulants currently in clinical trials are striving for If a thrombus has been formed and has been in place in situ
ease of administration and efficacy with low risk of bleeding for a short period of time, precluding organization, logic
complications. Melagatran and Ximelagatran are dipeptide would dictate that accelerating the lysis of the clot may help
mimics of fibrinopeptide A’s binding region to thrombin’s preserve function in the vascular bed served by the vessel.
active site. Melagatran is administered subcutaneously but Thrombolytic therapies using urokinase and streptokinase
Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy 689
QUESTIONS
Case Study 2 1. What is the initial test that should be ordered to evaluate
the patient’s prolonged APTT? What can this test tell
A 28-year-old gravida 2, para 0 woman desires to have chil-
you about the clotting abnormality?
dren. She seeks advice because she has miscarried two prior
2. An equal mix of the patient’s plasma with normal
pregnancies and is concerned about her ability to have a suc-
plasma demonstrates an APTT of 62 seconds. What can
cessful pregnancy.
you conclude regarding the cause of the prolongation of
Current Medications: Patient reports taking aspirin for
her APTT?
headaches but otherwise takes no medications.
3. Further laboratory testing reveals a prolonged dilute
Family History: Parents and siblings are alive and well.
Russell’s viper venom time, which normalizes when
excess phospholipid is added to the plasma. Based on
PERTINENT HISTORY the results of these tests, what is the probable cause of
Hematopoietic: No history of anemia, bleeding disorder, her prolonged APTT?
jaundice, or easy bruising. Two years ago she had a clot in 4. What further testing should be performed to help con-
her left leg that was treated with anticoagulants for firm these results?
6 months. The leg has a small amount of swelling by the end
of the day. She has never received a blood transfusion. ANSWERS
Menstrual History: Unremarkable menstrual cycle,
1. The 1:1 mixing study with normal plasma; the test will
although somewhat irregular. Last menstrual period:
differentiate between a coagulation inhibitor or factor
39 days ago.
deficiency.
2. The 1:1 mixing study fails to completely correct the
PERTINENT PHYSICAL FINDINGS APTT, indicating the presence of an inhibitor.
Vital Signs: Blood pressure, 125/80 mm Hg; respirations, 3. She has a lupus anticoagulant and history consistent
16/min; pulse, 72/min, and regular. with the antiphospholipid syndrome (recurrent sponta-
Skin: Mild livedo reticularis is noted on distal arms and legs. neous abortion, mild thrombocytopenia, and deep
Pelvic and Rectal Examination: The cervix is parous and venous thrombosis).
soft. 4. Repeat testing for lupus anticoagulant in 6 to 8 weeks to
Bones, Joints, and Muscle: No edema is apparent in the left show persistence and correlate with clinical history.
leg at the time of the examination.
LABORATORY FINDINGS
Hgb, 13.5 g/dL; WBC, 8000/uL; differential WBC:
Neutrophils, 5100/uL; lymphocytes, 2200/uL; monocytes,
500/uL; eosinophils, 200/uL; platelet count, 100,000/uL.
PT, 12 seconds; INR, 1.0; APTT, 70 seconds (1): fibrinogen,
269 mg/dL; thrombin time, 18 seconds.
continued
a oi one
1. Which characteristic of endothelial cells suppresses te. Fibroblast
fibrinolysis? d. Eosinophil
a. Platelet adhesion e. Erythrocyte
b. Production of t-PA inhibitor
4. A 28~year-old man suffered from recurrent deep venous
c. Secretion of von Willebrand’s factor thrombosis and pulmonary embolus. Studies in the labo-
d. Release of tissue factors
ratory show that his plasma is unable to inhibit factors
2. Which of the following statements concerning warfarin Villa and Va, indicating the plasma is deficient in which
is true? naturally occurring inhibitor/anticoagulant?
a. It inhibits production of contact coagulation factors. jot). Protein C
had had one prior deep venous thrombosis. She has no c. Factor V Leiden
children but has had two spontaneous abortions. Which d Mutated forms of factors V and VII
laboratory test do you expect to be often abnormal?
8. What is the most significant inhibitor of tissue plasmino-
a. Thrombin time
gen activator?
oa . Activated partial thromboplastin time
a. Plasminogen activator inhibitor- |
. Assay for factor V
b. Plasminogen activator inhibitor-2
. Assay for plasminogen
c. Plasminogen activator inhibitor-3
CO
ee. Assay for antithrombin
d. Plasminogen activator inhibitor-4
6. A 25-year-old woman presents with a thrombus in her
9. What is the major inhibitor of thrombin and factor Xa?
right leg. She had a similar event 2 years ago in the left
a. Protein C
Jeg. Her mother, a paternal uncle, and her sister have all
b. Antithrombin
had episodes of thrombosis, and one of these was a pul-
c. Protein S
monary thromboembolus. Of the following potential eti- d. Heparin cofactor II
ologies; which is the most likely?
a. Protein S deficiency 10. Which of the following is not a thrombolytic agent?
b. Antithrombin deficiency a. Tissue plasminogen activator
c. Hyperhomocysteinemia b. Urokinase
d. Factor V Leiden ‘ c. Streptokinase
e. Factor XI deficiency d. a,-Antiplasmin
7. What is the most common cause of activated protein C See answers at the back of this book.
resistance?
a. Functional PS deficiency
b. Inhibitors of activated protein C
w Heparin-induced thrombocytopenia is described as a sud- w Vitamin K is a cofactor for a carboxylase that inserts an
den decrease in the platelet count 5 days after heparin additional carboxyl group on the glutamic acid residues in
therapy is initiated and is caused by the interaction of the amino terminal end of coagulation factors I, VII, IX,
platelet factor 4 (PF4) on the platelet membrane. and X.
w Unfractionated heparin is an unbranched polysaccharide mw Warfarin therapy or oral anticoagulation therapy acts by
that is prepared from bovine lungs or porcine intestines with interfering with the recycling of vitamin K and depressing
a range of molecular mass from 4000 to 35,000 daltons. It the activities of factors II, VII, LX, and X; PC; and PS.
acts by accelerating the rate at which AT is capable of w Warfarin therapy is monitored by the prothrombin time
binding thrombin. Heparin therapy is monitored by the (PT) and international normalized ratio (INR); the INR is
APTT, with a target value of one-and-a-half times the maintained between 2 and 3 for patients on anticoagulant
upper limits of the reference range for the APTT. therapy.
@ Low-molecular-weight heparin (LMWH) is_ prepared wg Thrombolytic agents include t-PA, urokinase, and strep-
from unfractionated heparin by fractionation or depoly- tokinase. Hemostatic evaluation includes PT, APTT,
merization, producing heparins of 1000 to 12,000 daltons. fibrinogen assay, thrombin time, and platelet count.
Acknowledgment
The authors wish to express their deep gratitude to
John D. Olson, MD, PhD, for his assistance in preparing this
chapter. His knowledge of and commitment to the subject
were inspiring and invaluable. Without his guidance this work
would not have been possible.
. Golino, P, et al: Role of tissue factor 39. Demers, C, et al: Thrombosis in patients symptomatic for pulmonary
pathway inhibitor in the regulation of tis- antithrombin III-deficient persons: embolism. Thromb Haemost 89(1):198,
sue factor-dependent blood coagulation Report of a large kindred and literature 2003.
Cardiovasc Drug Rev20(1):67, 2002. review.Ann Intern Med 116(9):754, 56. Kyrle, PA, et al: High plasma levels of
24. Crandall, DL, et al: Modulation of adi- OOF factor VIII and the risk of recurrent
pose tissue development by pharmaco- 40. Heparin-induced decrease in circulating venous thromboembolism. N Engl J Med
logical inhibition of PAI-1. Arteriosclero antithrombin-IIf. Lancet 17;2(8038):581, 343(7):457, 2000.
Thromb Vasc Biol 2006 NYY. . Cristina, L, et al: High plasma levels of
Oct;26(10):2209-15. 41. Sheffield, WB, et al: Antithrombin struc- factor VII and risk of recurrence of
. Bergheim, I, et al: Metformin prevents ture and function. In High, KA, and venous thromboembolism. Br JHaema-
alcohol-induced liver injury in the Roberts, HR (eds): Molecular Basis of tol 124(4):504, 2004.
mouse: Critical role of plasminogen acti- Thrombosis and Hemostasis. 58. O’ Donnell, J, et al; Elevation of FVII: C
vator inhibitor-1. Gastroenterology Marcel Dekker, New York, 1995, p 355. in venous thromboembolism is persistent
130(7):2099, 2006. . Menache, D, et al: Antithrombin II: and independent of the acute phase
. Bouma, BN, and Meijers, JC: Thrombin- Physiology, deficiency, and replacement response. Thromb Haemost 83(1):10, 2000.
activatable finbrinolysis inhibitor. therapy. Transfusion 32:580, 1992. ay). Kamphuisen, PW, et al: Increased levels
J Thromb Haemost 1:1566, 20034. . Poort, SR, et al: A common genetic vari- of factor VIII and fibrinogen in patients
Pf Dahlback, B, et al: Familial throm- ation in the 3”-untranslated region of the with venous thrombosis are not caused
bophilia due to a previous unrecognized prothrombin gene is associated with ele- by acute phase reactions. Thromb
mechanism characterized by poor antico- vated plasma prothrombin levels and an Haemost 81(5):680, 1999.
agulant response to activated protein C: increase in venous thrombosis. Blood 60. Koster, T, et al: Role of clotting factor VIII
Prediction of a cofactor to activated pro- 88:3698, 1996. in effect of von Willebrand factor on
tein C. Proc Natl Acad Sci USA 44, Kapur, RK, et al: A prothrombin gene occurrence of deep-vein thrombosis.
90;1004, 1993. mutation is significantly associated with Lancet 345(8943):152, 1995.
. Bertina, RM, et al: Mutation in blood venous thrombosis. Arterioscler Thromb 6 pant. O'Donnell, J, et al: High prevalence of
coagulation factor V associated with Vasc Biol 17:2875, 1997. elevated factor VIII levels in patients
resistance to activated protein C. Nature 4S. De Stefano, V, et al: Inherited throm- referred for thrombophilia screening:
369:64, 1994. bophilia: Pathogenesis, clinical syn- Role of increased synthesis and relation-
. Williamson, D, et al: Factor V Cambridge: dromes, and management. Blood ship to the acute phase reaction. Thromb
A new mutation (Arg 306 Thr) associ- Siies oll, WOES), Haemost 77(5):825, 1997.
ated with resistance to activated protein 46. D’ Angelo, A, and Selhub, J: Homocys- . Patel RK, et al: Risk factors for venous
C. Blood 91:1140, 1998. teinic and thrombotic disease. Blood thrombosis in the black population.
. Ridker, PM, et al: Mutation in the gene KO, UT Thromb Haemost 90(5):835, 2003.
coding for coagulation factor V and the 47. Jensen, R, and Ens, GE: Tissue factor . Scanu, AM: Atherothrombogenicity of
risk of myocardial infarction, stroke, and pathway inhibitor. Clin Hemost Rev 6:1, lipoprotein (a): The debate. Am J Cardiol
venous thrombosis in apparently healthy LOD 82:26Q, 1998.
men. N Engl J Med 332:912, 1995. 48. Bombeli, T, et al: Decreased anticoagu- 64. Djurovic, S, and Berg, K: Epidemiology
1S) — . Dahlback, B: Inherited thrombophilia: lant response to tissue factor pathway of Lp(a) lipoprotein: Its role in
Resistance to activated protein C as a inhibitor in patients with venous throm- atherosclerotic/thrombotic disease.
pathogenic factor of venous thrombo- boembolism and otherwise no evidence ChinGeneho22 31 OTe
modulin. Blood 85:607, 1995. of hereditary or acquired thrombophilia. 65. Marcovina, SM, and Koschinsky, ML:
WwtO . Dahlback, B, et al: Resistant to acti- Thromb Haemost 91(1):80, 2004. Lipoprotein (a) as a risk factor for coro-
vated protein, the FV:Q 506 allele, and 49. Lean, SY, et al: The effects of tissue fac- nary artery disease. Am J Cardiol
venous thrombosis. Ann Hematol tor pathway inhibitor and anti-beta- SZ 7URIOOS:
72:166, 1996. 2-glycoprotein-I IgG on thrombin gener- 66. Sha, J, et al:. Apo(a) promotes thrombo-
83: Tait, RC, et al: Prevalence of protein C ation.. Haematologica. 91(10):1360, sis in a vascular injury model by a mech-
deficiency in the healthy population. 2006. anism independent of plasminogen.
Thromb Haemost 73:87, 1995. 50. Tollefsen, DM: Laboratory diagnosis of J Thromb Haemost 3(10):2281, 2005.
34. D’ Angelo, SV, et al: Relationship antithrombin and heparin cofactor I 67. Vormittag, R, et al: Lipoprotein (a) in
between protein C antigen and anticoag- deficiency. Semin Thromb Hemost patients with spontaneous venous throm-
ulant activity during oral anticoagulation 16:162, 1990. boembolism. Thromb Res 120(1):15,
and in selected disease states. J Clin . Ebert, RF: Index of Variant Human Fib- 2007.
Invest 77:416, 1986. rinogens. CRC Press, Boca Raton, FL, 68. van Hylckama Vlieg, A, et al: High levels
Sy Engesser, L, et al: Hereditary protein C 1991. of factor IX increase the risk of venous
deficiency: Clinical manifestations. Nni) . Dawson, NA, et al: Acquired dysfibrino- thrombosis. Blood 95(12):3678, 2000.
Ann Intern Med 106:677, 1987. genemia. Am J Med 78:682, 1985. 69. Lowe, GD: Factor [X and thrombosis.
. Comp, PC: Congenital and acquired . Kraaijenhagen, RA, et al:High plasma Br JHaematol 115(3):507, 2001.
hypercoagulable states. In Hull, R, and concentration of factor VILc is a major 70. Meyers, JC, et al: High levels of coagu-
Pineo, GF (eds): Disorders of Thrombo- risk factor for venous thromboembolism. lation factor XI as a risk factor for
sis. WB Saunders, Philadelphia, 1996, Thromb Haemost 83(1):5, 2000. venous thrombosis. N Engl J Med
i) 31339). 54. Tsai, AW, et al: Coagulation factors, 342(10):696, 2000.
7
StF Rodeghiero, F, and Tosetto, A: The epi- inflammation markers, and venous — . Eichinger, S, et al: Thrombin-activatable
demiology of inherited thrombophilia: thromboembolism: The longitudinal fibrinolysis inhibitor and the risk for recur-
The VITA Project/Thromb investigation of thromboembolism etiol- rent venous thromboembolism. Blood
Haemost78(1):636, 1997. ogy (LITE). Am J Med 113(8):636, 103(10):3773, 2004. Epub 2004 Jan 22.
38. Melissari, E, et al: Congenital throm- 2002. (2 . van Aken, BE, et al:Interleukin 8 and
bophilia among patients with venous Dh Bombeli, T, et al: In patients symptomatic venous thrombosis: evidence for a role
thromboembolism.,Blood Coagul Fibri- for deep-vein thrombosis factor VIII of inflammation in thrombosis. Br J
nolysis 3(6):749, 1992. elevation is found twice as frequent as in Haematol 116(1):173, 2002.
694 Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy
. Lisman, T, et al: Reduced plasma fibri- 89. Roubey, RAS: Autoantibodies to phos- presence of lupus anticoagulants. Arch
nolytic potential is a risk factor for pholipid-binding plasma proteins: A Pathol Lab Med 111:120, 1987.
venous thrombosis Blood 105(3):1102, new view of lupus anticoagulants and 104. Brandt, JT, et al: Laboratory identifica-
2005. other “antiphospholipid” autoantibod- tion of lupus anticoagulants: Results of
74. Miyakis, S, et al: International consensus ies. Blood 84:2854, 1994. the second international workshop for
statement on an update of the classifica- 90. Rand, JH, and Wu, X: Antibody- identification of lupus anticoagulants.
tion criteria for definite antiphospholipid mediated disruption of the annexin-V Thromb Haemostas 74:1597, 1995.
syndrome: Report of an international antithrombotic shield: A new mecha- 5. Lazarchick, J, and Kizer, J: The labora-
workshop. J Throm Haemost 4:295, nism for thrombosis in the antiphospho- tory diagnosis of lupus anticoagulants.
2005. lipid syndrome. Thromb Haemostas Arch Pathol Lab Med 113:177, 1989.
. McNeil, HP, et al: Immunology and clin- 82:649, 1999. 106. Curtin, N, et al: Extensive evaluation of
ical importance of antiphospholipid anti- OIF Horbach, DA, et al: Lupus anticoagu- the instrumentation laboratory IL test
bodies. Adv Immunol 49:193, 1991. lant is the strongest risk factor for both D-Dimer immunoturbidimetric assay on
76. Triplett, DA, et al: The relationship venous and arterial thrombosis in the ACL 9000 determines the D-Dimer
between lupus anticoagulants and anti- patients with systemic lupus erythe- cutoff value for reliable exclusion of
bodies to phospholipid. JAMA 259:550, matosus. Thromb Haemostas 76:916, venous thromboembolism. Lab Hema-
1988. 1996. tol 10(2):88, 2004.
Ue Male, C, et al: Clinical significance of OD Coulam, CB, et al: American Society 107. Goodacre, S, et al: Variation in the
lupus anticoagulants in children. J Pedi- for Reproductive Immunology report of diagnostic performance of D-dimer for
atr 134:199, 1999. the committee for establishing criteria suspected deep vein thrombosis. QJM
78. Bowie, EJW, et al: Thrombosis in sys- for diagnosis of reproductive autoim- 98(7):513, 2005.
temic lupus erythematosus despite circu- mune syndrome. Am J Reprod 108. Paneesha, S, et al: High D-dimer levels
lating anticoagulants. J Lab Clin Med Immunol 41:121, 1999. at presentation in patients with venous
62:416, 1963. 8). Brandt, JT, et al: Criteria for the diagno- thromboembolism is a marker of
1. Harris, EN: Syndrome of the black swan. sis of lupus anticoagulants: An update. adverse clinical outcomes.
Br JRheumatol 26:324, 1987. Thromb Haemostas 74;1185, 1995. Br JHaematol 135(1):85, 2006.
80. Harris, EN, et al: Anticardiolipin anti- 94. Arnout, J, et al: Optimization of the 109. Bounameaux, H, et al: Plasma measure-
bodies: Detection by radioimmunoassay dilute prothrombin time for the detec- ment of D-dimer as diagnostic aid in
and association with thrombosis in sys- tion of the lupus anticoagulant by use suspected venous thromboembolism: an
temic lupus erythematosus. Lancet of a recombinant tissue thromboplastin. overview. Thromb Haemost 71(1):1,
ANIL, WS, Br JHaematol 87:94, 1994. 2004.
81. Loizu, S, et al: Measurement of anticar- . Thiagarajan, P, et al: The use of the 110. Lockshin, MD: Pregnancy loss in the
diolipin antibodies by enzyme-linked dilute Russell viper venom time for the antiphospholipid syndrome. Thromb
immunosorbent assay: Standardization diagnosis of lupus anticoagulants. Haemostas 82:641, 1999.
and quantitation of results. Clin Exp Blood 68:869, 1986. iit. Greinacher, A, et al: Clinical features of
Immunol 62:738, 1985. 96. Exner, T, et al: A sensitive test demon- HIT including risk factors for thrombo-
82. Galli, M, et al: Anticardiolipin antibodies strating lupus anticoagulant and its sis: a retrospective analysis of 408
(ACA) directed not to cardiolipin but to behavioral patterns. Br J Haematol patients. Thromb Haemost 94:132, 2005.
a plasma cofactor. Lancet 335:1544, 40:143, 1978. ili, Warkentin, TE: Clinical presentation of
1990. OFF Norbis, F, et al: Diluted Russell’s viper heparin-induced thrombocytopenia.
. Matsuurra, E, et al: Anticardiolipin venom time and colloidal silica clotting Semin Hematol 35(Suppl 5):9, 1998.
cofactor(s) and differential diagnosis of time for the identification of the . Nand, S, et al: Heparin-induced throm-
autoimmune disease. Lancet 336:177, phospholipid-dependent inhibitors bocytopenia with thrombosis: Inci-
1990. of coagulation. Thromb Res 85:427, dence, analysis of risk factors, and
84. McNeil, HP, et al: Antiphospholipid anti- QO. clinical outcomes in 108 consecutive
bodies are directed against a complex 98. Triplett, DA, et al: A hexagonal (II) patients treated at a single institution.
antigen that includes a lipid binding phase phospholipid neutralization assay Am J Hematol 56:12, 1997.
inhibitor of coagulation: 82 Glycopro- for lupus anticoagulant identification. 114. Warkentin, TE, and Kelton, JG: A
tein I (apolipoprotein H). Proc Natl Acad Thromb Haemostas 70:787, 1993. 14-year study of heparin-induced throm-
Sci USA 87:4120, 1990. 99. Triplett, DA, et al: Laboratory diagno- bocytopenia. Am J Med 101:502, 1996.
85. Oosting, JD, et al: Lupus anticoagulant sis of lupus inhibitors: A comparison of itil, Chong, BH: Heparin-induced thrombo-
activity is frequently dependent on the the tissue thromboplastin inhibition pro- cytopenia. Br JHaematol 89:431, 1995.
presence of Beta2-Glycoprotein I. cedure with a new platelet neutraliza- 116. Van Cott, EM: How to diagnose and
Thromb Haemostas 67:499, 1992. tion procedure. Am J Clin Pathol manage heparin induced thrombocy-
86. Bevers, EM, et al: Lupus anticoagulant 79:678, 1983. topenia. CAP Today March:50, 1997.
IgG’s (LA) are not directed to phospho- 100. Sletnes, KE, et al: Preparation of NY, Amiral, J, et al: Platelet factor 4 com-
lipids only, but to a complex of lipid- plasma for the detection of lupus anti- plexed to heparin is the target for anti-
bound human prothrombin. Thromb coagulants and antiphospholipid anti- bodies generated in heparin-induced
Haemostas 66:629, 1991. bodies. Thromb Res 66:43, 1992. thrombocytopenia. Thromb Haemostas
87. Permpikul, P, et al: Functional and bind- 101. Olson, JD: Addressing clinical etiolo- 68:95, 1992.
ing studies of the roles of prothrombin gies of a prolonged aPTT. CAP Today 118. Amiral, J, et al: Presence of autoanti-
and Beta2Glycoprotein | in the expres- Ssh, IE). bodies to interleukin-8 or neutrophil-
sion of lupus anticoagulant activity. 102. Adcock, DM, and Marlar, RA: Acti- activating peptide-2 in patients with
Blood 83:2878, 1994. vated partial thromboplastin time heparin-associated thrombocytopenia.
88. Galli, M, et al: Different anticoagulant reagent sensitivity to the presence of Blood 88:410, 1996.
and immunological properties of antipro- the lupus anticoagulant. Arch Pathol 119. Warkentin, TE, et al: Laboratory testing
thrombin antibodies in patients with Lab Med 116:837, 1992. for the antibodies that cause HIT: How
antiphospholip antibodies. Thromb 103. Brandt, JT, et al: The sensitivity of dif- much class do we need: J Lab Clin Med
Haemostas 77:486, 1997. ferent coagulation reagents to the 146:341, 2005.
Chapter 28 Introduction to Thrombosis and Anticoagulant Therapy 695
120. Suh, J-S, et al: Characterization of the using flow cytometry. Br JHaematol 147. Doyle, DJ, et al: Adjusted subcutaneous
humoral immune response in heparin- 95:724, 1996. heparin or continuous intravenous
induced thrombocytopenia. Am J 134. Warkentin, TE, et al: The pathogenesis heparin in patients with acute deep
Hematol 54:196, 1997. of venous limb gangrene associated venous thrombosis. A randomized trial.
. Kelton, JG, et al: Heparin-induced with heparin-induced thrombocytope- Ann Intern Med 107:441, 1987.
thrombocytopenia: Laboratory studies. nia. Ann Intern Med 127:804, 1997. 148. Basu, D, et al: A prospective study of
Blood 72:925, 1988. IS) Haas, S, et al: Heparin-induced throm- the value of monitoring heparin treat-
. Cines, DB, et al: Immune endothelial- bocytopenia: The role of platelet activa- ment with the activated partial throm-
cell injury in heparin-associated throm- tion and therapeutic implications. boplastin time. N Engl J Med 287:324,
bocytopenia. N Eng J Med 316:581, Semin Thromb Hemostas 25(Suppl 1): NOs
1987. (7, NOY). 149. Fairweather, RB, et al: College of
123. Amiral, J, et al: Generation of antibod- 136. Warkentin, TE, and Greinacher, A: HIT: American Pathologists Conference
ies to heparin-PF4 complexes without recognition, treatment, and prevention: XXXI on Laboratory Monitoring of
thrombocytopenia in patients treated The seventh ACCP Conference on Anticoagulant Therapy. Laboratory Mon-
with unfractionated or low-molecular- Antithrombotic and Thrombolytic itoring of Oral Anticoagulant Therapy.
weight heparin. Am J Hematol 52:90, Therapy. Chest 126(Suppl):331S, 2005, Arch Pathol Lab Med 122:768, 1998.
1996. Chest 127:416, 2005. 150. Hirsh, J, et al: Oral anticoagulants:
124. Brandt, JT, et al: On the role of platelet . Hellgren, M, et al: Resistance to acti- mechanism of action, clinical effective-
FeyRIla phenotype in heparin-induced vated protein C as a basis for venous ness, and optimal therapeutic range.
thrombocytopenia. Thromb Haemostas thromboembolism associated with preg- Chest 114(5 Suppl):445S, 1998.
74:1564, 1995. nancy and oral contraceptives. Am J isl. Rieder, MJ, et al. Effect of VKORC1
12D: Burgess, JK, et al: Single amino acid Obstet Gynecol 173:210, 1995. haplotypes on transcriptional regulation
mutation of Fey receptor is associated . Bokarewa, MI, et al: Arg 506-Gln and warfarin dose. N Eng] J Med
with the development of heparin-induced mutation in factor V and risk of throm- 352 C2) 22802005:
thrombocytopenia. Br JHaematol bosis during pregnancy. Br JHaematol 2. Bauer, KA: Their disadvantages are
ONE GlE OOS: 92(2):473, 1996. cost, a narrow therapeutic window and
126. Arepally, G, et al: FetRIa polymorphism, . Olivieri, O, et al: Resistance to acti- no effective antidote. New Anticoagu-
subclass-specific IgG anti-heparin/PF4 vated protein C in healthy women tak- lants Hematol 450, 2006.
antibodies and clinical course in patients ing oral contraceptives. Br J Haematol IID}, Warkentin, TE: Heparin-induced throm-
with heparin-induced thrombocytopenia 91:465, 1995. bocytopenia: Pathogenesis, frequency,
and thrombosis. Blood 88(Suppl 1, 140. Harris, RC, and Ismail, N: Extrarenal avoidance and management. Drug
abstr):28la, 1996. * complications of the nephrotic syn- Saliety/o:3255 1997.
WALT Warkentin, TE, et al: Heparin-induced drome. Am J Kidney Dis 23:477, 1994. 154, Laposata, M, et al: College of American
thrombocytopenia in patients treated 141. Priest, JR, et al: A syndrome of throm- Pathologists conference XXXI on labo-
with low-molecular-weight heparin or bosis and hemorrhage complicating ratory monitoring of anticoagulant ther-
unfractionated heparin. N Engl J Med L-asparaginase therapy for childhood apy: The clinical use and laboratory
Sez SISK), WEE). acute lymphoblastic leukemia. J Pediatr monitoring of low-molecular weight
128. Isenhart, CE, and Brandt, JT: Platelet 100:984, 1982. heparin, danaparoid, hirudin and related
aggregation studies for the diagnosis of . Goodnough, LT, et al: Increased inci- compounds, and argatroban. Arch
heparin-induced thrombocytopenia. dence of thromboembolism in stage IV Pathol Lab Med 122:799, 1998.
Am J Clin Pathol 99:324, 1993. breast cancer treated with a five-drug SW). Lubenow, N, et al: Lepirudin in patients
1293 Sheridan, D, et al: A diagnostic test for chemotherapy regimen. Cancer with HIT.-results of the third prospec-
heparin-induced thrombocytopenia. 54:1264, 1984. tive study (HAT-3) and a combined
Blood 67:27, 1986. 143. Burris HA: Low-molecular-weight analysis of HAT-1, HAT-2 and Hat-3.
. Walenga, JM, et al: Laboratory tests for heparins in the treatment of cancer- J Throm Haemost 32428, 2005.
the diagnosis of heparin-induced throm- associated thrombosis: a new standard . Greinacher, A, et al: Anaphylactic and
bocytopenia. Semin Thromb Hemostas of care? Semin Oncol 33:53, 2006. anaphylactoid reactions associated with
25(Suppl 1):43, 1999. 144. Geerts, WH, et al: A prospective study of lepirudin in patients with HIT. Circula-
. Walenga, JM, et al: Laboratory test for venous thromboembolism after major tion 108:2062, 2003.
heparin-induced thrombocytopenia: A trauma. N Engl J Med 331:1601, 1994. . Lewis, BE, et al. Argatroban anticoagu-
multicenter study. Semin Hematol 145. Brandt, JT (ed): College of American lant therapy in patients with HIT. Circu-
36(Suppl 1):22, 1999. Pathologists Consensus Conference lation 103:1838, 2001.
NB, Stewart, MW, et al: Heparin-induced XXXI on Laboratory Monitoring of . Lewis, BE, et al: Argatroban anticoagu-
thrombocytopenia: An improved Anticoagulant Therapy. Arch Path Lab lation in patients with HIT. Arch Intern
method of detection based on lumi- Med. 122:765, 1998. Med 163:1849, 2003.
aggregometry. Br JHaematol 91:173, 146. Dalen, JE, and Hirsh, J (eds): Fifth . Magnani, HN, and Gallus, A: HIT: a
1995. American College of Chest Physicians report of 1,478 clinical outcomes of
. Lee, DH, et al: A diagnostic test for Consensus Conference on Antithrom- patients treated with danaparoid
heparin-induced thrombocytopenia: botic Therapy. Chest 114(Suppl):439S, (Orgaran) form 1982 to mid-2004.
Detection of platelet microparticles 1998. Thromb Haemost 95:967, 2006.
Ow}
— —
a
Ss
‘
a i
—_—
f
~
—
a
7
—
‘=
"
~ ~
a
7
4
| | “—
— aS =
tinge A MS OED
i j :
a
a
-
| i |
7
: a
iera Fs
4 es oy
_ |
= gs in 7 ;
— , 2G: ome Oh|ne
ett are
—— . ee ae
| *
} = 7 Pint ete | =—
moving Cl) is G.ipwst ie.
i re ivi
]
|
| | 7 disest@id
iter
ey,
af
tit wi
Came ews 7
align 7 |
oe f - ‘
2 Sull, We :
(ai! ; D
i >. 4
a
ein Sian }
Dian durun)‘s _" ir >
i ee
“o F |
: -
'6
i ; 2ir~w 0 A
aie) @ =
;
debddin
i te
; am
_
7 —
As he ; ®
: 7 =
[le Cie
ae ie Pe: 1 ly a in : ‘7 r _——_
i j 7 e
=
tou (
a 7 Ry ais enhate
oh a ee a a) Gevls 5 > '
ei Oo west
7 ¢® Gi za Dat
wo | lee )cous
tpegyhj
Fi pe ).<)~— oa LyGan a ae
_ 7 = a Dic + ’
; ; >
eee =i
rh)
= =
ee ae
-_ a 4 di» ' P ip tf ar
¥
PART 5
LABORATORY METHODS
Chapter ')
Quality Management,
Quality Assurance,
and Quality Control
Kim A. Przekop, BS, MT(ASCP)
introduction OBJECTIVES
Quality Management At the end of this chapter the learner should be able to:
Definition of Quality
Management and Quality . Define a quality laboratory test result.
Testing . List four divisions of quality management and their role in laboratory quality.
Legal Implications
“Quality Management . Explain three quality approaches.
Improves Bottom Lines” . Describe two general quality control activities.
Quality Management Plans
Quality Approaches . Recall quality questions to ask when choosing a new method.
Divisions of Quality . Illustrate the difference between accuracy and precision.
Management
Quality Software . Calculate the mean, standard deviation, and coefficient of variation from a set of data.
Sigma-metrics
Troubleshooting Quality
Control Problems
Peer Group Quality Control
Hematology Laboratory
Applications
Quality Plan Example
Method Validation Studies
Quality Control
Laboratory Quality Updates
Quality Meeting Launches
New Organization
Equivalent Quality Control
Option 4
Case Study 1
Case Study 2
Quality Management technical personnel can be held liable for harm if the patient
can show that the laboratory person acted negligently. '
It is most important that top management be quality-minded.
In the absence of sincere manifestation of interest at the top,
little will happen below. “Quality Management Improves
Bottom Lines”
—Joseph M. Juran
Companies that were reluctant to include Total Quality
Definition of Quality Management Management (TQM) in their policies in the 1970s and 1980s
and Quality Testing were reassured of the positive financial impact on their
bottom lines with several studies. One study looked at the
Quality Management (QM) comprises all of the processes winners of quality awards versus control companies.
that affect the quality of a product (laboratory test result) from The extent to which award-winning companies signifi-
beginning to end. What is quality (Fig. 29-2)? A quality test cantly outperformed the controls during the period following
result is free of defects (accurate and precise), gives the clin- TQM implementation is shown in Figure 29-3. Award-winning
ician the information he or she needs to make a medical deci- companies experienced an average 91% growth in operating
sion (customer service), and is within a reasonable cost (cost income compared to 43% for the controls; 69% increase in
efficient). How does a laboratorian ensure quality results? sales versus 32%; 79% increase in total assets versus 37%;
Processes are instituted at every step of the way to comply 23% increase in number of employees versus 7%; 8% rise in
with regulatory guidelines and best practice models. Quality return on sales versus no improvement for the controls; and 9%
tools are used correctly to find all types of errors and apply improvement in return on assets versus 6%. They sum up their
that knowledge to the control systems. Everyone is involved findings with three guidelines for companies implementing
in decision-making, including the physicians who will be quality practices:
ordering the test. From the patient’s posture (sitting vs. lying
1. TQM is a good investment. (“Don’t give up on TQM.
down) and manner of collection, to the analysis of a tube of
When implemented effectively, it improves financial per-
blood on an instrument and the acceptance of results in a
formance dramatically.”’)
computer; there are a myriad of variables to consider and to
2. Be patient. (“The benefits of TQM are achieved over a long
control.
period.... Even after effective implementation, it still takes
a couple of years before financial performance starts to
Legal Implications improve.)
3. Be realistic. (“Set realistic expectations on the potential
There are possible legal costs to laboratories and to personnel impact of TQM. Organizational characteristics such as
when quality management procedures are not instituted or size, capital intensity, extent of diversification, and the
followed. When a laboratory professional fails to adhere to maturity of the TQM implementations influence the gains
“that which is acceptable,” errors may occur that lead to from TQM. These and other factors should be considered
adverse consequences for patients. In general, laboratories are in setting expectations.”)*
more likely to be liable for regulatory violations than to be
sued for negligence (medical malpractice) or other legal rea-
sons. However, litigation can occur and individual laboratory Quality Management Plans
The Centers for Medicare and Medicaid Services (CMS) regu-
late all laboratory testing (except research) performed on
Accurate &
humans in the United States through the Clinical Laboratory
precise
Improvement Amendments of 1988 (CLIA-88). These amend-
ments have been updated five times and were finalized in 2003.
According to CLIA’s “Survey Procedures and Interpretive
Guidelines for Laboratories and Laboratory Services (Appen-
dix C) Part 493, Laboratory Requirements,” all laboratories
that perform patient testing need a quality management plan.’
Quality
laboratory This chapter is mostly concerned with non-waived testing
test result (moderate and highly complex tests). To assist laboratories, the
College of American Pathologists (CAP) has incorporated reg-
ulations from CLIA into their checklist questions on quality.
‘Customer Cost Even if a lab is not registered with CAP, the checklists are
service efficient
available (www.cap.org). There are examples of key indicators
Figure 29-2 m The elements of a quality laboratory result. *Reprinted with permission from www.asq.org © 2004 American Society for
Quality.
700 Chapter 29 Quality Management, Quality Assurance, and Quality Control
100
@ Quality award winners
B Control firms
Oo©
40
30
performance
in
change
%
20
10
-
Operating Sales Total Employees Return on Return on
income assets sales assets
Figure 29-3 ® Quality award-winning companies outperform control firms. (Reprinted with permission from Hendricks and Singhal, “Don’t
Count TQM Out.” Quality Progress, April, 1999, p. 38.)
to monitor, how to interpret statistical data, how to measure the world-class company (not necessarily another laboratory) to
effectiveness of a plan, etc. The following are a few of the per- learn what can be done better. For example, if a laboratory wants
tinent questions: to raise the standard of its courier service to excellent, it might
compare its current service to UPS or FedEx, both considered
* Does the Quality Management (QM) plan cover all aspects
best in class, and implement its quality practices. °
of the laboratory service?
¢ Are appropriate corrective and/or preventive actions taken
ROOT CAUSE ANALYSIS
when opportunities for improvement are identified?
¢ Has the QM plan been implemented as designed? This approach is used after a problem occurs to investigate
* Does the laboratory summarize and review its records of errors what happened, how it happened, and why it happened. There
and incident reports at defined intervals to identify trends and are four steps to a Root Cause Analysis:
initiate corrective/preventive actions as appropriate? 1. Data collection
* Does the laboratory have a procedure for reporting device- 2. Charting of each sequence of events for each contributing
related adverse patient events, as required by FDA (Food factor
and Drug Administration)? * 3. Root Cause Mapping to uncover how a factor contributed
A laboratory can follow any currently accepted quality-related to the problem
plan, or establish its own plan. A quality management plan is not 4. Recommendations and Implementation
designed or expected to eliminate all errors in a laboratory. The
primary objective should be patient safety based on quality test INTERNATIONAL STANDARDS ORGANIZATION (ISO)
results. Every plan should contain the following elements: The ISO organization has members from more than 156 coun-
tries. Their common goal is to standardize quality products
* Climate of patient safety and quality
around the globe and provide a common technological lan-
¢ Risk assessment
guage to transfer knowledge. ISO 9000:2000 is an industry
¢ Control measures to monitor high-risk processes
standard for QM systems. 7
¢ Data management; customer service
¢ Continuous improvement.*?
SIX SIGMA
This approach focuses on reducing waste by reducing varia-
Quality Approaches tion. The term “Six Sigma” refers to a calculation for the
Sigma scale; a quality process would have a Sigma score
There are numerous books on approaches to laboratory qual- between 3 and 6 Sigma. To achieve Six Sigma (world-class
ity management plans. However, to simplify the process, sev- quality), a process must not produce more than 3.4 defects per
eral popular quality approaches are defined below. million opportunities. Many organizations, from the govern-
ment to private and public companies, are now using Six
BENCHMARKING Sigma practices. New York-Presbyterian Hospital began their
World-class quality cannot be obtained by simply comparing Six Sigma program in 2003. Today it is ranked 7th in U.S.
one laboratory’s process to another laboratory’s process. Bench- News & World Report's Best Hospitals Honor Roll (before
marking requires that performance be measured against another implementing Six Sigma they were ranked 14th.) *!°
Chapter 29 Quality Management, Quality Assurance, and Quality Control 701
RISK MANAGEMENT AND FAILURE MODE and results. The president of the United States gives the award
AND EFFECTS ANALYSIS annually to companies in varying sectors who apply. Since
Risk Management came from the insurance and financial sec- 1988, 65 companies have been honored with this award. The
tors, and basically asks, “What can go wrong, and what can first Healthcare Award was given in 2002 to the Franciscan
we do about it?” Failure Mode and Effects Analysis (FMEA) Sisters of Mary Healthcare System in the Midwest, which
began in the aerospace industry in the 1960s and is a proac- includes 21 hospitals and 3 nursing homes in 4 states.'°
tive statistical approach used to identify possible failures of a
product or service and then determine the frequency and
impact of the failure. For example, a test result would be the Divisions of Quality Management
product, and a failure could be an incorrect test result that
In a clinical laboratory, there are 12 major divisions of Qual-
could potentially harm the patient. A Hazard score can be
ity Management, also called Quality System Essentials. They
obtained by combining a rating of the severity of harm with
are Organization, Facilities and Safety, Information Manage-
the frequency that the harm might occur. The relationship
ment, Occurrence Management, Customer Service, Person-
between cause and effect is at the heart of FMEA. Six Sigma
nel, Equipment, Documents and Records, Assessments,
includes the discipline of FMEA." ?
Purchasing and Inventory, Process Control, and Process
Improvement (Fig. 29-4). There are many steps that can
LEAN
affect the quality of a patient test result, and all steps need to
Maximizing customer value is the main goal of Lean. The
be subjected to quality standards.
ideas of Lean were pioneered by the Toyota car maker after
WWII, and some expressions of Lean still have Japanese
ORGANIZATION
names today. The Lean method identifies seven types of
All personnel should commit to quality at every step of every
waste. Examples of laboratory waste include:
process.
1. Defects: Errors in laboratory results or reports, missing
reports, late reports. FACILITIES AND SAFETY
2. Waiting: Time periods where laboratory personnel are The layout of buildings, elevators, walkways, and storage
waiting for the patient or the next batch of samples at rooms should facilitate easy access and short time frames for
receiving or testing. transporting specimens and supplies. Work areas should
3. Extra processing: Rework, such as redraws, retesting, incorporate a workflow approach to reduce turnaround times
rehandling, or resending that occur because of defects in and wasted movement. Temperatures are critical in a labora-
the original effort. tory and should be stable. The most up-to-date protective
4. Transport: Unnecessary movement of samples, supplies, equipment and procedures should be in use to protect work-
paper, or staff, and retrieval of lost, moved, or misplaced ers from injury and illness; unscheduled time off from work
items needed in the work. affects quality. Nonworkers are also considered in any safety
5. Motion: Extra steps taken by staff to accommodate and infection control plan.
“workarounds” arising from inefficient layout of sample
paths, instrumentation, supplies, storage, or information. INFORMATION MANAGEMENT
6. Inventory: Extra inventory of laboratory reagents, supplies, There are multiple concerns for the laboratory concerning the
paper, and other materials that is not directly required for control of information. Patient data is readily available, in
the current work. paper and electronic form, and must be protected from inter-
7. Overproduction: Performing unnecessary laboratory test- nal and external threats. Statistical reports and company
ing for any reason. information on employee computers is vulnerable. Regardless
of the cost, it is imperative that every healthcare facility
A person can be certified as a Lean Green Belt. The Mayo review the risks and implement procedures to protect vital
Clinic recently adopted Lean principles in its operations.'>"* information from attack or abuse.
Customer
Service
Purchasing
Documents
and Inventory
and Records
Information
Management
Personnel
Process Facilities/Safety
Improvement
Process Control
Figure 29-4 ™ The 12 divisions of Quality Management (QM), also called Quality Systems Essentials (QSE).
who will then translate the result into an action for a Procedures should follow current guidelines and need to be
patient. The best quality test result that tells a physician noth- written for a newly hired employee who would read the pro-
ing useful is useless. Answering the phone properly, coopera- cedure and then be able to perform the test with minimal
tion among laboratory departments, and calling a critical result supervision. An excellent source for writing laboratory
promptly are other examples of excellent customer service. procedures can be found in the Clinical and Laboratory
Standards Institute’s (CLSI) “New Laboratory Documents:
PERSONNEL Development and Control; Approved Guideline-Fifth
Employees should be hired for attitude and competence; qual- Edition” (GP2-A5), updated May 2006. (In 2005, CLSI
ity results start with quality people. All employees deserve to changed its name from National Committee for Clinical
be trained correctly and consistently. Performance appraisals Laboratory Standards, i.e. NCCLS). Records show what has
and competency assessments are tools for praise, corrective been done; instrument records serve as a history of the
action, and learning.'’ Licensure of all laboratory personnel is instrument. Appropriate records are stored according to
a hot topic; currently 11 states require licensure.'* Because rules of several agencies and state and federal regulations
technology is swiftly moving forward, continuing education and change frequently. The storage time to use is the longest
must be a priority, whether a state is licensed or not. time of all the regulations that affect a department.
EQUIPMENT ASSESSMENTS
When purchasing new equipment, a careful review of all There are internal assessments (1.e., correlations between
currently available instruments and their specifications is similar instruments) and external assessments (1.e., surveys,
warranted: cost, accuracy, precision, risk, usefulness, ease- inspections, correlations between two labs). CLIA, JCAHO
of-operation, maintenance schedule, consumables required, (Joint Commission on Accreditation for Healthcare Organiza-
test menu, etc.'? Consider future needs and technological tions), and CAP requires correlations every 6 months on
advances because most equipment is replaced every 5 years instruments reporting the same result; a laboratory must
or so. Method validation studies must be done correctly to determine the limits of correlation.*°' If there are multiple lab
uncover any hidden biases so that the foundation for a qual- sites within a health-care system, one site is deemed the
ity monitoring system is solid. “core” site, and the others must perform correlations every
6 months with the core site if they are reporting the same
DOCUMENTS AND RECORDS automated results as the core lab. Physicians and patients may
Documenting any variations in normal operations is essen- be using more than one lab in the system; the test results need to
tial to understanding what processes need to be monitored. be comparable. There may be several agencies that can inspect
Chapter 29 Quality Management, Quality Assurance, and Quality Control 703
a laboratory, i.e., AABB, CAP, JCAHO, CMS, FDA, plus state complaints, and errors. Any QC program can be used to
agencies; the laboratory management team must be knowledge- improve a process (see “Quality Approaches’’).°
able of all regulatory requirements that pertain to them.
follows the route of a specimen through the laboratory. The method’s errors, probability of error detection and false rejec-
standards for quality systems are under the Analytic Systems tion. control rules, and number of control measurements. For
section of subpart K: more information on QC statistics, go to www.westgard.com.
A. Accuracy B. Precision
= Sra
i - a)
Sy eee
ne dh
10d
IE
as
a Vtats
¥ “ aed :
Seger
Se) > ee
COMPARISON OF METHODS EXPERIMENT reject the method or to identify and eliminate the causes of the
A comparison of methods experiment is primarily used to errors. The laboratory director should be consulted when the
estimate the average systematic error observed between two data are difficult to interpret.
similar methods, but can also reveal the constant or propor-
tional nature of that error. The results of patient specimens are RECOVERY EXPERIMENT
compared to determine the analytical errors between the
A recovery experiment can be used when there is no compar-
methods. A minimum of 40 well chosen patient samples
ison method. The purpose is to uncover proportional system-
should be tested over a minimum of 5 working days. These
atic error. Commercial solutions of high concentrations of the
samples should be distributed one-third in the low to low-
analyte to be measured are obtained and added in a linear
normal range, one-third in the normal range, and one-third in
fashion to patient samples, and the samples are run two or
the high abnormal range. The correlation coefficient (r) is a
more times to reduce the random error. The results are aver-
useful statistical tool for comparing two sets of data. A perfect
aged and compared, and a percent difference is calculated.
linear relationship between the data would result in a correla-
The total error defined by CLIA is compared to the percent
tion coefficient of 1.0. Figure 29-9 is an example of a corre- difference: if the difference is greater than the total error al-
lation study between identical hematology analyzers. Each lowed, the method or test is not acceptable.
lab must establish its own correlation acceptable limits for CLSI also provides a series of documents that provide
each assay. Statistical software, such as Microsoft Excel, can
extensive information about individual experiments:
produce a graph along with the correlation coefficient.
Regression statistics are another mathematical comparison of * EP5-A. Evaluation of precision performance of clinical
two sets of data; systematic differences can be detected more chemistry devices.
readily with this method. (For a complete discussion of labo- ¢ EP6-P. Evaluation of the linearity of quantitative analytical
ratory statistics for method validation log on to http:// methods.
westgard.com). Once the data have been collected, method ¢ EP9-A. Method comparison and bias estimation using
acceptability should be judged on the basis of the sizes of the patient samples.
random, systematic, and total analytical errors. If these errors ¢ EP10-A. Preliminary evaluation of quantitative clinical lab-
are small compared to the amount of error that would invali- oratory methods.
date the use and interpretation of a test result, the method is ¢ EP14-P. Evaluation of matrix effects.
acceptable. If the errors are too large, it may be necessary to * NRSCL12-P. Source book of reference methods.
LHi = LH2
HGB HGB
88 88
92 92
93 92 HGB CORRELATION: LH1 vs. LH2
94 92
96 96
96 95
gore 10.1
OtnmnttOvd
10.5 10.4
10.6 10.7
10.7 10.6
AOE AO MRS
(Creag 2 ae
Sc hy mS
ie dan d=
idto® Pipa es
1A-7ieahts6
W767
(Sale ees
(DOME 4241
124 ei22
125 12.4
127 12:8
12ST
12 lige st3.0
182 ~~«18.1
13.6 13.5
187 1) 126 Sample number
13.8 13.6 Correlation Coefficient = 0.9978
13.9 13.8
14 an
Lav AA
15.0 15.0
Figure 29-9 @ Correlation study of hematology analyzers (hemoglobin).
Chapter 29 Quality Management, Quality Assurance, and Quality Control 709
TREND
A trend is a slow change in QC values on a QC chart, either
Box 29-5 Minimum QC Requirements
rising or falling steadily across several days or weeks. ((@BF...)
¢ Run two controls each day of testing for quantitative tests.
ELECTRONIC QC * Run controls after changing all reagents, preventive mainte-
Some analyzers and test systems have internal electronic nance, or replacement of a critical part or component.
checks such as clot detectors, abnormal plasma/serum detec- ¢ Rotate QC among the staff.
tors (lipemia, hemolysis), or a mechanism to detect insuffi- * Test QC the same as patients.
cient samples. There could be monitors for temperatures, light e Establish or verify the QC ranges.
source outputs, moving parts sensors, etc. For calibration, ¢ Establish a mean and SD for new unassayed QC before
there might be verification rules in the software for acceptable going live.
runs, or statistical control rules for QC. Electronic QC usually * Do not report patients unless QC is within limits.
monitors variables during the analytical phase only. The dif- ¢ Document all QC procedures performed.
ferent types of QC procedures in the laboratory and the errors haere renee nintninaneievnenttn nA NAST CNRS ARIN LARS RRA
the manufacturer’s package insert ranges. The SDs can be in-house and calculations for quality (accuracy and precision).
entered into the instrument for future use with each new lot of Figure 29-1] is an example of a L-J graph for WBC QC
controls. To test the new ranges, run the controls a few times normal level.
every day for a week. If the control values fall outside the new
ranges, then the new ranges may be too “tight” and need to be SHIFTS AND TRENDS
widened a bit. Multiply the SD by 2, then 3, then 4, and so on The L—J graph is a visual view of how the QC is performing,
until the controls are within the new range. Document the and therefore, how the method is performing. According to
process adequately for inspection purposes. Note: some dif- the Gaussian distribution, data points should occur equally
ferential parameters, such as basophils and eosinophils, nor- around the mean, and this is true for most analytes. A shift in
mally have a high SD due to relatively low actual values. the values represents a sudden and consistent bias from the
mean, 1.e., a group of data points are clustered on one side of
Levy-Jennings Graphs the mean. A shift is representative of systematic error. It is
commonly accepted that six points on one side of the mean
Levy and Jennings introduced the idea of control graphs in constitutes a shift; however, every lab should determine how
1950.°*° The Levy—Jennings (L-J) graph used today consists of many data points constitute a shift. Causes of a shift include
individual data points plotted on a graph of the mean and the improperly prepared reagents or controls, reagent deteriora-
control limits of the particular control level. The L-J graph tion, change in lot numbers, and failure of an instrument com-
historically has consisted of 1, 2, or 3 SD ranges. The roots of ponent (Fig. 29-12).
the 2 SD phenomenon can be traced back to Carl Frederick A trend is a gradual change in the same direction, either
Gauss (1777-1855), who introduced the concept of the normal increasing or decreasing (Fig. 29-13). A trend is also system-
curve. Because 95% of the points in a normal data set for a atic error. Again, each lab must determine how many data
particular parameter are expected to be within 2 SD of the points constitute a trend. Trends can be caused by the same
mean, it seemed appropriate to use + 2 SD limits for labora- items as a shift, but because the data are slowly changing the
tory QC. Therefore, when Levy and Jennings started using a root cause may be more difficult to identify. Look for items
chart for QC, the 2 SD range was implanted and has remained that can change over time, such as reagent or lamp deteriora-
ever since. *! However, laboratories must establish their own tion, temperature fluctuation, calibration shift, and so forth.
mean and limits for each assay based on experiments run Use a systematic logical troubleshooting approach in isolating
WBC
1000
x
Figure 29-11 mA typical Levy-Jennings graph. Each data point is plotted so an overall view of the data is possible.
+2SD
Mean
& a
WBC
1000
x —2SD
Figure 29-12 m™ Example of a shift in QC values on a Levy-Jennings graph. Notice that between day 8 and day 20 the data has abruptly
shifted above the mean.
712. Chapter 29 Quality Management, Quality Assurance, and Quality Control
WBC
1000
x
Figure 29-13 m Example of a trend in QC values on a Levy-Jennings graph. Notice that between day 8 and day 20 there is a slow trend
above the mean.
the cause, making only one change at a time and document- ¢ 1,, This rule refers to one data point exceeding the + 3 SD
ing each action taken. limit.
Another type of change commonly seen on an L-J graph ¢ 2,, This rule refers to two consecutive data points exceeding
is an increase in imprecision. The data points become further the + 2 SD limit.
away from the mean, and the SD becomes larger. If there is ¢ R,, This rule refers to one data point exceeding the +2 SD
contamination of the diluent water used in coagulation recon- limit, and another close data point exceeds the —2 SD limit
stitution, the QC material may display such a dispersion of (opposite directions). An example of this rule is illustrated
results.*Other causes of imprecision include poor mixing of in Figure 29-14.
the control material, pipetting errors, and so forth. * 2of3,, This rule refers to two out of three control points exceed-
ing either the +2 SD or the —2 SD limit on the same side.
Westgard MultiRule Quality Control ¢ 7, This rule refers to seven consecutive data points trending
in the same direction, either progressively lower or higher.
When QC data points have been run and are entered onto a ¢ 4,, This rule refers to four consecutive control measurements
L-J QC chart, the results must be reviewed. When is a data exceeding the +1 SD or —1 SD limit on the same side of the
point too far out? What if all the points are above the mean? graph.
Dr. James O. Westgard, the originator of “Westgard Rules,” ¢ 10, This rule refers to 10 consecutive control measurements
has answered these questions and more. Westgard Rules is a falling on one side of the mean.
series of decision criteria (control rules) to decide whether an
analytical run is in-control or out-of-control. There are rules CHOOSING WESTGARD RULES
for two levels or three levels of control material. To improve Consider these factors when choosing Westgard Rules for a
sensitivity, it is suggested that at least two rules be used method:
together for every control material level. Here is a list of the
* Is the method highly automated, a new method, or a manual
most commonly used Westgard Rules:
method? The type of method, automation, and reliability
¢ 1,, This rule refers to one data point exceeding the +2 SD will have an effect on the rules chosen.
limit. * Select a combination of rules having at least one rule that
¢ 1,;, This rule refers to one data point exceeding the + 2.5 SD responds to random error and one rule that responds to sys-
limit. tematic error (Table 29-4)
WBC
1000
x
Figure 29-14 mA Levy-Jennings graph showing a violation of the Westgard Rule R,..
Chapter 29 Quality Management, Quality Assurance, and Quality Control 713
* Assess the probabilities of rejecting a control value for that method is performing, which in turn can assist in choosing
combination of rules. how to control that method (Box 29-6). A Sigma refers to pre-
* Automatically eliminate the rules that normally have a high cision expressed as a standard deviation of the measurement
probability of rejection procedure. In laboratory testing, a measurement procedure is
the sequence of events from obtaining a sample to reporting a
Rejection refers to false rejection. A technologist does not
result. A method can be any number between 2 and 6 Sigma.
want to be notified by the controls when there is nothing
For Six Sigma quality, the ideal is to have the smallest method
wrong (false rejection). However, she does want to be notified
SD so that six times the SD will fit into a process’s perfor-
if there is a problem with the method. The information in the
mance and produce a high-quality result. The process should
table below was interpolated from a critical-error graph,
be judged, not just the QC material.
which shows whether or not a rule would detect a medically
To provide some perspective on the observed sigma-
important error (see Table 29-4). From the table, it is obvious
metrics for production processes, a number of benchmarks
that the 1,, rule has the highest false rejection rate of any of
are often cited:
the rules—around 9% for a two-control system. This means
that 9 times out of 100 runs a technologist would be respond- * “World class quality” is represented by 3.4 DPM (defects
ing to a false alarm. If three control levels are being analyzed, per million opportunities in a process) or 6 Sigma
the false alarms go even higher. Yet it is the most commonly performance.
used rule in laboratories today. For highly automated systems, * Airline baggage handling shows a 0.4% or 4000 DPM mis-
a better rule would be 1, ;, or 1,,. handling rate, which corresponds to a 4.15 Sigma process.
Each method has a different accuracy and precision; the * Airline travel has a very low fatality rate: 0.43 deaths per
control rules are chosen to match them. If a method is perform- million passenger flights, which is better than a 6 Sigma
ing well (low CV and SD), fewer rules are needed. If the method process.
is a problem method (moderate to high CV and SD), more rules * Typical process performance in business and industry is
are needed to maintain a higher quality of patient results. The often suggested to be about 4 Sigma.
accuracy (bias) and precision (CV) of a method are known from ¢ Firestone production of tires for Ford Explorers appears to
the method validation studies. To review an existing method, the be near the 5 Sigma level on the basis of available informa-
control material CVs for precision (in percent) and CLIA profi- tion on accident rates in the popular press.
ciency testing results for that method can be used to estimate
To find out how well a method is performing, use the follow-
accuracy (bias in percent). The CV and bias can be used in a
ing basic formula (percentages for all):
metric (calculation) which makes it easier to determine how
much and which type of control rules are needed, based on how Sigma-metric = (TE — bias)/SD
well the method is performing. The method’s performance can
To obtain a Sigma-metric, subtract the method bias from
be plotted on the graph in Figure 29-15 to find your Six Sigma
the TE (use the CLIA TE for the method) and divide the prod-
Scores
uct by the method SD. This number shows statistically if the
method is high quality or not as long as the method remains sta-
Sigma-metrics ble. That is why it is crucial to run quality control materials—
to detect instances of instability in the method that could
Six Sigma is not new; it has been around since the 1980s and produce an erroneous result.
has revolutionized many other industries and saved compa- The minimum acceptable quality for a Six Sigma
nies billions of dollars. The principle is based on the Gauss- process is 3 Sigma. If a method is below 3 Sigma, the method
ian distribution and has been updated to correspond to a is not a quality method. You first need to make changes in the
process. Now it is time to start applying this proven quality process and repeat the experiments until your method has
tool in the laboratories. Six Sigma can quantify how well a attained a 3 Sigma level. For a 3 Sigma method, the maxi-
mum number of QCs and rules must be applied; a poorer
quality method requires more controls to maintain the maxi-
mum quality possible, and the opposite is true of a higher
Table20-4 Westgard MultiRules:
quality method. It is a win-win situation when a method is a
Bhilai aed of Detecting —
Errors, False’ Rejections
High Probability High Probability of
D sox 25-6SirSees
Error False Rejection of Error Detection
(atGa 20%) ¢ Well-documented principles
Condition ‘Gt feastOe)
* Useful way to gauge method quality
No errors I. ¢ Easy to calculate
Random Ly 55, I36,13 55,Ras
e Easy to understand
error
¢ Based on Gaussian curve
Systematic 296, 41s, 20f39,, 31,, 10,
error ¢ Use for further quality tests
714 ~— Chapter 29 Quality Management, Quality Assurance, and Quality Control
12.0
6.0
%)
in
(bias
inaccuracy
Observed
0.0
0.0 2.0 3.0 4.0 6.0
quality method. Not only are the patient results more precise Operator suspects a problem when the control was run: short
and accurate, QC material can be kept to a minimum. sampling, mix up of control levels, etc. Do not run patients
For a 3 Sigma or less method, the maximum error detec- until you are satisfied that the problem has been solved or a sys-
tion with low false rejection rate would be a combination of tem is in place to monitor the method for future reoccurrence.
these rules: 1,;, with six controls and the 1,/2,/ R,/3,/6, The first step of troubleshooting QC is to determine whether the
MultiRules with six controls. If you are using a trilevel mate- error is systematic or random. How can the operator distinguish
rial (3 levels) with a 4 Sigma process, you could choose a rule between them? Review Figure 29-14. Which rule was violated?
such as 2o0f3,, for systematic error, and another rule for ran- Does that rule do a better job of detecting random or systematic
dom error, such as 1;,. For higher Sigma methods, possibly error? There is also a third kind of error, “flyers,” that are diffi-
only a single rule needs to be used (choose a rule for detect- cult to catch because they are caused by a bubble or a one-time
ing systematic error).** problem. The type of error should be related to the potential
causes for that error. Remember: Matching the type of error to
CALCULATING QUALITY the potential causes makes good troubleshooting sense.
To support the interest in easy calculations for quality proce-
dures, computer programs are available to automatically SYSTEMATIC ERRORS
select the QC rule(s), number of control levels, and frequency If a multitest system is being run (i.e., automated CBC), how
needed to maintain a particular level of quality desired. many of the parameters were out of control? If more than one
EZ Rules3° by Westgard QC, Inc. (http://www.westgard. parameter was affected, then the troubleshooting should be
con/ezrules.html) is the latest version of the popular Validator® logically aimed at processes that the parameters have in com-
software. A demo of the program can be downloaded for mon. For example, the WBC counting chamber also deter-
free. A SigmaMetric score can also be calculated with this mines the Hgb parameter. Another route to investigate is any
program. The data gathered from the method validation recent changes to the system, reagents, calibration, and main-
experiments are entered into the forms along with the tenance, as they cause many systematic errors.
desired number of control levels and rejection rate.
Several other QC planning tools are available on www.
RANDOM ERRORS
westgard.com: OPSpecs Charts, Critical-Error Graphs, and
The first and easiest way to troubleshoot random errors is to in-
Power Function Charts. These programs use new grids and
spect the system while it is operating. Watch every process that
charts for calculating the necessary QC procedures for individ-
can be visually accessed. Look for loose seals, bubbles, leaks,
ual analytes using imprecision and inaccuracy of the method.
faulty delivery volumes, clogs, and any mechanical parts and
reagents. If there is nothing visually wrong, consult the opera-
Troubleshooting Quality Control Problems tor’s manual and troubleshooting guide for assistance. Ask key
operators to take a look; review the error log for clues. Call
The method validation is performed, the Sigma-metric is calcu- Technical Service for suggestions or to have an engineer inspect
lated, and the Westgard Rules have been chosen for the method. the instrument. Run a precision check of at least 10 replicates of
If all of these procedures were performed correctly, there is a a fresh patient sample to rule out imprecision. If no problems
low false rejection rate for the control values. Thus if a control can be detected, add another random error Westgard Rule to
value falls outside the QC rules, there is a high probability that help detect a future error and monitor the system (tell all staff
the control is doing its job: detecting random or systematic about the problem also). Always document all investigations
error. Rerunning the control should be considered only if the and findings in a problem log for the method or instrument.
Chapter 29 Quality Management, Quality Assurance, and Quality Control 715
Peer Group Quality Control and less if possible. If the ranges were adjusted every month to
match the peer mean, the QC process would be negated in
Most of the larger companies that manufacture instruments favor of looking good on paper.
and/or QC products have a peer group QC program. The daily As with any quality review, written documentation must
or monthly data are sent to the company to compare it to other be noted on the peer group report that it was reviewed and no
labs running the same instrument or QC material for the same problems were seen with the data. If any assays require inves-
assays. This serves two purposes: to satisfy one of the CLIA tigation, the details should be written as if an inspector were
requirements for outside assessments and to provide feedback reading them. Always date any reviews and list any corrective
about a laboratory’s methods versus peers. Review the group- or preventive measures. Writing “will monitor” 1s unaccept-
ing criteria to be sure that the instrument is in the correct able these days.
group according to reagent lot number or control lot number.
Knowing how a method compares with peers gives laborato-
rians One more tool in their quality toolbox. Hematology Laboratory Applications
STATISTICS
Quality Plan Example
The manufacturer will probably provide peer statistics in their Laboratory Alpha decides to combine selections from several
report containing one or more of the following: Quality Management approaches to form their own unique
SDI (Standard Deviation Index): This calculation describes Quality Plan. From the Lean approach, the staff follows a
the systematic error or bias of a method as a multiplier of the CBC, a PT, and a urinalysis sample from collection through
standard deviation for the group: result reporting to the physician. Samples are chosen from
SDI = (Lab mean — Group mean)/ Group SD different areas, 1.e., outpatients, inpatients, clinics, to cover as
many scenarios as possible. Staff observe the motions of the
An SDI of 0.0 indicates that the lab mean is exactly the sample and the employees to document extra waiting, extra
same as the group mean. A SDI of 2.0 or higher requires steps, and unnecessary movements. If a defect or extra pro-
investigation: the method is in danger of systematic inaccu- cessing step occurs, these will be noted. The laboratory must
racy. An advantage to using SDI is a person can scan a peer define how often the study will be performed, how the data
group report and view the SDI of all the assays fairly quickly. will be collected and presented, and ongoing review and cor-
CVI (CV Index): The CV of the lab and the CV of the group rective actions. Many quality entities are covered with this
compared: plan, making it comprehensive. However, this would be only
CVI = Lab CV/Group CV a portion of a total Quality Management Plan. The staff also
decide to use Six Sigma for automated processes; the Sigma
A CVI of 0.0 indicates the laboratory’s CV is the same score is calculated for all assays and changes are instituted to
as the group CV. Any CVI greater than 0.0 suggests that the raise the Sigma score to at least a 3 Sigma. Once they are con-
laboratory’s imprecision is less than that observed for the fident that most of the processes in the laboratory are at peak
group. A CVI greater than 1.0 suggests the lab’s imprecision efficiency, the staff decide to apply for the Malcolm Baldrige
is higher than that of the group (Box 29-7). National Quality Award, which will help focus improvements
Z-score. Measures how many standard deviations the in several customer service areas. There are regulatory
mean of an analyte is from the mean of the peer group. A requirements from several agencies that should be included in
Z-score has no units; however, the score can be positive or the plan: turnaround time for critical tests, participation in a
negative. proficiency testing program, continuous competency assess-
Z-Score = Lab mean — Peer mean/SD
ments, appropriate quality control procedures for each assay,
reviewing errors and correcting the causes, and others. A
Hematology Department must also have its own plan,
REVIEW OF PEER GROUP DATA
although the general Laboratory plan may cover most of the
While reviewing peer data, the laboratory’s assay mean should
elements.
be compared to the peer’s mean: is the lab’s mean consistently
above or below the peer mean every month, but the lab’s SDI
and CVI are acceptable? Is it time to consider an adjustment in Method Validation Studies
the ranges? QC ranges need a little adjustment now and then
Once a new method or new instrument has been chosen and
as part of the normal instrument and QC material process. A
is installed, the method validation studies must be run. The
good rule of thumb is to adjust QC ranges only every 90 days,
focus is to introduce about the same amount of error that
would be present on a daily basis in the normal running of the
test or instrument. If the test will be performed on three shifts,
Box 29-7 SDI and CY
validation samples should be performed on three shifts. Allow
e sDI (SD Index), compares your mean to the group mean | each technologist who will be performing the test to also
* CVI (CV Index), compares your CV in the group’s CV | analyze some of the validation samples. For normal patient
}
i CSS SNe
| ener ein oe oop parent ne ae studies, the goal is to duplicate the same types of patients
716 Chapter 29 Quality Management, Quality Assurance, and Quality Control
tested every day. A plan is necessary to organize the samples Atlanta and created a new organization, The Institute for
needed for each study, the time required for each study, and Quality in Laboratory Medicine (IQLM). This consortium
the personnel who will be conducting the studies. will bring together more than 70 groups from payers, govern-
ment agencies, accrediting organizations, laboratory profes-
EXAMPLE OF METHOD VALIDATION STUDIES sionals, health systems, device manufacturers, clinicians, and
Hematology Laboratory Alpha has just installed a new auto- patients to discuss how to improve patient safety through lab-
mated hematology instrument. After the basic functions of the oratory testing.** **
instrument are verified by the manufacturer, the validation
studies begin. A replication experiment is planned in two
phases, preliminary and comprehensive. The preliminary
Equivalent Quality Control Option 4
phase is completed in one day with one run of 20 replicates of This term was penned by CMS in their Surveyor’s Guidelines
the same sample (within-run random error). The comprehen- in the final CLIA-88 rule. Equivalent Quality Control (QC)
sive phase includes analyzing quality control material of dif- describes situations in which a laboratory could run QC
ferent levels on all shifts by several technologists for at least materials Jess often than the previous CLIA rules of two levels
20 days (between-run random error). The mean, SD, and CV per run. There was much discussion of EQC in laboratory
are calculated for all parameters for both phases and are organizations, and most of them agreed that the level of quality
acceptable. A reportable range experiment is performed with is not yet high enough to allow QC to be run less often than cur-
a commercial CBC and reticulocyte linearity kit that covers as rent practice. In 2004, AdvaMed went as far as to propose a
much of the analytical range as possible (proportional system- new Option 4 where the manufacturer performs a risk assess-
atic error and linearity). The manufacturer of the kit receives ment, recommends a quality plan to monitor and detect the
the data and calculates the statistics; the laboratory personnel risks, and produces data to prove that the quality plan really
review the data and decide on a reportable range. Because the works. This information would be reviewed and approved by
new automated analyzer is similar to the current one, a small the FDA. Laboratories could then implement the quality plan in
normal range study is performed with 20 carefully screened place of one of the current CLIA options. Since the final CLIA-
adult patient samples from both sexes, and the previous range 88 rule has eliminated FDA clearance of manufacturer’s QC
is verified. For pediatric normal values, a decision is made to instructions, the laboratory now must validate all quality con-
transfer the ranges by calculation from the pediatric sample trol procedures, even manufacturer’s guidelines.*°
portion of the comparison of methods study. The comparison
of methods experiment is performed with 40 adult and
40 pediatric patient samples, using normal and abnormal
patients across the entire analytical range. The results of all
the studies are acceptable. Case Study 1
A technologist has just run the three levels of controls on a
Quality Control hematology analyzer. The low level for WBC has been
within 1SD of the mean for the past 3 weeks; however,
Now that the instrument performance has been investigated today the value is greater than 4 SD of the mean. The lab
and deemed acceptable, the monitoring of the quality required uses the following Westgard Rules: R,., 20f3>,, 77, 10,.
needs to be planned and executed. The first step is to calculate
the Sigma-metric score for the instrument. For a hematology QUESTIONS
analyzer with multiple parameters, a critical parameter might
be chosen, or a parameter with the highest SD; the laboratory 1. What should be done next?
2. Is this a random or systematic error?
must decide this. Suppose the Sigma-metric score was 4.0 for
3. Do all the patients need to be rerun since the last low
hemoglobin. With this information, the supervisor uses the
level was within limits?
EZRules3 software to determine which MultiRules to use
with the analyzer. After entering in the information into the
ANSWERS
calculator, the MultiRules given are 1,;, and R,, with two
levels of controls (example only). 1. Most control material for hematology analyzers is pack-
aged as a set of three levels: low, normal, and high. The
Westgard Rule of R,, has been violated; action is required.
Laboratory Quality Updates Errors from improper mixing of the control material
should be ruled out first; the control is then rerun once.
Quality Meeting Launches New If the WBC low level is still outside 4 SD, a new control
Organization vial is opened, mixed, and rerun. A background check is
also appropriate for troubleshooting problems with low
In recent years, many health-care organizations have con- levels of controls. If the background check is acceptable
and the control is still outside 4 SD, observe the instru-
vened to discuss improving the health of the public with
ment during all phases of testing: are there any leaks,
better use of laboratory tests. In April 2003, the Centers for
continued
Disease Control and Prevention (CDC) hosted a conference in
Chapter 29 Quality Management, Quality Assurance, and Quality Control 717
Di cctiont x
# Troubleshooting control value problems is easier when a instrument, and a report is sent with useful statistical
specific rule(s) is matched to a specific method. information, such as SDI, CVI, and the means and SD’s
m Systematic errors affect an entire method (proportional or from the past few months.
constant); random errors affect a part of the process. m The next few years should be exciting as laboratory
m Peer group data is a great tool for quality management. A groups discuss where quality is going.
method is compared to others with the same method or
Introduction OBJECTIVES
Types of Body Fluids At the end of this chapter, the reader should be able to:
and Anatomy
Pericardial, Pleural, L List the types of body fluids from closed body cavities studied in the hematology
and Peritoneal (Serous) laboratory.
Fluids . Identify the type of procedure used to obtain each type of fluid.
Nw
Cerebrospinal Fluid
Synovial Fluid . Describe the formation of serous (peritoneal, pericardial, and pleural), cerebrospinal,
and synovial fluids.
Specimen Collection
and Preparation . Identify the cell types normally found in serous, cerebrospinal fluid, and synovial fluids.
Collection
. Define effusion, transudate, and exudate and describe their characteristics.
Preparation
. Describe the laboratory methods for body fluid analysis.
Cellular Components
of Body Fluids eS
eC. Describe
SCN the principle, advantages, and disadvantages of the cytocentrifuge method
Neutrophils for body fluid morphological examination.
Lymphocytes
. Describe the common cellular artifacts introduced by cytocentrifugation.
Monocytes
Tissue Cells . Differentiate normal from reactive or malignant cells in body fluids.
Eosinophils, Basophils, Mast
Cells 10. Associate morphologic abnormalities in body fluids with their reactive and malignant
causes.
Pleural, Pericardial, and
Peritoneal (Serous) Fluids lidbs Describe the importance of distinguishing a traumatic lumbar puncture from a true
Effusions: Transudates and CNS hemorrhage.
Exudates . Explain the clinical importance of detecting leukemia, lymphoma, and metastatic
Cellular Responses, cancer cells in body fluids.
Microorganisms, and
Malignant Cells in Serous . Describe the purpose of synovial fluid analysis.
Fluids . Describe the types of crystals found in synovial fluids.
Types of Effusions,
. Associate the types of crystals found in synovial fluid with the types of joint
Laboratory Analysis, and
diseases.
Clinical Correlations
Pleural and Pericardial . Define birefringence and relate it to crystal analysis.
Effusions
Peritoneal Effusions
Cerebrospinal Fluid
Specimen Collection and
Processing
720
Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis WaN
pleura. This fluid provides lubrication for the membranes dur- reabsorbed at the same rate by the arachnoid villi of the
ing respiration while preventing the formation of any space arachnoid mater. The arachnoid mater penetrates the inner
between the lungs and chest wall. This maintains the lungs in dura and its venous sinuses. The choroid plexus epithelium
an expanded state. and the endothelium of the capillaries in contact with the
The abdominal cavity is lined by a membrane also CSF form the blood—CSF barrier.'! This barrier controls
composed of a single layer of mesothelial cells known as the the concentrations of solutes between the blood and CSF.
peritoneum. It is the largest and most complex serous mem- The normal total volume of CSF in adults is 90 to 150 mL,
brane in the body. The parietal peritoneum covers the inner and 10 to 60 mL in neonates.
surface of the abdominal wall. The visceral peritoneum cov-
ers the surfaces of the stomach, small and large intestines,
liver, and the superior aspect of the urinary bladder and
Synovial Fluid
uterus. The kidneys, pancreas, duodenum, some lymph Joints, also known as articulations, are the junctions between
nodes, and the abdominal aorta are posterior to (not two or more bones. The freely movable limb joints of the
enclosed by) the peritoneum, and are known as retroperi- body (e.g., shoulder, elbow, knee, hip, etc.) are enclosed by
toneal. The potential space created by the two layers of peri- strong fibrous tissue for support and alignment of the bones,
toneal membranes is known as the peritoneal cavity. The known as the joint capsule. It consists of an outer fibrous
parietal peritoneum secretes a serous fluid that prevents fric- layer and an inner membrane with a rich blood supply, called
tion between the membranes from the constant motion of the the synovium or synovial membrane, composed of a single
abdominal organs, and is absorbed by the visceral peri- layer of mononuclear synovial cells. Articular cartilage covers
toneum. Normal pleural, pericardial, and peritoneal cavities the bony surfaces of the joint that permits smooth motion dur-
do not contain appreciable amounts of fluid, and as such, are ing movement. The synovium also surrounds the tendons and
not true cavities until a disease state causes the accumula- free margins of the ligaments and articular cartilage, but does
tion of fluid. not extend over the surface of the intra-articular cartilage. An
ultrafiltrate of plasma, known as synovial fluid (or mucin; an
older terminology), is secreted by the synovial cells. This fluid
Cerebrospinal Fluid
fills the synovial (joint) cavity, the space that exists between the
The central nervous system (CNS) consists of the brain and bones and enclosed by the synovium and intra-articular carti-
spinal cord, enclosed by the bony structures of the skull and lage. The synovial cells also produce a mucopolysaccharide
vertebrae of the spine. These structures are lined by special known as hyaluronic acid. This gives the fluid a viscous
membranes known as the meningeal membranes or consistency providing lubrication and facilitation of movement.
meninges. The meninges consist of three layers of tissue: a Synovial fluid also transports nutrients to the articular cartilage.
relatively thick outer membrane known as the dura mater, Synovial cells are also phagocytic, removing cellular debris that
which provides the major protection for the brain and spinal exists at the surface of the membrane. The large joints may nor-
cord, a thinner middle membrane called the arachnoid mally contain approximately 1 mL of synovial fluid.
mater, and an inner membrane that lies directly on the sur-
face of the brain and spinal cord known as the pia mater.
The area between the arachnoid mater and pia mater is Specimen Collection and Preparation
known as the subarachnoid space. It contains a selective Collection
ultrafiltrate of plasma known as cerebrospinal fluid (CSF),
which protects and supports the brain and spinal cord, and A properly collected specimen is crucial to proper diagnosis.
maintains a constant ionic environment by circulating nutri- Body fluids are collected only by a physician or properly
ents and removal of waste products. These functions are trained medical personnel because these procedures are
controlled by the blood-brain barrier, which consists of the invasive and may potentially cause harm to the patient.
endothelial cells of brain capillaries wrapped by supporting Specimens are always obtained by aseptic technique for
cells of the brain known as astrocytes. These cells permit the diagnostic or therapeutic purposes. They are potentially
passage of essential substances such as water, oxygen, and infectious and must be handled with universal precautions.
carbon dioxide, and exclude the passage of large molecules They must also be handled with great care, as they
such as proteins, peptides, and many drugs. frequently are nonretrievable, especially in neonates. The
The CSF circulates through the ventricular system of aspirated fluid is placed into evacuated, anticoagulated,
the cerebrum, cerebellum, and brain stem. Masses of spe- and/or sterile tubes to be sent to the laboratory. Details about
cialized capillaries in the pia mater, known as the choroid the collection of each body fluid type are discussed in more
plexus, project into the ventricles and are the main source detail later in the chapter.
of production of CSF. The ventricles and central spinal
canal are lined by a layer of epithelial cells with villous
Preparation
projections and cilia known as ependymal cells, or the
ependyma. Together, the ependyma and choroid plexus On receipt in the laboratory, the body fluid specimen must be
produce approximately 20 mL of CSF per hour, which is processed immediately. The cells within a fluid are usually in
Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis 728,
a lower concentration than peripheral blood and more fragile; cells within a fluid suspension directly onto a glass slide as a
their viability deteriorates once they are removed from monolayer. The method utilizes a small funnel apparatus that is
the body. held against a glass slide by a clamp that fits into the cytocen-
First, the specimen is evaluated for the appropriate trifuge. Between the funnel and the slide is a fitted strip of
collection container and visually inspected for volume, filter paper with a hole under the funnel outlet. Cytocentrifuga-
color, clarity, viscosity, and/or the presence of fibrin clot. tion is based on the principle that cells are concentrated by slow
Next, the sample is set up to perform the cell count. The centrifugation with simultaneous absorption of the noncellular
automated methods currently in use for peripheral blood portion of the fluid into a filter paper. The filter preferentially
cell counts are not recommended for use on body fluids for absorbs the fluid portion leaving the cells in a concentrated but-
several reasons: ton on a microscope slide. This method markedly improves the
1. The peripheral blood cell-counting instruments are not quality of the cell morphology obtained as compared to the
standardized for a body fluid medium. direct smear technique. It is superior for differential analysis
2. Cell size variation in fluids is greater than in peripheral blood. over the performance of an older technique known as the hema-
3. Background debris and clots are often present in serous cytometer chamber differential count, which had the following
fluids. limitations:
4. Certain fluids (e.g., synovial) are quite viscous. ¢ Chamber differential counts only provided the clinician
Despite its imprecision, manual cell counting using a hema- with cell categorization limited to polymorphonuclear or
cytometer is the preferred method for total cell counts on mononuclear leukocytes; the mononuclear category included
body fluid specimens (see Chap. 31). Unlike peripheral many different cell types but was usually interpreted as
lymphocytic.
blood, and unless the specimen is obviously bloody or
cloudy, the fluid is counted undiluted. If it is necessary to * Cell differentials were done on a limited number of cells,
i.e., the number present in the chamber.
dilute the sample, the diluent used must be appropriate for
the type of specimen (i.e., synovial fluid diluent must not ¢ The cells were either unstained or minimally stained to
determine nuclear detail.
contain acetic acid; see discussion on Synovial Fluid.) The
¢ Malignant or reactive cells were impossible to differentiate
diluent must be filtered and free of any background debris
from other cells.
by microscopic verification. Separate dilutions may be
¢ No permanent preparation could be retained for future
required for the RBCs and WBCs to reduce error. The num-
review.
ber of squares (area) to be counted is determined by the
concentration of cells present:* The College of American Pathologists has recommended
¢ If more than 200 cells are seen in a square millimeter of the cytocentrifugation as the preferred method for hematologic
chamber (e.g., a WBC square), the cells in the five RBC analysis.* The obvious advantages include (Table 30-1):
squares of the center square should be counted. * Its relative ease and speed of preparation
¢ If more than 200 cells are seen in the entire ruled area (..e., * Cell differentiation is done on a concentrated preparation
all nine squares), the cells in each of the four corner WBC (good cell recovery).
squares should be counted. * Cell differentiation is determined via Romanowsky (Wright;
¢ If fewer than 200 cells are seen in the entire ruled area, the Wright-Giemsa) staining.
cells in all nine squares should be counted. ¢ Normal, reactive, and malignant cells can be identified with
Neutrophils e
Lymphocytes
Lymphocytes are often seen in all types of fluids in variable
numbers. They can vary in appearance from small to large in
size, and normal to reactive in their nuclear morphology. The
nuclei are artifactually more prominent in cytocentrifuge
preparations. The shape of the nucleus may be irregular
and the cytoplasm may have artifactual projections (see
Figs. 30-2 and 30-3).* Lymphocyte morphology in malig- a
nancyis determined by the type of neoplasm involved, el
i.e., leukemia or lymphoma. The lymphocytes seen in these
Figure 50-5 @ Macrophages. Also a lymphocyte (A) and a plasma
conditions will appear homogeneous, as they are clonal in cell (B). Wright stain, x1000 magnification. (Courtesy of Judith
origin. Plasma cells, if present, are usually seen only in Brody, M.D., North Shore-Long Island Jewish Health System Labora-
chronic inflammatory disorders. tories, Lake Success, New York.)
726 Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
Tissue Cells
Synovial cells have a similar appearance to mesothelial
Benign tissue cells are seen in all fluids and must be differen-
cells, although the cytoplasm appears denser than the
tiated from malignant cells. Pleural, pericardial, and peri-
mesothelial cell. Cartilage cells (chondrocytes) may be seen
toneal fluids all contain benign mesothelial cells, as they are
in a variety of arthritic conditions. These cells contain a cen-
normally sloughed from the membrane. These appear as large
tral, small, round, pyknotic nucleus surrounded by a clear
cells with moderate to abundant cytoplasm that may be
zone and a distinct burgundy colored cytoplasm.°
stained a light or dark blue and may contain phagocytosed
debris or granules. The nucleus is eccentric with a smooth
nuclear outline, and a fine, homogeneous chromatin pattern. Eosinophils, Basophils, Mast Cells
Nucleoli may be large and prominent, usually uniform in size
and shape (Figs. 30-7 and 30-8). The appearance of mesothe- Eosinophils, basophils, or mast cells may be present in
lial cells can vary within the same fluid specimen, which may small numbers in pericardial, pleural, peritoneal, or syn-
cause some difficulty in identification (Fig. 30-9). ovial fluids. Increased numbers can be seen in various dis-
CSF tissue cells (choroid plexus cells, ependymal cells) orders, and their presence may or may not be correlated
tend to form clusters. They may have cytoplasmic granules with their concentration in the peripheral blood. Mast cells
and slightly irregular nuclei. Arachnoid mater cells are fre- can be distinguished from basophils by having a round
quently seen as a syncytium: a mass of cytoplasm containing nucleus (not segmented) and a higher number of cytoplas-
several nuclei. These benign cells are usually only seen in the mic granules that are smaller in size than those usually seen
CSF from infants or adults who have had recent neurosurgery in basophils.*
or an implanted reservoir to deliver chemotherapeutic agents
or antimicrobials directly into the CNS.*
Pleural, Pericardial, and Peritoneal laboratory parameters most frequently characterize a fluid as a
(Serous) Fluids transudate:
Form as a an ‘of7 Goillacy hydride pressure Form as a eae of ihee Secneapninesor : emipnare
or J plasma oncotic pressure resorption
Clear, pale yellow; do not clot Cloudy, turbid, purulent, bloody; clots on standing
WBC: <1000 cells/uL WBC: > 1000 cells/uL
Specific gravity: =1.015 Specific gravity: >1.015
Total protein: =3.0 g/dL Total protein: >3.0 g/dL
Fluid/serum TP ratio: <0.5 Fluid/serum TP ratio: >0.5
LDH: <200 IU/L LDH: >200 IU/L
Fluid/serum LDH ratio: <0.6 Fluid/serum LDH ratio: >0.6
Glucose: ~serum/plasma glucose Glucose: <serum/plasma glucose
Cholesterol: <60 mg/dL Cholesterol: >60 mg/dL
Fluid/serum cholesterol ratio: <0.3 Fluid/serum cholesterol ratio: >0.3
728 Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
frequently clot on standing owing to the presence of fibrino- categorized as benign/reactive when the cellular abnormalities
gen. If a portion of the sample is centrifuged, a clear super- are caused by infection, inflammation, or other sterile reactive
natant indicates the presence of an abundance of leukocytes processes such as pulmonary infarction or cirrhosis of the liver.
or cellular debris; a white supernatant is caused by the pres- Serous fluids associated with bacterial infections will demon-
ence of lipids. strate an inflammatory cellular response that, on differential
If the fluid has a milky or opaque appearance that analysis, consists predominantly of segmented neutrophils.
remains in the supernatant after centrifugation, this is known These cells may reveal Dohle bodies, toxic granulation, and/or
as a chylous effusion, an exudate resulting from leakage or a vacuolization. Viral, fungal, or mycobacterial infections may
blockage of the lymphatic vessels. It is rich in chylomicrons, be associated with a predominance of lymphocytes or show
has an elevated triglyceride level (greater than 110 mg/dL), a mixed inflammatory response. These lymphocytes are
and contains predominantly lymphocytes. Chylous effusion frequently reactive and transformed, resembling immunoblasts.
most often results from a malignancy such as lymphoma or Benign/reactive lymphocytes consist of a heterogeneous
carcinoma, or from trauma. population of cells with varying nuclear shape, amount of
An exudate unrelated to the lymphatics, known as a cytoplasm, and degree of cytoplasmic basophilia, whereas
pseudochylous effusion, results from a persistent, chronic malignant lymphoma cells would appear as a homogeneous
effusion due to diseases such as tuberculosis (TB) and population, most with the same nuclear and cytoplasmic fea-
RA-associated inflammation of the pleura (i.e., rheumatoid tures. Eosinophilic responses can occur with parasitic or fungal
pleuritis). Unlike a chylous effusion, this exudate is caused infections, hypersensitivities, pneumothorax, some immune dis-
by the breakdown of cellular lipids, does not contain chy- orders, and peritoneal dialysis. Many will have an inflammatory
lomicrons, and usually has a low triglyceride level (less cell response that is not diagnostic for any specific disorder;
than 50 mg/dL). Its appearance is caused by cellular debris these would be referred to as nonspecific, reactive cellular
and cholesterol crystals, and contains a mixed reactive cell responses (Table 30-6).
population with many inflammatory and necrotic cells In rare cases, lupus erythematosus (LE) cells form spon-
(Table 30-5). taneously. An LE cell is a phagocyte, either neutrophil or
Trauma, aneurysm, malignancy, pancreatitis, or TB may monocyte, that has phagocytosed a naked nucleus showing a
also produce a bloody, or hemorrhagic effusion. homogeneous, smooth chromatin pattern. This finding is sus-
The aforementioned analyses are not always able to picious but not diagnostic of SLE; other autoimmune disor-
definitively distinguish transudates from exudates when the ders may also show this phenomenon.
results are equivocal; the patient’s clinical history and Mesothelial cells, as previously described, can have a
physical examination can provide further information. The wide variation in morphology. They may show nonspecific
finding of an exudate must always be followed by more reactive changes that include multinuclearity, the presence of
extensive tests to determine its cause. nucleoli, mitotic activity, and sometimes an increase in cell size
(Figs. 30-10 and 30-11). They have been classified into cate-
gories, based on their diverse morphology as quiescent; hyper-
Cellular Responses, Microorganisms, trophied; epithelioid; phagocytic; and senescent* (Table 30-7).
and Malignant Cells in Serous Fluids Quiescent mesothelial cells are the normal constituents of the
membrane that are shed into the fluid (see Fig. 30-7). Hyper-
Effusions can occur as a result of a wide variety of local or sys- trophied cells are larger, with more prominent nucleoli, and can
temic disease processes. They may be categorized as normal be seen in reactive processes that cause cellular hyperplasia
cellular, benign/reactive, or malignant, based on the cell types (Fig. 30-12). Epithelioid mesothelial cells are seen as clumps
present. Normal cellular effusions, as their name implies, are or sheets, owing to the disrupted membrane surfaces in reactive
neither reactive nor malignant; the cell morphology is normal processes (Fig. 30-13). Phagocytic mesothelial cells are indis-
regardless of the cause of the effusion. A serous fluid is tinguishable from macrophages or histiocytes (Fig. 30-14).
° Cryptococcus spp.
¢ Histoplasma spp.
The senescent mesothelial cell is degenerated, with a single ¢ Candida albicans
large vacuole forming a signet ring configuration (Fig. 30-15). * Candida tropicalis
The nucleus may show pyknosis and karyorrhexis: one or more
spherical, densely staining nuclear fragments. All except quies- Malignancy is a major cause of serous effusion; therefore,
cent mesothelial cells can closely resemble malignant cells. pleural, pericardial, and peritoneal fluids may contain malig-
Extreme caution must be taken to avoid misinterpretation; nant cells from a variety of neoplasms. Their identification is
referral to pathology and/or cytology is required. critical for accurate diagnosis. A malignancy may already be
Most types of pathogenic bacterial and fungal organisms known to exist, but its presence in the fluid demonstrates
will stain with Wright and Wright-Giemsa stain, and are metastasis; it may also be an initial diagnosis. The laboratory
detectable on a routine cytocentrifuge preparation. Bacteria professional must look at the entire cellular area of the slide
will stain blue regardless of the Gram’s stain reaction under low power to detect suspicious clusters of cells. It is not
(Fig. 30-16). If the organisms are intracellular (Fig. 30-17), possible to distinguish malignant cells from benign reactive
discernible as being in the same plane of view as the cell via
Pigure 50-12 ® Hyperplastic mesothelial cells. Wright stain, Figure 30-15 @ Senescent mesothelial cell (right); becoming
400 magnification. (Courtesy of Judith Brody, M.D., North signet-ring configuration. Wright stain, *1000 magnification.
Shore-Long Island Jewish Health System Laboratories, Lake Success, (Courtesy of Judith Brody, M.D., North Shore-Long Island
New York.) Jewish Health System Laboratories, Lake Success, New York.)
Figure 50-14 @ Phagocytic macrophages/mesothelial cell. Note Figure 30-17 m Bacteria in body fluid: Neisseria species; note the
the ingested RBCs (Jeft). Wright stain, x1000 magnification. intracellular diplococci in neutrophils. Wright stain, 1000 magnifi-
(Courtesy of Judith Brody, M.D., North Shore-Long Island Jewish cation. (From Best, M: Body Fluids Examination. ASCP Workshop
Health System Laboratories, Lake Success, New York.) 9294, Boston, 1990, with permission.)
Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis 731
Figure 350-18 @ Clusters of malignant cells. Wright stain, x1000 Figure 50-21 ® Nuclear molding. Wright stain, 1000 magnifica-
magnification. (From Best, M: Body Fluids Examination. ASCP Work- tion. (From Best, M: Body Fluids Examination. ASCP Workshop
shop 9294, Boston, 1990, with permission.) 9294, Boston, 1990, with permission.)
Large, frequently >50 um
Giantism
High/variable N:C ratio (21:1)
Nuclear Multinucleation; variable size
Pleomorphic, nonuniformity
Irregular nuclear membrane;
herniation into cytoplasm; clefts
Nuclear hyperchromasia
Unevenly distributed chromatin
Nuclear molding
Figure 350-22 ™ Abnormal mitotic activity; acinar (glandular) Prominent nucleoli with irregular
arrangement of malignant cells. Wright stain, x1000 magnification. size and shape
(From Best, M: Body Fluids Examination. ASCP Workshop 9294, Increased/abnormal mitotic
Boston, 1990, with permission.) activity
Cytoplasmic Uneven staining of cytoplasm
Bizarre vacuolization or large,
singular vacuole
Abnormal inclusions
Fused cytoplasmic membranes
Cannibalism
Cellular Arrangement Multilayered formations
3-D forms
Spherical aggregates
Cell clumps lacking, or having
- poorly defined margins
Wraparound, rosette or acinar
formations
Single cells
can occur with atelectasis [collapsed lung]), can lead to the for- The abnormal accumulation of fluid in the pericardial
mation of a pleural effusion. The presence of large amounts of space is known as a pericardial effusion. This is most fre-
fluid may restrict lung expansion, causing dyspnea and mild quently caused by damage to the lining of the cavity and
hypoxemia. Pleural effusions caused by congestive heart fail- increased capillary permeability. The function of the normal
ure, nephrotic syndrome, or cirrhosis tend to be bilateral, pericardium is to oppose dilatation of the heart; this is
whereas unilateral effusions are caused by diseases that reflected by the mean central venous pressure. Interference
develop below the diaphragm, such as cirrhosis with ascites, with pericardial venous and lymphatic drainage, which occurs
hepatic abscess, pancreatitis, and tumors.° in acute pericarditis, can lead to the formation of an effusion.
The volume of fluid, the rate of its formation, and the elastic-
SPECIMEN COLLECTION AND PROCESSING ity of the pericardial membrane will determine what effect the
Analysis of a pleural effusion begins with the removal of fluid effusion will have on cardiac function. A small effusion may
by a procedure known as thoracentesis (Table 30-9). Portions have no effect and not produce any symptoms; a large effu-
of the pleural or thoracentesis fluid, as it may also be classi- sion that develops very slowly may also not cause any symp-
fied, are placed into heparinized tubes for chemical analysis, toms provided that the pericardium has the ability to stretch.
sterile tubes for microbiological cultures and Gram and/or A severe complication of pericarditis or a traumatic injury can
acid-fast stains, and tubes containing ethylenediaminete- cause a condition known as cardiac tamponade. This occurs
traacetic acid (EDTA) for cell counts and differential analysis. when pericardial fluid or blood within the pericardial space
Additional nonadditive evacuated tubes may also be collected under increased pressure restricts the motion of the heart. This
for cytology and immunophenotyping studies. Thoracentesis produces a state of critical cardiovascular dysfunction, caus-
may also be used for therapeutic purposes. ing decreased cardiac output and hypotension, and if left
The gross appearance of the pleural fluid provides untreated will cause death. Treatment requires the aspiration
important diagnostic clues to the nature of the disease of the pericardial fluid in a procedure known as pericardio-
process. Pure blood in the pleural cavity, a hemothorax, can centesis (see Table 30-9). A portion of the aspirate is sent to
result from severe chest injuries, stab or gunshot wounds, or the laboratory for analysis as discussed earlier for thoracente-
surgical procedures. A bloody, or hemorrhagic, effusion in the sis. A wide variety of diseases and disorders can produce peri-
absence of trauma almost always suggests the presence of cardial effusions, such as neoplastic disease, infectious
malignancy, or occasionally a pulmonary infarction. Leakage agents, cardiovascular disease, renal disease, collagen vascu-
of the thoracic duct causes a chylothorax, most commonly lar diseases, and hemorrhagic events (e.g., trauma, aneurysm,
caused by a malignancy such as lymphoma or carcinoma.'° or anticoagulant therapy.)
This produces a milky-white, opaque pleural fluid that
remains opaque after centrifugation. Infection of the pleural LABORATORY ANALYSIS AND CLINICAL
space by bacterial pneumonia or a ruptured lung abscess will CORRELATIONS
cause empyema: the collection of pus in the pleural cavity. QUALITATIVE ANALYSIS The hematology laboratory will
This fluid will be turbid to opaque and have an extremely receive the body fluid in tubes containing EDTA, which pre-
elevated WBC count of 25,000/uL or higher. After centrifuga- vents clotting and preserves the cell morphology for analysis.
tion, the supernatant of this type of fluid will be clear due to If the fluid is completely clotted no analysis can be per-
the removal of the cellular debris. formed. If the specimen is partially clotted, which can occur
if the specimen is not properly mixed after collection, the cell
count would be inaccurate, but an attempt should be made at
performing the morphological examination for the presence
‘tabieso-0 Body Fluid = —t™S of reactive or malignant cells, with the required physician
a nd Collection notification and comment on the report.
_ Procedure Gross visual inspection of the fluid is initially per-
formed. A normal pleural fluid is pale yellow in color and
Nomenclature> transparent. Abnormal findings would be cloudy, turbid,
bloody, yellow, green, or chylous. If the fluid is grossly
Body Cavity/ Collection
Region Fluid Name Procedure bloody, and a microhematocrit performed on the specimen
has a packed cell volume (PCV) greater than 50% of the
Pleural cavity/lungs Pleural (serous) |Thoracentesis peripheral blood PCV, this would indicate the presence of a
Pericardial cavity/ Pericardial Pericardiocentesis hemothorax.
heart (serous)
Peritoneal cavity/ Peritoneal, Paracentesis
A normal pericardial fluid is pale yellow in color
abdomen ascites and transparent. Turbidity can be caused by infection or
(serous) malignancy. Grossly bloody fluid may result from trauma
CNS; subarachnoid — Cerebrospinal Lumbar puncture; or disease, or the possibility that intracardiac blood was
space/brain spinal tap
aspirated into the specimen during pericardiocentesis. The
and spinal cord
Arthrocentesis findings of a chylous fluid are rare, but can be caused by
Synovial cavity/ Synovial
joints the leakage of lymphatic vessels from trauma, lymphoma,
or carcinoma.
734 Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
into the pleural space. By itself, it is not diagnostically signif- having a more pronounced irregularity in size and shape of
icant; it may also be manifested in parasitic or fungal the nuclei and nucleoli, uneven chromatin distribution with
diseases, malignancy, connective tissue disorders, hypersensi- hyperchromasia, and irregular cellular arrangements (see
tivities, or pulmonary infarction. The presence of eosinophilia Table 30-7).
in an exudative effusion is an indication that the condition is Increased concentrations of mesothelial cells are seen in
probably not malignant or tuberculous. Mast cells and pneumonia, pulmonary infarction, and malignant disorders. A
basophils often accompany eosinophils, and 5% to 10% may marked increase, especially if the fluid is bloody or
be seen in pleural fluid eosinophilia’ (see Table 30-6). eosinophilic, suggests the presence of a pulmonary embolism.
Mesothelial cells in small numbers are normally However, they are usually markedly reduced in number in
sloughed into the serous cavities. During inflammatory cases of TB of the pleura (i.e., tuberculous pleurisy) or if there
processes, they proliferate and are shed in greater numbers. is massive infection of the pleural cavity with pyogenic (pus-
They often vary in appearance, manifesting reactive or atypi- producing) organisms; this may be related to the fibrinous
cal changes, which can create difficulty during the differential exudate that covers the lining of the cavity, trapping the cells?
examination (see Figs. 30-12 and 30-13); familiarity with (see Table 30-6).
these issues is necessary when examining the slides.
Mesothelial cells may appear as single cells, in clusters, or Peritoneal Effusions
sheets. Clustering can be an artifact caused by cytocentrifuga-
tion that may produce a resemblance to malignant cells. SPECIMEN COLLECTION AND PROCESSING
Clumps, or loose aggregates of benign mesothelial cells, can A peritoneal effusion is an accumulation of fluid in the peri-
be differentiated from malignant cells by comparing the toneal space, clinically termed ascites. The procedure to with-
appearance of those in the clump with other more easily dis- draw this fluid is known as paracentesis (see Table 30-9). The
tinguished mesothelial cells in the same smear. A uniform, collected fluid can be called peritoneal, ascitic, or paracente-
regular arrangement of cells that display fenestrations: open- sis fluid. Paracentesis is most commonly performed to rule
ings or windows between the cytoplasmic membranes of the out bacterial peritonitis and malignancy. Aspiration may be
cells, usually indicates that they are benign. Spheroidal or combined with /avage: a flushing of the peritoneal space with
multilayered cell formations are more likely to be malignant lactated Ringer’s solution. Evidence of blood in the lavage
(see Figs. 30-18 and 30-19). The cells are large, with scant to fluid is an indication for immediate surgical exploration
abundant light gray to deep blue cytoplasm, and may present (laparotomy) of the abdominal cavity for internal bleeding.
an area of perinuclear pallor; as such, they may resemble Because the abdominal cavity is large and distendable,
large plasma cells. There may also be variably sized cytoplas- more than approximately 500 mL of fluid must usually be
mic vacuoles. The nucleus occupies one third to one half the present before the effusion can be detected by radiologic or
cell’s diameter, and may be round to oval in shape with a physical examination. Diagnostic abdominal paracentesis,
smooth nuclear membrane. The chromatin distribution is uni- with the removal of 50 to 100 mL of fluid, is essential to
form, with a stippled, dark purple Wright stain effect. There determine a differential diagnosis. A portion of this is sent to
may also be one to three spherical nucleoli. Mesothelial cells the laboratory for analysis in the same manner as discussed
that could be classified as “reactive” have slightly irregular previously for pleural and pericardial fluids.
nuclei with prominent nucleoli (Fig. 30-27). Atypical The causes of peritoneal effusions are the same as those
mesothelial cells more closely resemble malignant cells, involved in pleural and pericardial effusions: increased capillary
736 Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
pressure or permeability, abnormal colloid osmotic pressure, Chylous fluid is rare, but can be caused by the leakage of lym-
poor lymphatic drainage, or cardiac abnormalities. Additional phatic vessels due to trauma, hepatic cirrhosis, TB, lymphoma,
causes include abdominal conditions that do not directly involve or carcinoma.
the peritoneum, such as hepatic cirrhosis, intrahepatic and por-
tal venous obstruction, hypoalbuminemia, renal function, ovar- QUANTITATIVE MICROSCOPIC AND MORPHOLOGIC
ian disease, pancreatic disease, or parasitic infection. Malignant ANALYSIS The total RBC and WBC counts are useful for
disease and alcoholic cirrhosis are the most common causes of diagnosis in peritoneal lavage, as they improve the accuracy
peritoneal effusion." and specificity of the diagnosis. A positive lavage fluid would
Patients in renal failure, who must undergo dialysis to be visibly bloody, and considered as such when the total RBC
cleanse the blood of waste products, can be treated with a pro- count is greater than 100,000/uL (or greater than 50,000/uL in
cedure known as continuous ambulatory peritoneal dialysis known penetrating trauma such as stab or gunshot wounds).
(CAPD). This procedure utilizes the natural properties of the The total WBC count would be greater than 500/uL. How-
peritoneal membrane and the infusion of a dialyzing fluid into ever, when indeterminate by eye, a lavage would be consid-
the cavity to remove the impurities in the bloodstream. This ered positive when the total RBC count is 50,000 to
procedure requires laboratory monitoring to measure its 100,000/uL (10,000 to 50,000/uL in cases of penetrating
effectiveness, and carries an inherent risk of recurrent peri- trauma).'! The total WBC count would be 100 to 500/uL. A
tonitis. The used dialysis fluid (dialysate) can be sent to the negative lavage fluid would show a total RBC count less than
laboratory for analysis. 50,000/uL (or less than 1000/uL in penetrating trauma). The
total WBC count should be less than 100/uL.
LABORATORY ANALYSIS AND CLINICAL Cell counts on nonlavage peritoneal fluids, specifically the
CORRELATIONS total WBC count, are of limited value in differential diagnoses,
QUALITATIVE ANALYSIS The hematology laboratory will but are useful in distinguishing peritoneal transudates from
receive’ the fluid in tubes containing EDTA, which prevents exudative effusions. A WBC count greater than 300/uL is con-
clotting and preserves the cell morphology for analysis. A sidered to be abnormal. It is extremely necessary to make a
clotted fluid cannot be analyzed. As with pleural and pericar- correct diagnosis as early as possible in suspected spontaneous
dial fluids, a partially clotted specimen would not provide an bacterial peritonitis (SBP) to reduce the morbidity and mortality
accurate cell count, but morphological examination for the associated with it. A WBC count greater than 500/uL is useful
presence of reactive or malignant cells should be performed, presumptive evidence in distinguishing between an exudate in
with the required physician notification and comment on the bacterial peritonitis and a transudate of cirrhosis. Serial WBC
report. counts and cultures are helpful in assessing the adequacy of
The criteria previously described to differentiate transu- treatment in patients with SBP and also in differentiating SBP
dates and exudates may not fully apply for ascitic fluid as they from nonperforation secondary bacterial peritonitis."
do for pleural and pericardial fluids. The total protein and LDH A Wright-stained cytocentrifuge preparation is used for
ratios may be better indicators in most cases (see Table 30-4). the differential examination. The same procedure, as described
A more reliable method is known as the serum-ascites albu- earlier in the discussion of pleural and pericardial effusions, is
min gradient (SAAG), calculated by subtracting the ascitic followed for the morphologic examination. An exudative peri-
fluid albumin concentration from the simultaneously col- toneal effusion characteristically contains a variable number of
lected serum albumin concentration.'*? The SAAG is signifi- neutrophils, lymphocytes, eosinophils, basophils, macrophages,
cantly greater than 1.1 g/dL in transudates (high-gradient and mesothelial cells.
ascites) than in exudates.'* It can also provide valuable infor- The presence of more than 25% segmented neutrophils is
mation to assist in the differential diagnosis of ascites: considered abnormal and suggestive of bacterial infection. The
e.g., patients with high-gradient ascites include those with absolute neutrophil count may also be helpful; counts greater
cirrhosis, alcoholic hepatitis, cardiac-related ascites, or mas- than 500 neutrophils/uL are a fairly sensitive indicator of spon-
sive liver metastasis. Low-gradient ascites (SAAG less than taneous or secondary bacterial peritonitis. In CAPD, peritonitis
1.1 g/dL) are associated with peritoneal malignancies and is defined by a WBC count of greater than 100/uL with more
nonmalignant diseases such as TB, pancreatitis, nephrotic than 50% neutrophils, or if microorganisms are seen on the
syndrome, biliary disease, or connective tissue diseases.'* cytocentrifuge preparation and/or Gram stain of the dialysate."!
Normal or transudative peritoneal fluid is pale yellow or straw Chronic, long-standing effusions may show neutrophils with
colored, and transparent. The visible appearance of the fluid decreased cytoplasmic granules, and nuclear pyknosis and kary-
may be helpful in determining the cause of the effusion. orrhexis as in pleural or pericardial effusions.
Abdominal blunt trauma, postoperative complications, rupture Lymphocytes transform in response to various stimuli
of the liver or spleen, intestinal infarction, pancreatitis, or malig- and exhibit a variety of morphologic features. Differentiation
nancies can cause the production of a grossly bloody fluid. of benign lymphocytes from malignant lymphoma in effusions
Effusions resulting from perforation of the gall bladder or may occasionally be difficult. A predominance of lymphocytes
intestines, duodenal ulcers, cholecystitis, or acute pancreatitis can be seen in transudates from patients with congestive heart
will appear green owing to the presence of bile. Exudative effu- failure, cirrhosis, or the nephrotic syndrome; they may also be
sions will be cloudy or turbid if they contain elevated protein seen in chylous effusions, tuberculous peritonitis, and other
concentrations, increased leukocytes, and/or microorganisms. malignant disorders.
Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis 137
Eosinophilia in peritoneal fluid is less commonly seen CSF collection is commonly performed by intervertebral
than in pleural fluid. Eosinophilic ascites is rare; when present, puncture in the lumbar region of the spinal column, between
it contains greater than 50% eosinophils. It is associated with vertebrae L3 and L4, known as a lumbar puncture (LP) or
congestive heart failure, vasculitis, and malignant lymphoma.'° “spinal tap” (see Table 30-9). The procedure must be per-
Patients with CAPD may exhibit peritoneal eosinophilia, usu- formed aseptically and without trauma. In patients with coagu-
ally greater than 10% or as high as 95%.'° The cause is lopathies or infection at the puncture site, LP is contraindicated.
unknown, but may be caused by hypersensitivity to the foreign If the patient has a normal intracranial pressure, determined at
material, or the possibility that air entered the cavity during the initiation of the LP, at least 20 mL of fluid can be removed
the connection of the catheter line to the infusion set!’ (see in an adult without complication. Significantly lower volumes
Table 30-6). Small numbers of basophils and mast cells may may be collected from infants, children, and neonates, or if the
also be seen in cases of peritoneal eosinophilia. intracranial pressure is elevated. Approximately 2-4 mL of fluid
It is possible to find choroid plexus cells from the CNS is collected into each of three to five sequentially numbered,
in the peritoneal fluid of a patient with a ventriculoperitoneal sterile, nonadditive tubes. The tubes must be filled in numerical
shunt: a drainage device inserted into the ventricles of the order. The first tube is used for chemical analysis, because it
brain to remove excess CSF in cases of hydrocephalus, neo- typically would be contaminated with peripheral blood and cel-
plastic conditions, or head injury. The catheter is placed under lular debris from the initiation of the puncture, and requires
the skin, from the skull to the abdomen, allowing the excess centrifugation before analysis. The second tube is used for
fluid to drain into the peritoneal cavity where it is reabsorbed. microbiological analysis. The third tube is sent to hematology
A one-way valve controls the flow of the fluid. for the cell count and differential analysis. Additional tubes
Mononuclear phagocytes are present in variable num- may be used for further chemistry analyses, serology, cytology,
bers. These cells, as well as mesothelial cells, may have a flow cytometry, immunocytochemistry, or molecular genetic
variable appearance. As discussed previously under pleural analysis. An LP may also be performed for therapeutic pur-
and pericardial fluid analysis, differentiation from malignant poses, such as to reduce intracranial pressure, or to administer
cells may be difficult. Atypical mesothelial cells, resembling anesthetics, radiographic contrast media, or antifungal or
malignant cells, are especially seen in chronic effusions and chemotherapeutic agents directly into the CNS. A whole blood
in ascitic fluid associated with cirrhosis.'* LE cells have also sample is also collected at the time of LP for a complete blood
been reported. Referral to pathology and cytological exami- count (CBC) and chemical analysis of the serum.
nation must be performed if a malignancy is suspected. On receipt in the laboratory, the CSF, also known as
“spinal fluid,’ must be processed immediately (STAT); analysis
must occur within | hour of collection owing to cellular degra-
Cerebrospinal Fluid dation and lysis, which will lead to falsely lower WBC and RBC
counts. The lower protein content of CSF leads to destabiliza-
The CSF is a selective ultrafiltrate of plasma, under the con- tion of the cell membranes. Any specimen remaining after
trol of the blood-brain and blood—CSF barriers, and differs analysis must be refrigerated in case additional tests are neces-
from serous fluids in that a greater volume is present in the sary. An exception to this procedure applies only to specimens
normal state. It is also actively secreted by cells in the ventri- for microbiological examination: these must not be refrigerated,
cles and subarachnoid space. Examination of CSF is an as some microorganisms (e.g., Niesseria meningiditis) are
extremely important diagnostic procedure, and its acquisition destroyed by low temperatures. It is preferable to place them in
is not without risk to the patient. All specimens must be han- a 37°C incubator until testing is performed, especially during
dled with universal precautions and never discarded until all off-hours when the bacteriology department may not be staffed.
testing has been completed.
the analysis of CSF is distinguishing between a true CNS removal of large volumes of CSF during radiologic proce-
hemorrhage versus a traumatic LP. If the bloodiness decreases dures, hyperthyroidism, leukemias, and in normal children
with each successive tube, this would suggest a traumatic tap. 6 months to 2 years of age.
If the bloodiness is similar for all tubes, this could indicate a Albumin, a constituent of TP, is not synthesized in the
CNS hemorrhage. However, this method is not always CNS, but is transported across the blood—CSF barrier. A
reliable.*' method to evaluate the integrity (degree of permeability) of
A clotted CSF is an abnormal finding; it suggests an the blood—CSF barrier is to calculate a ratio of the serum
increased fibrinogen and/or protein concentration from periph- albumin to the CSF albumin, which should normally be less
eral blood contamination, an infectious condition within the than 9. If greater than normal, the albumin originated from the
CNS, or a markedly increased protein concentration in the fluid. blood due to a damaged blood—CSF barrier, intracerebral
The presence of any color should be noted. If the speci- bleeding, or traumatic LP. Electrophoretic analysis can
men is bloody, the color of the supernatant is interpreted after also be used to visualize and quantify the individual protein
centrifugation. This must be performed as soon as all the cell fractions.
counts are completed. Xanthochromia, a pink, orange, or yel- The IgG concentration is measured to determine the
low color of the supernatant, is caused by the breakdown of amount of production within the CNS, normally about 3 mg/
hemoglobin. It is usually thought to indicate a true CNS hem- day. It is the predominant immunoglobulin in the CSE. It is eval-
orrhage, and is present in more than 90% of patients within uated to assist in determining a diagnosis of MS or other
12 hours of subarachnoid hemorrhage onset.** Lysis of RBCs demyelinating diseases. The IgG may originate from the blood
present in the CSF begins approximately | to 2 hours after a if the integrity of the blood—CSF barrier is disrupted, or from
subarachnoid hemorrhage. Hemoglobin is converted to biliru- increased synthesis by lymphocytes and plasma cells within the
bin within 24 hours of RBC lysis, giving the supernatant a CNS. To correct for possible increased permeability, increased
yellow tint. If the fluid supernatant is clear and colorless, this serum IgG and albumin concentrations, and to isolate intrathe-
could suggest that either the LP was traumatic,, the CNS cal IgG production, the CSF IgG index is calculated.
hemorrhage occurred less than 2 hours before the LP, or min-
_ CSF IgG X serum albumin
imal RBC lysis has occurred. Xanthochromia will occur, acre eat
however, if a bloody fluid from a traumatic LP is not
processed within | hour of collection. Other causes of xan- All components must be from simultaneously collected
thochromia include jaundice (serum bilirubin levels of 10 to samples. An approximate normal index value would be less
15 mg/dL), extremely elevated CSF protein concentrations than or equal to 0.7; this is highly dependent on the methodol-
(greater than 150 mg/dL), and hyperbilirubinemia in prema- ogy and normal cut-offs for the four analytes. An elevated
ture infants who have an immature blood—CSF barrier. index greater than 0.7 would be seen with increased intrathecal
IgG synthesis, which could occur in MS, subacute sclerosing
BIOCHEMICAL ANALYSIS pan-encephalitis, bacterial or viral meningitis/meningoen-
Biochemical analysis of CSF routinely includes the measure- cephalitis, Guillain-Barré syndrome, neurosyphilis, SLE affect-
ment of the total protein (TP) and glucose levels, with com- ing the CNS, or other neurologic conditions. In addition, more
parison to the simultaneously collected serum levels. Other than 90% of patients with MS have two or more discrete bands
analyses may include albumin, IgG, and LDH; the clinical sit- in the gamma-globulin fraction on CSF electrophoresis which
uation would determine when these would be performed. is not present on serum protein electrophoresis. These are
There are many other more complex tests and electrophoretic known as oligoclonal bands; however, they are not specific for
procedures that can be performed, but these are beyond the MS. Diagnosis also requires neurologic history and physical
scope of this chapter. examination.
Protein enters the CSF by diffusion across the blood— Glucose in the CSF is entirely derived from the blood,
CSF barrier. Some CNS synthesis of protein also occurs, but is normally only about 60% to 70% of serum levels. It
mostly in the form of IgG. The normal CSF TP level in adults enters almost completely by facilitated diffusion in the
is approximately 15 to 45 mg/dL, which is less than 1% of the choroid plexus.'? Changes in the blood glucose concentration
serum level. Higher levels (up to 100 mg/dL) are possible in may require 2 to 4 hours to equilibrate with the CSF. Ideally,
neonates owing to immaturity of the blood—CSF barrier. CSF and blood specimens should be drawn in the fasting
These values are also method dependent and each laboratory state; however, this is not always clinically possible. The
must determine its own range. An increased TP is the most analysis must be performed immediately to avoid artifactual
frequent, although nonspecific, abnormality found in CSF decreases caused by the presence of leukocytes and/or bacte-
analysis. Abnormally increased CSF TP is seen in meningeal ria. Without knowing the actual serum glucose level, an
and CNS disease (e.g., increased permeability of the blood— acceptable range for CSF glucose should be 50 to 80 mg/dL,
CSF barrier, meningitis, traumatic LP, CNS hemorrhage, assuming a normal fasting serum glucose range of 65 to
multiple sclerosis [MS], obstruction to CSF circulation, 110 mg/dL.** A traumatic LP will cause a false elevation due
neoplasm, or cerebral infarction). As little as 0.1 mL of blood to the introduction of peripheral blood. Patients with hyper-
in | mL of CSF can increase the TP by 400 mg/dL. Low TP glycemia may have higher CSF glucose values depending on
levels are associated with CSF leakage due to skull fracture, the timing of the last carbohydrate load; however, the kinetics
increased intracranial pressure due to water intoxication, of glucose transport changes as the blood glucose level
Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis 739
Figure 30-31 m Reactive (plasmacytoid) lymphocytes in CSF in Figure 50-32 ™@ Siderophages in CSF: hemosiderin pigment in
‘multiple sclerosis. Wright stain, x1000 magnification. (From Best, M: macrophages (A). Wright stain, 400 magnification. (From Best, M:
Body Fluids Examination. ASCP Workshop 9294, Boston, 1990, with Body Fluids Examination. ASCP Workshop 9294, Boston, 1990, with
permission.) permission.)
742 > Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
Figure 50-55 @ Siderophage with hematoidin (hematin) Figure 30-34 @ Eosinophils (A) and reactive lymphocytes (8) in
pigment. Wright stain, 1000 magnification. (From Best, M: Body CSF; ventricular shunt. Wright stain, 1000 magnification.
Fluids Examination. ASCP Workshop 9294, Boston, 1990, with (Courtesy of Judith Brody, M.D., North Shore-Long Island
permission.) Jewish Health System Laboratories, Lake Success, New York.)
Plasma cells are normally not present in CSF. In inflam- sheets or as single cells are helpful features in their identifica-
matory conditions associated with lymphocytic reactions, tion. Occasionally, the nucleus may be pyknotic and eccentri-
transitional stages of reactive lymphocytes, plasmacytoid cally placed. The cytoplasm is moderate to abundant, stains a
lymphocytes, and plasma cells can be seen. These would gray-blue with Wright stain, and may contain vacuoles;
include acute viral infections, and chronic inflammatory the cytoplasmic borders may be indefinite, and cilia may
states; TB, syphilis, sarcoidosis, Guillain-Barré syndrome, occasionally be present.'? When found in cytocentrifuged
and MS. preparations, they may resemble lymphocytes or monocytoid
Other granulocytes that may be seen include eosinophils cells. However, if the fluid contains an increased number of
and basophils. Eosinophils are rarely seen in a normal CSF. lymphocytes and/or monocytes, artifactual clusters of these
They may be increased in a variety of infectious and non- cells may resemble choroid plexus cells.'? Situations or con-
infectious disorders. Eosinophilic meningitis is defined ditions in which these cells may be seen include CSF obtained
when the total WBC count is greater than 10% eosinophils; by ventricular tap, following traumatic brain injury, brain
a parasitic infection should be suspected with this find- surgery, ischemic brain infarction, radiologic procedures, and
ing.° Idiopathic eosinophilic meningitis, without any in children with hydrocephalus and ventricular shunts.'* *? It
evidence of a pathogen, has also been described.*' Other is important to recognize these cells because they can be mis-
infectious causes include viral, fungal, or rickettsial infec- takenly identified as malignant cells; however, they have no
tions. Noninfectious causes include malignancy, intrathe- diagnostic significance.
cal therapy, radiographic contrast media, and systemic LE cells are rarely seen in CSR.**
drug reactions. Malignant cells can be seen in the CSF. Every Wright-
Eosinophilia is also associated with malfunctioning ven- stained CSF slide must be searched for the presence of
tricular shunts or foreign body reactions (Fig. 30-34).
Basophils are normally not seen in CSF. There are conditions
in which they can be found in small numbers, such as inflam-
matory diseases, foreign body reactions, parasitic infections,
convulsive disorders, and chronic myelogenous leukemia
(CML)."°
Ependymal or choroid plexus cells that line the cerebral
ventricles and choroid plexus, sometimes referred to as neu-
roectodermal cells, may be seen in CSF under certain circum-
stances. They are difficult to differentiate, but it is believed
that the majority of the cells seen in the fluid are choroid
plexus cells. They are rarely, if ever, seen in normal CSF
obtained by LP in adults; however, they may occasionally be
found in infants.'° These cells are medium in size, and may
appear in papillary clusters or sheets, or sometimes individu-
ally. The nuclei are the size of a small lymphocyte, round to
Figure 30-35 @ Ependymal or choroid plexus cells (neuroepithe-
oval in shape, with delicate, finely granular, and evenly dis-
lial tissue) in sheet formation. Wright stain, 400 magnification.
tributed chromatin. Nucleoli are not present (Figs. 30-35 and (From Best, M: Body Fluids Examination. ASCP Workshop 9294,
30-36). Their uniformity of nuclear size and appearance in Boston, 1990, with permission.)
Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis 743
Synovial Fluid
Normal synovial fluid is transparent, colorless, viscous, and
does not clot. The protein content is approximately one fourth Appearance Transparent, colorless to straw
that of plasma; glucose, electrolytes, and uric acid are similar to colored
Physical Qualities Viscous; does not clot
plasma in their concentrations. Age, as well as many inflamma-
WBC Count <200/uL
tory and pathologic joint disorders, can alter the volume and Differential Neutrophils: <25%; remainder as
composition of the synovial fluid affecting its function. An lymphocytes, monocytes,
increased volume of synovial fluid is considered to be an effu- synovial cells, macrophages
sion, and classified as inflammatory or noninflammatory, septic, RBC Count None
or hemorrhagic. Inflammatory responses caused by mechanical, Total Protein 1-3 g/dL
Glucose <10 mg/dL serum-fluid difference
chemical, immunological, or bacterial damage change its cellu- Crystals None
lar and chemical constitution. The impaired function of the fluid Uric Acid Equivalent to normal serum level
may play a role in the development of degenerative joint
Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
heparinized tube for microbiological culture and Gram stain; formation. Viscous fluids represent normal or noninflam-
a third portion placed into a sodium heparin-anticoagulated matory states; viscosity decreases with increasing inflam-
evacuated tube for chemical and/or crystal analysis; a mation, caused by lytic enzymes that depolymerize the
fourth portion into a liguid EDTA-anticoagulated tube for hyaluronic acid. This reduces the lubricating ability of the
cell counts and/or crystal examination (powdered EDTA fluid, promoting damage to the joint. If the physician
and lithium heparin can produce crystalline structures that requires, fluid viscosity would be assessed at the time of
can be mistaken for synovial crystals). A normal fluid does aspiration (not in the laboratory), by allowing the fluid to
not clot; this must be centrifuged as soon as possible to drip from the needle onto a microscope slide or by rapidly
remove all the cells. The supernatant fluid can be assayed lifting the needle from the drop of fluid on the slide and
for rheumatoid factor, antinuclear antibody, complement, observing the length of the “string” of fluid that forms. A
glucose, total protein, lactate, uric acid, and other compo- normal fluid forms a string of 4 to 6 cm in length.** As the
nents. Testing must be initiated within one hour of collec- protein—hyaluronic acid complexes are degraded by lytic
tion; cellular degradation and chemical changes occur with enzymes, the viscosity decreases and no string forms.*’
time. Refrigeration is required if any delay in testing is There is no clinically useful information derived by mea-
anticipated. For some analytes, the supernatant may need to suring the fluid’s viscosity.
be frozen if testing cannot occur immediately. If a sufficient The volume of the specimen bears little correlation
volume of fluid is submitted, a routine analysis should to the severity of the joint disease. Some effusions, though
include an examination of a wet preparation for crystals, apparently significant by the amount of swelling present,
a cell count and differential analysis, a Giam stain and may be difficult to aspirate if they contain considerable
culture, and chemical analysis for protein and glucose debris or fibrin content. If a limited volume of fluid is
concentrations. collected, only the most clinically relevant laboratory tests
are requested by the physician, such as a Gram stain and
Laboratory Analysis and Clinical culture, and microscopic examination for leukocytes and
crystals.°
Correlations
The color of the fluid changes with the disease
QUALITATIVE ANALYSIS process; it is dependent on the quantity of albumin, biliru-
The hematology laboratory will receive the fluid in tubes con- bin, cells, and other debris present. A normal fluid may be
taining EDTA, which should prevent clotting, although very colorless to straw colored. Noninflammatory, inflamma-
viscous fluids may form small clots if they are bloody and/or tory, and infectious fluids can have a yellow color, due to
not well mixed. If the specimen is partially clotted, the cell hemoglobin breakdown of RBCs that enter the synovial
count would be inaccurate, but an attempt should be made at cavity during inflammation, or from chromogenic products
performing the morphological examination for the leukocyte of bacteria within the joint cavity. A traumatic aspirate will
differential, with the required physician notification and com- show streaks of blood in the fluid. Hemarthrosis, hemor-
ment onthe report. If the fluid is completely clotted, no analy- rhage within a joint, produces a homogeneously bloody
sis can be performed. fluid. This can be caused by disease (e.g., hemophilia) or a
Once the specimen is received by the laboratory, it traumatic injury. Centrifugation is necessary to determine
is examined grossly for volume, color, clarity, and clot the origin of the blood. A xanthochromic supernatant
746 Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
(similar to CSF) indicates the breakdown of RBCs has glucose in the fluid has no diagnostic significance, whereas
occurred; a bloody or dark red-brown supernatant is sug- the difference between the fluid and plasma concentration
gestive of hemarthrosis rather than a traumatic aspirate or can be clinically important. Normally, synovial fluid glu-
injury (see Table 30-12). cose is in equilibrium with the plasma level during a fast-
Fluid clarity also changes with the disease process; ing, or at least 6 hours postprandial state; the fluid glucose
an inflammatory fluid may be translucent or opaque. is usually equal to or not greater than 10 mg/dL less than
Turbidity increases with inflammation, which may be the plasma level. Generally, patients with noninflammatory
caused by leukocytosis, crystals, or cartilage debris. Septic joint disorders have fluid glucose levels that are within
arthritis will produce a purulent fluid, caused by the 25 mg/dL less than the plasma glucose level. Differences
presence of bacteria and pus. The presence of crystals can greater than 25 mg/dL lower than the plasma level are
create a cloudy, turbid, fatty, or milky appearance (see indicative of inflammatory or septic disorders, due to
Table 30-12). Large quantities of degenerated synovial consumption by microorganisms or increased cellular
lining cells can create the appearance of pus, commonly metabolism (see Table 30-12). Overlap of expected glucose
seen in patients with RA. differences exist between the different types of disorders
Spontaneous clotting of the fluid, the fibrin clot test, is and may not be present in all patients with a particular
observed in inflammatory conditions. Fibrinogen and other disorder.
clotting proteins are acute phase reactants, which can be pre- The concentration of protein in synovial fluid is
sent in synovial fluid. A grossly bloody fluid caused by a trau- dependent on several factors that include the synovial
matic aspiration or traumatic injury will produce spontaneous membrane permeability, the molecular weight of various
clotting due to plasma fibrinogen; a hemorrhagic effusion protein molecules, the plasma level of protein, local synthe-
would not clot. sis, and local consumption. Normal synovial fluid protein
A test that is now obsolete, but of historical interest, levels range from bk to 3 g/dL. Clinically, increased protein
was known as the mucin clot or Ropes test.’ This test levels (greater than 3 g/dL) simply indicate the presence of
was used when a very limited aspirate was obtained, to inflammation, which increases membrane permeability.
determine if the aspirated fluid was synovial, subcutaneous There may also be local synthesis of immunoglobulin
tissue fluid, or local anesthetic. It also was used to discern that increases the amount of total protein present. Elevated
if the effusion was inflammatory or noninflammatory by protein is commonly associated with RA, gout, septic
evaluating the integrity of the molecular interactions arthritis, SLE, or other systemic disorders that can affect
between the hyaluronic acid and protein in the fluid. A the joints.
few drops of the fluid were added to a solution of 2% to Uric acid levels in normal serum and synovial fluid are
5% acetic acid. After a few minutes, a clot formed (mucin essentially in equilibrium. The levels are increased in both
clot) if the fluid is synovial, due to the polymerization of the serum and synovial fluid of patients with gout: a
the hyaluronic acid. The density and friability of the metabolic disorder in which excess uric acid precipitates in
clot after shaking evaluated the molecular interactions. A the synovial cavity causing an arthritic condition. The
dense clot that remained intact when shaken indicated a measurement of uric acid in synovial fluid is not the recom-
normal or noninflammatory (e.g., osteoarthritic) fluid. mended method for the diagnosis of gout; definitive diag-
A clot that fragmented or shredded easily on shaking indi- nosis is achieved by the detection of monosodium urate
cated that enzymatic degradation of the hyaluronic acid (MSU) crystals in the fluid by polarizing microscopic
occurred; this is associated with inflammatory effusions. examination (see discussion on crystals). In some cases,
Use of this test has been discontinued because it is nonspe- however, synovial fluid uric acid levels may be diagnosti-
cific, there are no standard criteria for comparison, and cally significant even if no crystals are observed; values
end-point interpretation is subjective. significantly greater than the upper reference range for
serum uric acid may be diagnostic.** *° It may also verify
the presence of MSU crystals in laboratories without the
Quantitative Analysis: Biochemical Analysis proper polarizing microscopic equipment or if the analyst is
and Microscopic Examination inexperienced.°
examined by routine microscopy for cells, crystals, and and histiocytes. In RA, synovial fluid may contain many
other particulates. plasma cells and moderate numbers of lymphocytes, histio-
A total WBC and RBC count must be performed man- cytes, synovial cells, cartilage cells, and rarely, multinucleated
ually in a hemocytometer; debris within the specimen may giant cells.
clog the flow cells of automated analyzers, and crystals or Certain diagnoses can be suspected when comparing
fat globules may be erroneously counted as cellular mater- the total WBC count with the percentage of segmented neu-
ial. Viscous fluids are difficult to pipette. They must be pre- trophils present. If the differential count reveals 90% or
treated with either 0.05% hyaluronidase in phosphate buffer more segmented neutrophils, then an infectious agent is
(i.e., one drop per milliliter of fluid), or with a pinch of most likely present (see Table 30-12). A Gram stain and
lyophilized hyaluronidase, to liquefy the sample. Fluids culture must be obtained; microorganisms can be seen in
should be counted undiluted and allowed to settle in the synovial fluid if present in sufficient numbers, and should
counting chamber for at least 30 minutes. If the appearance be seen as intracellular organisms. However, if bacteria
of the fluid suggests that dilution is necessary (i.e., bloody are absent on Gram stain this would not exclude the possi-
and/or turbid), only isotonic (0.85% or 0.9%) or phosphate bility that there is sepsis in the synovial cavity. Bacterial
buffered saline must be used as the diluent. Other diluents organisms are more common; pathogenic yeasts are seen
may contain acetic acid that will precipitate the hyaluronic only rarely. If fluids have a total leukocyte counts in the
acid, trapping cells and falsely lowering the WBC count. If range of 2000 to 200,000/uL, with greater than 50% neu-
the fluid is bloody, it may be necessary to lyse the RBCs trophils, a possible diagnosis of RA, SLE, or Reiter’s
with 0.3% saline or 0.1 N hydrochloric acid in order to syndrome should be considered. Reiter’s syndrome is a
count the WBCs more accurately.” The reason is that a suf- reactive arthritis caused by intestinal bacteria that also
ficient dilution of the RBCs may overdilute the WBCs; the affect the skin, eyes, and muscles. The presence of LE cells
greater the dilution factor, the more error that may be intro- is suggestive of SLE but not diagnostic, as they can also be
duced. A separate count must then be performed for the seen in RA.
RBCs. A normal synovial fluid does not contain red blood Eosinophilia of the synovial fluid, defined as greater than
cells; however, an RBC count less than 2000/uL is consid- 2% of the total WBC count, can be associated with RA,
ered normal; the concentration of RBCs is rarely of clinical rheumatic fever, metastatic carcinoma, parasitic infestation of
significance unless there is a need to determine if a trau- the joint, Lyme disease, radiologic procedures, and radiation
matic tap has occurred. therapy.°
The WBC count is necessary to classify the type of Other types of cells that can be observed in inflammatory
effusion present, but by itself is not diagnostic. A normal synovial fluids are known as RA cells and Reiter’s cells. RA
fluid will have less than 200 WBC/uL. Noninflammatory cells are associated with RA but are not specific for a diagnosis
effusions may contain 200 to 3,000 WBC/uL. Inflammatory of RA. These cells may also be called “ragocytes” or “inclusion
effusions may contain 3000 to 75,000 WBC/uL. Infectious body cells”. These are neutrophils that may contain from | to
disorders may contain 50,000 to 200,000 WBC/uL and the 20 granules in the cytoplasm, and are known to contain
crystal-induced disorders are associated with WBC counts immune complexes such as IgG, IgM, complement, and
of 500 to 200,000/uL.° There is obviously a considerable rheumatoid factor. Reiter’s cells, also nonspecific and not
amount of overlap to the leukocyte counts among disorders. diagnostic for Reiter’s syndrome, are vacuolated macrophages
The counts must be considered in the context of the clinical with intracytoplasmic inclusions or debris of ingested neu-
presentation; and most importantly, the differential cell trophils, which may appear as unrecognizable blue material by
count and morphology, which is the most important factor Wright stain.
in the differential diagnosis in synovial fluid analysis (see Synovial lining cells are mononuclear cells with mor-
Table 30-12). phology resembling that of the mesothelial cells in serous
Morphologic examination is performed using Wright- fluids. Their nuclei may have small, regular nucleoli. They
stained cytocentrifuge preparations. If a cytocentrifuge is not may become proliferative in a reactive setting similar to
available, or if fluids have extremely high cell counts, standard mesothelial cells. Reactive synovial cells may be multinu-
push smear or slide-on-slide methods may be used. The fluid cleated and may occur in clusters. Synovial lining cells may
should be liquified and appropriately diluted, as previously also be difficult to differentiate from monocytes and histio-
mentioned, within | hour of collection owing to cellular degra- cytes. Their presence does not have any specific diagnostic
dation and the risks of misdiagnosis. The cells that are normally significance.
present include lymphocytes, monocytes, macrophages, and Malignant cells can be seen in synovial fluid, although
synovial cells. Neutrophils are also present, but do not exceed this is extremely rare; they are usually derived from metasta-
25% of the total cells. Abnormal synovial fluids can contain tic disease.
neutrophils, lymphocytes (normal and reactive), plasma cells,
monocytes, eosinophils, histiocytes, macrophages, synovial Crystal Examination
cells, and LE cells. In acute inflammatory conditions, synovial
fluid can contain many neutrophils and a moderate number of Every synovial fluid sent to the laboratory for cell counts
synovial cells; bacteria may also be present. In chronic inflam- must be examined for crystals, especially if infection is
mation, synovial fluid contains lymphocytes, plasma cells, not a consideration. This aspect of fluid analysis has the
748 Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
greatest impact on diagnosis and therapy; if incorrect or present in the crystals. A drop of the stain is added to a drop of
improperly performed it can result in permanent disability the synovial fluid, placed on a slide, and cover slipped. Crys-
for the patient. As previously mentioned, the specimen tals containing calcium appear orange with light microscopy
must have been collected into liguid EDTA or sodium or bright red under polarized light.
heparin, to avoid confusion with crystalline structures Laboratories that perform crystal analysis and identifica-
caused by the powdered forms of these anticoagulants. tion must use appropriate quality control materials, to align
Evaluation should be performed on a wet preparation; how- the polarized microscopes properly and for comparative pur-
ever, examination of the fluid by standard centrifugation or poses in identification.
cytocentrifuge preparation is also necessary. The cytocen-
trifuge concentrates the fluid; a sample that is negative by a TYPES OF CRYSTALS, ANALYSIS, AND CLINICAL
wet preparation may actually show crystals on the cytocen- CORRELATION
trifuge slide. If sufficient numbers of crystals are present, The causes of crystal deposition are not well understood,
they can be seen with plain light microscopy on the Wright- but certain predisposing factors may exist such as increas-
stained slide. It is important to note, however, that some ing age, the presence of joint damage, and familial inheri-
Wright staining techniques may result in the dissolution of tance. Others include metabolic disorders, osteoarthritis
some crystals. Extra cytocentrifuge slides should be pre- (OA), and SLE (see Table 30-12). The most common types
pared and left unstained to determine if crystals can be seen of crystals that may be present in synovial fluid include
before staining. If few crystals are present, phase-contrast monosodium urate (MSU), calcium pyrophosphate dihy-
or polarized light microscopy may be necessary to see them drate (CPPD), basic calcium phosphates (BCP), oxalates,
initially by wet preparation or cytocentrifugation. Synovial cholesterol and lipids, and steroids. Less common crystals
fluids should be thoroughly examined for crystals, for and artifacts also occur, which may be confused with clini-
at least 15 minutes, before they are reported as negative. cally significant synovial fluid crystals or interfere in their
Artifaéts such as cell clumps or fibrin strands may obscure detection.
or trap the few crystals that may be present. Excellent images of synovial fluid cells and crystals are
Polarized light must be used for the detection and con- readily available on the Internet. The reader is urged to visit
firmation of birefringence, the ability of a particular mater- the following Web site(s) for useful educational material:
ial to refract light. This can be determined with a polarized
www.archrheumatol.net/ASF/index.html
light microscope that contains a rotating filter, known as a
“polarizer” situated below the slide stage in the light path. www.palmslab.com.au/.../ Infolink/issue9.shtml
A fixed filter, a second polarizer known as the “analyzer,” is http://www-medlib.med.utah.edu/WebPath/
located above and between the objectives and the oculars. BONEHTML/BONE063.html
Both filters will allow only light of one direction or polarity
to exit. When the polarizer is rotated 90 degrees with MONOSODIUM URATE CRYSTALS The presence of MSU
respect to the analyzer, it creates a block, or maximum crystals is pathognomonic for gout, causing a condition
extinction, of the polarized light, yielding a dark field. If known as gouty arthritis. These crystals are seen in the ma-
the specimen contains birefringent material, the direction of jority of patients during acute attacks. Between attacks,
the light is refracted, allowing this light to pass through the they may be seen in approximately 75% of patients.> They
analyzer and is seen as a bright particle or crystal against may also be present, occasionally, in cases of inflammation.
the dark field. Not all materials possess birefringent charac- MSU crystals are typically long, thin, and needle-like with
teristics, but this attribute assists in identification of the pointed ends. They may be seen singly or in aggregates;
particles present in the synovial fluid. Another filter, known rarely, they may appear as spherules that are composed of
as a “red compensator,” changes the velocity of the trans- clusters of many individual needle-shaped crystals.2 Some
mitted light, allowing birefringent crystals to display differ- Wright staining methods can cause MSU crystals to dis-
ent colors depending on their position in relation to the axis solve, which is why the wet preparation must always be
of the compensator. This is used to further identify and examined first. These crystals may be found within neu-
confirm the existence of certain types of crystals. The com- trophils or macrophages during attacks of acute gouty
pensator is placed at a 45-degree angle to the axis of the arthritis.” It is important to report their appearance as intra-
polarizing lenses, which causes the background to become and/or extracellular; phagocytosis of crystals suggests that
a reddish color when properly aligned. The slide (or the they are responsible for the acute arthritis.2* MSU crystals
stage, depending on the microscope’s design) is rotated to are strongly birefringent and appear white in polarized
bring a crystal into a parallel orientation with the compen- light, however, they lose their birefringence with compen-
sator. The color is noted; then the crystal is oriented perpen- sated polarization. These characteristics are sufficient to
dicular to the axis of the compensator and the color change identify them as MSU crystals. If these crystals are parallel
is noted. If many crystals are present, only one crystal to the axis of the compensator they will appear yellow (neg-
needs to be found that fits the identification criteria for that ative birefringence); if perpendicular they will appear blue.
particular crystal. As previously mentioned, an increased concentration of
In addition, if unknown crystals are seen, alizarin red S uric acid in the serum and/or synovial fluid, even without
can be used, which selectively stains any calcium that may be the presence of MSU crystals, is diagnostic of gout.
Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis 749
CALCIUM PYROPHOSPHATE DIHYDRATE CRYSTALS CHOLESTEROL AND LIPID CRYSTALS Cholesterol crys-
CPPD crystals are characteristically present in a group of tals can be present in chronically inflamed joints, and are
disorders known as CPPD deposition disease (CPDD).*' It not diagnostic of any particular disorder. They may also be
is also known as pseudogout or chondrocalcinosis. These found in chylous effusions which contain a high concentra-
crystals are a common cause of arthritis, most frequently seen tion of lipids. Although rarely observed, they are most
in the elderly, and in patients with degenerative arthritis. often seen in cases of patients with long histories of RA,”
Patients with hereditary forms of CPDD, as well as meta- OA, and ankylosing spondylitis.** Crystal formation is
bolic disorders, may also possess CPPD crystals. The thought to involve the cholesterol from the cell membranes
symptoms of this condition may imitate gout or RA. Occa- of degenerating cells associated with impaired drainage of
sionally, a synovial fluid may contain both MSU and CPPD the joint.° They have characteristically large, rectangular
crystals; the presence of one does not exclude the other. notched-plate shapes that are easier to see by regular light
This may be an indication of septic arthritis, especially if microscopy, although strongly birefringent with polariza-
the WBC count is extremely elevated. These crystals may tion. However, they may also resemble the needle or rhom-
also be seen intra- and/or extracellularly. They can be boid shapes of MSU or CPPD crystals; this can lead to
more difficult to identify, typically appearing as short misidentification. Compensated polarization is not reliable
rectangular shapes, but they may also assume multiple as a method of differentiation because cholesterol crystals
three-dimensional forms such as rods and rhomboids. are variable in their appearance. The presence of lipophages
Needle-shaped forms are also possible, causing misidentifi- (lipid-laden macrophages) during the microscopic exami-
cation as MSU crystals. They are more easily seen with nation may provide additional evidence of the type of
light microscopy; they are only weakly birefringent, crystal present.
and therefore may be difficult to see with polarized light. Lipids can exist in crystalline and noncrystalline forms
With compensated polarization, these crystals have the and are typically found in both chylous and chronic effu-
opposite appearance of MSU. They appear blue if parallel sions.** Noncrystalline lipids occur as fat globules that
to the axis (positive birefringence), and yellow if perpen- are round and nonbirefringent. Lipid crystals are round
dicular, thereby differentiating them from MSU crystals. when seen by light microscopy. With polarized light,
The use of alizarin red S staining will indicate the presence the crystalline forms appear as strongly birefringent
of calcium. ‘ Maltese crosses. Under compensated polarization, they
appear as red and blue crystals. Lipid crystals can be mis-
BASIC CALCIUM PHOSPHATE CRYSTALS The BCP crys- taken for MSU spherules by an inexperienced analyst.
tals are a group of compounds consisting mainly of calcium However, observation under high-power magnification
hydroxyl phosphate, also known as hydroxyapatite (HA); (< 100) would reveal they are not composed of numerous
others include octacalcium phosphate, and less frequently, individual crystals.
tricalcium phosphate. These crystals are very small in
size and form aggregates, which are occasionally visible CORTICOSTEROID CRYSTALS Patients with known non-
by light microscopy on a wet preparation as shiny infectious joint disorders, such as OA and RA, may occasion-
irregular cytoplasmic inclusions. These crystals may also ally receive treatment with corticosteroid injections into the
appear extracellularly. They will appear as purple inclu- joint cavity or surrounding connective tissue, to reduce the
sions on a Wright-stained smear. They are too small and inflammatory response. Knowledge of a history of intra-
too weakly birefringent to be identified by polarized articular steroidal injection is important in the evaluation of
light microscopy. Positive staining does occur with alizarin synovial fluid. The synthetic corticosteroids form artifactual
red S, but this is nonspecific. HA crystals are not crystals of varying sizes, shapes, and birefringent qualities.
pathognomonic for any specific disorder, but they are They may persist in the synovial fluid for several weeks or
associated with arthritic syndromes such as RA and OA; months after injection. Steroids do not have characteristic
these involve calcification in and/or around the joint, with crystalline shapes. They may be amorphous with irregular
periarticular inflammation, calcific deposits, and the pres- outlines and ragged edges, or they can resemble MSU and
ence of HA in the synovial fluid. Most typical clinical CPPD crystals. They can be strongly birefringent or not at all.
laboratories will not be able to definitively identify these They can also be phagocytosed by leukocytes and/or synovial
crystals. However, diagnosis, prognosis, and treatment are cells, appearing intracellularly. These multiple factors
not dependent on it. should alert the analyst to the probability that they are
steroidal crystals, one of the more common artifacts that can
OXALATE CRYSTALS Crystals of calcium oxalate may be be found in synovial fluid.
found in the synovial fluid of patients with primary oxalosis,
arare inborn error of metabolism, but is more commonly seen HEMATIN OR HEMATOIDIN CRYSTALS Red blood cells
in patients with chronic renal failure who undergo hemodial- that are present in the synovial cavity are phagocytosed by
ysis. The deposition of these crystals causes arthropathy. The macrophages. Enzymatic degradation of the hemoglobin
crystals appear as small, bipyramidal or pleomorphic shapes, results in the formation of hemosiderin, which may subse-
with a wide variation in birefringence and stain positively quently form hematin or hematoidin crystals. Their presence
with alizarin red S. suggests that a significant hemarthrosis has occurred. These
750 Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
crystals are rhombohedral shaped, but occasionally may between the layers of the pericardium, pleura, and peri-
be irregularly shaped. They appear gold in color with regu- toneum, respectively.
lar light microscopy and are not birefringent. It is possible The central nervous system (CNS) consists of the brain
for these to be mistaken for CPPD crystals by an inexperi- and spinal cord, which are lined by the meningeal mem-
enced technologist; however, their appearances differ with branes, Or meninges, that consist of three layers: the dura
polarization. mater, arachnoid mater, and pia mater. Between the arachnoid
mater and pia mater is the subarachnoid space, containing the
ARTIFACTS Numerous particulates can appear in synovial cerebrospinal fluid (CSF).
fluid during microscopic analysis that are clinically Joints (articulations) are junctions between two or more
insignificant and/or not originally of synovial origin. These bones. They are enclosed by the joint capsule and lined by the
are known as artifacts and must be differentiated from clin- synovial membrane, or synovium, composed of a single layer
ically significant synovial particulates. Most can be of mononuclear synovial cells. Synovial fluid is contained
avoided with the use of clean microscope slides and cover- within the synovium and joint capsule.
slips, such as dust, glass fragments, lint, paper fibers, and Manual cell counts of WBCs and RBCs are the methods
so forth. Cartilage fragments, collagen fibrils, or fat glob- used for body fluids. Slides for morphologic analysis are pre-
ules may be present due to a disease process or as a result pared via cytocentrifugation. Advantages and disadvantages
of the arthrocentesis; metallic fragments may arise from of this method are summarized in Tables 30-1 and 30-2.
articular prostheses. As previously mentioned, therapeutic The most common types of cells encountered in body
intra-articular steroid injections can cause the formation of fluids are neutrophils, lymphocytes, monocytes, and tissue
crystalline structures. Wright or Wright-Giemsa-—stained cells. Other cells less frequently seen include eosinophils,
slides may contain stain precipitates. Artifactual crystals basophils, mast cells, plasma cells, and malignant cells.
typically have variable appearances by regular light and An abnormal* collection of fluid is known as an
polarized light microscopy. They do not have definite crys- effusion. Effusions that accumulate due to systemic dis-
tal morphology. Clinically significant crystals have regular eases are known as transudates; those that accumulate
outlines with smooth, parallel edges. Polarized light due to pathology within a compartment are known as
microscopy can create confusion in differentiating artifact exudates. The differences between the two are outlined in
from pathologic crystals, most commonly when starch Table 30-4. Effusions can occur due to a wide variety of
granules enter the specimen from surgical gloves. They local or systemic disease processes. They may be further
appear as variably to strongly birefringent Maltese crosses, described as normal cellular, benign/reactive, or malignant,
similar to lipid crystals. In addition, they can be mistaken based on the cell types present. The types of effusions, their
for MSU spherules. However, starch granules have irregu- locations, and the methods of collection are described in
lar outlines and a central depression visible by light Table 30-9.
microscopy. The CSF is a selective ultrafiltrate of plasma. Indica-
tions for its analysis include suspicion of meningeal infec-
Summary tion, subarachnoid hemorrhage, malignancy within the
CNS, or demyelinating diseases. CSF collection is per-
The analysis of fluids from normally sterile body compart- formed by lumbar puncture (LP) or “spinal tap.” A normal
ments is an important element in the diagnosis and treatment CSF is clear, colorless, sterile, and watery in consistency. A
of disease. Body fluids are a diverse group of non-blood, non- common problem in the analysis of CSF is distinguishing
urine fluids that are found in closed body cavities. The types between a true CNS hemorrhage and a traumatic LP.
of cells, their concentrations, and the presence and types of Grossly bloody specimens may result from a traumatic tap,
crystals are useful in disease diagnosis. It is important for the or the presence of a subarachnoid or an intracerebral hem-
laboratory professional to be able to distinguish benign from orrhage. Xanthochromia, a pink, orange, or yellow color of
reactive or malignant cells and appropriately refer these find- the CSF supernatant, is caused by the breakdown of hemo-
ings to the pathologist. All stained fluid slides with atypical, globin. Other causes of xanthochromia include jaundice,
suspicious, or malignant cells must be reviewed by a pathol- extremely elevated CSF protein, and hyperbilirubinemia in
ogist prior to reporting. premature infants.
The aspirated fluid is placed into evacuated, anticoagu- Biochemical analysis of CSF routinely includes the mea-
lated, and/or sterile tubes to be sent to the laboratory. The surement of the total protein (TP) and glucose levels. An
specimens must be processed immediately. The cells are usu- increased TP is seen in meningeal and CNS disease. Glucose
ally less numerous than in peripheral blood and they begin to in the CSF is normally about 60% to 70% of serum levels.
deteriorate once they are removed from the body. Decreased CSF glucose is characteristic of bacterial, tubercu-
Fluids from the thoracic and abdominal cavities are lous, or fungal meningitis. Viral meningitis, however, is not
referred to as serous fluids. The heart and lungs within the associated with a decreased CSF glucose. Normal findings in
thoracic cavity, and various organs in the abdominal cavity, CSF are summarized in Table 30-10.
are lined by a membrane composed of mesothelial cells. CSF WBC counts are clinically significant; even low
Pericardial, pleural, and peritoneal fluids are contained counts are suggestive of pathology. The types of WBCs
Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis = 751
present are most important in the interpretation of a CSF, and leukocytes. The cells normally present include lymphocytes,
their relationship to the clinical findings. The morphologic monocytes, macrophages, synovial cells, and neutrophils,
evaluation of the CSF must be performed regardless of a which should not exceed 25% of the total nucleated cells
normal cell count. present. Every synovial fluid must have an examination for
Normal synovial fluid, aspirated by arthrocentesis, is crystals. Polarized light must be used for the detection and
transparent, colorless, viscous, and does not clot. Impaired confirmation of birefringence. The most common types of
function of the fluid may play a role in the development of crystals that may be present in synovial fluid include
degenerative joint diseases. Synovial fluid is most often ana- monosodium urate (MSU), calcium pyrophosphate dihydrate
lyzed when there is a suspicion of infective (septic) arthritis (CPPD), basic calcium phosphates (BCP), oxalates, choles-
or crystal-associated diseases. Analytes routinely measured terol and lipids, and steroids.
in the evaluation of joint disorders include glucose, protein, The procedural name, collection requirements, routinely
and uric acid. The microscopic examination of synovial ordered tests, and storage conditions for each body fluid type
fluid is crucial to determining the presence of crystals and are summarized in Table 30-13.
Tube Types
Fluid Type Procedure(s) Collected* Tests Storage
Serous:
Pleural Thoracentesis 1. EDTA—lavender top 1. Cell counts, morphology, 4°C
differential count
Pericardial Pericardiocentesis 2. Heparin—green top 2. Color, clarity, glucose, total 4°C
Peritoneal Paracentesis protein, LDH, cholesterol,
(Ascites) bilirubin, specific gravity
3. Sterile, heparinized 3. Culture, Gram stain, AG
acid-fast stain
4. Nonadditive tube; plain 4. Clot formation; gross 4°C
red top examination
Cerebrospinal Lumbar puncture Sterile, nonadditive, Tube 1: Chemical/Serological 4°C
sequentially numbered Studies
tubes Color, clarity, glucose, Total
Protein, IgG, Albumin
Tube 2: Microbiological SHC
Studies
Culture, Gram Stain, India Ink
Preparation
Tube 3 or 4: Cytology or 4°C
Hematology
Cell counts, morphology,
differential count
Synovial Arthrocentesis 1. Liquid EDTA— 1. Cell counts, morphology, AXE
lavender top differential
count, crystal
analysis
2. Sodium heparin— 2. Color, clarity, glucose, AL@
green top total protein, uric acid,
lactate, rheumatoid factor,
complement, ANA, crystal
analysis
3. Sterile (heparinized) 3. Culture, Gram stain AG
4. Plain, nonadditive 4. Clot formation; gross 4°C
examination
*A whole blood specimen for serum preparation must be drawn at the time of fluid collection for comparative studies.
**Microbiological specimens must be kept at 37° C until cultures are prepared; thereafter, remaining specimen must be stored
at 2-8°C until all testing is completed.
ANA = antinuclear antibody.
oe Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
ANSWERS QUESTIONS
1. A fluid collection in the chest cavity is known as a . What type of procedure was performed to obtain the
pleural effusion. joint fluid and what terminology applies to the speci-
2. The procedure to withdraw a pleural effusion is known men?
as a thoracentesis. 2. Into what types of collection tubes should the aspirated
3. The pleural effusion collected is considered to be an specimen be placed?
exudate, because the WBC count is greater than eS). What abnormal results are present, if any, and what
1000/uL, the total protein is greater than 3 g/dL, the could they indicate?
total protein ratio is greater than 0.5, and the LDH ratio 4. What additional tests should be performed to aid in
is greater than 0.6. diagnosis?
4. This specimen is not chylous, as the predominant cell type
in the differential examination are neutrophils, whereas
ANSWERS
chylous effusions predominantly contain lymphocytes. It
could be considered pseudochylous by its appearance, but 1. The procedure to obtain fluid from a joint space is
the laboratory studies for triglycerides are not available. known as arthrocentesis; the fluid collected is known as
5. Further tests to be performed should include a Gram synovial fluid.
stain and culture. 2. The specimen should be placed into sterile nonadditive,
6. These findings are representative of an acute reactive sodium heparin, and liquid EDTA tubes.
process, which present with greater than 50% neu- 3. The child’s peripheral blood WBC count and erythrocyte
trophils on differential examination. sedimentation rate are elevated but consistent with the
presence of an acute upper respiratory infection or other
inflammatory process. However, the fluid analysis
reveals abnormalities in the clarity, cell counts and dif-
ferential, and the glucose level. These parameters indi-
cate the presence of an active inflammatory process: a
normal synovial fluid would not contain RBCs, there
would be less than 200 WBC/uL, and neutrophils would
Case Study 3 not exceed 25% of the WBCs. The fluid WBC count
falls within the Group II/Inflammatory disease category,
A 2-year-old male child was brought to the emergency room however, other parameters fit into the Group III/
to determine why he suddenly was refusing to walk and Infectious disease category. The presence of neutrophilia
complaining of pain in his right leg. There was no history of (greater than 80%) would be highly suggestive of septic
trauma. He had been seen by his pediatrician earlier in the arthritis, regardless of the WBC count. Results can over-
day due to the presence of fever and an upper respiratory lap disease categories, and it is possible for more than
infection; he was diagnosed with otitis media, prescribed an one type of disease process to be present simultaneously.
antibiotic, and sent home. Clinical presentation and the microscopic findings, most
Physical exam in the emergency room revealed pain and importantly the differential cell count and morphology,
tenderness of the right ankle. There was also some edema will determine what process is involved and what course
and erythema. X-ray exams of the lower extremities did not of action should be taken. The glucose level is not in
reveal any fractures or displacements. Laboratory tests were equilibrium with the plasma; differences greater than
ordered on his peripheral blood, and a fluid aspirate was 25 mg/dL lower than the plasma level are indicative of
taken from the ankle. The results were as follows: inflammatory or septic disorders, due to consumption by
microorganisms or increased cellular metabolism.
Peripheral blood: 4. Additional testing must include examination of a Gram
_ WBC: 26,000/uL stain and culture of the fluid to identify the presence of
Differential: Neutrophils = 75% bacterial organisms. If positive, this would point to a
Bands = 2% diagnosis of septic arthritis.
Lymphocytes = 19%
Monocytes = 4%
_ RBC parameters: Normal
Erythrocyte sedimentation rate: Elevated
Plasma glucose:
Fluid analysis:
96 mg/dL
Case Study 4
Color: Yellow
Clarity: Purulent A 50-year-old man with a history of alcohol abuse was seen
WBC: 22,000/uL in the emergency room for symptoms of nausea, vomiting,
Differential: Neutrophils = 95%
and abdominal pain. On physical exam, the abdomen
Macrophages = 5%
appeared distended. A computerized tomographic (CT) scan
RBC: 375,000/uL
Fluid glucose: 39 mg/dL of the abdomen revealed a collection of fluid. About
1200 mL of fluid was drained and portions were sent to the
continued continued
754 Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
ANSWERS
Case Study 4. conta
1. Paracentesis is the procedure to drain and collect fluid
from the peritoneal cavity. This collection of fluid is
laboratory for analysis. Peripheral blood was collected
known as a peritoneal effusion, or ascites. The collected
simultaneously. The laboratory findings were as follows:
fluid may be referred to as peritoneal fluid, paracentesis
fluid, or ascitic fluid.
Serum values:
. This fluid is a transudate.
Glucose 99 g/dL
_ Transudative ascitic fluids can be caused by increased
Why
Total protein 7 g/dL
Albumin 4.0 g/dL capillary pressure or permeability, abnormal colloid
LDH 60 U/L osmotic pressure, poor lymphatic drainage or cardiac
Fluid analysis: Clear, pale yellow abnormalities. Additiona! causes include abdominal con-
Glucose 100 mg/dL ditions that do not directly involve the peritoneum, such
Total protein 1.9 g/dL as hepatic cirrhosis, intrahepatic and portal venous
TP ratio 0.3 obstruction, hypoalbuminemia, renal function, ovarian
LDH 25 U/L disease, pancreatic disease, or parasitic infection. Alco-
LDH ratio 0.4 holic cirrhosis is one cause of peritoneal effusion, which
Albumin 2.4 g/dL appears to be the case in this patient. Malignant disease
SAAG 1.6
can also produce an effusion.
RBC 500/uL
WBC 60/uL 4. The SAAG, or serum-ascites albumin gradient, is a
Differential: Neutrophils = 10% value calculated by subtracting the ascitic fluid albumin
Lymphocytes = 80% concentration from the simultaneously collected serum
Monocytes/Macrophages = 10% albumin concentration. This value is useful in differenti-
‘ Atypical mesothelial cells seen ating whether the fluid is transudative or exudative in
nature; transudative ascitic fluids have an SAAG signifi-
cantly greater than 1.1 g/dL, whereas exudative fluids
QUESTIONS are found to be less than 1.1 g/dL. In addition, patients
1. What type of procedure was performed to drain and col- with high-gradient ascites include those with cirrhosis,
lect the fluid, what body cavity was involved, and what alcoholic hepatitis, cardiac-related ascites, or massive
terminology applies to the specimen? liver metastasis. Low-gradient ascites are associated
2. Is the fluid a transudate or an exudate? with peritoneal malignancies and non-malignant diseases
3. What process(es) could cause the production of this type such as TB, pancreatitis, nephrotic syndrome, biliary
of fluid? disease, or connective tissue diseases.
4, What is the SAAG and what is its significance? 5. The presence of atypical mesothelial cells are especially
5. What does the presence of atypical mesothelial cells seen in chronic effusions and in ascitic fluid associated
indicate? with cirrhosis.
continued
Acknowledgments
The author gratefully acknowledges the contributions of
Judith P. Brody, M.D., and Michele L. Best, MT (ASCP).
Direct iome
1. A specimen is sent to the laboratory labeled “synovial 3. All of the following are serous fluids except:
fluid.” What procedure was used to obtain the specimen? a. Synovial
a. Thoracentesis b. Peritoneal
b. Paracentesis c. Pleural
c. Arthrocentesis d. Pericardial
d. Lumbar puncture
4. The anticoagulant used for hematological body fluid
i) . The presence of MSU crystals is pathognomonic for analysis Is:
which disorder? a. Heparin
a. Pseudogout b. Oxalate
b. Osteoarthritis c. EDTA
c. Rheumatoid arthritis d. Citrate
d. Gout
Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis 755
ARY cH
d ms SUMM @ Synovial fluid is secreted by the synovial cells, also pro-
ducing hyaluronic acid.
mw Fluids from the thoracic and abdominal cavities are re- m The large joints may normally contain approximately
ferred to as serous fluids; they are ultrafiltrates of plasma. 1 mL of synovial fluid.
gw The pleural, pericardial, and peritoneal cavities are lined by gwAll body fluids are potentially infectious and they
a membrane composed of a single layer of mesothelial cells. frequently are nonretrievable.
m Normal pleural, pericardial, and peritoneal cavities do not maImportant terms: Thoracentesis = removal of pleural
fluid; Pericardiocentesis = removal of pericardial fluid;
contain appreciable amounts of fluid.
Paracentesis = removal of peritoneal fluid; Lumbar
= The central nervous system (brain and spinal cord) is lined puncture = collection of CSF; Arthrocentesis =
by the meningeal membranes/meninges. removal of synovial fluid; Effusion = an abnormal col-
mThe subarachnoid space contains cerebrospinal fluid lection of fluid in a body cavity; Transudates = effusions
(CSF), which is a selective ultrafiltrate of plasma. caused by a systemic disease state; Exudates = effusions
caused by a primary pathologic state within the compart-
gw The normal total volume of CSF in adults is 90 to 150 mL,
ment; Chylous effusions = exudates resulting from leak-
and 10 to 60 mL in neonates.
ing or blocked lymphatic vessels.
w Joints (articulations) are enclosed by a joint capsule; the syn-
ovium or synovial membrane is composed of synovial cells.
continued
756 Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
m CPPD crystals are characteristically present in a group of Cholesterol crystals have characteristically large, rectan-
disorders known as CPPD deposition disease (CPDD), gular notched-plate shapes and are strongly birefringent.
also known as pseudogout or chondrocalcinosis. CPPD
g Lipid crystals are round when seen by light microscopy.
crystals typically appear as short rectangular shapes, but
With polarized light, the crystalline forms appear as
they may also assume multiple three-dimensional forms
strongly birefringent Maltese crosses.
such as rods and rhomboids. CPPD crystals are more eas-
ily seen with light microscopy; they are only weakly bire- m Patients with OA and RA may occasionally receive treat-
fringent. ment with corticosteroid injections into the joint cavity or
surrounding connective tissue.
= The BCP crystals are a group of compounds consisting
mainly of hydroxyapatite (HA). @ Steroids do not have characteristic crystalline shapes.
g HA crystals are not pathognomonic for any specific disor- @ Septic, inflammatory, and noninflammatory effusions are
der, but they are associated with arthritic syndromes classified by the quantity and types of leukocytes present;
such as RA and OA. HA crystals are too small and too the crystal-induced joint diseases are characterized by the
weakly birefringent to be identified by polarized light presence of crystals.
microscopy. @ A normal fluid will have less than 200 WBC/uL and does
= Calcium oxalate crystals may be found in the synovial not contain red blood cells; the concentration of RBCs is
fluid of patients with primary oxalosis, a rare inborn error rarely of clinical significance unless there is a need to
of metabolism. determine if a traumatic tap has occurred.
REFERENCES Kjeldsberg, CR, and Knight, J: Body Guide to the Cells of Pleural, Pericardial,
Fluids: Laboratory Examination of Peritoneal, and Hydrocele Fluids.
—_— . Cutler, RWP, and Spertell, RB. Cere-
Amniotic, Cerebrospinal, Seminal, Kluwer Academic, Boston, 1989.
brospinal fluid: A selective review. Ann
Serous and Synovial Fluids,ed 3. ASCP —_19. Kjeldsberg, CR, and Knight, J: Body
to
Newall 282 Press, Chicago, 1993, pp. 159-222. Fluids: Laboratory Examination of
; Chogyoji, M, et al: Laboratory Bit ds, 10. Ray, PH, et al: The problem of chylotho- Amniotic, Cerebrospinal, Seminal,
Appendix A. In: Kjeldsberg CR, Knight rax. Mayo Clin Proc 42:457, 1967. Serous and Synovial Fluids, ed 3. ASCP
J. Body Fluids: Laboratory . 11. Kjeldsberg, CR, and Knight, J: Body Press, Chicago, 1993, pp. 65-157.
Examination of Amniotic, Cerebrospinal, Fluids: Laboratory Examination of 20. Seehusen, DA, et al: Cerebrospinal Fluid
Seminal, Serous and Synovial Fluids, Amniotic, Cerebrospinal, Seminal, Analysis. September 15, 2003. Online.
ed 3. ASCP Press, Chicago, 1993, Serous and Synovial Fluids, ed 3. ASCP Available: http://www.aafp.org/
pp. 303-348. Press, Chicago,; 1993, pp. 223-264. afp/20030915/1103.html (February 28,
ios)
: Rabinovich, A, and Cornbleet, PJ: Body 12. Pare, P, et al: Serum-ascites albumin 2008).
fluid microscopy in U.S. Laboratories: concentration gradient: a physiologic 21. Edlow, JA, and Caplan, LR: Avoiding
Data from two College of American approach to the differential diagnosis of pitfalls in the diagnosis of subarachnoid
Pathologists surveys, with practice rec- ascites. Gastroenterology 85:240, 1983. hemorrhage. N Engl J Med 342:29, 2000.
ommendations. Arch Pathol Lab Med 13. Rector, WG, and Reynolds, TB: Superi- 22. Fishman, RA: Cerebrospinal Fluid in
118:13, 1994. ; oe. ority of the serum-ascites difference over Diseases of the Nervous System, ed 2.
. Clare, CN: Morphologic Analysis of the ascites total protein concentration in WB Saunders, Philadelphia, 1992.
Body Fluids in the Hematology Labora- separation of “transudative” and “exuda- 23. Krieg, AF: Cerebrospinal fluid and
tory. In: McKenzie, SB (ed). Clinical tive” ascites. Am J Med 77:83, 1984. other body fluids. In Henry JB (ed),
Laboratory Hematology. Upper Saddle 14. Broussard, CN, and Carey, CD: Ascites: Clinical Diagnosis and Management
River: Pearson Education, 2004, Diagnosis and Management. Practical by Laboratory Methods. WB Saunders;
pp. 623-651. | Gastroenterology
16, 1998. Philadelphia, 1979, pp. 635-679.
. Kjeldsberg, CR, and Knight, J: Body 15. Adams, HW, and Maimz, DI: 24. Menkes, JH: The causes for low spinal
Fluids: Laboratory Examination of Eosinophilic ascites. Am J Digest Dis fluid sugar in bacterial meningitis:
Amniotic, Cerebrospinal, Seminal, 22:40, 1977. another look. Pediatrics44:1, 1969.
Serous and Synovial Fluids, ed 3. ASCP 16. Chan, MK, et al: Peritoneal eosinophilia 25. Knight, JA, et al: Early (chemical)
Press, Chicago, 1993, pp. 265-301. in patients on continuous ambulatory diagnosis of bacterial meningitis: Cere-
. Hamm, H, et al; Cholesterol in pleural peritoneal dialysis: a prospective study. brospinal fluid glucose, lactate, and lac-
effusions: a diagnostic aid. Chest 92:296, Am J Kidney Dis 11:180, 1988. tate dehydrogenase compared. Clin
1987. : 17. Daugirdas, JT, et al: Induction of peri- Chem 27:1431, 1981.
. Valdes, L, et al: Cholesterol: a useful pa- toneal fluid eosinophilia by intraperi- 26. Krieg, AF, and Kjeldsberg, CR: Cere-
rameter for distinguishing between toneal air in patients on continuous am- brospinal fluid and other body fluids. In:
pleural exudates and transudates. Chest bulatory peritoneal dialysis. N Engl J Henry JB, (ed): Clinical Diagnosis and
99:1097, 1991. Med 313:1481, 1985. Management by Laboratory Methods.
. Sun, NCJ, et al: Morphologic Interpreta- 1g. gpriggs, AT, and Boddington, MM: WB Saunders, Philadelphia, 1991,
tion of Body Fluids. ASCP Workshop Atlas of Serous Fluid Cytopathology: A pp. 445-473.
Publications, 1986, p. 1.
758 Chapter 30 Body Fluid Examination: Qualitative, Quantitative, and Morphologic Analysis
Ls Krieg, AF, and Kjeldsberg, CR: Cere- 34. Chen, KTK, and Moseley, D: Cartilage 4l.— Moskowitz, RW: Diseases associated
brospinal fluid and other body fluids. In: cells in cerebrospinal fluid. Arch Pathol with deposition of calcium pyrophos-
Henry JB (ed): Clinical Diagnosis and Lab Med. 114:212, 1990. phate in hydroxyapatite. In Kelley, WN,
Management by Laboratory Methods. 35: Rippey, JH: Synovial fluid analysis. Lab et al(eds); Textbook of Rheumatology,
WB Saunders, Philadelphia, 1991, Med 10:140, 1979. ed 3. Pa: WB Saunders, Philadelphia,
p. 641. 36. Young, LL: Aspirating the Ankle Joint: 1989, pp. 1449-1467.
. Oehmichen, M: Cerebrospinal Fluid Radiology Cases in Pediatric Emergency 42. Ettlinger, RE, and Hunder, GG: Synovial
Cytology: An Introduction and Atlas. Medicine. 1995. Online. Available: effusions containing
WB Saunders, Philadelphia, 1976. http://www.hawaii.edu/ cholesterol crystals: Report of
. Oehmichen, M, et al: Origin, prolifera- medicine/pediatrics/pemxray/v3c06. 12 patients and review. Mayo Clin Proc
tion and fate of cerebrospinal fluid cells. html (July 24, 2008). 54:336, 1979.
J Neurol 27:145, 1982. SHE Wener, MH: Synovial fluid analysis. 43. Fam, AG, et al: Cholesterol crystals in
. Weller, PF, and Liu, LX: Eosinophilic 1999. Online. Available: osteoarthritis joint effusions.
meningitis. Semin Neurol 13:161, 193.. http://www.rheumatology.org/ J Rheumatol 8:273, 1981.
. Kuberski, T: Eosinophils in the cere- publications/primarycare/number6/ 44. Small, DM: Liquid crystals in living and
brospinal fluid. Ann Intern Med 91:70, hrh0033698.asp (March 17, 2008). dying systems. J Colloid Interface Sci.
1979. 38. Ropes, MW, and Bauer, W: Synovial Syporell, WOT.
. Wilkins, RH, and Odom, GL: Ependymal- Fluid Changes in Joint Disease. Harvard 45. Kahn, CB, et al: Corticosteroid crystals
Choroidal cells in cerebrospinal fluid: University Press, Cambridge, MA, 1953. in synovial fluid. JAMA211:807, 1970.
Increased incidence in hydrocephalic 3h). Teloh, HA: Clinical pathology of synovial
infants. J Neurosurg 41:555, 1974. fluid. Ann Clin Lab Sci 5:282, 1975.
33 Nosanchuk, JS, and Kim CW: Lupus 40. Reeves, B: Significance ofjoint fluid
erythematosus cells in CSF. JAMA uric acid levels in gout. Ann Rheum Dis
25:2883, 1976. 24:569, 1965.
Chapter
Hematology Methods
Virginia C. Hughes, MS, MT(ASCP)SBB, CLS(NCA)I
Introduction OBJECTIVES
Specimen Collection At the end of this chapter, the learner should be able to:
Patient Identification
Safety if Calculate a manual white blood cell count, red blood cell count, and platelet count.
Verification of Laboratory 2 . Calculate a percentage of reticulocytes and an absolute/corrected reticulocyte count.
Requisitions
Labeling the Blood 3 . Calculate a reticulocyte production index (RPI).
Specimen . List errors that could occur in the performance of a centrifugal microhematocrit
Specimen Accessioning determination.
Method 1. Venipuncture
. Caiculate the mean corpuscular volume, mean corpuscular hemoglobin, and a mean
Method 2. Capillary Blood
corpuscular hemoglobin concentration.
Collection
Unopette Technology— . Identify factors that affect the erythrocyte sedimentation rate.
Manual Counts . Identify hemoglobins separated at alkaline and acid pH in hemoglobin electrophoresis.
Method 3. Red Blood Cell
. Name a method for quantifying hemoglobin A, and hemoglobin F.
Counts
Method 4. White Blood Cell . Describe the differential solubility test for hemoglobin S.
Counts
. Describe a procedure for detecting Heinz bodies.
Method 5. Platelet Counts
Evaluation of Peripheral . Identify conditions that show increased and decreased osmotic fragility.
Blood Smear . Describe the basic principle for the Ham’s test and the sugar water test for
Method 6. Slide Preparation paroxysmal nocturnal hemoglobinuria.
and Wright Stain
Method 7. The White Blood . Explain the basic principle of the cyanmethemoglobin method for hemoglobin
Cell Differential determination.
Methods Used in Detection . Calculate a correct white blood cell count for a peripheral blood smear containing
and Monitoring of more than 10 nucleated red blood cells per 100 white blood cells.
Anemia . Differentiate between a venipuncture and capillary puncture.
Method 8. Hemoglobin
Determination . Identify the appropriate site for capillary puncture on an infant.
Method 9. Microhematocrit
. Compare and contrast hereditary spherocytosis and pyruvate kinase deficiency in the
Determination autohemolysis test.
Method 10. Red Blood Cell
Indices
Method 11. Reticulocyte
Counts
Method 11A. Reticulocyte
Counts Using the Miller
Disc
Standard Methods for
Specific Anemias
Method 12. Sickle Screen
Method 13. Helena SPIFE®
Alkaline Hemoglobin
Electrophoresis
159
760. Chapter 31 Hematology Methods
3. Date of birth or age patient’s results can be detrimental and sometimes fatal.
4. Gender Many hospitals and laboratories have implemented preprinted
5. Name of physician patient labels that contain the information in Table 31-1. An
6. Location or room number example of a patient label is depicted in Figure 31—1. There
All of this information should be cross-checked with the lab- are many varieties of barcode printers. One example is the
oratory requisition form or laboratory orders for each patient. A-50 by Argox (Fig. 31-2). The A-50 is a direct thermal
printer compatible with 128 and Codabar.* The tubes in which
If the patient is not wearing a wristband, a nurse should be
blood is collected should be labeled after the blood is col-
notified to verify patient identification.
lected and not before; this alleviates labeling another tube if
OUTPATIENTS
blood cannot be collected successfully on the first venipunc-
ture. If preprinted labels are not available, the patient’s first
For outpatients who have blood collected, the phlebotomist
and last name and social security number should be written
should ask the patient to recite his or her full name, address,
indelibly on the tube of blood.
social security number, and date of birth. The phlebotomist
must check this information with the laboratory requisition
form or physician’s record of laboratory orders. Specimen Accessioning
Safety Once the specimen has been collected, it is accessioned and
delivered to the hematology or appropriate department of the
Universal precautions should be in practice at all times when
collecting blood specimens.' This means that all specimens
should be handled as if they were infected with an infectious AGE, GENDER DEPT y PATIENT ID#
agent (e.g., human immunodeficiency virus). Such safety pre- PATIENT —DOEJOHN M51YM ER 123450 02/2299
—— DATE OF DRAW
cautions include wearing a new pair of latex gloves for each
patient, washing hands between patients, and disposing of
needles, and contaminated supplies in a puncture-resistant
UNIQUE BAR CODE
biohazard container (sharps).
laboratory to be analyzed. In specimen accessioning, the _ Assemble necessary venipuncture equipment (Fig. 31-4).
information on the printed label is transmitted to the LIS, _Apply tourniquet 3 to 4 inches above phlebotomy site.
Oo
Ww
which can be done manually or with the aid of abarcode scan- Proper application of tourniquet is illustrated in Figure 31—5.
ner (Fig. 31—3).* The patient identifiers as well as the time the . Locate the vein using the index and middle fingers and
specimen was collected and received in the laboratory are gently palpate the vein The most common vein entered
permanently entered into the LIS as well as the initials of is the median cubital vein (Fig. 31-6). Remove the
the phlebotomist. tourniquet.
5. Cleanse the venipuncture site using 70% isopropyl alcohol.
Method 1: Venipuncture Cleansing is done using a circular motion, starting at the
inside of the venipuncture site to the outside area.
The most common anticoagulant used in the hematology lab- 6. Once the alcohol has dried the tourniquet is reapplied to
oratory is EDTA and the most common technique for obtain- the patients arm as illustrated in Figure 31—5. The plastic
ing the specimen is venipuncture using evacuated tubes, which cap of the needle is removed and fastened to the vacu-
allow for collection of multiple tubes. tainer holder. The phlebotomist’s nondominant hand
holds the skin taut while the dominant hand holds the nee-
Principle dle apparatus using the thumb, index, and middle fingers.
After aseptic cleansing of the antecubital fossa, a vacutainer The needle is inserted bevel up using an angle of 15 to
needle is inserted into the patient’s vein. An evacuated tube is 30 degrees.* Once the vein is entered, the evacuated tube
placed into the vacutainer holder breaching the rubber sheath is pushed completely into the tube holder. Allow tubes to
needle which facilitates flow of blood. fill completely because “short draws” may lead to erro-
neous results. The tourniquet is removed before the tube
Specimen or all tubes are filled. Remove the last tube from the
EDTA-anticoagulated blood needle holder and invert all tubes several times for proper
mixing. Place a sterile gauze pad over the site and remove
Equipment the needle in a quick motion. Place the clean gauze pad on
Vacutainer tubes the venipuncture site and ask the patient to hold it on his
Tourniquet or her arm.
Vacutainer holder
Alcohol pad
Vacutainer needle
A sterile
Adhesive bandage multiple-use needle
Procedure
1. Properly identify patient by cross-checking unique identi-
fiers (e.g., social security number) found on phlebotomy req-
uisition form with unique identifiers on patient wristband.
A holder
An evacuated
glass tube
Specimen
EDTA anticoagulated blood or capillary blood
Equipment
Microtainer tubes
Alcohol pad
Automated puncture device
Adhesive bandage
Procedure
|. Properly identify patients by cross-checking unique identi-
fiers on requisition form with those found on patient wrist-
3 4 band or ankle band (infants).
2. Cleanse the selected site with 70% isopropyl alcohol.
Figure 351-5 @ Application of the rubber tourniquet. (From Perform this task using a circulation motion and allow to
Wedding,
L ME,t and Toenjes, SA: Medical; Laboratory Procedures, air dry. For infants, the most common site used is the
FA Davis, Philadelphia, 1998, p 102, with permission).
plantar surface of the heel (Fig. 31—7) and for pediatrics,
adolescents, and adults the third and fourth fingers on
7. Label the blood tube or tubes. This inust be done prior to the palmar side fleshy surface of the distal phalanx
leaving the patient’s side. Most blood specimens are (Fig. 31-8).
labeled with printed bar code labels (see Fig. 31-1) con-
taining patient demographic and test information.
8. Apply an adhesive bandage to the patient’s arm and deliver HEEL PUNCTURE SITES
specimens to the appropriate laboratory department.
Cephalic vein
Basilic vein
Median
cubital vein
Accessory
cephalic vein
Basilic vein
CALCANEOUS
Cephalic vein (HEEL BONE)
Median
antebrachial vein
esa iiiiim ee
beneath puncture site and proceed to fill the tube by touch-
ing the tip of the Microtainer™ to the drop of blood. When
eT
|
1.0mm
tube is filled, remove the tip and secure the cap on the
= ea
Microtainer™ tube. Invert tube six to seven times to allow
proper mixing with anticoagulant.
4. Label Microtainer ™ tube appropriately and deliver the
specimen to respective laboratory department.
innHIMHIN
yun IAA
see! ae
MMs
Figure 31-10 @ Spencer Bright-Line double counting system with
improved Neubauer ruling. This represents an enlarged view of one
of the two ruled squares of the hemacytometer. The four corner pri-
mary squares are used for counting white blood cells. The arrows in
the upper left corner square represent the suggested counting
pathway of cells. Five secondary squares (labeled RBC) of the center
primary square are used for counting red blood cells. In platelet
enumeration, all 25 squares of the center primary square are
counted. (From Wedding, ME, and Toenjes, SA: Medical Laboratory
Figure 31-9 @ Microtainer tube containing EDTA.
Procedures. FA Davis, Philadelphia, 1998, p 277, with permission.)
Chapter 31. Hematology Methods 765
HEMACYTOMETER
Side View
the cells within the square and those touching the innermost
line are counted. If the boundary line is triple, all of the cells Figure 31-13 @ Unopette system, consisting of a reservoir
containing a diluent, a pipet used to deliver the specimen, a
within the squares and those touching the middle line inward
reservoir, and a pipet shield used to puncture the plastic seal on
are counted. reservoir, and serve as a cap for the pipet to prevent evaporation.
Hemacytometers and coverslips should meet the specifi-
cations of the National Bureau of Standards and are so Specimen
marked by the manufacturer. A standardized coverslip must EDTA-anticoagulated blood
be used that has been ground to fit the specifications of the
hemacytometer, ensuring a uniform depth and, therefore, a Equipment
constant volume. An ordinary coverslip must not be used. Unopette reservoir No. 5851
Manual cell counts are most often performed in cases of
Sodium chloride 8.5 ¢g
thrombocytopenia.
The Unopette system consists of calibrated pipets, Sodium azide O.l g
reagent-filled reservoirs to ensure precise dilutions, and a Distilled water 1000 mL
pipet shield to puncture the Unopette reservoirs (Fig. 31-13). Capillary pipet (10-uL)
Hemacytometer and coverslip
Method 3: Red Blood Cell Counts Microscope
Limitations
The hemacytometer must be properly filled to avoid erro-
neous results.
Figure 51-15 @ Unopette conversion to dropper assembly by Figure 31-17 @Red blood cells are illustrated at <400 using
withdrawing pipet from reservoir and securing it in reverse position Unopette methodology.
Chapter 31 Hematology Methods 767
4. Place the hemacytometer in a Petri dish lined with moist fil- stain, is the most common dye. A well prepared blood smear
ter paper and let sit for 10 minutes to allow platelets to settle. provides valuable information concerning the patient’s hemato-
5. Under 40X magnification using bright-light or phase logic status and is a vital component of the CBC (complete
microscopy, platelets are counted in all 25 small squares blood count) in the event a slide preparation is warranted.
within the center primary square (see Fig. 31-10). Count both
sides of the hemacytometer and average the results. Platelets Principle
appear as dense, dark bodies and can be round, oval, or rod- Blood from an EDTA-anticoagulated specimen or capillary
like, sometimes showing dendritic processes (Fig. 31-18). blood is applied to a standard glass slide using the slide-
6. Multiply the average number of platelets counted on both to-slide method. The blood smear is then stained using Wright
sides of hemacytometer by 1000 to get total platelet stain. Blood smears are fixed using methanolic fixative solu-
count/mm*. This step simplifies multiplication of dilution tion to stabilize cellular components. Eosin Y is an anionic
factor (100) and depth factor (10), which is equal to 1000. dye that stains cytoplasm pink to yellowish-red and methyl-
For example, if an average of 200 platelets are counted, the ene blue, which stains the nucleus and RNA blue.°
number per mm? is 200,000, while the number per liter is
200 X 10°.
Specimen
EDTA-anticoagulated blood
Limitations
A delay in mixing blood sample (anticoagulated sample) may
result in platelet clumping and subsequent erroneous results. Reagents/Equipment
The hemacytometer must be properly filled; underfilling or Clean glass slides
overfilling may lead to erroneous results. In the event of Diff-Quik® Stain Set®
platelet satellitosis (neutrophils ringed with adhesive platelets), Fixative Solution: 1.8 mg/L of triaylmethane dye in
obtain correct platelet counts by collecting a fresh specimen methyl! alcohol.
with sodium citrate as the anticoagulant.
Solution I: 1 g/L of xanthene dye, buffer and sodium
Reporting Results azide 0.01%
Results are expressed as number of platelets per uL or mm* Solution II: 1.25 g/L of thiazine dye mixture
(0.625 g/L of azure A and 0.625 g/L of
Normal Values methylene blue) and buffer
150,000—450,000/uL 150-450 x 10°/L Coplin jars
Immersion oil
Evaluation of the Peripheral Blood Smear
Procedure
Method 6. Slide Preparation 1. Place a small drop of EDTA-anticoagulated blood or
and Wright Stain capillary blood on the end of a clean glass slide
(Fig. 31-19A).
The wedge blood smear is the most common smear preparation
in the hematology laboratory and Wright stain, a Romanowsky
2. Place a second slide (spreader) in front of the drop of blood
at a 30 to 45 degree angle (Fig. 31-198).
3. Draw the spreader slide into the drop of blood and push the
spreader slide forward, spreading blood across the length of
the first slide. The smear at the opposite end of where the
drop of blood was placed should have a feathered edge
appearance (Fig. 31-19D). An example of an improperly
made blood smear is illustrated in Figure 31—19E. An unac-
ceptable unstained blood smear is shown in Figure 31-19C.
Acceptable and unacceptable stained smears are shown in
Figure 31-19F.
4. Discard the spreader slide into an appropriate biohazard
sharps container.
5. Gather 3 Coplin jars. Label as follows: fixative, solution I,
solution II.
6. Dip slide in Diff-Quik® fixative solution five times for
about | second each. Allow excess to drain.
7. Dip slide in Diff-Quik® solution I five times for about
Figure 351-18 ™ Photograph of platelets loaded onto a hemacy- | second each. Allow excess to drain.
tometer using a x40 objective (phase contrast). The platelets
appear as dense, refractile, dark bodies that can be round, oval, or Dip the slide in Diff-Quik® solution II five times for about
rod shaped with a diameter of approximately 2 to 4um. 1 second each. Allow excess to drain.
Well-made peripheral blood smear
4
vad
Figure 31-1 A. A drop of blood is placed on glass slide. B. A spreader slide is situated at a 30- to 45-degree angle. C. Draw back the
slide against the drop of blood and spread the blood smoothly. D. Adequate blood smear with feathered edge. £. Unacceptable blood smear.
F. Acceptable and unacceptable stained smears.
770. Chapter 31 Hematology Methods
Limitations
Only precleaned slides should be used for staining blood
smears. Characteristic staining patterns are described in
Table 31-2.
A popular automated slide stainer used in the hematol-
ogy laboratory is the Hema-Tek Slide Stainer (Fig. 31—20).
This automated slide stainer has positions for 25 slides on a
metal platen. Slides are stained at a rate of one slide per
minute. This instrument has excellent reproducibility and is
nearly maintenance free. The Wright’s stain is contained in a
“stain-pak” from a variety of manufacturers and sits within
the instrument connected by stylets and tubing. The Wright
stain is triggered to pump and deliver a precise amount of
solution to the slide via capillary space on the instrument.
Once stained, the slide is rinsed and dried on the platen and
eventually drops into a separate compartment.
Principle
One hundred white blood cells are counted and classified
on a Wright-stained peripheral blood smear using 1000x Pigure 351-20 m The Hema-Tek Slide Stainer and Stain-Pak.
magnification. (Courtesy of the Bayer Corporation, Tarrytown, NY.)
Reagents/Equipment
Microscope
Differential counter a. Scan the edges using low power (10X objective) and
Hand counter center of the slide to be sure there are no clumps of
RBCs, WBCs, or platelets.
Procedure b. Scan the edges for abnormal cells.
1. Determine the overall staining quality of the blood smear. 3. Find an optimal area for the detailed examination and enu-
2. Determine if there is a good distribution of the cells on the meration of cells.
smear. a. The RBCs should not quite touch each other.
b. There should not be areas containing large amounts of
broken cells or precipitated stain.
c. The RBCs should have a graduated central pallor.
4. Switch to high power (40X objective). Determine the
WBC estimate.
: ott a. Count the number of white blood cells in ten fields and
GalalacCanctitient divide by 10. Multiply this number by 2000. This factor is
peg SIRE ea based on the fact that each WBC seen in a high power field
ee Sones (hpf) is equivalent to approximately 2000 cells per uL.
Picts roePee Correlate the WBC estimate with the WBC counts per
MEAG oi ese oT ; mm’. Other methods correlate the average number of
Metamyelocyte cytoplasm i WBCs/hpf with a range of WBC counts from a previously
Neutrophil cytoplasm i established table found in the procedure manual used in
Lymphocyte cytoplasm the laboratory.
ee pte 5. Perform a 100 WBC differential count.
Bone Pink Evaluate RBC anisocytosis, poikilocytosis, hypochroma-
Basophilic granules Purple-black sia, polychromasia, and RBC inclusions (see Chap. 5).
Neutrophilic granules Pink-purple 6. Perform a platelet estimate and evaluate platelet
Platelet granules Pink-purple morphology.
fo cece pee purple a Count the number of platelets in 10 oil immersion fields.
Howell—Jolly bodies Purple b. Divide by 10.
Cabot rings Pink-purple c. Multiply by 20,000/mm? if a wedge smear is
used; multiply by 15,000 if it is an automated smear.
Chapter 31. Hematology Methods 771
Report platelet estimate as adequate, increased, or Drabkin’s reagent: Dissolve | g of sodium bicarbonate,
decreased. 0.2 g of potassium ferricyanide, 0.05 g of sodium
7. Correct any total WBC count per mm} that has greater than cyanide in distilled water, qns to | liter.
10 nucleated red blood cells (NRBCs) per 100 WBCs. Hemoglobin standard
a. When performing the WBC differential, do not include
NRBCs in the count; NRBCs are reported as the number Procedure
of NRBCs per 100 WBCs. 1. Acquire reference material for hemoglobin at an approxi-
b. Use the following formula to correct a WBC count: mate value of 20 g/dL. Make four dilutions of reference
Corrected WBCs/mm* = WBC/mm? X 100 material using Drabkin’s reagent so that hemoglobin values
100 + No. of NRBCs/100 WBCs are four to six increments apart (e.g. 6, 10, 16, and 20 g/dL
of hemoglobin). Mix the contents of tubes and analyze at
8. The absolute WBC count may be determined by multiply- 540 nm using a spectrophotometer. Plot hemoglobin values
ing the differential (relative) count (%) by the total WBC versus absorbance on standard graph paper.
count.’ For example, to determine the absolute count of 2. For patient testing, add 5 mL of Drabkin’s reagent to a
segmented neutrophils when 60% of segmented neu- clean test tube.
trophils are counted in the differential and the total WBC 1SS). Add 20 uL of EDTA-anticoagulated whole blood to the tube.
count is 10,000/uL: 4. Mix contents of tube and allow to stand at room tempera-
Absolute segmented neutrophils = 10,000/uL < 60%/100 = ture for 5 minutes. Read absorbance at 540 nm on a spec-
6000/uL trophotometer.
5. Determine patient hemoglobin using standard curve pre-
Normal Values pared in step 1.
Relative counts
Lymphocytes 20-44% Limitations
Drabkin’s reagent should be kept away from direct sunlight.
Monocytes 2-9%
Adding Drabkin’s reagent to samples containing hemoglobin
Neutrophils 50-70% C or S will yield a turbid suspension and will fail to lyse the
Bands 2-6% red cells. To circumvent this, add 5 mL of water to mixture
Eosinophils 04% and incubate at room temperature for 5 minutes. Read off of
the spectrophotometer and double the result.
Basophils 0-2%
Absolute counts Reporting Results
Lymphocytes 1.2-3.4 XX 10°/uL 1.2-3.4 XX 10°/L Results for hemoglobin are reported in units of g/dL.
Monocytes 0.11-0.59 XK 10°/uL 0.11-0.59 x 10°/L
Normal Values
Neutrophils 1.4-6.5 x 10°%/uL s1.4-6.5 10°/L Newborn: 17-23 g/dL
Bands 0-0.7 X 10?/uL 0-0.7 X 10°/L Infant child: 9-14 g/dL
Eosinophils 0-0.5 X 10°/uL 0-0.5 X 10°/L Adult men: 14-18 g/dL
Basophils 0-0.2 X 10°/uL 0-0:2 x 107/L Adult women: 12-16 g/dL
Limitations
If capillary blood is used, the first drop should be wiped away
in capillary blood collection to avoid contamination with
interstitial fluid, which will dilute out the sample.
The buffy coat should not be included in the packed red
cell volume reading.
If the patient is on chloramphenicol or penicillin, a
decreased hematocrit level may be found.’
Reporting Results
The hematocrit is reported as a percentage of red cells to Figure 31-235 ™ Microhematocrit rotor. (Photo courtesy of Inter-
total blood volume. national Equipment Co.)
Chapter 31 Hematology Methods 773
Normal Values
MCV 80-94 fL
MCH 28-31 pg
— Buffy coat MCHC 32-36%
Interpretation
Determination of the MCV, MCH, and MCHC gives valuable
information that helps to characterize RBCs. According to the
— Red blood cells
MCV, erythrocytes may be classified as normocytic, micro-
cytic, or macrocytic. Based on the MCHC, erythrocytes may
be classified as normochromic or hypochromic (Table 31-3).
The MCH expresses only the mean weight of hemoglobin per
erythrocyte.
So that with a Hgb of 14 g/dL and an RBC count of 4.2 MEAN PLATELET VOLUME
million/mm*, the MCH = 33 pg. The mean platelet volume (MPV) represents the average size
of platelets. It is determined from the arithmetic mean of the
platelet histogram provided by the hematology analyzer (See
chapter 32). The normal values may range from 7.4 to 10.4 fL.
The MPV may be increased in idiopathic thrombocytopenic
purpura (ITP) and in hypoxic states of chronic obstructive pul-
monary disease (COPD).'°
Normal Values
Reticulocyte Count
Newborn: 2.5-6.0%
Adult: 0.5-2.0%
Absolute reticulocyte count: 24-80 X 10°/L
RPI 3 or greater
Maturation Time
1 day
1.5 days
2 days
Figure 31-26 @ Photograph of a reticulocyte using new methyl- 3 days
ene blue stain.
Chapter 31 Hematology Methods 775
Principle Reagents/Equipment
Using the Miller ocular disc, reticulocytes and mature Reaction vials prefilled with sodium hydrosulfite
red cells are separated and counted in separate squares of
Phosphate buffer
the disc. Using a factor of 9, reticulocytes can be easily
calculated. SickleScreen control set
Deionized water
Reagents and Equipment Tube reading rack
New methylene blue (NMB)
Miller ocular disc Procedure
Microscope slides 1. Bring all reagents to room temperature.
2. Label one reagent tube for each patient and control.
10 X 75 mm test tubes
3. Place tubes in reading rack and add phosphate buffer to
6-inch capillary tubes each tube until it reaches the “Fill to this level” mark.
Hand counter 4. Add 50 ub of patient sample and controls to respective
tubes.
Procedure 5. Incubate at room temperature for 10 to 20 minutes.
1. Dissolve 10 g of NMB and 8.9 of NaCl (0.152 mol/L) in
1000 mL of distilled water. Filter through Whatman No. | Interpretation
filter paper before use. Store at room temperature. Positive: If hemoglobin S or another sickling hemoglobin is
2. Add an equal number of drops of thoroughly mixed EDTA present the solution will be turbid and the dark lines of the
anticoagulated blood and NMB mixture to a glass 10 X reading rack will not be visible when viewing through the
75 mm tube. tubes.
3. Make a conventional wedge smear and let air dry. Do not Negative: If no sickling hemoglobin is present, the solu-
counterstain with Wright’s stain. tion will be clear to slightly cloudy and the lines of the tube
4. Use a 100X objective and a 10 ocular secured with reading rack will be visible through the tubes. Both positive
Miller disc or similar apparatus and count the reticulocytes and negative reactions are illustrated in Figure 31—27.
and RBCs on the smear. As in differential enumeration of
WBCs, choose a section of the smear where cells are close Limitations
to each other but not touching or overlapping. Count a If the hemoglobin level is 8 g/dL or less, a false-negative
total of 20 fields. result may occur.
5. In each field, count RBCs in the smaller square and reticu- Patients with multiple myeloma or other plasma cell
locytes in the larger square. Determine percent reticulo- dyscrasias may yield false-positive results
cytes by the following formula: Elevated hemoglobin F levels may yield false-negative
results and therefore this test should not be performed on
% Reticulocytes = Total reticulocytes in larger square _
infants 6 months of age or younger.'*
Total RBC in smaller square X 9 oe
For example, if 10 reticulocytes are counted in the larger
square and 900 mature RBCs in the smaller square, the %
reticulocytes would be:
10 X 100 = 0.12%
900 x9
Principle
Red blood cells are lysed by a surfactant and the released
hemoglobin is reduced by sodium hydrosulfite. While reduced
hemoglobin A is soluble in this medium, hemoglobin S is insol-
uble and forms a turbid suspension in phosphate solution. Both
sickle cell disease and sickle cell trait can be detected with the Figure 351-27 @® Positive (left) and negative (right) results of sickle
SickleScreen® test. solubility test.
776 Chapter 31 Hematology Methods
A patient that has been recently transfused may yield 5. Remove one disposable applicator blade assembly from
false-negative results. the packaging. Remove the protective guard from the
Patients with hemoglobin variants such as hemoglobin blade by gently bending the protective piece back and
Charlem OF Coeorgetown May yield positive reactions (Table 31-5). forth until it breaks free.
6. Place the applicator blade into the vertical slot numbered
Reporting Results 2 in the applicator assembly. Note that the blade assembly
Results are reported as positive or negative. will fit into the slots in the applicator assembly only one
way; do not try to force the applicator blade into the slots.
7. Slide the disposable sample cup strips into the top chan-
Method 13. Helena SPIFE® Alkaline nel of the cup tray (numbered | to 20).
Hemoglobin Electrophoresis 8. Pipet 17 uL of patient or control hemolysate into the
disposable cups. Cover until ready for use.
Principle
Small samples of hemolysates prepared from packed cells are
Gel Application
automatically applied to the SPIFE alkaline hemoglobin gel."* 9. Remove the gel from the protective packaging and dis-
The hemoglobins in the sample are separated by electrophore-
card the overlay.
sis using an alkaline buffer and are stained with acid blue
10. Using a REP blotter C, gently blot the entire gel with each
stain. The patterns are scanned on a densitometer and the rel-
blotter using slight fingertip pressure on the blotter.
ative percentage of each band is determined.
Remove the blotter.
11. Dispense approximately 2 mL of REP prep onto left side
Specimen
of SPIFE chamber.
EDTA-anticoagulated blood
12. Place the left edge of the gel over REP prep, aligning the
round hole on the left pin. Gently lay the gel down on
Reagents/Equipment
the REP prep, starting from the left side and ending on the
SPIFE Alkaline hemoglobin gel
right side, fitting the obround hole over the right pin. Use
Acid blue stain paper towel or absorbent paper to wipe around the edges
REP/SPIFE hemolysate reagent of the gel backing, especially next to electrode posts to
Citric acid destain remove excess REP prep. Make sure that the gel lays flat
in the chamber and that no bubbles remain under the gel.
AFSC Hemo Control
13. Clean the electrodes with deionized water and wipe with
AA, Hemo Control lint-free tissue before and after use.
SPIFE instrument 14. Place a carbon electrode on the outside ledge of each gel
block outside the magnetic posts.
Procedure 15. Press the Test Select/Continue buttons located on the elec-
1. Centrifuge anticoagulated blood for 10 minutes to separate trophoresis and stainer sides of the instrument until the
cells from plasma. alkaline hemoglobin option appears on the displays.
2. Remove plasma.
3. Wash packed cells three times by resuspending in 5 to Electrophoresis/Staining
10 volumes of normal saline solution (0.85% NaCl), cen- 16. With alkaline hemoglobin on the display, press the start/stop
trifuging and aspirating supernatant as before. button. An option to either begin the test or skip the opera-
4. After washing the samples, prepare the samples by mixing tion will be presented. Press start/stop again to begin.
10 uL of sample with 100 uL of hemolysate reagent. 17. The auto applicator speed should be set at 4. Place the
Vortex-mix or shake vigorously for 15 seconds. REP auto applicator assembly onto the tray alignment
pins. Lower the applicator tips down into the sample wells
by flipping the toggle switch to the down position. Press
test select/continue to time sample loading.
18. After 30 seconds, lift the applicator tips out of the wells
by flipping the toggle switch to the up position.
19. Carefully lift the entire applicator assembly away from the
Positive Negative cup tray and immediately place it onto the alignment pins
HAbCyartem HbA
located on the SPIFE electrophoresis chamber. Lower the
HbS HbF applicator blades down onto the gel by flipping the toggle
ADS Gravis HbC switch to the down position. Lower the chamber lid.
HbCziguinchor HbD
20. Press the test select/continue button. Sample application
HbG
HbE will be timed for 30 seconds.
HG resee
21. After sample application is complete, raise the lid and
HbA, carefully lift the applicator blades by flipping the toggle
switch to the up position.
Chapter 31. Hematology Methods 777
22. Remove the applicator assembly from the electrophoresis CELLULOSE ACETATE pH 8.4
chamber. Discard the applicator blade and sample cups as
biohazardous.
oaNormal
23. Close the chamber lid, and press the test select/continue
Sickle trait
button to start electrophoresis. SPIFE will beep when
electrophoresis is complete. Hemoglobin D trait
24. After electrophoresis is complete, use the Gel Block SC disease
Remover to remove both gel blocks from the gel. Lift the
SE disease
gel holder from the stainer chamber. Attach the gel to
the holder by placing the round hole in the gel mylar over Normal cord blood
the left pin on the holder and the obround hole over the C Harlem trait
right pin on the holder. Control
25. Place the gel holder with the attached gel facing back-
wards into the stainer chamber. OoCA;A2 S FA
26. With alkaline hemoglobin on the display, press the C D
E G
start/stop button. An option to either begin the test or skip
O
the operation will be presented. Press start/stop again to
begin. The instrument will stain, destain, and dry the gel. CITRATE AGAR pH 6.0-6.2
27. When the process is complete, the instrument will beep. QaES
Normal
Remove the gel holder from the stainer. Take the gel off
of the holder and replace the holder. Sickle trait
Hemoglobin D trait
Interpretation
The hemoglobin gels may be inspected visually for the presence SC disease
of hemoglobin bands. The Helena Hemo controls provide a SE disease
marker for the band identification. The controls AA, and AFSC
Normal cord blood
Hemo Control should be used as markers for the ___location of
hemoglobin bands. Hemoglobin electrophoresis on cellulose C Harlem trait
acetate and citrate agar is depicted in Figure 31—28. In addition,
Control
the relative percentage of each hemoglobin band by scanning
the dried gels in the densitometer using a 595-nm filter.
o = Designates origin
Limitations
Citrate agar electrophoresis may be necessary to confirm
abnormal hemoglobins
Anion-exchange column chromatography is a more
Figure 31-28 ™ Comparative hemoglobin electrophoresis. Hemo-
accurate method for quantitating HgbA).
globin electrophoresis on cellulose acetate and citrate agar, indicat-
ing patterns of mobility. The width of the band is not indicative of
Normal Values hemoglobin concentration.
Birth 100% Hgb F
Adult 98% Hgb A
3.5% Hgb A; Reagents/Equipment
<2% Hgb F SPIFE Acid Hemoglobin gel
Acid blue stain
REP/SPIFE Hemolysate Reagent
Method 14. Helena SPIFE® Acid
Citric acid destain
Hemoglobin Electrophoresis
AFSC Hemo Control
Principle SPIFE instrument
Very small samples of hemolysates prepared from packed
cells are automatically applied to the SPIFE Acid Hemoglo- Procedure
bin gel.'* The hemoglobins in the sample are separated by 1. Centrifuge anticoagulated blood for 10 minutes to separate
electrophoresis using a citrate buffer and are stained with acid cells from plasma.
blue stain. 2. Remove plasma.
3. Wash packed cells three times by resuspending in 5 to
Specimen 10 volumes of normal saline solution (0.85% NaCl),
EDTA-anticoagulated blood centrifuging and aspirating supernatant.
7718 Chapter 31 Hematology Methods
4. After washing samples, prepare the samples by mixing a Lower the Applicator blade down onto the gel by flipping
10-uL sample with 100 uL of hemolysate reagent. Vortex- the toggle switch to the down position.
mix or shake vigorously for 15 seconds. 22. Press the test select/continue button. Sample application
Nn . Prepare control (AFSC) mixing | part contro! to | part will be timed for 30 seconds.
hemolysate reagent. _ After sample application is complete, carefully lift the
. Remove one disposable applicator blade assembly from Applicator blade by flipping the toggle switch to the up
the packaging. Remove the protective guard from the position.
blade by gently ending the protective piece back and forth 24. Remove the Applicator assembly from the electrophoresis
until it breaks free. chamber. Discard the applicator blades and sample cups
. Place the applicator blade into the numbered vertical as biohazard waste.
slots in the applicator assembly. Note that the blade _ Close the chamber lid, and press the test select/continue
assembly will fit into the slots in the applicator assem- button to start electrophoresis. SPIFE will beep when
bly only one way; do not try to force the applicator electrophoresis is complete.
blade into the slots. . After electrophoresis is complete, use the gel block
. Raise the applicator blades by flipping the toggle switch remover to remove the gel blocks. Place one electrode
to the up position. Using the adjustment knob, set the across each end of the gel to prevent curling during
Auto Applicator speed to 4. drying.
. Slide the disposable sample cups strip into the center . Close the chamber lid and press the test select/continue
channel of the cup tray (numbered 21 to 40). button to dry the gel.
. Pipet 17 uL of patient or control hemolysates into each of . After the gel has been dried, carefully remove the gel
the disposable cups. Cover until read for use. from the electrophoresis chamber.
Oe Remove the gel from the protective packaging and discard . Remove the gel holder from the stainer chamber. Attach
the overlay. the gel to the holder by placing the round hole in the gel
I, Using a REP Blotter C, gently blot the entire gel with each mylar over the left pin on the holder and the obround hole
blotter using slight fingertip pressure on the blotter. over the right pin on the holder.
Remove the blotter. . Place the gel holder with the attached gel facing back-
. Dispense approximately 2 mL of REP Prep onto left side wards into the stainer chamber.
of SPIFE chamber. Si Press the test select/continue button until acid hemoglobin
. Place the left edge of the gel over REP Prep, aligning the appears on the display.
round hole on the left pin. Gently lay the gel down on 52) With acid hemoglobin on the display, press the start/stop
the REP Prep, starting from the left side and ending on button. An option to either begin the test or skip the oper-
the right side, fitting the obround hole over the right pin. ation will be presented. Press start/stop to begin. The
Use paper towel or absorbent paper to wipe around the instrument will stain, destain, and dry the gel.
edges of the gel backing, especially next to electrode . When the gel has completed the process, the instrument
posts, to remove excess REP Prep. Make sure that the will beep. Remove the gel holder from the stainer. Take
gel lays flat in the chamber and that no bubbles remain the gel off the holder and replace the holder.
under the gel.
. Clean the electrodes with deionized water and wipe with Interpretation
lint-free tissue before and after use. The hemoglobin gels should be inspected visually for the
. Place a carbon electrode on the outside ledge of each gel presence of abnormal hemoglobin bands (see Fig. 31-28).
block outside the magnetic posts. Glycated hemoglobin migrates with HbF. The Helena AFSC
. Press the test select/continue buttons located on the elec- Hemo Control provides a marker for band identification. This
trophoresis and stainer sides of the instrument until the control should be run on each SPIFE Acid Hemoglobin gel.
acid hemoglobin option appears on the display. The control verifies all phases of the procedure and acts as a
. With acid hemoglobin on the display, press the start/stop marker to aid in the identification of the hemoglobins in the
button. An option to either begin the test or skip the oper- unknown samples.
ation will be presented. Press start/stop to begin.
. The Auto Applicator speed should be set at 4. Place Limitations
the REPAuto Applicator Assembly onto the tray align- Anion-exchange column chromatography is the most accurate
ment pins. Lower the applicator tips down into the method for quantitating HgbA,. Low levels of HgbF may be
sample cups by flipping the toggle with to the down accurately quantitated using radial immunodiffusion.
position. Press Test Select/Continue to time sample
loading. Normal Values
. After 30 seconds, lift the applicator tips by flipping the Birth 100% Hgb F
toggle switch to the up position.
. Carefully lift the entire Applicator Assembly away from
Adult 98% Hgb A
the Sample Tray and immediately place it onto the align- 3.5% Hgb A,
ment pins located on the SPIFE electrophoresis chamber. <2% Hgb F
Chapter 31 Hematology Methods 779
Method 15. Hemoglobin A, Determination aspirate the undrained supernatant (making sure not to
disturb the resin) and discard.
Principle 6. Slowly and carefully apply 100 uL of the patient
The anion-exchange resin is a preparation of cellulose cova- hemolysate to the column. During application, do not
lently coupled to small positively charged molecules. The pos- allow the sample to form bubbles or run down the side of
itively charged cellulose attracts negatively charged molecules. the tube. Excessive force used during application will
Proteins, such as the hemoglobins, contain many positive and disturb the resin and may cause erroneous results.
negative charges due to the ionizing properties of the compo- 7. Immediately after sample application to the column, add
nent amino acids. In the anion-exchange chromatography of 100 uL of the same patient hemolysate to a Total Fraction
HbA,, buffer and pH levels are controlled to cause different collection tube. QS the tube to the scribed line using
hemoglobins to possess different net negative charges. These deionized water. Total volume = 15 mL.
negatively charged proteins are attracted to the positively 8. Allow the sample to completely absorb into the resin. The
charged cellulose and bind accordingly. After binding, the pro- hemolysate will have a glossy appearance when viewed
teins are removed selectively from the resin by altering the pH from above until the sample is completely absorbed by the
or ionic strength of the elution buffer. Because of the pH of the resin. On complete absorption, the top of the resin will
resin and the ionic strength of the HbA, developer, HbA, does have a dull, mat-like appearance.
not bind to the positively charged cellulose and is eluted as the 9. Following the absorption of the sample into the resin bed,
developer moves through the column. The other normal and slowly apply 2.5 mL of HbA, developer to the column.
most abnormal hemoglobins are retained by the resin. The Excessive force when applying the developer may disturb
HbA, fraction is compared to a total hemoglobin fraction by the resin and cause invalid results.
determining the absorbance of each using a spectrophotometer 10. Allow all of the developer to pass through the column into
and then calculating the percentage of HbA,.'° the HbA, collection tube (approximately 30 minutes to
2 hours). The eluate contains the HbA,.
Specimen 11. QS the small collection tube to the scribed line with
EDTA-anticoagulated blood deionized water. Total volume = 3 mL.
12. Invert both collection tubes several times to mix contents
Reagents/Equipment thoroughly.
Beta-Thal HbA, Quik Columns 13. Determine the HbA, percentage in each sample using a
HbA, Developer standard spectrophotometer.
A. Adjust the wavelength to 415 nm
Hemolysate Reagent C
B. Zero the instrument with deionized water.
Spectrophotometer C. Read and record the optical density (O.D.) of both the
HbA, and TF collection tubes.
Procedure D. Determine the HbA, % as follows:
1. For each patient quantitation, obtain a Quik Column, HbA,
Absorbance of HbA, fraction X 100
Collection tube, and total fraction tube. = HbA, %
5 (Absorbance of TF solution)
.
i) Allow the appropriate number of columns and reagents to
come to room temperature. where HbA2% = percentage of HbA2 in sample
3. Prepare patient samples by placing 50 uL of whole blood Absorbance of HbA2 fraction = absorbance of the con-
into a small laboratory test tube. Add 250 ul of hemolysate tents of the small collection tube at 415 nm (HbA2 fraction).
reagent C to test tube. Absorbance of the TF solution = absorbance of the con-
4. Shake the tube to achieve complete hemolysis of sample. tents of the large collection tube at 415 nm (other hemoglobin
Complete lysis of the sample is essential for accurate fractions).
results. Allow the sample to stand 5 minutes before use. 5 = dilution factor (15 mL of TF tube/3 mL of HbA2
5. Preparation of columns. tube 5 5)
A. Upend each column twice to remove any resin from the 100 = percentage conversion factor
top cap closure. Remove the top cap closure and gently
resuspend the entire contents of the column using a Pas- Interpretation
teur pipet with a small rubber bulb. Make sure all the Results of HbA, should be interpreted in conjunction with
resin is resuspended above the filter. patient history, total hemoglobin values, and other clinical
B. Immediately after resuspension of each column, hold and lab findings. Any value between 3.5% and 8% is consid-
the column over a sink or absorbant paper and remove ered indicative of beta (8)-thalassemia trait. Values above
the bottom tip closure, allowing the buffer to drain. 8% indicate the presence of additional hemoglobin variants
If the column is allowed to stand with the bottom tip such as HbC, E, O, D, G, S, or S—G hybrid.
closure in place, resuspension must be repeated.
C. As the resin repacks, you will see an interface slowly Limitations
move up the tube. As soon as the slurry settles to form 1. Failure to completely resuspend the contents of the column
an interface of resin and remaining supernatant above, may cause slow flow and erroneous results. Time must be
780. Chapter 31 Hematology Methods
allowed after resuspension for the formation of a distinct Hemolysate reagent: 0.07% potassium cyanide and
interface between the resin and supernatant. Any trapped 0.005 M EDTA
bubbles may be removed with a Pasteur pipet. AFSC Control (cat. no. 5331)
2. The bottom tip closure must be removed immediately after
AA, Control (cat. no. 5328)
resuspension. Resuspension must be repeated if the column
is allowed to site with the bottom closure in place after A rapid electrophoresis (REP) unit used to elec-
resuspension. Failure to do so may cause slow flow and trophorese, dry, and scan gels
erroneous values.
3. As soon as the resin repacks, the remaining supernatant Procedure
must be aspirated and discarded. 1. Prepare a 1:4 dilution with IEF hemolysate reagent. Mix
4. To avoid back pressure in the column, do not remove the 1 part (25 uL) of whole blood with three parts (75 wL)
bottom tip closure before removing the top cap closure. hemolysate reagent.
Any bubbles trapped in the column resin may slow or stop 2. Make a 1:8 dilution by mixing one part (10 uL) packed
the flow rate, leading to erroneous results.
cells to 7 parts (70 uL) hemolysate reagent.
5. If the developer ceases to flow through the column the col- 3. Place three 1/8-inch diameter discs into a test tube.
umn must be discarded and the quantitation repeated with Reconstitute with 25 uL of hemolysate reagent. 2 uL must
a fresh column. be applied by hand to the gel.
6. Any disturbance of the resin during the procedure may 4. Vortex-mix briefly and allow the sample to stand at least
cause erroneous results. No more than 5 minutes should 5 minutes before use. The cells should be completely
elapse from the time the column stops flowing until the lysed before gel application.
developer is added so that the resin does not dry out. 5. Place 30 sample cups into the sample tray. Place 75 uL of
7. Some of the abnormal hemoglobins (HbS, C, E, O, E, G, sample or control lysates into appropriate sample cups.
S-G hybrid) are eluted with HbA, in this method. The pres- 6. Place REP blotter A on the sample tray in area adjacent to
ence of the abnormal hemoglobins should be confirmed by the sample cups. Place approximately 4 mL of Sureprep®
electrophoretic techniques. HbF does not interfere with this into outside wash well of sample tray. Place approxi-
method. mately 4 mL of water into the inside wash well of the
sample tray.
Normal Values 7. Remove the gel from the protective packaging and dis-
Normal HbA, range = 2.2% to 3.3% card overlay. Use the REP prepper to remove excess
moisture from the sample wells and trough.
8. Place left edge of gel over the chamber, aligning the round
Method 16. Isoelectric Focusing hole on the left pin. Gently lay the gel down on the cham-
ber starting from the left side and ending on the right side,
Principle fitting the obround hole over the right pin.
The isoelectric point (pI) is the pH at which a molecule carries 9. Insert the IEF electrode adapter marked (Front) between
no net electrical charge. In order to have a sharp isoelectric the two magnetic posts located at the front of the chamber
point, a molecule must be amphoteric, meaning it must have floor. Insert the IEF electrode adapter marked (Back)
both acidic and basic functional groups. Proteins and amino between the two magnetic posts located at the back of the
acids are common molecules that meet that requirement. Pro- chamber floor.
teins can be separated according to their isoelectric point in a 10. Clean and wipe the IEF electrodes with a lint-free tissue.
process known as isoelectric focusing (IEF). At a pH below the 11. Place an electrode into the slots created by the adapter—
pl, proteins carry a net positive charge. Above the pI they carry one on each outside edge and one in the middle. Be sure
a net negative charge. The pH of an electrophoretic gel is deter- all three electrodes are seated firmly against the electrode
mined by the buffer used for that gel. If the pH of the buffer is gel blocks.
above the pI of the protein being run, the protein will migrate 12. The REP unit will automatically apply samples and elec-
to the positive pole. If the pH of the buffer is below the pI of trophorese and dry the gel.
the protein being run, the protein will migrate to the negative 13. Remove the gel from the REP unit for evaluation. If the
pole of the gel. If the protein is run with a buffer pH that is gel is to be used for future evaluation, it will be necessary
equal to the pl, it will not migrate at all. By using ampholyte to dry down the electrode gel blocks by placing the gel
buffers in the pH range of 6 to 8, the separation and identifica- into an I.0.D. at 60°C for 10 to 15 minutes.
tion of abnormal hemoglobins are possible based on migration 14. Gels may be visually examined or scanned using a
of proteins based on their isoelectric points." 415-nm interference filter.
Specimen
Interpretation
EDTA-anticoagulated blood or heparinized blood
HbE and Hb,-Arab migrate slightly anodal to HbA,. HbG-
Reagents/Equipment Philadelphia and HbD-Los Angeles are separated from HbS
REP Hemoglobin IEF gel: each gel contains 1% wt/vol as 18 Hby epore-HbA, HbF are clearly separated from each other,
agarose, 5.3% vol/vol carrier ampholytes, and allowing identification between sickle cell trait, sickle cell
0.01% thymol as a preservative. anemia, and HbS/f-thalassemia (Fig. 31-29). The AFSC
Chapter 31 Hematology Methods 781
Specimen
EDTA-anticoagulated specimen.
Reagents/Equipment
Fetal cell fixing solution (80% reagent alcohol)
Fetal cell buffer solution (citrate buffer, 0.081 M)
Fetal cell stain (erythrosin-B, fast green)
Microscope
Glass slides
0.85% saline
Ey ve G Philadelphia Glass test tubes
Coplin jars
~S D Los Angeles
Procedure
HbA»
O-Arab, E 1. Mix the blood sample well by gentle inversion.
pe > —— 2. Place three drops of 0.85% saline and two drops of mater-
Constant
Spring
pring — A> nal blood into a glass test tube. Mix by gentle agitation.
3. Place one drop of diluted blood on a glass slide near one
pH 8.0 Cathode end. Prepare thin film by drawing the edge of another
slide through the drop of blood, and across the slide.
a - denatured Hb
b - ferrous/ferric hybrid 4. Air dry the slide at room temperature.
c - ferrous/ferric hybrid 5. Place the smears in a clean vessel containing sufficient
d - methemoglobin fetal cell fixing solution to cover the smears. Let stand for
5 minutes. Note: the slides should be placed in the groove
Pigure 51-29 ™ Schematic of isoelectric focusing pattern. (Helena of the Coplin jars singly, so that the smear is adequately
Laboratories, Beaumont, TX.) exposed to solution.
6. Remove the smears and rinse thoroughly in distilled
water. Allow the slides to drain dry.
Control and AA, control should be run with each gel. Dilute 7. Place the smears in a clean vessel containing sufficient fetal
the AFSC Control 1:2 with hemolysate reagent before use. cell buffer solution to cover the smears. Allow to remain in
solution at room temperature for 8 to 10 minutes.
Limitations 8. Remove the slides from the solution and immediately
1. HbGgaiveston ANd HbGy, 44, Cannot be distinguished from HbS. place in a clean vessel containing fetal cell stain. Stain for
2. HDpammersmithy Hbgethesdas Hbgrigham Carmot be distinguished 3 minutes.
from HbA. 9. Remove the slides from the fetal cell staining solution and
. HbE, HbCyaiem, HbO,,., and Hbx,;, cannot be separated.
rinse thoroughly in distilled water. Dry slides at room
temperature.
WwW HbNopattimore and HbL;,.,., cannot be separated.
5. HbCC cannot be distinguished from HbC/B-thalassemia and 10. Examine slides under the oil-immersion lens. Fetal cells
HbEE cannot be distinguished from HbE/B-thalassemia. will stain a dark reddish-pink while adult cells contain-
6. If the gels exhibit drying under low ambient humidity, ing hemoglobin A will appear white to light pink with a
darker center (Fig. 31—30). Slides should be read within
bands will be skewed and exhibit uneven migration. A band
24 hours. A positive control may be prepared from a
will migrate in an arched manner if the concentration of
mixture of | part cord blood to 9 parts normal adult
hemoglobin is too high. A sample application of 1 uL
blood.
should rectify the problem.
7. Bands from fresh whole blood may electrophorese at a Interpretation
slightly slower rate than the control. Older samples may The common means of reporting fetal cells is as a percentage
electrophorese at a slightly faster rate than the control. If of normal adult cells. This ratio is achieved by randomly
controls do not perform as expected, test results should be observing 8 to 10 fields of cells. Count the number of adult
considered suspect or invalid. cells in each field and total them. Count the number of fetal
cells in each of the same fields and total the fetal cells. Deter-
mine the percentage of fetal cells to adult cells by dividing the
Method 17. Hemoglobin F Acid Stain
total number of fetal cells counted by the number of adult
Principle cells counted."
Red cells containing fetal hemoglobin are resistant to acid For example, 25 fetal red cells and 4000 adult cells are
elution whereas red cells containing hemoglobin A will counted. The percentage of fetal cells to adults cells is
appear as ghost cells on exposure to acid. 25/4000 X 100 = 0.6%.
782. Chapter 31 Hematology Methods
Reagents/Equipment
Glucose-6-phosphate (0.01 M)
Triphosphopyridine nucleotide (0.0075 M)
Digitonin solution
Potassium phosphate buffer
UV light
Whatman No. | filter paper
Procedure
|. Prepare 0.01 M glucose-6-phosphate by dissolving 42 mg
of glucose-6-phosphate in 10 mL of distilled water.
2. Prepare 0.0075 M triphosphopyridine nucleotide by dissolv-
ing 18.75 mg of triphosphopyridine nucleotide in 1.0 mL of
1% sodium bicarbonate solution. Figure 31-31 @ Heinz bodies.
Chapter 31 Hematology Methods 783
Reagents/Equipment
Phosphoenolpyruvate (trisodium salt) Methods to Detect Red Cell Membrane
0.03 M Adenosine diphosphate (ADP) Disorders
0.015M Nicotinamide-adenosine dinucleotide phos-
Method 21. Sugar Water Test
phate (NADH)
Magnesium sulfate Principle
PNH (paroxysmal nocturnal hemoglobinuria) red cells
0.25M Potassium phosphate buffer
hemolyze when incubated with compatible normal serum in a
UV light source sucrose solution of low ionic strength. The principle of this
Whatman No. | filter paper test is that sucrose provides a medium of low ionic strength
0.85% Sodium chloride that promotes binding of complement to red cells.*' PNH cells
undergo lysis in these conditions by virtue of their increased
Procedure sensitivity to complement.
1. Prepare phosphoenolpyruvate (PEP) by dissolving 0.6984 g
of PEP in 8.5 mL of distilled water. Neutralize with 0.25N Specimen
NaOH (pH 7.8) and dilute to 10 mL with distilled water. Clotted blood from patient
784 Chapter 31 Hematology Methods
Reagents/Equipment
Clotted blood from both patient and ABO compatible
donor
0.005 M Phosphate buffer (pH 6.1): Sodium dihydrogen
phosphate (NaH,PO,H,O): Dissolve 690 mg of
monobasic sodium phosphate in | L of distilled
water.
Procedure Specimen
|. Separate the serum from the clotted blood samples of both Defibrin
patient and control.
2. Prepare a 50% saline suspension of cells for each sample Reagents/Equipment
from the residual clots. 0.2 N Hydrochloric acid
3. Add to each of these tubes 0.85 mL of sucrose solution and 0.85% Sodium chloride
0.05 mL of unacidified autologous serum.
0.1-mL and |!.0-mL graduated pipets
4. Add 0.1 mL of the 50% suspension of patient’s red cells to
one tube. 37°C Water bath
5. Add 0.1 mL of the control cells to the second tube. 20 mL of patient’s defibrinated blood. Place two bent
oO. Mix both the tubes and incubate at 37°C for 30 minutes. paper clips in the bottom of a 50-mL Erlenmeyer
7. Centrifuge both tubes at 2000 rpm for | to 2 minutes and flask and swirl the flask continuously for 10 to
examine the supernatant for hemolysis. 15 minutes.
5. Mix all the tubes by shaking well, incubate at 37°C for Volumetric pipet
| hour, and centrifuge at 2000 rpm for 5 minutes. Examine 10 mL
the supernatants for hemolysis.
P y Volumetric flask
Interpretation 100 mL
A positive test is indicated by hemolysis in the tubes contain- Spectrophotometer
ing the patient’s cells with acidified
; serum (tubes
wy 2 and 3), the Heparinized venous blood
other tubes are unaffected (Fig. 31-33). A positive test is usu-
ally diagnostic of PNH. Procedure
1. Make a 1% saline from the stock by diluting 10 mL to
Limitations 100 mL with distilled water.
From 10% to 50% lysis is usually obtained in a positive test, 2. Make serial dilutions of the 1% working solution as
when lysis is measured quantitatively on the basis of liberated depicted below:
hemoglobin. The test can be made more sensitive by using a 1% Working Distilled % Final
young red cell population (reticulocytes) such as the upper saline (mL) water (mL) saline
red cell layer obtained by centrifugation.** 2.0 3.0 0.20
Dee) Tes 0.25
Reporting Results 3.0 7.0 0.30
Hemolysis is reported as a percentage. Qe 6.5 0.35
SD 625 O75
Method 23. Osmotic Fragility 4.0 6.0 0.40
4.25 2p //) 0.425
Principle 4.5 SS 0.45
The test determines the resistance of the red cells to hemoly- 4.75 D9) 0.475
sis In varying concentrations of hypotonic saline at room 5.0 5.0 0.50
temperature. ae) 4.5 05
6.0 4.0 0.60
Specimen 6.5 SD 0.65
Fresh heparinized blood 7.0 3.0 0.70
fie) De) O75
Reagents/Equipment 8.0 2.0 0.80
Stock 10% sodium chloride, buffered (pH 7.4) 8.5 IES 0.85
Sodium chloride 180 g 3. Mix the saline dilutions well and add 0.1 mL of heparinized
whole blood to each tube.
4. Stopper the tubes and mix by inversion. Allow the tubes to
stand for 2 hours at room temperature and then remix the
same way.
5. Centrifuge the suspensions for 5 minutes at 2000 rpm. Cal-
culate the percent hemolysis in each of the supernatants us-
ing a spectrophotometer at 540 nm. Set up a 100% standard
by adding 0.1 mL of blood to 10 mL of distilled water.
6. Calculate % hemolysis as follows:
Interpretation
When red cells are placed in a hypertonic solution, they lose
Figure 31-35 @ Results of a Ham’s test. fluid until an equilibrium is set up with the surrounding fluid,
786. Chapter 31 Hematology Methods
Interpretation
100
Normal blood undergoes little spontaneous hemolysis when
incubated under sterile conditions for 48 hours. The addition
of either glucose or ATP reduces the degree of hemolysis. In
hereditary spherocytosis, the addition of glucose reduces the
degree of hemolysis, while in pyruvate kinase deficiency the
addition of glucose does not affect the degree of hemolysis
(Fig. 31-35).
Ooio)
Limitations
hemolysis
% The autohemolysis test is no longer used in the differential
diagnosis of nonspherocytic congenital hemolytic anemia,
because specific enzymatic assays are now available that pro-
vide better accuracy.
Reporting Results
Hemolysis is reported as a percentage.
140) Ok) CH O:7/ OG O5 Off 08 @2 @al ©
Sodium chloride (g/100 mL)
Normal Values
Figure 351-54 = Comparative osmotic fragility curve (blue line, Lysis at 48 hours: without added dextrose 0.2%
sickle cell anemia; purple line, hereditary spherocytosis). Normal
range is shaded. 7, normal biconcave disc; 2, disc-to-sphere trans- with added dextrose 0-0.9%
formation; 3, disc-to-sphere transformation; 4, lysis. with added ATP 0-0.8%
Chapter 31 Hematology Methods 787
Procedure
1. Collect whole blood anticoagulated with EDTA.
10 Pyruvate
kinase 2. Dilute whole blood with 0.5 mL of 3.8% sodium citrate or
0.85% sodium chloride.
[oS). Mix the blood—diluent solution by inversion.
4. Insert the Westergren tube into the plungeable vial cap of the
diluent mixture. Let blood draw to the top of the tube by cap-
illary action. Place the tube in the Westergren rack in a ver-
(%)
glucose
with
Lysis !
tical position and leave undisturbed for | hour (Fig. 31-36).
I
I Hereditary 5. After | hour has passed, record the distance in mm from the
Normal I spherocytosis top of the column of red cells to the plasma-red cell interface.
range
1
The buffy coat should not be included in the measurement.
i
Interpretation
ieee
15) e010 20 eas
Various diseases can yield an increased ESR. These are listed
Lysis without glucose (%)
in Table 31-6. An increased ESR can also be seen in cases of
Figure 31-55 @ Incubation hemolysis test. This test provides a hyperfibrinogenemia, patients who are able to form rouleaux,
further measure of cell resistance to hemolysis. Pyruvate kinase— and patients with hypergammaglobulinemia. Abnormal red
deficient blood demonstrates an abnormal rate of hemolysis that is cell morphology may be associated with a decreased ESR’
independent of the presence or absence of glucose in the incuba-
(Table 31-7).
tion media. In contrast, the blood from a patient with hereditary
spherocytosis shows more marked hemolysis when glucose is
absent. (From Hillman, RS, and Finch, CA: Red Cell Manual, ed 7.
Limitations
FA Davis, Philadelphia, 1996, p 124, with permission.) Low results can occur when the collected specimen is
allowed to stand for more than 3 hours. The Westergren
Reagents/Equipment
Westergren tubes Figure 31-56 ® Westergren methodology for ESR. (Courtesy of
Westergren rack Polymedco, Inc, Cortlandt Manor, NY.)
788 — Chapter 31 Hematology Methods
Reagents
tube must be situated perfectly vertical as any tilt can cause ESR-vacuum tube, 2.0 mL, containing 3.2% sodium
invalid results. citrate
Case Study
A 70-year-old woman diagnosed with multiple myeloma is
currently on melphalan and prednisone. Her CBC revealed
the following:
reer ome
1. Twenty microliters of blood are drawn into a Unopette 4. Which of the following factors affect the ESR?
system for a WBC count. Fifty cells are counted on one a. Increased fibrinogen
side and 52 on the other side of the hemacytometer b. Extreme poikilocytosis
(4 large squares). Calculate the WBC count. c. Use of heparin as an anticoagulant
dee. VOTE d. All of the above
Bel 2ae OL, 5. What type of hemoglobin electrophoresis would be best
Cole OC OWE to separate hemoglobins S and D?
dO L077 a. Cellulose acetate at alkaline pH
N . If a patient has a reticulocyte count of 8% with a hemat- b. Citrate agar at acid pH
ocrit of 18%, what is the corrected reticulocyte count? c. Either cellulose acetate or citrate agar may be used
a. 20% 6. What is the principle for the acid elution test for HbF?
be2 370 a. HbF is resistant to acid elution; it can be precipitated
Cas070 and stained.
d. 8% b. HbF is susceptible to acid elution; it can be dissolved
3. Which of the following errors will cause falsely elevated and measured photometrically.
results for a centrifugal microhematocrit? c. HbF is resistant to acid elution; it can be separated by
a. Reading of buffy coat with red cells aspirating the acid from the remaining hemoglobin.
b. Incomplete sealing of capillary tubes d. HbF is susceptible to acid elution; it can be destroyed
c. Allowing the centrifuge to