Sie sind auf Seite 1von 39

C HAPTER 60 

MYELODYSPLASTIC SYNDROMES
Christopher J. Gibson, Benjamin L. Ebert, and David P. Steensma

Myelodysplastic syndromes (MDS) are clonal hematopoietic disor- The third major component of MDS, and possibly the most
ders characterized by ineffective hematopoiesis and peripheral blood prominent, is its temporal positioning as a potential preleukemic
cytopenias, abnormal cell morphology on blood and bone marrow state. This was first conceptualized in 1942 as odo-leukemia (“odo”
examination (Fig. 60.1) and a risk of progression to acute myeloid from the Greek for edge or threshold) by a group from France led by
leukemia (AML). It has a variable clinical course, manifesting in some Paul Chevallier to describe a case of anemia evolving into leukemia,6
patients as indolent cytopenias necessitating occasional transfusions which they attributed to benzene exposure. A similar disorder was
and in others as aggressive diseases that rapidly evolve into treatment- termed preleukaemic anemia by a British hematologist, J.L. Hamilton-
refractory leukemias, with a broad spectrum of severity in between. Paterson, in 1949.7 This term was adopted and expanded by an
Despite this phenotypic variability, individual cases of MDS share American group based in Chicago in the early 1950s,8 providing one
many pathophysiologic mechanisms at the molecular and cellular of the clearest descriptions of a disorder clinically defined by both
levels, and our understanding of these mechanisms has evolved cytopenias and a risk of leukemia. Other groups in the 1950s and
substantially in the last decade. At the same time, although MDS also 1960s published similar descriptions under different terminologies.9
shares features with AML and other myeloid malignancies, mounting The 1970s saw some of the greatest strides toward our modern
evidence shows that it is appropriately conceived as a distinct class of conceptualization of MDS. In 1970, Dreyfus and colleagues coined
diseases, and that the older conceptualization of MDS as “preleuke- the term refractory anemia with excess blasts (RAEB, described in
mia” is overly simplistic. French as les anémies réfractaires avec excès de myeloblasts)10 and later
While understanding of MDS pathophysiology is improving, the attempted to further characterize components of RAEB based on
condition remains difficult to treat, and outcomes for patients with morphology and clinical characteristics.11 The term hematopoietic
MDS have unfortunately not improved substantially since the last dysplasia was used in the early 1970s to describe a heterogeneous
edition of this textbook. This chapter describes our current under- group of disorders distinct from AML, and the term was later simpli-
standing of the classification, pathobiology, clinical features, and fied to myelodysplasia.
treatment approaches for this heterogeneous group of disorders. One of the key events in the history of myeloid disease classifica-
tion was the formation of the French-American-British (FAB)
Cooperative Group in 1976, which in that year issued a comprehen-
HISTORY sive and influential categorization of AML12 based on the 1975
classification of Galton and Dacie.13 The FAB classification persisted
Central to an understanding of how MDS is diagnosed and classified as the dominant descriptive system for AML until the early 2000s.
is the concept of morphologic dysplasia, a pathologic term that was The original FAB AML formulation from 1976 included two types
originally introduced as a shortening of the phrase dysmorphology of of “dysmyelopoietic syndromes” that should not be confused with
neoplasia. Although specific criteria for diagnosis of MDS as a distinct AML: RAEB and chronic myelomonocytic leukemia (CMML),
clinical syndrome exist and are discussed subsequently, the term which had first been described in the late 1960s.14 However, it was
myelodysplasia refers more generally to an abnormal appearance of not long before MDS was recognized as a separate group of diseases
hematopoietic precursors during pathologic examination of bone and accorded a distinct classification system, described in more detail
marrow, which may result from many different causes (e.g., drug later.
toxicity, nutritional deficiency, viral infection). The process by which
our understanding of MDS has evolved from that of a morphologic
oddity to that of a distinct disease process has been a century in the CLASSIFICATION
making, propelled forward at several points by paradigmatic shifts in
pathologic and molecular characterization of hematologic diseases.1 The currently accepted classification scheme for MDS was initially
In describing the history of MDS as a distinctly categorized disease published by the World Health Organization (WHO) in 2001 and
entity, it is perhaps most useful to separately describe the history of most recently revised in 2008 (Table 60.1), with a third revision
its major conceptual components. The earliest recognized of these, not planned for 2016. Like the classifications of the FAB group, the
surprisingly, was that of dysplasia itself. Although studies of peripheral WHO classification is principally a morphologic system and classifies
blood in anemic patients were reported in the 19th century,2 the first disease chiefly based on the number of dysplastic lineages and the
real characterization of actual dysplasia was probably made by Giovanni percentage of marrow blasts, though it also includes one specific
di Guglielmo in Pavia in 1923, when he described abnormal erythroid cytogenetically defined subtype, MDS with isolated del(5q). The
forms in the marrows of patients with various types of cytopenias.3 A other WHO 2008 subtypes include refractory cytopenia with unilin-
second major concept, that of ineffective hematopoiesis, was developed eage dysplasia (RCUD), which is most commonly refractory anemia
in the 1930s with the description of refractory anemia in patients (RA); refractory cytopenia with multilineage dysplasia (RCMD);
unresponsive to iron pills or liver extract (the precursor to vitamin B12 refractory anemia with ringed sideroblasts (RARS); RAEB (subdi-
supplementation);4 in the 1950s, others expanded this to include vided into RAEB-1, 5%–9% blasts, and RAEB-2, 10%–19% blasts);
similarly refractory leukopenia and thrombocytopenia and termed the and unclassifiable MDS (MDS-U) for those cases that do not clearly
collective disorder “refractory cytopenias.”5 Importantly, however, fall into any other category. In addition, the WHO system classifies
ineffective hematopoiesis and marrow dysplasia were initially incom- cases with myeloproliferative features separately; this group includes
pletely linked, and the ineffective hematopoiesis of many early refrac- CMML and RARS with thrombocytosis (RARS-T).
tory anemia patients was probably rooted in other disorders, such as The WHO classification was intended to replace the prior FAB
anemia of inflammation caused by advanced rheumatologic disease or classification system, which was initially proposed in 1982 as an
nonmyeloid neoplasia. MDS correlate to the FAB system for classifying AML.15 The 1982

944
Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 945

A B C
Fig. 60.1  ELEMENTS OF MYELODYSPLASTIC SYNDROME. Myelodysplastic syndromes are generally
characterized by cytopenias (A) caused by ineffective hematopoiesis (B), which is related to multilineage
dysplasia (C). (A) This patient presented with a white blood cell count of 1500/µL, hemoglobin 8.9 g/dL,
and platelet count 47,000/µL. (B) The bone marrow was hypercellular, indicating ineffective hematopoiesis.
(C) Evidence of trilineage dysplasia was apparent on the peripheral smear. Anisocytosis with macroovalocytes
and poikilocytosis is seen in the red blood cells (C, top). The latter included the somewhat uncommon finding
of Cabot ring forms (right). A large proportion of the granulocytes were severely hypogranular (C, middle left)
compared to some normal forms still in the circulation (right). Platelets (C, bottom) were decreased in number,
and many were severely hypogranular (middle, barely visible) compared with residual normal platelets (left).

FAB system distinguished five subtypes of MDS, namely (1) RA, Future iterations of a classification system may incorporate elements
(2) RARS, (3) RAEB (defined as 5%–19% blasts in the marrow), of genetic or clinical data that better group MDS patients based on
(4) RAEB in transformation (defined as 20%–29% blasts in the disease biology, prognosis and natural history, or anticipated respon-
marrow), and CMML. siveness to treatment modalities.22 For now, however, this classifica-
The WHO system thus bears some similarities to the FAB scheme, tion scheme remains only one of several descriptive tools applied to
but there are several notable changes. First, the WHO system recog- patients with MDSs.
nizes that MDS may present with isolated leukopenia or thrombo-
cytopenia, albeit uncommonly, hence the change from RA to the
broader RCUD and the addition of a second distinct class for patients EPIDEMIOLOGY AND ETIOLOGY
with more than one affected cell line. Second, a mounting body of
evidence suggested that MDS with proliferative features is clinically Most available data suggest that MDS is one of the most common
and biologically distinct from MDS without such features and thus hematologic malignancies, although this claim has been difficult to
warrants separate designation.16,17 Third, the unique clinical syn- validate until recently because of confusing terminology and incom-
drome associated with isolated del(5q), in combination with its plete reporting of cases to registries.23,24 Cases of MDS were not
particular responsiveness to treatment with lenalidomide, warranted reported to the National Cancer Institute’s Surveillance, Epidemiol-
its designation as a specific entity.18 Finally, the cutoff for diagnosis ogy, and End Results (SEER) until 2001, and initial reports to the
of AML was changed from 30% to 20% bone marrow blasts or registry suggested an incidence of only about 10,000 new cases per
greater, rendering the designation of RAEB in transformation obso- year.25 Subsequent comparison of these data with Medicare and
lete, although this change was not without its detractors.19 insurance claims for MDS suggested that a substantial proportion of
The WHO revision improved on certain aspects of the FAB MDS cases went unreported to SEER, with an estimated age-
system. In particular, the FAB system required that patients display adjusted incidence of >5.3 per 100,000, compared to the SEER
at least 10% dysplasia in two different cell lineages to achieve even a estimate of 3.3.26
diagnosis of RA; this excluded patients with mild or unilineage The reporting difference was especially stark in patients age 65
dysplasia, many of whom had natural histories indistinguishable from and older, where it was estimated that the actual incidence of MDS
those who met the diagnostic criteria. While the WHO guidelines might actually be close to fourfold higher than what was captured in
eliminated the dual-lineage requirement and allows a diagnosis of the database (75 versus 20 per 100,000). Some of the underreporting
MDS in patients with unilineage dysplasia, it nevertheless draws was likely related to specific criteria for entry into the database (for
arbitrary lines of distinction between subclasses of a heterogeneous instance, myeloid malignancies could only be counted once, such that
disease, and patients with minimal dysplasia are still typically excluded patients presenting with a new diagnosis of secondary AML were
from the diagnosis.20 Artificial boundaries such as the 10% dysplasia, usually coded as AML, without mention of MDS). Much of the
15% ring sideroblasts and 5%/10%/20% blast cutoffs often prove underreporting, however, was likely because of the complexity of
themselves to be of variable relevance, both biologically and clinically, diagnosing and classifying MDS, as described earlier. The rate of
because they fail to capture a number of variables important in MDS, reporting appears to have recently improved, with annual incidence
including age, sex, and most cytogenetic and genetic data. in the 2007–2011 SEER database estimated at around 20,500 and
Although some of the FAB and WHO subgroups imply very an age-adjusted incidence of 4.9 per 100,000.27
rough prognostic information—for instance, patients with FAB Even the most accurate database, however, would probably under-
RAEB or WHO RAEB-2 are at higher risk of progression to AML estimate the total global burden of MDS, which is likely present in
than subgroups without excess blasts21—other systems specifically a substantial percentage of older patients with idiopathic cytopenias
dedicated to estimating disease risk, such as the International Prog- who never undergo bone marrow analysis.28 Although some propor-
nostic Scoring System (IPSS; see later), are better suited to this task. tion of this uncaptured population likely has biologically indolent

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
946 Part VII  Hematologic Malignancies

TABLE 2008 World Health Organization Classification of the TABLE Genes Recurrently Mutated in Myelodysplastic
60.1 Adult Myelodysplastic Syndromes 60.2 Syndrome
Refractory Cytopenia With Unilineage Dysplasia Gene Frequency (%) Notes
Dysplasia ≥10% of cells from a single lineage
Splicing Factors
Blasts <5% in marrow; <1% in peripheral blood; no Auer rods SF3B1 20–30 Strong association with RARS
Notes Includes refractory anemia (RA), refractory neutropenia, SRSF2 10–15 (MDS) Enriched in CMML
refractory thrombocytopenia; RA is by far the most 40 (CMML)
common subtype
U2AF1 5–12 Association with del(20q)
Refractory Anemia With Ring Sideroblasts
Epigenetic Modifiers
Dysplasia Isolated erythroid dysplasia
TET2 20–30 (MDS) Enriched in CMML
Blasts <5% in marrow; <1% in peripheral blood; no Auer rods 40–50 (CMML) Mutually exclusive with IDH
Notes ≥15% of erythroid precursors are ring sideroblasts DNMT3A 8–13
Frequently associated with SF3B1 mutations ASXL1 10–20 (MDS) Enriched in CMML
MDS With Isolated del(5q) 30–40 (CMML)
Dysplasia Normal or increased megakaryocytes with hypolobated EZH2 5–10 (MDS) Enriched in CMML
nuclei 20–30 (CMML) May be functionally involved
Blasts <20% (though usually much less) in 7q−
Notes del(5q31) must be sole chromosomal abnormality IDH1/2 <5 More frequent in AML
Refractory Cytopenia With Multilineage Dysplasia ATRX Rare Associated with acquired
Dysplasia ≥10% of cells from two or more myeloid lineages thalassemia
Blasts <5% in marrow; <1% in peripheral blood; no Auer rods Transcription Factors
Notes Peripheral monocyte count must be <1 × 109/L; ring RUNX1 10–15 Can be somatic or germline
sideroblasts may be present GATA2 Rare Mostly germline
Refractory Anemia With Excess Blasts ETV6 <5 Can be somatic or germline
Dysplasia No specific requirement
TP53 10–12 Association with complex
Blasts RAEB-1: 5%–9% in marrow, <5% in peripheral blood, AND karyotype, therapy-related
no Auer rods disease
RAEB-2: 10%–19% in marrow, 5%–19% in peripheral Kinases and Receptors
blood, OR Auer rods JAK2 <5 Enriched in RARS-T
Notes Old designation of RAEB-t (20%–30% blasts) now NRAS 5–10 Seen in progression to AML
considered AML CBL <5 Enriched in JMML
Unclassifiable MDS
PTPN11 <5 More common in JMML
Dysplasia Minimal, or not meeting criteria for another subtype
BRAF Rare Also seen in hairy cell
Blasts <5% in marrow; <1% in peripheral blood; no Auer rods
leukemia
Notes In presence of clonal cytogenetic finding considered Cohesin Complex
diagnostic of MDS STAG2 5–10 Cohesin class mutations
Note: excludes refractory cytopenias of childhood. MDS/myeloproliferative RAD21 <5 enriched in high-risk MDS
neoplasms such as chronic myelomonocytic leukemia and RARS with SMC3 <2 and secondary AML.
thrombocytosis are classified separately.
AML, Acute myeloid leukemia; MDS, myelodysplastic syndrome; RAEB, SMC1A <2
refractory anemia with excess blast. GCPR Complex
GNAS Rare Mutations recently described
GNB1 Rare in wide range of
hematologic malignancies,
including MDS.
disease that would never require therapy, more thorough cross-
sectional studies, particularly in older patients, would help clarify the AML, Acute myeloid leukemia; CMML, chronic myelomonocytic leukemia;
GCPR, G-coupled protein receptor; IDH, isocitrate dehdryogenase; JMML,
distinction between this end of the MDS spectrum and more aggres- juvenile myelomonocytic leukemia; MDS, myelodysplastic syndrome; RARS,
sive biology that brings patients to clinical attention. refractory anemia with ring sideroblasts; RARS-T, RARS with thrombocytosis.
As referenced earlier, MDS is by and large a disease of older adults
and reflects the inexorable acquisition of genetic mutations by aging
hematopoietic progenitor cells (Table 60.2). In the United States, the
median age at diagnosis is approximately 71 years,29 and in the
absence of a congenital disorder or exposure to radiation or cytotoxic anemia,36 dyskeratosis congenita and other telomeropathies, and
chemotherapy for another disease, diagnosis before the age of 50 is germline mutations in GATA2,37 RUNX1,38 and ETV6.39 In Fanconi
rare.30 Recent data suggest that a substantial proportion of older anemia and the telomeropathies,40 MDS during young adulthood
adults, perhaps as many as 10% of those over age 70, harbor hema- may be the initial presenting sign of the disease.
topoietic clones defined by the presence of mutations recurrently Most subtypes of MDS appear to be more common in men than
found in MDS and AML, and that this state of “clonal hematopoiesis women, with the most recent SEER data suggesting respective age-
of indeterminate potential” progresses to MDS at a rate of 0.5% to adjusted incidences of 6.7 versus 3.9 per 100,000.41 The one excep-
1% per year.31,32 MDS occurs in children much more rarely, at an tion to this is MDS with isolated del(5q), which most series show to
estimated annual rate of 1 per 1 million.33 Most cases are classified be more common in women.42 The reason for these sex differences
as oligoblastic myelogenous leukemia (essentially RAEB), and other is unclear; some have postulated a protective factor encoded on the
subtypes are even less common.34 Several genetic syndromes confer X chromosome, or a similar protective factor on the Y chromosome
an increased risk of MDS; these include Down syndrome,35 Fanconi (which is clonally lost in some cases of MDS). Others have suggested

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 947

that the increased incidence in men overall is likely caused by differ- or more hematopoietic compartments; this may or may not coincide
ences in occupational exposures, but this has never been clearly with genetic lesions that lead to ineffective hematopoiesis and cyto-
demonstrated. Indeed, the only toxic chemical exposure definitively penias, at which point MDS also becomes clinically evident.56
proven to cause MDS is benzene,43 which is now significantly less Proving that MDS is a clonal disease was not as straightforward
prevalent in industrial workplaces than it had been in the past. Other a proposition as it was for AML, in which examination of the bone
environmental exposures, including cigarette smoking, have been marrow typically identifies sheets of abnormal, morphologically
postulated but never definitively proven to predispose to MDS.44 identical blasts. In contrast, for MDS—particularly low-risk disease
MDS in the United States and Western Europe is epidemiologi- in which blasts are rare and the marrow architecture is disorganized
cally similar, but there are differences between these geographic and heterogeneous—a clonal origin was not immediately obvious
regions and other parts of the world. In Asia45 and Eastern Europe,46 based on morphology alone. Nonetheless, the clonal nature of MDS
for instance, the average age at MDS diagnosis is younger, and the was established in the 1980s by studies that showed skewed inactiva-
frequency of both severe cytopenias and risk of progression to leuke- tion of glucose-6-phosphate dehydrogenase (G6PD), an X-linked
mia may differ.47 Some of these differences are likely caused by gene, in the hematopoietic cells of female MDS patients heterozygous
variance in environmental exposures. In Japan, for example, broad for G6PD deficiency.57 More recent studies have used deep sequenc-
population-wide exposure to ionizing radiation from the atomic ing techniques to track the clonal evolution from MDS into AML
bombings of Hiroshima and Nagasaki in the 1940s continued to and have confirmed that in these cases, the preexisting MDS is as
influence MDS incidence well into the 1990s.48 However, the cause highly clonal as the resulting secondary AML.58
of differences in MDS subtypes, such as the low incidence of RARS That MDS is a disorder of stem or early progenitor cells has been
in Japan compared with the West, remain unclear. more difficult to prove.59 Some of this difficulty is a reflection of the
Indeed, the two exposures most consistently associated with elusiveness of human HSCs themselves: the ability of different human
subsequent development of MDS are ionizing radiation and cytotoxic cell populations to self-propagate after xenotransplantation into
chemotherapy, and MDS arising in these settings, known as therapy- immunodeficient mice, considered the functional hallmark of “stem-
related MDS or t-MDS, is frequently characterized by TP53 muta- ness,” varies with the exact degree of immunodeficiency of the mice
tions,49 multiple large-scale chromosomal abnormalities including into which the cells are transplanted.60 Moreover, xenotransplant
complex karyotypes (most commonly defined as ≥3 clonal chromo- experiments using immunophenotypically defined hematopoietic
somal anomalies),50 and frequent transformation to treatment- stem and progenitor cells (HSPCs; classically CD34+ CD38− Lin−)
refractory AML.51 In the United States, radiation is most frequently from MDS patients have not shown a striking proliferative or self-
encountered as treatment for other cancers, and radiation fields that renewal advantage for the MDS cells compared to normal controls,61
include the hips or pelvis, the most active sites of hematopoiesis in and the degree to which these experiments accurately depict the
adults, probably pose the greatest risk.52 Less commonly, exposure to clonal dynamics of MDS in humans is unclear. Some have further
radiation can occur as the result of occupational exposures (e.g., been troubled by the observation that lymphoid clonal expansion,
workers at nuclear reactors) or industrial accidents. Among chemo- which should occur with near-equal frequency to myeloid clonal
therapeutic agents, there is a substantial difference in the risk of expansion if MDS is indeed a disorder of very early hematopoietic
subsequent MDS. In particular, alkylating agents (e.g., cyclophospha- progenitors, is only rarely observed.62,63 However, more recent studies
mide and melphalan)53 appear to carry the greatest risk, while the risk combining immunophenotypic analysis with deep sequencing of
with nucleoside analogues is lower. Of particular note, the rapidly clonal mutations in MDS cells have shown that the mutations appear
progressive AML seen in association with topoisomerase inhibitors to originate exclusively in the most primitive, stem-cell-like compart-
(e.g., doxorubicin, etoposide) is not typically preceded by MDS.54 ment,64 and others have shown that differential expansion of specific
progenitor compartments may vary between different phenotypes
and risk profiles of MDS.65 This evidence has contributed to the
PATHOBIOLOGY conclusion that MDS is, in fact, a disorder of transformed HSCs.66
The conceptualization of MDS as a stem cell disorder explains,
The past 10 years have witnessed significant advances in our under- in large part, why it is so refractory to most attempts at conventional
standing of the rich and complex pathobiology underlying MDSs. therapy. Both normal HSCs and leukemia stem cells remain quiescent
MDS is now recognized to arise from interactions between acquired for much of their lifetimes, and during these periods they are largely
genetic mutations in hematopoietic precursor cells, alterations in the impervious to any agents, such as most chemotherapeutic drugs, that
microenvironment of the bone marrow, and dysregulated immune exert their effects during active DNA replication.67 Overcoming the
surveillance. Broadly speaking, the acquisition of sequential muta- intrinsic resistance to therapy conferred by the existence of quiescent
tions in precursor cells drives the development of a malignant clone, reservoirs of disease is one of the central challenges in developing
and alterations in the microenvironment and the immune response effective treatments for MDS.68
allow that clone’s expansion. This section details our current under-
standing of these concepts.
Genetic Alterations
Myelodysplastic Syndrome Stem Cells Like other cancers, the core hypothesis underlying MDS pathogenesis
is that the originating clone of MDS becomes increasingly abnormal,
One of the central challenges in understanding the pathogenesis of and ultimately malignant, through the sequential accumulation of
MDS has been isolating the cell of origin and understanding that acquired genetic or epigenetic abnormalities.69 Improvement in our
cell’s mechanisms of self-renewal and propagation, both of which are understanding of how these abnormalities are acquired, how they
necessary for the establishment of a malignant clone.55 In theory, the interact with each other, and their impact on pathways affecting
exact state of hematopoietic differentiation from which a malignant proliferation, self-renewal, and differentiation, has been one of the
clone arises could vary between cases of MDS, but the capacity for major advancements in the study of MDS over the last decade.
self-renewal implies that the origin cell was either a hematopoietic Several different classes of genetic abnormalities may be found in
stem cell (HSC), and thus possessed intrinsic self-renewal capabilities, MDS. The first to be recognized were cytogenetic abnormalities on
or it was a more differentiated myeloid progenitor that acquired the standard karyotyping,70 which are present in about 50% of patients.71
ability to self-renew. Clonal expansion then occurs through the A second category of cryptic chromosomal aberrations, including
acquisition of new mutations or epigenetic alterations that either microdeletions and copy number-neutral loss of heterozygosity, are
enhance proliferation or confer resistance to apoptosis. MDS presum- too small to be detected by karyotype but may be found with fluo-
ably becomes morphologically apparent when the dominant clone rescence in-situ hybridization (FISH)72 or single nucleotide polymor-
acquires a subsequent genetic lesion that leads to dysplasia within one phism (SNP) arrays.73 The most common type of abnormality,

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
948 Part VII  Hematologic Malignancies

mutations in single genes, has been increasingly well characterized,


and clinical tests for recurrent mutations are becoming increasingly Splicing Factor Mutations
available, though as discussed later, it appears that not all mutations
have equal clinical relevance.74 Finally, epigenomic alterations—global Genes encoding splicing factors, which excise introns to create mature
aberrations in histone and chromatin modification—are common in messenger RNA (mRNA) transcripts, are the most commonly
MDS, and are often, though not always, associated with mutations mutated class of genes in MDS, with between half and two-thirds of
in genes involved in epigenetic regulation.75 patients harboring such a mutation.83 As opposed to most of the other
classes of mutations discussed here, which occur in other malignan-
cies with significant frequency, splicing factor mutations are rarer in
Somatic Mutations other cancers than MDS, and some have unique associations with
specific morphologic phenotypes.84 Splicing factor mutations tend to
Of the types of genetic abnormalities found in MDS, acquired muta- be mutually exclusive within MDS clones, and most of the affected
tions in individual genes are the most recently recognized; they are genes encode proteins that comprise the E/A splice site recognition
also the most frequent, currently estimated to be present in around complex that acts at the 3′ end of pre-mRNA, suggesting that the
80% of MDS patients.69 Although certain environmental exposures mutations converge on a shared biologic pathway.85 Exactly how
increase the risk of acquiring potentially deleterious mutations, most splicing factor mutations predispose to the development of MDS,
are acquired randomly, either spontaneously (e.g., deamination of and whether they can help predict aspects of natural history or
cytosine to uracil), during DNA replication before cell division, or treatment response, remain areas of active investigation.
during DNA repair.
One of the central challenges of understanding the genetics of
MDS has been determining which mutations contribute to the SF3B1
pathogenesis of the disease and which do not. Accumulating evidence
has shown that HSCs acquire nonsynonymous exonic mutations with SF3B1 encodes the U2 small nuclear riboprotein complex (snRNP)
translational consequences at a rate of about one mutation per responsible for 3′ branch site recognition and is the most frequently
decade.76 Since the incidence of MDS increases with age, HSPCs mutated splicing factor gene. SF3B1 mutations can be found in
from an average MDS patient should typically contain between 5–10 about 20% to 30% of all MDS patients; there is a particularly
such mutations (though the actual number varies widely between strong association with ring sideroblast morphology, with mutations
patients), against a background of hundreds of noncoding single found in 60% to 85% of patients with RARS or RARS-T and
nucleotide variations. The vast majority of these mutations, both substantial percentages of other MDS subtypes in which ring
coding and noncoding, are of no pathogenic consequence and are sideroblasts can be found.86,87 The most common mutations are
thus termed passenger mutations, while a minority, the so-called driver missense substitutions that change lysine to glutamate at codon
mutations, actually contribute to the development of the disease.77 700, with other smaller hotspots in the same vicinity. These muta-
Distinguishing driver mutations from passenger mutations is not tions all occur within a cluster of 26 nonrepeating HEAT domains
always trivial. Passenger mutations, without conferring clonal advan- that are thought to be involved in binding of SF3B1 to other
tage, tend not to recur with any significant frequency in cohorts of members of the U2 snRNP.88 As with other splicing factor muta-
MDS patients; however, whereas a few driver mutations are relatively tions, they tend to be heterozygous and imply a gain of function.
common in MDS, many have been found in only a small fraction How exactly the mutations affect MDS pathogenesis is not com-
(<5%) of cases, suggesting novel mechanisms of disease pathogenesis.78 pletely clear, but they are often acquired early in disease develop-
Similarly, whereas noncoding mutations are unlikely to contribute to ment, tend not to be associated with complex karyotypes, and tend
pathogenesis and can thus be ignored when analyzing MDS genomes, not to be associated with poor-prognosis mutations.89 SF3B1 muta-
the majority of coding mutations, even those present within a major- tions are clinically associated with a distinct phenotype of isolated,
ity of cells in the malignant clone, are also nonpathogenic and are transfusion-dependent anemia with preserved white blood cell and
simply artifacts captured by an aging HSPC. Classifying a mutation platelet counts relative to SF3B1-wildtypes, as well as a lower risk
as a driver thus involves accumulating evidence that the mutation of progression to AML.90 Indeed, in larger studies of MDS cohorts,
occurs recurrently in MDS, that its putative function could plausibly SF3B1 mutations appear to confer an improvement in relative
contribute to MDS pathogenesis, and, in the best-case scenario, that survival, making them somewhat unique among MDS-associated
this function can be recapitulated using in vitro or in vivo models. mutations.80
Large-scale genomic studies of MDS cohorts have shown that many
of the recurrently mutated genes can be segregated into one of a few
functional categories,78–81 and this functional characterization has in SRSF2
turned revealed insights about how an MDS clone develops over time.
Genes affecting RNA splicing (SRSF2, SF3B1, U2AF1) are the most SRSF2 encodes a member of the serine/arginine (SR)-rich family of
commonly mutated and tend to be early events in MDS pathogenesis. pre-mRNA splicing factors that interacts with the U2 and U1 com-
Genes affecting epigenetic regulation (TET2, ASXL1, EZH2, ponents of the spliceosome. After SF3B1, it is the second-most
DNMT3A) are the next-most commonly mutated and also tend to commonly mutated splicing factor, with mutations present in 10%
occur early. On the other hand, mutations in genes for growth factors to 15% of MDS91 and 40% of CMML.92 The overwhelming majority
(NRAS, JAK2) tend to occur late and in subclonal populations. of mutations are heterozygous missense substitutions for proline at
These observations have contributed to an overarching model of codon 95, implying a gain of function. A small minority of in-frame
how sequential somatic mutations cooperate to bring about MDS. insertions or deletions affects the same region. SRSF2 mutations
In this model, early mutations in splicing factors and epigenetic genes co-occur with several other mutations, many of which are also fre-
do little to affect proliferation or differentiation on their own, but quently found in CMML, including TET2, ASXL1, CUX1, IDH2,
rather create a permissive environment for the acquisition of subse- and STAG2. In contradistinction to patients with SF3B1 mutations,
quent mutations in genes that confer proliferative advantages or patients with SRSF2 mutations tend to have more dysplasia in the
blocks in differentiation. The latter group of mutations contributes granulocytic lineage and less in the erythroid lineage, and patients
more directly to the clinical features of MDS, but the former is consequently tend to have a less prominent transfusion requirement,
responsible for its tendency towards genomic and epigenomic insta- more enrichment in the RAEB subtypes, and, at least in some studies,
bility, which in turn may contribute to the disease’s poor response to a greater risk of progression to AML.93 This phenotype is more het-
treatment. This section is a brief summary of our current understand- erogeneous than that associated with SF3B1, but most studies have
ing of some of the most important genes and pathways that are nevertheless shown that SRSF2 mutations appear to confer an inferior
recurrently deranged in MDS.82 prognosis.91

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 949

many patients are either compound heterozygotes or have uniparental


U2AF1 disomy (UPD) at chromosome 4q, leading to effective abrogation of
TET2 function.101 Indeed, patients with TET2 mutations have been
U2AF1 encodes an auxiliary factor in the U2 spliceosome that is shown to have globally altered methylation profiles.102 TET2 muta-
responsible for recognizing the AG splice acceptor dinucleotide at the tions often occur early in MDS pathogenesis and are thought to alter
3′ end of introns. U2AF1 mutations occur in about 12% of patients HSC homeostasis, a theory supported by the fact that HSCs in TET2
with MDS.94 Similar to other commonly mutated splicing factor knockout mice display enhanced self-renewal and repopulation
genes, the two most common mutations are both heterozygous mis- capacity. In humans, however, TET2 mutations do not appear to
sense substitutions, again implying a gain of function, and both themselves confer a specific phenotype;79 rather, they may create an
appear to alter sequence specificity of pre-mRNA binding and splic- epigenomic environment permissive to the acquisition of other muta-
ing.95 The two mutations are in separate zinc finger DNA binding tions that are responsible for determining these factors. Recent evi-
domains, one at codon 34 toward the N-terminal of the protein, and dence suggests that TET2 mutations appear to predict a favorable
one at codon 157 toward the C terminal. Some studies have shown response to hypomethylating agents, particularly in the setting of
an increased tendency of MDS with U2AF1 mutations to evolve into wildtype ASXL1.103 TET2 mutations were not previously felt to
secondary AML,96 but given the small numbers of patients in these confer prognostic information, but newer data suggest that, at least
studies, the strength of this effect is unclear. in patients undergoing stem cell transplant, they predict an inferior
outcome.104

Epigenetic Modifier Mutations


DNMT3A
Epigenetic changes are biochemical modifications that affect chroma-
tin structure, and thereby gene expression, without actually altering DNMT3A is a member of the family of DNMTs, which catalyze the
the DNA sequence itself. The two types of epigenetic changes most addition of methyl groups to cytosine residues of CpG dinucleotides.
relevant to MDS are DNA methylation and histone modification.75 These dinucleotides tend to cluster in 5′ promoter regions upstream
DNA methylation involves the addition of a methyl group to the of genes, and increased methylation of these CpG islands is associated
cytosine residues of cytosine-guanine pairs by members of the DNA with decreased expression of the associated downstream gene.105 The
methyltransferase (DNMT) family of enzymes. These cytosine- observation that many cancers often display aberrant methylation
guanine pairs frequently cluster together in “CpG islands,” which are relative to healthy tissue has led to the hypothesis that hypermethyl-
typically located just upstream of promoter regions. CpG islands tend ation, particularly in the promoters of tumor suppressor genes, plays
to be unmethylated at baseline, but their progressive methylation a role in cancer pathogenesis. This hypothesis has been somewhat
leads to transcriptional silencing of the downstream genes. Studies supported by the efficacy, albeit imperfect, of so-called hypomethylat-
have shown that many MDS patients display aberrant methylation ing agents like decitabine and 5-azacitidine in MDS and AML (see
patterns compared to healthy controls, with isolated hypermethyl- section on Therapy, later).
ation in the promoters of critical tumor suppressors despite global The DNMT3A gene consists of 29 exons and encodes a 908-
hypomethylation elsewhere. This phenomenon has been hypothesized amino acid protein that, along with DNMT3B, is one of the two
to play a role in the pathogenesis of MDS, but attempts to glean enzymes responsible for de novo CpG methylation independent of
prognostic or predictive information from specific methylation pat- replication, whereas a third methyltransferase, DNMT1, is respon-
terns in MDS patients have been largely unsuccessful, and the success sible for maintenance of baseline hemimethylation during active
of so-called hypomethylating agents such as 5-azacitidine and replication. Only DNMT3A mutations, however, have been found to
decitabine (which may in fact not truly act by reducing global occur recurrently in myeloid malignancies, perhaps suggesting dif-
methylation) has been variable. ferential expression in hematopoietic cells. Analysis of DNMT3A
A second type of epigenetic regulation involves the biochemical mutations in patients with MDS has shown a preponderance of
modification of histones, the structural protein complexes that form missense single nucleotide variations predicted to alter protein func-
scaffolding for chromatin packaging. The interaction between his- tion, although nonsense mutations, insertions, and deletions (indels)
tones and chromatin represents an additional level of transcriptional have been observed as well. The mutations occur throughout the
control, in which unwinding of chromatin is required for the tran- gene, although a mutational hotspot at R882H, in the methyltrans-
scription machinery to physically access DNA. The dynamics of ferase domain, has been described in a minority of MDS patients.106
chromatin-histone interactions are largely mediated by complex In AML, the R882H mutant protein has been shown to inhibit
biochemical modifications of specific histone amino acids. In MDS wildtype DNTM3A, suggesting a dominant negative mechanism.107
and other myeloid disorders, these modifications can either be In earlier studies, DNMT3A-null HSCs transplanted into mice
affected directly by mutations in genes coding for histone-modifying displayed aberrant global methylation patterns, increased self-renewal,
enzymes or indirectly by the permutation of biochemical pathways and impaired differentiation capacity compared to wildtype HSCs,
that regulate the balance between open and closed chromatin. but the mice themselves did not develop dysplasia or other hemato-
logic malignancies.108 In more recent studies, however, mice trans-
planted with DNMT3A-null HSCs had shortened overall survival
TET2 and developed a spectrum of hematopoietic malignancies similar to
that seen in humans, including leukemia and MDS.109 The different
TET2, which encodes a member of the Ten-Eleven Translocation outcomes in these two studies is thought to be caused in large part
gene family, is the most commonly mutated epigenetic regulator in by the fact that mice in the older experiments were serially trans-
MDS.97 TET2 mutations occur in 20% to 30% of all MDS and are planted at 18 weeks, before they had a chance to develop overt
particularly enriched in CMML, where they can be found in 40% hematologic disease, whereas mice in the later experiments were
to 50% of cases.98 The TET2 protein is a methylcytosine oxy- observed for 6 months. The long latency to development of malig-
genase responsible for converting 5-methylcytosine (5mC) into 5- nancy in these experiments may speak to the subtlety of the epig-
hydroxymethylcytosine (5hmC) using iron and α-ketoglutarate enomic abnormalities initially conferred by acquisition of DNMT3A
(α-KG, produced by IDH1 and IDH2), and for further oxidizing mutations.
5hmC to 5-formyl- and 5-carboxycytosine.99 These reactions are In humans, the significance of DNMT3A mutations in the
thought to contribute to active demethylation through base excision pathogenesis of MDS is not clear. They occur with somewhat less
repair back to unmodified cytosine.100 Mutations in TET2 tend to be frequency than the most common recurrent mutations (between 8
inactivating frameshift or nonsense mutations or specific missense and 13% in most studies80,110 although some studies have quoted
substitutions predicted to lead to abrogation of protein function, and much lower frequencies below 5%).111 On the other hand, clonal

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
950 Part VII  Hematologic Malignancies

DNMT3A mutations have recently been shown to exist with modest deleterious cytogenetic occurrence in MDS,123 though not all 7q
frequency in healthy older adults and are in fact significantly more deletions affect the EZH2 locus.124 Mutations in EZH2 itself are
common than mutations in any other single gene,31 with an overall found in 5% to 10% of patients with MDS80 and 20% to 30% of
frequency that is significantly higher than their aggregate representa- patients with CMML.125 Consistent with the model of EZH2 as a
tion among hematologic malignancies. This discrepancy could either negative regulator of pluripotency and survival, mutations tend either
reflect the same long latency of DNMT3A-mutated hematologic to be inactivating frameshift or nonsense mutations, or missense
malignancy that was observed in mouse models, or it could suggest mutations concentrated in the gene’s SET domain, which is critical
that DNMT3A mutations have a relatively less potent pathogenic for DNA binding.126,127 EZH2 mutations confer a negative prognosis
effect than some other genes (e.g., TET2 mutations, which are less in MDS that appears to be independent of other prognostic factors,
frequent in the general population but more frequent in MDS). including the IPSS score, in part because they tend not to be associ-
DNTM3A mutations appear to be negatively correlated with ated with specific clinical characteristics that are incorporated into
mutations in certain other genes, particularly SRSF2 and ASXL1,80 these systems.79
and in low-risk MDS they seem to have a positive correlation with
SF3B1 mutations.112 There is evidence that DNMT3A mutations
confer a poor prognosis in cytogenetically normal AML,106 but that IDH Genes
prognostic significance so far has not been convincingly shown to
apply to DNMT3A-mutated MDS. The isoforms of isocitrate dehydrogenase, encoded by IDH1 and
IDH2, are responsible for the conversion of isocitrate to α-KG, which
as above is used by TET2 in the conversion of 5mC to 5hmC.
ASXL1 Mutations in IDH1 and IDH2 lead to enzymes with neomorphic
activity that convert α-KG to d-2-hydroxyglutarate, which accumu-
ASXL1 codes for a polycomb chromatin-binding protein and is lating data suggests is an oncometabolite that can inhibit both TET2
involved in epigenetic regulation of gene expression. It acts as a and other epigenetic enzymes, including prolyl hydroxylases and a
co-activator of the retinoic acid receptor and directly interacts with number of histone demethylases.128 Mutations in IDH1/2 and other
chemical modifiers of histones (e.g., NCOA1, a histone acetyltrans- members of the IDH pathway (including WT1) have repeatedly been
ferase, and LSD1, a histone demethylase). ASXL1 mutations occur shown to be important in AML,129 but IDH mutations are relatively
in about 10% to 29% of total MDS and myeloproliferative neoplasm infrequent in MDS, occurring in 5% or fewer of patients.79,80 Both
(MPN),113 17% of AML, and 40% of CMML.114 IDH1 and IDH2 mutations can cooccur with most other recurrent
The specific mechanisms by which ASXL1 mutations affect the mutations besides TET2, with which they are essentially mutually
development of MDS are not clear. The first mice engineered to have exclusive.
constitutive ASXL1 germline insufficiency survived to adulthood
with relatively mild lymphopenia and modest splenomegaly, and did
not develop myelodysplasia.115 Subsequently, ASXL1 mutations were Transcription Factor Genes
found to confer global reduction H3K27 trimethylation by disrupt-
ing normal recruitment of polycomb recessive complex 2 (PRC2), Transcription factors represent a third class of genes commonly
which places the H3K27 mark in vivo.116 This was followed by a more mutated in MDS. Similar to epigenetic and splicing genes, mutated
physiologic attempt at conditional ASXL1 knockout in murine transcription factors can have pleiotropic effects on a number of gene
hematopoietic cells, which did induce abnormal myeloid differentia- targets, and these mutations are indeed also often early events in
tion that was compounded by additional loss of TET2. In this study, MDS pathogenesis.
ASXL1-deficient cells displayed differential expression of a set of
genes largely related to hematopoietic differentiation.117
In humans, most MDS-associated ASXL1 mutations affect the RUNX1
C-terminal portion of the protein, specifically the plant homeo
protein interaction domain.118 This observation has led to speculation RUNX1 (formerly known as AML1) is the transcription factor gene
that the mutant protein retains DNA-binding activity and thus exerts most commonly mutated in MDS, and its biology is complex. It
a dominant-negative effect on wildtype ASXL1, which may not have encodes the alpha subunit of the core binding transcription factor
been captured in the mouse experiments. In studies of MDS cohorts, and is involved in determining the lineage fate of HSCs.130 RUNX1
investigators have observed that ASXL1 mutations co-occur less fre- was initially identified as one of the genes involved in two different
quently with certain other recurrent genetic lesions, particularly common pathogenic translocations: t(8;21), found in AML, and
DNTM3A and JAK2.119 On the other hand, ASXL1 mutations have t(12;21), found in acute lymphoblastic leukemia.131,132 Subsequently,
been found to cooccur with both RUNX1 and TET2 abnormali- germline point mutations in RUNX1 were identified in autosomal
ties.120 The most recent rigorous studies suggest that ASXL1 muta- dominant familial platelet disorder with propensity for AML,38 and
tions have a modestly poor prognosis, but tend to occur in patients later as somatic events in both sporadic AML and MDS.133 Reflecting
with low IPSS scores who would otherwise be felt to have indolent the complexity of RUNX1 biology, mutations occurring throughout
disease, perhaps suggesting a more profound pathogenic effect the gene, can be either monoallelic or biallelic, and can be frameshift
than might be suspected.79 insertions or deletions or nonsense or missense substitutions.134
However, most mutations appear to have an inactivating effect on
RUNX1 function, either by affecting the DNA-binding RUNT
EZH2 domain or by disrupting the C-terminal protein interaction domain.135
Many of the remaining mutations outside these regions appear to
EZH2 encodes the catalytic subunit of PRC2, which promotes the affect RUNX1 interactions with epigenetic regulators like MLL,
di- and trimethylation of lysine 27 on histone 3 (H3K27). Locally, thereby affecting histone methylation.136 Both clonal and subclonal
methylated H3K27 results in closed chromatin and transcriptional RUNX1 mutations appear to confer a poor prognosis in MDS
repression, and global H3K27 trimethylation in particular is associ- patients, irrespective of other prognostic factors.78
ated with reduced pluripotency and cellular senescence, suggesting a
role for EZH2 in regulation of cell fate.121 Recent studies have shown
that α-KG also regulates relative levels of methylation at this locus, ETV6
suggesting a possible convergence with the TET2/IDH pathway.122
EZH2 resides on the long arm of chromosome 7, and its loss has ETV6 encodes an ets-like transcription factor with mostly repressive
been hypothesized to be at least part of the reason 7q− is such a activity.137 It is situated on the short arm of chromosome 12, and its

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 951

role in hematologic malignancy has been best characterized by its tyrosine kinase mutations found in MDS,156 but still only occur in
involvement in recurrent translocations, including t(3;12)(q26;p13)138 about 5% of cases and as mentioned earlier, tend to begin as sub-
and deletions of 12p.139 In MDS, however, both missense and inac- clonal, proproliferative events that frequently drive the transition to
tivating frameshift point mutations have been described as well. AML.157 Best known for its role in MPNs, JAK2 is mutated in about
There is some data that the missense mutations, which mostly cluster 3% to 5% of MDS, particularly RARS-T and CMML.158 JAK2
in the DNA-binding ETS domain, have a dominant negative effect mutations in MDS tend not to be associated with overproduction of
on the wildtype allele.140 However, whether malignant transformation mature hematopoietic cells as they are in MPNs, likely as a conse-
requires ETV6 activity below a critical threshold remains unclear, as quence of concomitant biologic defects contributing to ineffective
not all frameshift mutations occur with loss of heterozygosity (and hematopoiesis.159
some frameshifts can confer dominant negative function as well).
ETV6 mutations are relatively rare events in MDS, occurring in at
most 5% of cases;79 recently, familial cases of MDS and AML because Cohesin Complex Genes
of inherited ETV6 mutations have also been described.39
Genes encoding members of the cohesin complex family, including
RAD21, STAG2, SMC3, and SMC1A, are each mutated in a small
GATA2 minority of MDS cases, but collectively, cohesin mutations can be
found in about 10% of MDS.160 Their physiologic role is in the
GATA2 encodes a transcription factor with pleiotropic effects in early maintenance of chromatid structural fidelity, particularly during
hematopoietic progenitor cells.141 In contrast to most other genes mitosis. How cohesin complex mutations affect MDS pathogenesis
described here, acquired mutations in GATA2 are quite rare, but there is not completely understood, since they do not appear to directly
are a number of clinical syndromes associated with germline GATA2 promote chromosomal instability. Clinical studies, however, have
mutations, several of which involve a risk of developing MDS and shown that they appear to be disproportionately associated with
AML. These include Emberger syndrome (congenital lymphedema multilineage dysplasia, and confer inferior survival and an increased
and risk of AML),142 autosomal dominant monocytopenia and risk of progression to AML.161
mycobacterial infection syndrome,143 and dendritic cell, monocyte, B
and natural killer (NK) lymphoid deficiency syndrome.144 In addi-
tion, GATA2 mutations have also been described in kindreds with no Other Genes
accessory phenotype beyond a strong history of early-onset MDS/
AML.37 Several phenotypic components appear to be dependent on The genes described previously collectively account for the majority
the type of mutation. For instance, nearly all patients who develop of mutations found recurrently in MDS, but this list is certainly not
MDS or AML have mutations in or immediately 5′ to the second exhaustive. Even very recent, broad surveys of patients with MDS
zinc finger domain, which tend to be missense substitutions predicted using the most updated panels of gene mutations and high-resolution
to affect DNA binding.145 On the other hand, a second group of FISH and karyotyping have shown that between 10% and 20% of
patients with truncating frameshift mutations in the N-terminal patients lack a detectable genetic abnormality.78–81 There are multiple
region of the gene tend to present at younger ages with more pro- possible explanations for this observation. For example, some of
nounced immune deficits, but have a lower risk of MDS or AML. these patients may have other types of aberrations not captured by
Although the mechanism by which these patients develop MDS is sequencing or karyotyping, such as small copy number abnormali-
not completely clear, the subset of patients with GATA2 germline ties that might be detected by SNP arrays, which are not routinely
mutations who develop MDS or AML has been observed to acquire performed in clinical practice. More likely, however, many of these
ASXL1 mutations with frequency much greater than would be patients probably have mutations in genes that are less frequently
expected to occur by chance.146 altered in MDS pathogenesis. Some of these mutations are in path-
ways represented by other, better-known genes; for example, rare
mutations have been described in PTPN11, a tyrosine phosphatase
TP53 that acts as a downstream effector in the RAS pathway,162 and in
BRAF, better known for its association with melanoma and hairy
TP53 mutations occur in about 10% of patients with MDS, and as cell leukemia.163 Similarly, rare mutations have been described in
in other settings imply a poor prognosis and response to therapy.79 EED and SUZ12, other components of the PRC2 that affiliate with
They are closely associated with a complex karyotype and tend not EZH2 and collectively interact with ASXL1.164 Other times, MDS
to cooccur with other recurrent driver mutations.147,148 They fre- can occur in the setting of mutations in genes better known for being
quently cooccur with del(5q) and may represent a progression associated with other diseases, such mutations in CBL, a gene better
pathway for patients with 5q− syndrome.149 They are also frequently known for its association with juvenile myelomonocytic leukemia
found in t-MDS and AML.49 Recently, a small series of patients with that encodes an E3 ubiquitin ligase responsible for degrading several
t-AML showed that TP53 mutations present in the leukemia were tyrosine kinases.165 Finally, mutations are occasionally described in
detectable in samples collected 3–6 years earlier, which in at least two relatively novel gene classes, including genes encoding the G-protein
patients predated their original chemotherapy.150 This suggests that subunits GNAS166 and GNB1167 and the DNA repair enzymes
the expansion of rare preexisting clones harboring TP53 mutations hOGG1, XRCC3, and XPD.168 Some patients may have mutations
may be the leukemogenic mechanism in at least some patients with in other genes that have yet to be described. While it is unlikely
therapy-related disease, rather than accumulation of DNA damage that a major, frequently mutated pathway remains undiscovered
induced by the chemotherapy itself. in MDS, study of these low-frequency, “long-tail” mutations may
yield valuable further insights into the molecular pathogenesis of the
disease.
Tyrosine Kinases and Growth Factor Receptors
Mutations in tyrosine kinase and growth factor receptor genes are Karyotypic Abnormalities
typically associated with proproliferative signals and occur in a wide
range of myeloid malignancies, including AML (FLT3151), MPNs Chromosomal abnormalities, larger-scale genetic aberrations that can
(JAK2152 and MPL153), and mast cell disorders (KIT).154 While they be detected on either karyotype or FISH, are also common events in
do occur in MDS, they are usually late, subclonal events that often MDS.169 The most common types of abnormalities in MDS are
mark progression to secondary AML.155 A few of these deserve specific deletions or duplications of very large chromosomal regions; as
mention. Activating mutations in NRAS are the most frequent opposed to some other hematologic cancers, translocations and

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
952 Part VII  Hematologic Malignancies

inversions are less common. Several of the most common karyotypic


abnormalities have been shown to have prognostic value, and one, Trisomy 8
del(5q), has enough unique biologic features to be its own subclas-
sification within the WHO criteria. Trisomy 8 is present in about 5% of MDS patients and can be found
in a wide range of other myeloid disorders, including AML, MPNs,
and aplastic anemia. In MDS, it can often be seen as a late, subclonal
del(5q) event.187 Although its contribution to pathogenesis is incompletely
understood, MDS patients with trisomy 8 appear to upregulate WT1,
Interstitial deletion of the long arm of chromosome 5 [del(5q)] is the an oncogene that can be mutated in AML (but very rarely in MDS),
most common chromosomal abnormality in MDS,42 and del(5q) is which may behave as a neoantigen that stimulates the expansion of
the only karyotypically-defined subtype recognized by the WHO.170 oligoclonal CD4+ and CD8+ T cells.188 Some evidence suggests that
Although the specific region affected varies between patients, there these populations may contribute to impairment of hematopoiesis in
are two commonly deleted regions (CDRs), one on 5q31.1 and the MDS patients with trisomy 8,189 which may explain why some patients
other at 5q32-33.3, with most patients having a deletion that includes with isolated trisomy 8 can have substantial responses to immune
both CDRs.171 MDS patients in whom del(5q) is the sole karyotypic suppression with antithymocyte globulin (ATG).190 Trisomy 8 has also
abnormality often display the “5q-minus syndrome,” which is clini- been associated with the development of paraneoplastic autoimmune
cally characterized by anemia, normal or elevated platelet count, phenomena, such as Behçet disease,191 again reflective of the immune
female predominance,172 lower risk of transformation to AML, and dysregulation characteristic of this cytogenetic abnormality.
a striking response to lenalidomide.173
Our understanding of the pathobiology underlying del(5q) MDS
has improved substantially over the past several years; a key observa- del(20q)
tion was the lack of recurrent point mutations in genes located on
5q in other patients with MDS, which, coupled with the fact that del(20q) is an infrequent chromosomal aberration in MDS, occurring
most patients with del(5q) retain one normal chromosome 5, sug- in about 2% of patients.192 Patients with del(20q) frequently have
gested that the pathobiology could be best explained by haploinsuf- prominent thrombocytopenia, may have concomitant mutations of
ficiency of deleted genes.174 Indeed, it now appears that different U2AF1, and appear to have an intermediate prognosis,193 although
genes lost in the CDRs are responsible for different aspects of the 20q loss can also be a late event that indicates clonal progression of
5q− phenotype. For instance, haploinsufficiency of RPS14, a ribo- disease.194 The best candidate driver gene lying within the common
somal subunit gene located at 5q31.2, leads to p53 activation in deleted region is a tumor suppressor gene known as MYBL2,195 but
erythroid progenitors and is responsible for the dyserythropoiesis seen recent studies suggest that in myeloid models, a reduction in MYBL2
in the syndrome,175 and deletion of a key microRNA, miR-145, is levels below what would be predicted for classic haploinsufficiency is
responsible for the megakaryocytic component of the phenotype.176 required to drive clonal expansion.196 Although ASXL1 resides on
Separately, haploinsufficiency of CSNK1A1, which is located at 5q32 20q, it sits outside the CDR,197 and most patients with 20q abnor-
and encodes casein kinase 1-alpha, is responsible for the sensitivity malities do not have concomitant ASXL1 mutations.
to lenalidomide, which accelerates the ubiquitination and degrada-
tion of remaining casein kinase 1-alpha through a cereblon-dependent
process.177 Other studies have suggested that additional genes on 5q, 17p Deletions
including EGR1, APC178, HSPA9179, NPM1180, and others181 may
contribute to features of the disease via a similar mechanism of Chromosome 17 abnormalities occur most often in MDS in associa-
haploinsufficiency in select cases. tion with complex karyotypes, which is most likely related to the fact
These points apply only to del(5q) as a sole karyotypic abnormal- that TP53 resides within the common deleted region on 17p.147 In
ity in MDS. When del(5q) is found with other chromosomal MDS, many patients with loss of 17p will have an inactivating
abnormalities, especially in the context of a complex karyotype (three mutation of their remaining copy of TP53, implying that haploinsuf-
or more karyotypic abnormalities, a finding often associated with ficiency is not in and of itself enough to drive pathogenesis.198 At the
TP53 mutations or 17p loss), the prognosis is poor and the response same time, patients almost never lose both copies of 17p as part of a
to lenalidomide seen in 5q− syndrome usually does not exist.149 larger chromosomal event, suggesting that some other gene or genes
Cooccurrence of del(5q) with TP53 mutation or 17p loss, in fact, in this region may be essential for hematopoietic cell survival. As with
occurs more frequently than would be expected by chance, suggesting TP53 point mutations, loss of 17p is an exceedingly poor prognostic
cooperativity of the two abnormalities. Del(5q) seen in the context factor in MDS, is frequently seen in cases of therapy-related disease,53
of AML, even if the sole karyotypic abnormality, is a universally poor and often presages the development of treatment-refractory AML.199
prognostic sign.182

Complex and Monosomal Karyotypes


Chromosome 7 Abnormalities
Complex karyotypes, meaning those with three or more cytogenetic
Deletion of one entire copy of chromosome 7 (i.e., monosomy 7) abnormalities, are one of the more common abnormalities in MDS.169
is also characteristic of MDS and portends a poor prognosis.183 The About half are associated with TP53 mutations; conversely, many, but
pathogenesis of chromosome 7 abnormalities is incompletely under- not all, patients with TP53 mutations have complex karyotypes.147 As
stood. Several genes recurrently mutated in MDS, including opposed to 17p abnormalities, which are reflective of the TP53 loss
EZH2,123 MLL3,124 and CUX1,184 lie on 7q, and it has been hypoth- itself, complex karyotypes are instead downstream effects of p53 loss
esized that loss of some or all of chromosome 7 contributes to MDS and represent the type of structural DNA damage that would have
pathogenesis via a haploinsufficiency mechanism similar to that seen triggered p53-mediated apoptosis in normal cells. The exceedingly
in del(5q). If such a mechanism exists, however, the evidence sup- poor prognosis associated with complex karyotypes is in fact probably
porting it has not yet been produced. Part of the difficulty in a proxy for p53 loss; the half of patients with complex karyotypes who
proving its existence lies in the fact that unlike del(5q), there is no have intact p53 function appear to have prognoses similar to patients
common deleted region on chromosome 7 in which to focus efforts with normal cytogenetics. Monosomal karyotypes, defined as complete
at driver gene discovery.185 It is important to note that in the revised loss of at least two entire chromosomes or one monosomy plus one
IPSS (IPSS-R), del(7q) is considered to be an intermediate-prognosis other abnormality, are also common and also tend to confer a poor
abnormality distinct from monosomy 7, which is classified as poor prognosis, particularly when associated with monosomy 7 or mono-
prognosis.186 somy 5.200 When monosomal and complex karyotypes occur together,

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 953

however, the monosomy does not appear to confer an additional poor into clinically evident cancers. Perturbation of this immune response
prognosis beyond that of the complex karyotype.201 presumably allows the persistence and propagation of abnormal
clones in MDS, but the extent to which this is pathogenically impor-
tant is not clear.217
Other Karyotypic Abnormalities Several aspects of innate immunity have been shown to be abnormal
in MDS patients.218 Some toll-like receptors, including TLR4, are
A number of other cytogenetic abnormalities have been repeatedly overexpressed on MDS progenitor cells,219 and increased TLR signaling
described in MDS, including –Y,202 del(13q),203 del(11q),139 del(12p) has been associated with higher degrees of apoptosis and the develop-
or t(12p),204 del(9q),192 idic(X)(q13),192 t(11;16)(q23;p13.3),205 t(3;21) ment of cytopenias. In patients with del(5q), loss of miRs 145 and 146a
(q26.2;q22.1),206 t(1;3)(p36.3;q21),207 t(2;11)(p21;q23),205 inv(3) from the common deleted region has been shown to upregulate tumor
(q21q26.2)207 and t(6;9)(p23;q34).208 Most of these do not have spe- necrosis factor (TNF) receptor–associated factor 6, a TLR adaptor
cific phenotypes or associated with them, but some do confer prognostic E3 ubiquitin ligase, as well as Toll IL-1 receptor domain-containing
information and are included in the IPSS-R. In particular, –Y and adaptor protein (TIRAP), two important downstream effectors of
del(11q) are designated “very good” prognosis; del(12p), del(20q), TLR4.220 del(5q) may also lead to the upregulation of CD14 on granu-
isolated del(5q), and normal karyotypes are considered “good’ ” trisomy locytes via heterozygous loss of mDia1, a scaffolding protein involved
8, del(7q), isochrome 17(q), trisomy 19, and trisomy 21 are considered in actin polymerization, which appears to confer abnormalities of the
“intermediate;” monosomy 7, double deleted (7q), derivative (3q), and innate immune response.221 More recently, myeloid-derived suppressor
complex karyotype with exactly three abnormalities are considered cells have been shown to be significantly expanded in some patients
“poor,” and complex karyotypes with more than three abnormalities with MDS, and increased signaling in these cells via interaction with
are considered “very poor.” In addition, finding any of these in the S100A9, an important inflammatory mediator, has been shown to
appropriate clinical context, regardless of whether they are known to similarly stimulate inappropriate apoptosis in hematopoietic precursor
carry prognostic information, is typically enough to warrant a diagnosis cells.222 Coincident with these findings, population-level data have
of MDS, even in the absence of clear morphologic dysplasia. suggested that patients with chronic inflammatory stimulation appear
to be at increased risk of developing MDS and AML.223
Abnormalities in the adaptive immune system also exist in MDS.
The Microenvironment in Myelodysplastic Syndrome These have been particularly well described in hypoplastic variants,
which have similarities to aplastic anemia at both the cellular and
The malignant transformation of hematopoietic progenitors in MDS genetic level, and both can respond to immune suppression.224 More
occurs not in isolation, but within the rich microenvironment of the broadly, oligoclonal T-cell receptor gene rearrangements can be found
bone marrow stroma, which has come to be known as the “hemato- in MDS patients of all subtypes,225 and clinically detectable abnor-
poietic niche.”209 Evidence has shown that under normal circum- malities of NK-cell, B-cell, and T-cell number and function can be
stances, hematopoiesis is in part regulated by paracrine signals released detected in a subset of patients as well.226–228
from nonhematopoietic mesenchymal cells,210 and evolving data
suggest that some of these processes are dysregulated in MDS. Early
circumstantial clues came from findings that patients with MDS and Abnormal Apoptosis
other hematologic malignancies have abnormal circulating levels of
cytokines known to affect hematopoiesis, including monocyte-colony Although MDS is typically characterized by normal or increased
stimulating factor, interleukin (IL) 1a, and granulocyte-monocyte marrow cellularity, abnormalities of apoptosis are frequently described,
colony stimulating factor (GM-CSF),211 and other studies have particularly in low-risk variants, where increased apoptosis has been
shown that hematopoietic cells from MDS patients respond abnor- hypothesized to contribute to the development of ineffective hema-
mally to these cytokines compared to cells from healthy controls.212 topoiesis.229 There are two major mammalian apoptosis pathways.
More recent studies have shown that hematopoietic progenitor cells The death receptor pathway, also known as the external pathway, is
transplanted into older mice display a narrower range of clonal expan- triggered by ligation of TNF family members, which leads to the
sion than identical cells transplanted into younger mice, suggesting recruitment and activation of caspases at the cell surface.230 There is
that an aging niche might exert selective pressure on dominant ample evidence of abnormalities in this pathway in MDS; for
hematopoietic progenitor cell clones.213 Furthermore, it has been instance, TNF-α levels have been shown to be increased in all MDS
discovered that conditional deletion of the gene encoding dicer1, a subtypes,231 and other proapoptotic cytokines, including IL-8, trans-
microRNA processing enzyme, in osteoprogenitor cells can induce forming growth factor β, interferon-γ, and Fas ligand, have been
myelodysplasia and secondary AML in mice;214 concurrently, other shown to be elevated as well.232 The other major apoptotic pathway,
studies have shown altered expression of DICER and DROSHA, known as the BCL-2 or intrinsic pathway, involves a complex balance
another microRNA-processing enzyme, in mesenchymal cells of between proapoptotic (BAX, BAK, BAD, PUMA, and others) and
MDS patients.215 Even more intriguing, it has recently been shown antiapoptotic molecules (BCL-2, BCL-XL, MCL-1, and others),233
that introduction of an activating B-catenin mutation in mouse and has also been shown to be abnormal in MDS. For example, ratios
osteoblasts can induce the development of AML through induction of proapoptotic (Bax/Bad) to antiapoptotic (Bcl-2/Bcl-XL) molecules
of the Notch ligand jagged1, which leads to Notch activation in are elevated in patients with lower-risk subtypes of MDS (RA and
HSCs.216 As tantalizing as these findings are, the extent of their RARS), but the ratio reverses in RAEB and secondary AML, primar-
pervasiveness across all MDS patients, and how microenvironmental ily driven by increased BCL-2 expression.234 Coordinate with this
alterations interact with the well-validated recurrent mutations found observation, but somewhat paradoxically, inhibition of apoptosis by
in hematopoietic cells, has yet to be determined. It is worth noting BCL-2 appears to prevent leukemic transformation in murine models
that HSC transplantation, which involves only the transplantation of of MDS.235 Preclinical studies further suggest that inhibition of
hematopoietic elements and not stroma, can be curative for some BCL-2 and other antiapoptotic molecules may delay progression of
patients with MDS, implying that at least in these patients, the niche disease236 and may sensitize MDS cells to hypomethylating agents
is not sufficient to sustain disease. such as azacitidine, but clinical trials combining these approaches
have not yet been performed.237

Immune Dysregulation
Transformation to Acute Leukemia
In normal human tissues, both the innate and adaptive immune
systems play important roles in identifying, isolating, and destroying Although MDS and AML are often uttered in the same breath and
early clones with malignant potential before they are able to transform conceptualized as two facets of a similar disease process, this is an

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
954 Part VII  Hematologic Malignancies

overly simplistic view that ignores the complexity and heterogeneity marrow microenvironment and beyond. Second, it will also be neces-
of each disease on its own.82 Although MDS can and often does sary to integrate these biologic storylines with clinical information,
transform into secondary AML, other types of de novo AML are in the hope that a fuller understanding of MDS biology may uncover
biologically quite distinct from MDS, and conversely, some low-risk new treatment approaches and better predictive and prognostic
forms of MDS are biologically distinct from secondary AML. At the models than do single biologic features alone. Such work is already
same time, however, there is a continuum between the two diseases beginning; for instance, a recent report suggested that an algorithm
that is only now starting to become fully understood. Recently, for integrating sequencing for common driver mutations with principal
instance, it has been shown that cases of AML harboring mutations component analysis of gene expression better predicts some clinical
in any of eight genes (SF3B1, SRSF2, U2AF1, ZRSR2, ASXL1, features of MDS than sequencing or expression data considered in
EZH2, BCOR, or STAG2) are highly likely to have evolved out of isolation, and the algorithm outperformed the IPSS in predicting
MDS, even when no antecedent stage of MDS was clinically overall survival (76% versus 64% accuracy).244 Thus far, however,
evident.148 The converse, that AML with driver mutations in other these types of integrative studies remain in relative infancy. Making
genes must have arisen de novo, is not necessarily true; NPM1 muta- significant gains in this type of deep understanding will be a substan-
tions appear to be restricted to de novo AML, but mutations in tial undertaking, requiring large, well-coordinated clinical trials with
DNMT3A, TET2, and IDH1/2 can occur in both secondary and de rigorous banking of serial blood and marrow samples, careful annota-
novo AML and thus do not appear to be ontogeny-specific. In this tion of clinical features and outcomes, and intensive collaboration
study, TP53-mutated AML comprised its own category and was between academics, industry, and clinicians in devising strategies for
associated with prior chemotherapy and complex karyotypes, consis- rationally targeting key elements of MDS biology.
tent with prior studies.
Understanding this shared genetic basis of MDS and secondary
AML has also shed light on the transition from one to the other. CLINICAL FEATURES OF MYELODYSPLASTIC SYNDROME
Although the eight genes defining secondary ontogeny (i.e., disease
arising from antecedent MDS) have different functions, they all tend Compared to other hematologic disorders, the clinical features of
to have broad, pleiotropic effects and for the most part have in mouse MDS are often underemphasized relative to laboratory aspects of the
models been shown to be insufficient for leukemogenesis when they disease. This is in part because of the fact that the major common
occur in isolation. The fact that they occur infrequently in de novo physiologic defect, ineffective hematopoiesis, usually leads to some-
AML may imply that they have effects strong enough to commit cells what vague signs or symptoms. Nevertheless, it is important to
to an MDS phenotype, with increased self-renewal and inhibitory understand these features within the context of the disease’s pathogen-
effects on differentiation and hematopoiesis, whereas TET2 and esis and management. Some patients with MDS have distinct clinical
DNMT3A mutations may have more subtle effects that increase phenotypes that are largely related to genetics or specific pathologic
self-renewal but do not in and of themselves predispose to a specific features. MDS can also coexist with other hematologic malignancies,
phenotype in the absence of specific secondary mutations. including multiple myeloma (even in previously untreated patients)245,
In most cases, progression of MDS to AML involves the acquisi- hairy cell leukemia,246 chronic lymphocytic leukemia (CLL),247 non-
tion of proproliferative mutations, which tend to be late, subclonal Hodgkin lymphoma,248 and large granular lymphocyte leukemia
events.238 These mutations frequently occur in tyrosine kinase genes (LGL).249 Parsing the components of presentation that could be caused
like FLT3,155 NRAS,239 or KIT,240 or in certain transcription factor by a coexisting process (e.g., in a patient with both MDS and CLL,
genes such as CEBPA,241 and lead to constitutively activated growth which is the primary contributor to the patient’s cytopenias?) is an
and proliferative pathways. As discussed elsewhere, patients who important task for clinicians and pathologists.
develop secondary AML from an antecedent MDS tend to have a
poorer prognosis, with both a lower rate of complete remission and
a poorer overall survival, independent of remission, than patients with Differential Diagnosis
de novo AML.242 In unselected populations, some of this difference
in outcome can be explained by proximate characteristics of the In the absence of clonal markers or excess blasts, MDS is a diagnosis
secondary AML cohorts, which tend to be older and have more of exclusion, requiring that other potential contributors to cytopenias
comorbidities, mirroring the demographics of the MDS population. and myelodysplastic morphology be ruled out to the greatest extent
However, patients with secondary AML have inferior outcomes even possible.250 Since cytopenias are the most common presenting clinical
compared to age-matched controls with de novo AML,235 which has feature, the differential diagnosis is in theory broad and includes
remained the case when redefining ontogeny based on the genetic numerous other entities covered in more detail elsewhere in this
stratification outlined earlier.243 This inferior outcome has been pre- book. In practice, however, all patients suspected of having MDS
sumed to reflect the intrinsic refractory nature of the antecedent should undergo a bone marrow aspirate and biopsy, since the diag-
MDS, and this presumption is now being proven true by sequencing nostic criteria are almost entirely pathologic and marrow findings are
bone marrow samples from secondary AML patients who morpho- critical for risk stratification and treatment planning.
logically appear to be in remission—in fact, the proliferative muta- Several nutritional deficiencies can cause cytopenias and morphol-
tions are often eliminated but the preexisting driver mutations ogy similar to MDS. Folate deficiency can cause macrocytosis and,
remain, suggesting that chemotherapy simply reverted the marrow to in extreme cases, megaloblastic morphology in the marrow.251 Vitamin
the preexisting clonal state.58,148 Overcoming the intrinsic resistance B12 (cobalamin) deficiency is a more classic cause of megaloblastic
to therapy that appears to be common to most patients with MDS changes, and patients with severe B12 deficiency can have bizarre
remains one of the central challenges of treating the disease. morphology with an abundance of early forms that can occasionally
be confused with evolving AML, especially erythroleukemia.252 Since
the hematologic changes of B12 deficiency can occur in the absence
Integrating the Pathobiology of   of classic neurologic symptoms, B12 and folate levels should be
Myelodysplastic Syndrome checked on all patients in whom a potential diagnosis of MDS is
being entertained, and methylmalonic acid and homocysteine levels
With the continuous, deepening accumulation of data describing the may help clarify whether deficiency is truly present in cases with
pathogenesis underlying MDS, one of the central future challenges borderline levels.253
will be integrating these data into coherent models that correctly Copper deficiency is less common than B12/folate deficiency, but
reflect fundamental aspects of the disease. The need for integration can also be mistaken for MDS. It classically arises in patients who
is twofold. First, there is a need to integrate disparate components of have had gastrectomies or certain forms of gastric bypass surgery,
the biology itself, including somatic mutations, changes in gene particularly biliopancreatic diversion,254 and can also develop in
expression, epigenetic disruption, and cell-cell interactions within the patients taking high doses of zinc, which competes with copper for

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 955

absorption in the small bowel255 (it is important to note that newer to medical attention.281 When present, fever is most commonly a sign
forms of gastric bypass, including gastric sleeve procedures and newer of infection, though a small number of patients may run low-grade
iterations of the roux-en-y, maintain small bowel absorption such that fevers in the absence of a discernible infection. Splenomegaly or
patients do not typically develop copper deficiency). Clinically, hepatomegaly are rare and should prompt consideration of either an
patients with copper deficiency develop an anemia that can be nor- alternate diagnosis or an MDS/MPN overlap syndrome.
mocytic or macrocytic and can occasionally be profound. Mild
neutropenia can also be seen, while clinically significant thrombocy-
topenia is rare.256 Such patients can also develop neurologic symptoms Dermatologic Manifestations
similar to those seen in vitamin B12 deficiency, including peripheral
neuropathy, myelopathy, demyelination, and rarely optic neuritis.257 Although dermatologic manifestations of MDS are uncommon,282 a
In the marrow, copper deficiency is classically associated with ring few deserve specific mention. Neutrophilic dermatosis (Sweet syn-
sideroblasts and erythroid and neutrophil vacuolization.258 In the drome) is characterized by painful plaques on the face, neck, or
right clinical setting, negative testing for somatic SF3B1 mutations extremities, often in association with fever and diffuse arthralgias.283
in the presence of definite ring sideroblasts should elevate the degree While neutrophilic dermatosis can be seen in any subclass of MDS,
of suspicion. it is classically associated with an impending transformation to AML.
Alcohol is a common cause of hematologic abnormalities, espe- It frequently responds to steroids or dapsone therapy, but may recur
cially macrocytic anemia, even in the absence of folate or vitamin B12 as steroids are tapered.284 Recently, Sweet syndrome in MDS patients
deficiency.259 Bone marrow examination often reveals sideroblastic has been associated with heterozygous mutations in MEFV, a gene
changes, and sometimes megaloblastic morphology, but frank dyspla- linked to Mediterranean fever.285
sia is uncommon.260 Patients with borderline marrow findings who Pyoderma gangrenosum (PG) is an ulcerative, necrotic lesion that
are suspected of significant alcohol intake should be advised to most frequently develops on the extremities.286 Histologically it is also
completely abstain from drinking (such advice is occasionally heeded), characterized by neutrophilic infiltrates; clinically, it may be associ-
with a repeat marrow examination in 2–3 months. ated with pathergy and frequently develops at the site of recent minor
Other drugs and toxins have also been associated with dysplastic trauma, such as intravenous (IV) catheter insertion sites. PG can be
changes in the marrow. Well-described offenders include valproic associated with a wide range of underlying conditions other than
acid,261 mycophenolate mofetil,262 ganciclovir,263 alemtuzumab,264 MDS. A high index of suspicion is necessary to distinguish it from
nucleoside analogues such as fludarabine265 and cytarabine, and the necrotizing infections, since treatment of PG involves systemic ste-
antimetabolites mercaptopurine and methotrexate.266 These tend to roids, and surgical debridement is contraindicated.
cause macrocytic anemias and can also cause neutropenia and throm- Other dermatologic manifestations of MDS include monocytic
bocytopenia. On the other hand, isoniazid267 and chloramphenicol,268 infiltrates, classically of the gingiva and other mucosal surfaces, a
and to a lesser extent cycloserine269 and pyrazinamide,270 have been process most frequently seen in CMML;287 the development of a
associated with modest sideroblastic anemia. The anemia associated chloroma or granulocytic sarcoma, which by definition implies pro-
with isoniazid, in particular, can often be reversed with high doses of gression to AML; and petechial lesions, which most frequently develop
vitamin B6 (pyridoxine).271 Chloramphenicol can separately cause an on the lower extremities and imply severe thrombocytopenia.
idiosyncratic pancytopenia that is particularly distinguished by
prominent vacuolization of erythroid precursors.272
Among infectious agents, HIV infection has specifically been Autoimmune Manifestations
associated with dysplastic changes in the marrow. Patients typically
have hypercellular marrows with evidence of trilineage dysplasia most A percentage of patients with MDS may have overlapping immuno-
predominant in the erythroid line.273 The erythroid hematopoiesis logic or rheumatologic features to their disease, which may in part
almost always is megaloblastic, and reticulated fibrosis is often seen arise from the immune dysregulation that occurs during disease
in bone marrow biopsies. The marrows also often have polyclonal pathogenesis.288 In a few patients, these may be the presenting
plasma cell expansion, lymphoid aggregates, and granulomas are complaint. Such manifestations can include episodes of seronegative
often seen. The differential diagnosis of erythrodysplasia in patients oligoarthritis or polyarthritis,289 cutaneous vasculitis,290 polymyosi-
with HIV infection includes the influence of medications, opportu- tis,291 or autoimmune peripheral neuropathies. Rare patients can
nistic infections, or a direct effect of HIV on the hematopoietic present with a lupus-like syndrome that can include fever, polyar-
progenitor cells.274 Since the US Preventive Services Task Force now thralgias, polychondritis, pleuritis, pericarditis, and cytopenias
recommends a one-time HIV test for all adults,275 essentially all (including hemolytic anemias). Other autoimmune phenomena have
patients in whom MDS is being considered should be tested to rule also been reported, including mucocutaneous ulcerations, iritis,
out HIV. polymyositis, inflammatory bowel disease, and erythrocyte aplasia.
Other primary hematologic disorders are also on the differential Many of these, which are essentially paraneoplastic syndromes,
diagnosis with MDS. Hypoplastic MDS, for example, can be difficult respond to the initiation of immunosuppressive agents such as
to distinguish from aplastic anemia and in the absence of character- corticosteroids.292
istic cytogenetic or genetic abnormalities.276 Similarly, some patients Some reports have additionally documented cases of patients who
with MDS may have a significant degree of co-existing fibrosis, which were diagnosed with rheumatologic conditions only weeks or months
can complicate the distinction from a myeloproliferative disorder.277 before they were found to have MDS, including relapsing polychon-
Again, genetic analysis may help make this distinction clearer.278 dritis,293 polymyalgia rheumatica or temporal arteritis,294 Raynaud
phenomenon, Sjögren syndrome, and autoimmune glomerulone-
phritis.295 However, since some of these conditions are relatively
Signs and Symptoms common themselves, particularly in older populations, whether they
represent a true association with MDS or are merely coincidental
Signs and symptoms of MDS are typically vague. Although some occurrences is not entirely clear.
patients profess to being asymptomatic, fatigue is extremely common,
as is a sense of general malaise.279 Rarely, patients may complain of
diffuse arthralgias that can lead to suspicion for an underlying rheu- Objective Findings: Erythroid Lineage
matologic disorder like systemic lupus erythematosus.
Smaller proportions of patients may present with severe or recur- The majority of patients with MDS present with some degree of
rent infections as a result of immune defects,280 most commonly anemia, which contributes significantly to fatigue and lethargy, the
neutropenia. Bleeding or easy bruising as a result of thrombocytope- most common presenting complaint.296,297 The anemia is often
nia or qualitative platelet defects also bring some patients with MDS macrocytic, and the peripheral blood smears of MDS patients

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
956 Part VII  Hematologic Malignancies

Fig. 60.2  ERYTHROID DYSPLASIA. Examples of dysplastic erythroid precursors (bottom) compared to
those with normal morphology in the sequence of erythroid maturation (top). The dysplastic forms include
(left to right) abnormal immature forms with multinucleation; maturing forms with multinucleation and
nuclear-to-cytoplasmic dyssynchrony; and more mature forms with megaloblastoid change, nuclear budding,
cloverleaf forms, cytoplasmic vacuolization, and cytoplasmic stippling. Ringed sideroblasts (far right) also are
evidence of erythroid dysplasia. Photomicrographs are from patients with refractory cytopenia with multilin-
eage dysplasia and ringed sideroblasts.

frequently show a variety of abnormal forms, including oval macro- Acquired hemoglobin H disease is a rare but well-described
cytes, and less commonly dacrocytes, elliptocytes, or acanthocytes. development in MDS that has been associated with acquired muta-
Anemia in MDS is usually caused by ineffective hematopoiesis. tions of ATRX,304 a gene encoding a chromatin-remodeling protein;
Consequently, the erythropoietin (EPO) level is typically normal or when mutated in the germline, ATRX has been associated with
modestly elevated, though in many elderly patients it is still lower X-linked alpha thalassemia/mental retardation (ATR-X) syndrome.
than would be expected for the degree of anemia, in some cases The pathogenesis of the syndrome is related to severe reduction in
because of subclinical renal insufficiency.298 The ineffective hema- synthesis of alpha globin chains, and indeed, patients with acquired
topoiesis causes abnormal iron utilization in a substantial number ATRX mutations have clinical features resembling alpha thalassemia,
of patients, many of whom have evidence of iron overload based including microcytosis, anisocytosis, poikilocytosis, target cells,
on ferritin or transferrin saturation.299 Studies have shown other schistocytes, dacrocytes, and beta-chain tetramers on crystal violet
abnormalities of erythrocytes as well, including increased levels of staining.305 As opposed to the macrocytosis seen in most MDS
fetal hemoglobin,300 aberrant surface antigens,301 increased osmotic patients, those with acquired ATRX mutations are typically pro-
fragility, and low levels of pyruvate kinase,302 although none of foundly microcytic, again similar to thalassemia.
these are routinely checked in clinical practice. Hemolysis is not
routinely observed in MDS patients, but may occur in those with
some of these latter abnormalities or with concomitant autoimmune Myeloid Lineage
disorders.
Morphologic changes in the erythroid lineage on bone marrow About one-half of patients with MDS are neutropenic at the time of
examination can vary widely from patient to patient. Many patients diagnosis.306 Many also have defective neutrophil function irrespec-
have megaloblastoid erythroid precursors that contain multiple nuclei tive of the absolute neutrophil count (ANC), and patients with MDS
or asynchronous maturation of the nucleus and cytoplasm (Fig. have been shown to have inadequate inflammatory responses to
60.2). It is this asynchrony, with continued membrane synthesis in infections. Many have diminished production of hematopoietic
the absence of normal nuclear maturation, which is thought to give growth factors like granulocyte colony-stimulating factor (G-CSF)
rise to the macrocytosis displayed by most patients. Ring sideroblasts, and GM-CSF, and their neutrophils often have reduced phagocytic,
erythroid precursors containing iron-laden mitochondria, are another chemotactic, or bactericidal capabilities.307–309 In place of neutrophils,
relatively common finding.303 They are defined by the presence of at the proportion of monocytes is often elevated, which can sometimes
least five granules that are positive with the Prussian blue reaction be shown to have been the case for months or even years before
and line at least one-third of the circumference of the cell nucleus. diagnosis. Such a history can be observed patients with pure MDS,
Ring sideroblasts are not in and of themselves diagnostic of MDS not just those with MDS/MPN overlaps like CMML.
and can be seen in a number of other disorders (including congenital The granulocytes of MDS patients also display frequent morpho-
sideroblastic anemias and nutritional deficiencies), but their presence logic abnormalities (Fig. 60.3). In the marrow, either myeloid
in association with other dysplastic changes is highly specific for hyperplasia or hypoplasia can occur, and there is often a prominent
MDS and, as described earlier, is tightly linked to acquired SF3B1 left shift towards immature forms. Myeloid precursors often display
mutations.88 Ring sideroblasts may be seen in all subtypes of MDS; asynchronous maturation of the nucleus and cytoplasm. In promy-
the specific designation of RARS is made when ring sideroblasts elocytes this can manifest as an early hypergranulation coupled with
comprise at least 15% of the cellularity, blasts are not increased, and reticulated nuclei and a prominent Golgi apparatus, but more mature
dysplasia in other lineages is minimal. myeloid forms are usually hypogranulated and hypolobated. The
In a normal bone marrow, myeloid precursors typically outnumber myeloid series may also be abnormally localized: rather than differ-
erythroid precursors by a ratio of 2–4 to 1. A special case arises in entiating in an organized fashion inwards from the endosteum,
cases of MDS in which this normal ratio is significantly skewed immature cells often cluster centrally in a morphologic process
towards the erythroid lineage, that is, 50% or more of the total cellular- known as abnormal localization of immature precursors (ALIP).
ity is erythroid. If in this case 30% or more of the remaining cellularity Morphologic characterization of the myeloid series is particularly
is comprised of myeloblasts, the case may be diagnosed as erythroleu- important with regard to the blast count, since ≥20% myeloblasts is
kemia (WHO AML, not otherwise specified, subtype acute erythroid characterized as AML using the WHO classification, and ≥5% quali-
leukemia, erythroid/myeloid type; formerly FAB-M6A).15,170 fies as RAEB. Even patients with lower blast percentages, however,

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 957

Fig. 60.3  GRANULOCYTIC DYSPLASIA. Granulocytic dysplasia is most evident in mature neutrophils
and can be contrasted to features of normal forms (far left, top), which are usually still present as a subpopula-
tion of the total cells in most cases. Granulocytic dysplasia is characterized by (left to right, starting at second
column) reduced cytoplasmic granulation, nuclear hypolobation (resulting in the binuclear or single-lobed
pseudo–Pelger-Huët forms), hypersegmentation, ringed forms (“rodent cells”), cells with nuclear twinning,
and cells with excessive nuclear excrescences. Photomicrographs are from a number of cases of refractory
cytopenia with multilineage dysplasia and refractory anemia with excess blasts.

Fig. 60.4  MEGAKARYOCYTIC DYSPLASIA. Dysmegakaryopoiesis is most obvious with the presence of
micromegakaryocytes and abnormal larger forms (panels 2–5). These are compared with a normal megakaryo-
cyte at same magnification (panel 1, far left). Micromegakaryocytes have single, two, or four small nuclei,
which indicate a low-ploidy level. Normal low-ploidy megakaryocytes can be seen in the bone marrow, but
these are immature forms and do not have mature granular cytoplasm with platelet material, as do the
micromegakaryocytes. Larger dysplastic megakaryocytes have multiple, small, widely spaced nuclei. Photomi-
crographs are from a number of cases of refractory anemia with excess blasts and refractory cytopenia with
multilineage dysplasia.

still have an increased risk of developing AML, and some have pro-
posed designating any MDS patient with more than 2% marrow Megakaryocytic Lineage
blasts as having “oligoblastic leukemia.”310 ALIP has been shown to
occur in most cases of RAEB and in about a third of cases in which Thrombocytopenia is present in 25% to 50% of MDS patients at
the blast percentage is less than 5%.311 In these latter cases ALIP has the time of presentation,296,306 and some patients with normal platelet
been shown to be an independent risk factor for subsequent develop- counts can have functional platelet defects.314 Laboratory abnormali-
ment of AML.312 ties include prolonged bleeding times, defective granulation, and
In the peripheral blood, visible morphologic abnormalities include abnormal platelet aggregation indices mediated by hypofunctional
so-called pseudo Pelger-Huët cells, which have condensed chromatin platelet glycoprotein IIb/IIIa, leading to what is known as a
and bilobed nuclei resembling an old-fashioned pince-nez (true Pelger- Glanzmann-type defect.315,316 These defects can occasionally manifest
Huët cells are a benign congenital abnormality seen in children). as spontaneous bleeding, or can be unmasked after trauma or surgery.
Granulocytes may display other nuclear abnormalities as well, includ- Thrombocytosis, by contrast, is relatively unusual in patients with
ing hypersegmentation reminiscent of vitamin B12 deficiency, and MDS, except in those with MDS/MPN overlap syndromes, 5q−
aberrant ring shapes. The hypogranulation visible in the marrow syndrome, or RARS-T.317 JAK2 mutations, in particular, are associ-
typically persists in the peripheral blood, and the left shift typical of ated with thrombocytosis. Thrombocytosis can occasionally be subtle,
MDS marrows is often, though not always, present to some degree especially in patients with advanced disease, in whom a normal
in the blood as well. platelet count may in fact represent a relative thrombocytosis coun-
Despite these quantitative and qualitative defects in leukocytes, teracted by dwindling megakaryocytic reserves.
only a minority of MDS patients have problems with recurrent In the marrow, megakaryocytes are most commonly present in
infections.313 When infections do occur, they tend to be bacterial and normal or increased numbers. A number of morphologic abnormali-
often arise from the lower respiratory tract, skin, and mucous mem- ties can be observed, including abnormally small forms (micromega-
branes. Patients without absolute neutropenia may still develop karyocytes), hypersegmentation, and nuclear hypolobation (Fig.
recurrent infections as a consequence of abnormal neutrophil func- 60.4).318,319 Megakaryocytes may also be abnormally distributed in
tion. Even though only some patients have recurrent infections, an clusters scattered throughout the marrow in MDS, rather than their
infection is the most common cause of MDS-associated death. normal parasinusoidal positioning.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
958 Part VII  Hematologic Malignancies

Lymphoid Lineage TABLE


Diagnostic Criteria for Myelodysplastic Syndrome
60.3
A number of abnormalities in the lymphoid lineage have been
A.  Presence of at Least One Unexplained Cytopenia for at Least 6
described in subsets of patients with MDS. Some patients have been Monthsa
shown to have global decreases in NK cells,226,320 some have deficient
Hemoglobin <11 g/dL, or
receptor localization on B cells,228 and some have abnormal T-cell
responses to mitogenic stimuli.227 Certain types of immune functions, Absolute neutrophil count <1.5 × 109/L, or
however, appear to be preserved, such as antibody-dependent cellular Platelet count <100 × 109/L
cytotoxicity.321 plus B.  Presence of One or More MDS-Qualifying Criteria:
The mechanism by which these abnormalities develop in MDS is >10% dysplasia in one or more hematopoietic lineage, or
unclear. One hypothesis proposes that they could be indirectly reflec-
5%–19% blasts in bone marrow, or
tive of shared somatic defects in a multipotent progenitor. Although
this has not been directly proven, it is theoretically possible given data MDS-defining cytogenetic abnormality, such as:
showing that most driver mutations in MDS patients are present in t(1;3)(p36.3;q21.1) t(2;11)(p21;q23) inv(3)(q21;q26.2)
the long-term hematopoietic stem cells (LT-HSC) compartment and
t(3;21)(q26.2;q22.1) −5 or del(5q) t(6;9)(p23;q34)
could therefore be passed both to myeloid and lymphoid cells.64
Alternatively, a number of immune functions, including both B-cell −7 or del(7q) del(9q)
and T-cell diversity, diminish as a function of normal aging,322,323 and del(11q) t(11;16)(q23;p13.3) del(12p) or t(12p)
in this context, deficiencies of immune function could develop as a −13 or del(13q) i(17q) or del(17p) idic(X)(q13)
consequence of lymphoid cells arising from and interacting with an
plus C.  Exclusion of Alternative Diagnoses
abnormally aged hematopoietic niche.
AML (i.e., <20% blasts, and no t(8;21), inv(16), t(16;16), t(15;17),
or erythroleukemia) or ALL
Other Objective Findings Other hematologic diseases (aplastic anemia, PNH, LGL, lymphoma,
myelofibrosis and other MPN)
Patients with MDS may have a variety of other laboratory abnormali- Viral infections (HIV, EBV, parvovirus)
ties as a consequence of their disease. Ineffective erythropoiesis may
Nutritional deficiencies (iron, copper, B12, folate)
lead to inappropriate iron deposition, resulting in abnormally elevated
ferritin and transferrin saturation.324 Nonspecific markers of cellular Medications (methotrexate, azathioprine, isoniazid, cytotoxic
turnover, such as elevations in lactate dehydrogenase,325 uric acid, chemotherapy)
and phosphate, may also be seen. Reflective of underlying immune Alcohol or other toxins
dysregulation, some patients may have abnormalities of immuno-
Autoimmune diseases (SLE, Felty syndrome, ITP, autoimmune
globulins (Igs), including hypogammaglobulinemia, polyclonal
hemolytic anemia)
hypergammaglobulinemia, and even monoclonal gammopathies.326,327
Whether this latter phenomenon is directly related to MDS or to an Congenital disorders (Diamond-Blackfan anemia, Shwachman-
unrelated, early plasma cell dyscrasia has not been definitively estab- Diamond syndrome, Fanconi anemia, and others)
lished, and the answer may indeed be different in different patients. a
Diagnosis can be made earlier than 6 months if no other cause is apparent for
cytopenias, or there are excess blasts or an MDS-defining cytogenetic
abnormality
ALL, Acute lymphoblastic leukemia; AML, acute myeloid leukemia;
DIAGNOSTIC SYSTEMS AND CLINICAL SYNDROMES EBV, Epstein-Barr virus; HIV, human immunodeficiency virus; ITP, immune
thrombocytopenic purpura; LGL, large granular lymphocyte leukemia;
As discussed throughout this chapter, MDS is heterogeneous in both MDS, myelodysplastic syndrome; MPN, myeloproliferative neoplasm;
morphology and clinical course, a characteristic that requires diag- PNH, paroxysmal nocturnal hemoglobinuria; SLE, systemic lupus
erythematosus.
nostic criteria sufficiently broad to capture the entire spectrum of
disease, and diagnostic criteria are typically left purposefully vague to
allow for inclusion of patients in whom a high suspicion of MDS
exists but who do not specifically meet these formal criteria (Table complexity of the criteria themselves, which can make them cumber-
60.3). Patients with MDS typically have at least one persistent, some for clinicians to use. Second, as described previously, there are
otherwise unexplained cytopenia (hemoglobin less than 11 g/dL, a number of conditions that can mimic aspects of MDS, and the
ANC <1,500/mm3, or platelet count less than 100,000/mm3), plus diagnostic criteria assume these diagnostic possibilities have been
one of the following: (1) ≥5% blasts in the bone marrow, (2) any of eliminated. Third, morphologic criteria are still based around strict,
several chromosomal abnormalities that are found recurrently in arbitrary percentages of dysplasia and blasts, which can be interpreted
MDS; (3) a meaningful degree of dysplasia, usually 10% or more, differently by different pathologists or may be vulnerable to sampling
detectable in at least one cell lineage on bone marrow examination, variations.328,329 Finally, although WHO criteria incorporate cytoge-
or (4) other evidence of clonal hematopoiesis.170 In the past this latter netics to a limited extent, the criteria have not yet been updated to
category has usually comprised either karyotype or FISH results include types of newer genetic results that have been shown to have
demonstrating chromosomal abnormalities missed by conventional diagnostic value, and it is likely that incorporating some of these
karyotype, but evidence of clonal hematopoiesis based on detection features will improve diagnostic clarity. A 2016 revision of the WHO
of recurrent somatic mutations may need to be considered as well. criteria eliminated the term “refractory anemia” so that, for example,
However, given that at least 10% of the population aged 70 or older RAEB-1 is now known as “MDS with excess blasts” (MDS-EB). This
harbor point mutations in genes associated with myeloid malignan- revision also noted that MDS with ring sideroblasts (MDS-RS) can
cies,31,32 detection of such a mutation in the absence of other diag- be diagnosed with 5% to 14% ring sideroblasts if a somatic SF3B1
nostic criteria cannot be considered equivalent to MDS. mutation is present; if SF3B1 is wild-type, at least 15% ring sidero-
As described elsewhere, the 2008 WHO criteria divide adult MDS blasts are needed.
into six general categories: RCUD (including RA, RARS, refractory
neutropenia, and refractory thrombocytopenia), RCMD, RAEB-1,
RAEB-2, MDS with isolated del(5q), and MDS-U. Despite the detail IPSS and IPSS-R
of the criteria, they still have a number of limitations, a fact that is
probably inescapable when attempting to categorize a disease with so Although the WHO classification system (and the FAB system before
many disparate manifestations. The most obvious limitation is the it) is the accepted standard for diagnosing MDS, it does not confer

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 959

TABLE 1997 International Prognostic Scoring System for TABLE 2012 Revised International Prognostic Scoring
60.4 Myelodysplastic Syndromes (IPSS) 60.6 System for Myelodysplastic Syndrome (IPSS-R)
Score Cytogenetic Included Karyotypic Abnormalities
Risk
Variable 0 0.5 1 1.5 Very good del(11q), −Y
Marrow blasts (%) <5 5–10 – 11–20 Good Normal, del(20q), del(5q) alone or +1 other
Karyotype Good Intermediate Poor – abnormality, del(12p)
Cytopenias 0–1 2–3 – – Intermediate +8, del(7q), i(17q), +19, +21
Any other single or double abnormality
Two or more independent clones
Poor der(3q), −7, double with del(7q), complex with
TABLE Survival Based on International Prognostic Scoring exactly 3 abnormalities
60.5 System for Myelodysplastic Syndromes (Percent)
Very poor Complex with >3 abnormalities
IPSS Risk Group # Patients 2 Years 5 Years 10 Years 15 Years Scoring Table
Low 267 (33%) 85 55 28 20 Parameter Category/Score
Cytogenetic Very good Good Intermediate Poor Very poor
Intermediate-1 314 (38% 70 35 17 12
risk 0 1 2 3 4
Intermediate-2 179 (22%) 30 8 0 –
Marrow blasts ≤2 3–4 5–10 >10
High 56 (7%) 5 0 – – (%) 0 1 2 3
Hemoglobin ≥10 8–9.9 <8
(g/dL) 0 1 1.5

much specific prognostic information, and other systems have been Platelet count ≥100 50–99 <50
developed expressly for this purpose. The most widely used has been (× 109/L) 0 0.5 1
the IPSS330 and its newer revised iteration, the IPSS-R.331 The original Neutrophil ≥0.8 <0.8
IPSS was first published in 1997 and was derived by analyzing count (× 0 0.5
baseline characteristics in over 800 newly diagnosed MDS patients 109/L)
from all diagnostic categories. Patients were scored based on three IPSS-R Risk Total % of Median 25% With
characteristics: the percentage of blasts in the marrow, the presence Group Score Patients Survival AML (Years)
of specific cytogenetic abnormalities, and the number of cytopenias (Years)
in the blood. They were then stratified based on total score into four Very low ≤1.5 19 8.8 NR
risk groups (low, intermediate-1, intermediate-2, and high) that were Low 2–3 38 5.3 10.8
shown to have prognostic value, both in estimating the chance of
progressing to AML and in estimating the duration of overall survival. Intermediate 3.5–4.5 20 3 3.2
This is shown in Table 60.4 and Table 60.5. The original IPSS High 5–6 13 1.6 1.4
excluded patients with t-MDS, patients with proliferative CMML, Very high >6 10 0.8 0.73
and patients who ultimately underwent allogeneic stem cell trans-
AML, Acute myeloid leukemia; NR, not reached.
plantation or other disease-modifying therapy.
The original IPSS had several limitations that ultimately neces-
sitated its revision. First, it was formulated 4 years before the adoption
of the first iteration of WHO classification criteria, and some of
changes in those criteria (for instance, changing the cutoff for AML and the serum ferritin, that have been shown to have independent
from 30% blasts to 20% blasts) effectively invalidated certain aspects prognostic value in dedicated studies. It does not capture the kinetics
of the IPSS. Furthermore, broad application of the criteria to larger of disease or comorbid conditions. In addition, similar to the WHO
populations of MDS patients began to reveal that the original IPSS classification criteria, IPSS-R does not include any molecular data
gave insufficient weight to some important clinical features, particu- other than cytogenetics.
larly the presence of severe cytopenias. The IPSS was only validated
in de novo disease, not t-MDS, and only in patients treated with
supportive care alone. In response to some of these evident limita- Other Risk Stratification Systems
tions, several alternative prognostic systems have been proposed over
the last decade, but none of them have been widely adopted.332–334 The limitations of the IPSS prompted attempts at more refined risk
The IPSS-R (Table 60.6), which was introduced in 2012, revised stratification using alternative models. Few of these have gained much
the MDS blast cutoff to 20% and assigned a point for each individual traction in clinical practice but retain value for use in clinical trials
cytopenia present at the time of diagnosis, stratifying on the basis of and retrospective studies. For instance, a need for better stratification
severity. Furthermore, it incorporated a broader range of cytogenetic of lower-risk patients (i.e., IPSS-low or Int-1) led to the creation of
abnormalities than the original IPSS, and stratified patients into five the Lower-Risk Prognostic Scoring System (LR-PSS) by the MD
risk categories instead of four. The resulting system thus incorporates Anderson group.332 The LR-PSS uses criteria similar to the IPSS but
more informative biology and offers better refinement of prognosis sets lower thresholds for point assignment (e.g., 2 points for a platelet
than its predecessor.335,336 count <50 × 109/L and 1 point for bone marrow blasts ≥4%). Sub-
The median survival times estimated by the IPSS and IPSS-R are sequent studies have confirmed the ability of LR-PSS to risk-stratify
largely reflective of natural history since they were originally derived patients, with those in the highest risk group having the greatest risk
in patients who did not receive any therapy for their MDS (Table of developing AML, and the poorest overall survival,339 and addition
60.5). However, IPSS-R stratifications have been shown to retain of genetic sequencing has shown that in these low-risk patients,
relevance when applied to treated populations, including risk strati- EZH2 mutations, though rare, portend a poor prognosis.112 Other
fication of patients receiving hypomethylating agents337 and those attempts to improve prognostic systems have included attempts to
who undergo allogeneic stem cell transplantation.338 incorporate formal measures of comorbidity into the IPSS,340 and an
Nevertheless, the IPSS-R still has prognostic limitations. It does attempt to combine the WHO classification system with the IPSS
not incorporate laboratory findings, such as lactate dehydrogenase that has been dubbed the WPSS.341 This latter system was formulated

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
960 Part VII  Hematologic Malignancies

before the introduction of the IPSS-R, but subsequent studies have typically remain transfusion dependent, and management of iron
suggested that the WPSS and the IPSS-R are about equal in their overload then becomes increasingly important.
accuracy of predicting prognosis.342 The designation of 5q− syndrome applies only to patients in
whom loss of 5q is the only cytogenetic abnormality. Patients in
whom 5q− is only one of several cytogenetic aberrations in fact tend
Specific Clinical Syndromes to have a worse prognosis than average, with a more rapid progression
to AML. As referenced previously, loss of 5q often occurs in tandem
Although MDS is a heterogeneous disorder, it contains a number of with TP53 mutations (or loss of 17p), another poor-prognosis
specific entities, either defined by cytogenetics, pathologic findings, combination.149
or clinical features, which possess distinctive clinical features or bio-
logic behaviors. Some of these are discussed in more detail later.
Hypocellular Syndromes
The 5q− Syndrome Most patients with MDS have hypercellular or normocellular
marrows. Hypocellular marrows, in contrast, are found in fewer
Patients with isolated loss of the long arm of chromosome 5 [so-called than 15% of patients, and delineate the entity of hypoplastic MDS
del(5q) or 5q minus syndrome] are a unique group whose biology is (Fig. 60.5D–E).224 These patients have substantial overlap, both
described in more detail earlier. Clinically, 5q− syndrome is character- morphologically and clinically, with other hypoplastic syndromes,
ized by a predominant anemia with preservation or even elevation of including aplastic anemia,345 paroxysmal nocturnal hemoglobinuria
platelet counts, striking pathologic feature is the presence of mono- (PNH),346 and T-cell LGL.249,347 In fact, distinction between these
nuclear micromegakaryocytes identified in the bone marrow biopsy entities can sometimes be difficult, as patients with MDS may
(Fig. 60.5A–C) and an indolent course with progression to AML in occasionally have PNH or LGL clones detectable by flow cytometry,
fewer than 25% of cases.343,344 For unclear reasons, it is more common though these are usual small and transient. Some of the resemblance
in women, who comprise 60% to 70% of cases. The anemia in lower may in fact reflect a shared pathogenesis; for instance, studies have
risk del5q MDS is usually very responsive to the initiation of lenalido- shown that a sizable minority of aplastic anemia cases harbor clonal
mide,173 which is discussed in more detail in the subsequent section somatic mutations in genes recurrently mutated in MDS, including
on Treatment. Those patients who become refractory to lenalidomide ASXL1 and DNMT3A,348 and that these patients have an inferior

A B C

D E F G
Fig. 60.5  SPECIFIC MYELODYSPLASTIC SYNDROMES. The 5q− syndrome (A–C); hypocellular
myelodysplastic syndrome (D and E), and myelodysplastic syndrome with fibrosis (F and G). The 5q− syn-
drome has specific morphologic correlates. There is a macrocytic anemia (A) and a cellular bone marrow
characterized by increased small monolobated megakaryocytes (B and C). The megakaryocyte nuclei have little
segmentation and are fairly round. Although some true micromegakaryocytes may be present (C, inset, same
magnification), the typical monolobated forms are not as tiny as the micromegakaryocytes. Hypocellular
myelodysplastic syndrome (D and E) can present a diagnostic problem and can be difficult to differentiate
from aplastic anemia. Dysplasia may be difficult to evaluate if the smears are paucicellular. The finding of
dysplastic megakaryocytes (note small widely separated nuclei, E) on the biopsy sample can be helpful.
Myelodysplastic syndrome with fibrosis (F) can be difficult to differentiate from myeloproliferative neoplasms
(MPNs). However, the lack of large megakaryocytes, which typically are see in the MPNs along with presence
of dysplasia in the circulating neutrophils (G) are useful clues to the correct diagnosis.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 961

prognosis compared to patients with mutations in PIGA and BCOR.349 As discussed elsewhere in this chapter, the mechanism of patho-
It is therefore possible that these studies are defining distinct disease genesis for t-MDS has long been thought to involve the acquisition
processes within a homogeneous-appearing morphology, with ASXL1 of severe, large-scale chromosomal damage, a theory that is circum-
and DNMT3A mutations defining disease more similar to MDS, and stantially supported by the mechanism of alkylators (which induce
BCOR and PIGA mutations defining disease more akin to true, double-stranded DNA breaks) and the fact that many patients with
immune-mediated aplasia. At the same time, finding somatic muta- t-MDS have complex karyotypes. On the other hand, it has previ-
tions characteristic of the alternative diagnoses (for instance, members ously been observed that some patients with t-MDS have point
of the telomerase family in aplastic anemia,40 or STAT3 mutations in mutations in TP53, which would not be expected to directly arise
LGL350) may clarify the picture as well. from alkylator-induced damage,360 but which can lead to the acquisi-
tion of chromosomal rearrangements even in the absence of chemo-
therapy. Recent deep studies of a small group of patients with
Myelodysplastic Syndrome/Myeloproliferative therapy-related AML harboring TP53 mutations have shown that
Neoplasm Overlap Syndromes clones harboring the mutations preceded the development of leuke-
mia by years, and in some cases could be detected before the original
Although previously included as a subtype of MDS, disorders with initiation of chemotherapy.150 This recapitulates older data showing
overlapping features of both MDS and MPN are no longer included that cytogenetic abnormalities present in the bone marrow of patients
in the WHO classification system and are now considered an entity who developed therapy-related leukemia after autologous transplant
unto themselves; they are discussed in more detail elsewhere. for lymphoma could be found in the stimulated autologous speci-
mens, which had been banked years before the development of
t-AML.365 These findings suggest an alternate method of t-MDS
Myelodysplastic Syndrome With Fibrosis pathogenesis, in which rare clones harbor preexisting mutations that
either confer a selective growth advantage during marrow reconstitu-
Marked fibrosis rarely occurs in cases of MDS (Fig. 60.5F–G) without tion, tolerance of DNA damage that would otherwise trigger apop-
myeloproliferative features, but when it does occur, it can be difficult tosis, or both, thus promoting clonal selection and expansion upon
to distinguish these cases from MPN.351 Evaluation for somatic muta- exposure to chemotherapy.
tions may be helpful, since finding a JAK2, CALR, or MPL mutations
is more common in pure MPNs than in MDS,352 while certain other
mutations, including TET2 and SRSF2, are somewhat enriched in Refractory Anemia With Ringed
MDS-MPN overlap syndromes.101,353 On the other hand, extensive Sideroblasts and Thrombocytosis
dysplasia is more suggestive of MDS. In addition, a few genetic
lesions appear to be more specific for pure MDS syndromes, includ- Although thrombocytopenia is the most common platelet abnormal-
ing SF3B1 mutations and complex cytogenetics. ity in MDS, rare patients present with marked thrombocytosis.317
Patients with overlapping MDS and fibrosis often have severe, This most frequently occurs in the setting of RARS, and in the second
progressive cytopenias with evidence of myelophthisis on peripheral iteration of the WHO criteria, these patients are classified as having
smear, but the splenomegaly associated with myelofibrosis in an RARS-T.366 On bone marrow examination, there are often features
MPN background is less common. Nevertheless, the prognosis for of both MDS, such as frequent ringed sideroblasts, and MPNs,
these patients appears to be relatively poor. such as megakaryocytic hyperplasia. Molecularly, patients often have
concomitant SF3B1 mutations, which drive the ring sideroblast
morphology,366 and JAK2 mutations, which drive the thrombocytosis
Therapy-Related Myelodysplastic Syndrome and are otherwise uncommon in MDS.367 Patients meeting criteria
for RARS-T are relatively uncommon and prognostic information
t-MDS is a well-recognized and feared consequence of cytotoxic is thus limited, but one small series of patients had better 5-year
chemotherapy for other cancers, but the overall incidence is difficult survival than matched patients with RARS, perhaps because the
to estimate.243,354 This is partly because of the fact that it is not usually proliferative drive of the JAK2 mutation helps preserve blood cell
reported as a distinct entity, and in part because it can be difficult to counts.368
establish causality in all patients who develop MDS following treat-
ment for a prior cancer. t-MDS has been best characterized in patients
with a prior history of breast cancer,52,355,356 lymphoma,357,358 and TREATMENT OF PATIENTS WITH  
myeloma,359 where the overall incidence is usually reported at around MYELODYSPLASTIC SYNDROMES
1%. On the other hand, t-MDS is rare following treatment for other
types of tumors, such as gastrointestinal and genitourinary cancers. Treating patients with MDS presents a number of challenges. First,
This difference is largely attributed to differences in the types and MDS patients are often elderly and thus frequently have serious
intensities of chemotherapeutic agents used to treat different tumor comorbid conditions and poor performance status. Second, the
types. In particular, high doses of alkylating agents, such as cyclo- protean nature of MDS, which is particularly heterogeneous even
phosphamide, ifosfamide, melphalan, and busulfan, have been associ- compared with other cancers, means that treatments appropriate for
ated with classic t-MDS,360 as has extensive radiation therapy.361 A some patients may be unhelpful for others. Third, a number of
distinct, well-characterized class of therapy-related myeloid malignan- biologic factors, most importantly the disease’s origin in a quiescent
cies occur after treatment with topoisomerase inhibitors but largely stem cell, make MDS highly refractory to most conventional treat-
consists of AML without an antecedent MDS phase.54,205,362 ments like intensive cytotoxic chemotherapy (which many MDS
t-MDS has a relatively distinct clinical behavior. It is typically patients are not healthy enough to receive anyway).
characterized by a latency of years and recurrent large-scale chromo- In addition, although the biology of MDS is increasingly well
somal abnormalities, especially of chromosomes 5 and 7 or 11q23. understood, most of the common derangements, like mutations in
Complex karyotypes are also common.363 These karyotypic abnor- splicing factors and epigenetic regulators, have wide-ranging, pleio-
malities, which occur in only about 10% to 15% of patients with de tropic effects that render narrowly targeted therapies infeasible. While
novo MDS, have a frequency of around 50% to 70% in t-MDS. some agents like hypomethylators and histone deacetylase inhibitors
They tend to have a more aggressive clinical course than patients with are matched to biologic processes that are frequently deranged in
de novo disease, with more frequent progression to acute leukemia. MDS, their usual clinical impact is modest at best. Allogeneic stem
They also tend to respond poorly to all classes of treatment, and even cell transplantation remains the only curative therapy for MDS, but
for those patients who undergo allogeneic stem cell transplantation, is unavailable to many because of age and comorbidities, and even
relapse is not infrequent.364 for those who undergo it, relapse is not uncommon.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
962 Part VII  Hematologic Malignancies

Despite these challenges, all patients with MDS should be offered 8–9 mg/dL or the patient has symptoms referable to anemia. More
some form of therapy. For elderly patients or those with serious restrictive transfusion strategies, such as those suggested by studies in
comorbid conditions who are too unwell to undergo active treatment, inpatient or critical care settings,374,375 may be appropriate in younger,
this may consist primarily of supportive care and possibly hospice or healthy patients, while some older patients, particularly those with
palliative care service referral. Other patients with lower-risk disease significant comorbid cardiac disease, may benefit from more liberal
may benefit from a period of observation, or judicious use of transfu- strategies targeting a hemoglobin of 9–10 mg/dL.376,377 Unless the
sion and growth factor support, balanced in carefully selected cases patient is an HSCT candidate (in which case irradiated blood is
with chelation therapy to prevent or relieve iron overload. Patients essential), there is no consensus about leukocyte depletion or irradia-
with higher-risk disease may derive benefit from treatment with tion of the product.378 Since many patients with MDS will receive
hypomethylating agents or other conventional therapies, and those hundreds of units of PRBCs over the course of their disease, preven-
patients with high-risk disease who meet other selection criteria tion and appropriate management of iron overload is also of substan-
should be referred for consideration of hematopoietic stem cell tial importance.379,380
transplantation (HSCT). Finally, patients should whenever possible
be encouraged to participate in clinical trials to the extent that this
remains consistent with their personal goals of care. Erythropoiesis-Stimulating Agents
Once the decision to pursue active treatment has been made, there
are at least three major points to consider. One is the choice of initial Transfusion dependency is a negative predictor of outcome in
treatment modality, which is guided in large part by risk stratification. MDS.381 While transfusion requirement may to some extent may be
This should typically start by computation of the IPSS-R, but IPSS-R a marker of more severe hematopoietic insufficiency and worse
score should not be the sole consideration in choosing therapy; for disease not captured by other prognostic markers, the poor outcomes
instance, a very young patient with otherwise lower-risk MDS that may also be a reflection of the adverse effects of repetitive transfusion,
has evolved quickly may still warrant consideration of a stem cell including accumulation of toxic iron species, immune modulation,
transplant, and an elderly patient with very high risk disease may and a risk of infection.382 Given these facts, the administration of
nevertheless opt not to pursue treatment with a hypomethylating ESAs has been heavily investigated in MDS in an attempt to spare
agent after a thorough discussion of the risks and potential benefits. patients from unnecessary transfusions.
Second, there are a number of special cases for which there is no clear More than 20 studies of ESAs have been conducted in MDS,
consensus about the single best treatment strategy; these include and 20% to 50% of patients experience meaningful, sustained
patients with lower-risk disease who have a dominant cytopenia other improvements in their hemoglobin level in response to ESA
than anemia, anemic patients with lower-risk disease who lack del(5q) supplementation.383–388 Predictors of response include lower pretreat-
but also have an elevated serum EPO, and patients with high-risk ment serum EPO levels (<500 U/L, but especially <100 U/L),389
disease who have progressed on a hypomethylating agent but are not lower IPSS scores,390 normal blast counts, normal or low-risk cytoge-
candidates for transplant. netics,391 and lower serum and marrow levels of inflammatory cyto-
Finally, since therapies other than allogeneic HSCT are rarely if kines.392 The serum EPO level should be evaluated in the context of
ever curative, defining what constitutes a meaningful response can be the degree of anemia; a trial of an ESA is usually reasonable in patients
a nontrivial endeavor. Objective criteria for response were proposed with EPO levels up to 200–300 U/L, which, while technically elevated
by an International Working Group (IWG) in 2000 and revised in on most laboratory reference scales, is still lower than would be
2006, and include hemoglobin, number of metaphases with abnormal predicted for most MDS patients’ degree of anemia. Patients with
karyotypes, and percentage of the marrow involved by blasts.369 EPO levels over 500 U/L, on the other hand, are unlikely to respond
However, improvements in these objective variables may not always to any dose or duration of ESA.
coincide with the ways patients perceive changes in their quality of There are two major formulations of ESA available in the United
life370,371 or MDS-related symptoms,279 and these perceptions may be States. Epoetin alfa, the shorter-acting of the two, can either be given
more likely to impact patients’ decisions to continue or discontinue at a dose of 150–300 U/kg three times weekly, or in a fixed dose of
active therapy for their disease. 40,000 to 60,000 units once weekly.388 Higher doses above 60,000 U/
weekly have not proven to be more effective.393 Darbepoetin alfa is
longer acting and is typically given in doses of 500 µg once every
Lower-Risk Disease 3 weeks.394 Retrospective comparisons of the two agents have not
shown a significant difference in efficacy.395 Other ESA formulations
Many patients with lower-risk MDS do not require active treatment are available outside the Unites States, such as epoetin zeta and bio-
directed at the underlying disease process, but this does not mean equivalent ESAs. Although there is some increase in response with
they require no treatment at all. In fact, providing appropriate sup- longer duration of therapy,396 in practical terms, a trial of 12 weeks
portive care is an important undertaking that can at times be complex is reasonable. Before starting therapy, other causes of anemia should
and time-consuming, and requires a detailed understanding of the be ruled out. In particular, response can be suboptimal if patients are
evidence behind the available supportive measures.372 absolutely or even relatively iron deficient, and oral or parenteral iron
supplementation may be reasonable in patients with ferritin levels at
the lower end of the normal range.
Management of Anemia One limitation of the numerous studies of ESAs in MDS patients
is the lack of a consistent definition of response, since most studies
Because most MDS patients become anemic at some point during predated uniform IWG criteria. Some studies define this based on a
their disease, appropriate management of their anemia is of critical hemoglobin increase (usually improvement by 1–2 g/dL),397,398 while
importance.373 Management can be broken down into two main others have defined response by transfusion reduction or quality of
components: transfusion support and use of erythropoiesis-stimulating life metrics.399 In practice, any of these improvements could be a
agents (ESAs). reasonable criterion for continuation of therapy. Of particular inter-
est, supplementation with ESA has not been shown to increase the
rate of progression to acute leukemia, and one comparative trial
Transfusion Support showed a nonsignificant trend toward a decreased risk of leukemia,
although the biologic basis for this is not clear.400 In addition, several
Many, if not most, patients with MDS will require packed red blood studies have shown an improvement in response in patients supple-
cell (PRBC) transfusion during the course of their disease. To mini- mented with a combination of ESA and G-CSF, especially in RARS,
mize the risks associated with repetitive transfusion, it is usually likely caused by pleiotropic effects of both growth factors on hema-
appropriate to reserve transfusion until the hemoglobin is below topoietic progenitors.399,401

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 963

ESAs are no longer routinely used for management of anemia in who show a predilection for bleeding. In general, patients should be
solid tumor patients, where studies have shown them to be associated transfused prophylactically rather than waiting for bleeding to occur.
with an increased risk of poor outcomes.402 Similar findings have also Although there are few large studies comparing transfusion strategies
been noted in patients without cancer receiving ESAs for anemia specifically for MDS patients, a recent study of patients with other
associated with renal failure.403 Such a risk has not consistently been hematologic malignancies showed that for most patients (including
shown in MDS, although most studies published to date have not those with AML), a prophylactic strategy using a trigger of 10 × 109
had long follow-up periods. The risk of thromboembolism has largely cells/L led to fewer significant hemorrhages than a therapeutic strategy
been correlated with the degree of improvement in hemoglobin, with in which patients were transfused only when bleeding.417
patients targeted to hemoglobins over 12 mg/dL at greatest risk. Thrombopoietin (TPO) analogs, which are standard therapeutic
Given this, the consensus recommendation in MDS is to initiate an options for refractory chronic idiopathic thrombocytopenia purpura
ESA at hemoglobin <10 g/dL and target a level between 11–12 g/ (ITP),418 are not yet approved by any regulatory agencies for use in
dL.404 MDS. The two major formulations approved in ITP are romiplostim,
a “peptibody” consisting of 14 amino acid peptides that bind the
extracytoplasmic domain of the TPO receptor fused to an IgG1 heavy
Management of Neutropenia and Infections chain, and eltrombopag, a small molecule without TPO homology
that nevertheless binds and activates the TPO receptor.419 Romiplos-
As referenced previously, patients with MDS often become neutro- tim is administered as intermittent subcutaneous injections, whereas
penic but also appear to have other qualitative immune defects that eltrombopag is an oral formulation that is taken daily.
are not accurately represented by the ANC,392,393 and infection is the Both agents have shown efficacy in terms of reducing platelet
leading cause of death in MDS.280 Despite this, supplementation transfusions and clinically significant bleeding events and increasing
with either G-CSF (filgrastim, tbo-filgrastim and others) or GM-CSF platelet counts in early-phase trials in MDS.420–424 In patients with
(sargramostim) has not consistently been shown to improve outcomes IPSS low or intermediate-1 risk MDS who had platelet counts less
in MDS patients.405–407 Nevertheless, it is reasonable to consider a than 50 × 109/L, romiplostim consistently led to significant improve-
trial of growth factor (usually G-CSF, because of less frequent adverse ments in platelet count, which were durable in about half of
effects) in lower-risk IPSS patients with significant neutropenia, typi- patients.421 A similarly designed phase I/II trial of eltrombopag is
cally defined as an ANC <1000/mm3. Supplementation in higher-risk ongoing.
patients, particularly those with any degree of excess blasts, is not However, concerns have been raised about the safety of TPO
usually recommended in the absence of chemotherapy, because of agonists in patients with MDS. First, a number of studies in ITP
lingering concerns about promoting expansion of leukemic clones.408 have shown an increase in marrow reticulin fibrosis in subsets of
Other therapeutic strategies for immune supplementation, including patients.425 While the absolute risk of fibrosis has not been deter-
granulocyte transfusion and cytokine administration, have not been mined, and fibrosis has been moderate in most patients, some have
shown to be helpful.409 speculated that the process could be accelerated within the abnormal
A thorough understanding of appropriate antibiotic use is an marrow microenvironment of MDS patients.426 Second, there is
important component in the care of MDS patients. Patients should concern about increased blast proliferation in the presence of TPO
be counseled to seek medical attention for any fever greater than agonists, since some blasts have functional TPO receptors. A random-
100.4°F/38.4°C, and those who are neutropenic should be hospital- ized controlled trial of romiplostim versus placebo (the only phase
ized and started on antibiotics while an appropriate search for infec- III study of either agent in MDS to date) was stopped early by the
tion is undertaken. A thorough discussion of treating febrile study’s safety monitoring committee because of interim analysis sug-
neutropenia can be found elsewhere in the text, but should begin gesting a higher rate of progression to AML in the romiplostim
with an antibiotic that covers a broad spectrum of gram-negative arm.427 At the time the study was stopped, 10 of 167 patients in the
organisms including Pseudomonas species.410 Although prophylactic romiplostim group had progressed to AML, compared to two of 83
antibiotics should not be started routinely, they may be appropriate in the placebo group. Since the study was far from completion of
for selected patients who demonstrate a pattern of recurrent infec- accrual at the time it was stopped, the risk of disease progression
tions.411,412 There is less consensus about prophylactic antivirals or could not be definitively assessed and the AML progression rate
antifungal medications, and the efficacy of the latter depends on local eventually nearly equalized; nevertheless, the manufacturer has now
microbiologic isolates and epidemiologic patterns. added a warning of risk of MDS progression to romiplostim’s label.
Until there is more conclusive evidence evaluating the link between
TPO analogs and the risk of disease progression, neither romiplostim
Management of Thrombocytopenia and Bleeding or eltrombopag can be recommended for routine treatment of MDS-
associated thrombocytopenia outside a clinical trial, although patients
Modest thrombocytopenia is common across many subtypes of MDS, with refractory bleeding who fail to respond to platelet transfusions
while severe depression of platelet counts is more frequently seen in could consider trying one of these agents palliatively.
patients with higher-risk IPSS scores or those undergoing active
treatment with hypomethylating agents or chemotherapy. Overall,
bleeding is the second most common cause of nonleukemic death in Management of Iron Overload  
MDS patients.413 Conventional wisdom and observational studies (Transfusional Hemosiderosis)
indicate that the risk of bleeding with trauma begins to increase at a
platelet count near 50 × 109 cells/L and that of spontaneous hemor- Although it is clear that MDS patients who undergo repeated blood
rhage does not increase significantly until the count is below 10–20 transfusions carry an increased risk of developing iron overload,381
× 109 cells/L. However, these cutoffs may underestimate the bleeding the importance of this relative to other disease-related risks, and how
risk in MDS patients, many of whom have qualitative defects in to assess and respond to it, remains a matter of some debate.382 MDS
platelet function that are not captured by the platelet count. A number is distinct from pediatric illnesses associated with iron overload,
of retrospective studies have evaluated the significance of thrombo- in which the benefit of chelation has been well established,428 in
cytopenia in MDS, all of which have found that thrombocytopenia that many MDS patients will not live long enough for the clinical
confers a poor prognosis caused by both the risk of bleeding and, in impact of end-organ iron deposition to be realized. Registry data
some cases, an elevated risk of transformation to AML.330,413–415 suggest that patients with higher transfusion requirements have a
As with red blood cells, management of thrombocytopenia can greater risk of complications,429,430 but registry data cannot prove the
either involve transfusions or use of growth factors. Many institutions direction of the relationship—that is, whether the transfusions lead
use a standard transfusion threshold of 10 × 109 cells/L,416 but as to complications or whether the need for transfusion is simply a
discussed earlier, a higher goal may be appropriate in selected patients marker of more severe disease or underlying comorbidities. Other

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
964 Part VII  Hematologic Malignancies

retrospective studies have provided circumstantial evidence for delete- inflammatory state, and is an imperfect marker of total body iron
rious effects of iron overload. For instance, one study has shown a burden. For quantitative assessment of hepatic iron overload, T2*-
higher rate of death from heart failure among MDS patients,431 and weighted magnetic resonance imaging of the liver has supplanted
another has shown increased rates of heart and liver failure among biopsy as the preferred method of validation.440
MDS patients with high lifetime numbers of transfusions,432 but
these observations again cannot prove causality.
There are two major formulations of chelating agent available in Other Therapies for Lower-Risk Disease
the United States, deferoxamine and deferasirox (a third agent,
deferiprone/L1, is available only for thalassemia patients via a The literature is replete with attempts to treat lower-risk MDS with
restricted distribution program). Deferoxamine is administered as a a variety of different drugs, especially so-called differentiating agents
continuous subcutaneous or IV infusion, a cumbersome mechanism such as retinoic acid or arsenic trioxide (ATO). With the exception
that has limited its applicability in widespread practice. Deferasirox, of lenalidomide for 5q− syndrome, most of these have been incon-
an oral formulation, is more convenient so is much more widely used, sistently effective at best.
but is quite costly.
Deferasirox is typically started at 20 mg/kg (14 mg/kg for the new
tablet form of deferasirox FDA approved in 2015) once daily and Lenalidomide
escalated to 30–40 mg/kg/day (28 mg/kg for the new tablet form of
deferasirox FDA approved in 2015), depending on iron kinetics and As described previously, lenalidomide is a derivative of thalidomide
patient tolerance. Although deferasirox has been shown to rapidly that is uniquely effective in reversing the severe anemia associated
mobilize stored iron and reduce labile plasma iron species, it is fre- with isolated del(5q). This was previously thought to be related to
quently discontinued either as a result of disease progression or caused poorly characterized immunomodulatory effects (as implied by
by side effects, which include gastrointestinal distress, renal impair- “Imid,” the name given to the class of thalidomide derivatives).441
ment, and rash. However, more recent work has shown that lenalidomide actually
To date, there have been no randomized controlled trials to exerts its effect in 5q− syndrome by mediating the binding of the E3
definitively evaluate the impact of iron chelation on outcomes in ubiquitin ligase cereblon to casein kinase 1α, thus triggering casein
MDS. The largest prospective trial to date, a single-arm cohort study kinase’s proteasomal degradation.442 Casein kinase 1α is an essential
of 1744 patients treated with deferasirox, included a prespecified kinase whose gene, CSNK1A1, lies within the minimally deleted
MDS subgroup of 341 patients. Over the study’s single year, median region on 5q.177 In normal cells, treatment with lenalidomide is
ferritin levels decreased significantly compared to baseline regardless insufficient to deplete casein kinase 1α to lethal levels, but the hap-
of whether patients were chelation naive or not, as did the median loinsufficiency of CSNK1A1 induced by loss of 5q renders 5q− cells
alanine aminotransferase, which was tracked as a marker of hepatic sensitive to the drug through a p53-dependent mechanism and allows
toxicity from iron overload. On the other hand, 48.7% of the MDS for repopulation of the marrow by wildtype hematopoietic progeni-
patients discontinued treatment because of side effects. Since there tors. This activity appears to be unique to lenalidomide and is not
was no control arm and the study period was short, no conclusions shared, for instance, by thalidomide. Unfortunately, the effect is not
could be drawn about the impact of chelation on more meaningful permanent and resistance eventually develops in most cases, often
clinical outcomes. Other studies have shown improvements in through inactivating mutations in TP53.443 Hopefully, however, these
hematopoiesis in patients started on chelation therapy,433,434 but these new insights will enable further studies of how these resistance
observations were not tied to longer-term outcomes either. mechanisms arise.
There are data from retrospective studies showing better out- Lenalidomide’s efficacy was first observed in MDS-001, a phase I
comes for patients who undergo iron chelation, but all of these study of 32 patients with symptomatic anemia who had not responded
are subject to patient selection bias. In one, a Canadian study of to ESAs.444 Twenty of the 32 patients became independent of transfu-
178 patients, only 18 received chelation therapy, suggesting that sion for some period of time, including 10 of 12 patents with del(5q).
these patients were carefully selected and destined to have a better Of the responders, those with del(5q) had a significantly longer
outcome anyway.435 In another, a French study of 97 patients in duration of response. This prompted further study of lenalidomide
whom 46% received chelation, the regimen and duration of chelation specifically in del(5q) patients in MDS-003, a phase II study in which
therapy varied considerably, and no baseline assessment of iron stores 76% of 148 enrolled patients had an improvement in anemia and
was performed.436 A recent metaanalysis of iron chelation studies 67% achieved transfusion independence, with a median improve-
in MDS found that use of chelation therapy was associated with ment in hemoglobin near 5 mg/dL. A second phase II trial, MDS-
an increase in length of survival (overall 61.2 months longer for 002, showed that lenalidomide was also effective in a substantially
patients receiving chelation),437 but since the analysis included only smaller subset of patients without del(5q), with an overall response
retrospective studies, it is very possible that these numbers are again rate of 43% and a transfusion independence rate of 26%. The factors
largely reflective of selection bias, with healthier or younger patients that determine lenalidomide responsiveness in non-del(5q) MDS
more likely to receive chelation therapy than those who were older or have not been determined.
sicker. The most conclusive evidence of lenalidomide’s efficacy in del(5q)
Despite the limitations of the available data, there are several comes from the phase III MDS-004 trial,173 a randomized, double-
published consensus opinion guidelines regarding the use of chelation blinded study in which patients were randomized 1 : 1:1 to receive
therapy in MDS.324,438 Although they differ with respect to their lenalidomide 10 mg/day on days 1–21 of a 28-day-cycle, lenalido-
specific recommendations, most operate on similar principles that the mide 5 mg/day on days 1–28, or placebo on days 1–28. Those
patients best suited to iron chelation therapy are either those with without an erythroid response at 16 weeks were unblinded and
lower-risk disease and significant anemia who presumably face a long became eligible for open-label treatment. At the conclusion of the
period of transfusion dependency, or those who could potentially be study, significantly more patients in both lenalidomide groups had
eligible for stem cell transplantation, since in the transplant setting achieved transfusion independence for at least 26 weeks than those
numerous studies have shown that a high ferritin is associated with in the placebo group (56% in the 10-mg group versus 43% in the
inferior outcomes.439 A third category of patients comprises those 5-mg group versus 5.9% in the placebo group), with those in the
with suspected end-organ damage as a result of iron overload. 10-mg group having a longer median duration of response (82.9
Some of the guidelines recommend waiting to start chelation weeks) than those in the 5-mg group (41.3 weeks). Intriguingly,
therapy until the ferritin is greater than 1000 µg/L or the patient has treatment with lenalidomide also appeared to lower the risk of
had more than 20–30 lifetime transfusions, but they also note that transformation to AML; at a median follow up of around 3 years,
neither of these criteria is supported by high-level evidence. Serum 36.4% of patients who had received placebo only and 30.4% who
ferritin is problematic since it is subject to variability based on had started with placebo and crossed over to lenalidomide had

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 965

developed AML, compared to 23.2% in the 5-mg group and 21.7% the complications of long-term steroid use in this population came
in the 10-mg group, though the crossover model prevented an assess- to outweigh the benefits. Nevertheless, immunosuppressive therapy
ment of statistical significance. may be reasonable for certain subsets of MDS patients in whom
Given recent progress in understanding of lenalidomide’s mecha- autoreactive T-lymphocytes contribute to the inhibition of effective
nism of action, it is perhaps unsurprising that it demonstrates con- hematopoiesis. Patients with hypoplastic MDS, in particular, have
siderably lower efficacy in MDS patients who do not have del(5q). been shown to have similarities to aplastic anemia, and may respond
In these patients, studies have consistently shown an overall response to treatment with ATG.458 Other studies have shown that patients
rate of about 25%, which on average lasts less than a year.445,446 It is with underlying PNH clones, which can be assessed by flow cytom-
not known whether the patients who respond have abnormalities of etry for PIG-anchored glycoproteins CD55 and CD59, may also
casein kinase 1α, or whether their sensitivity to lenalidomide occurs respond to ATG, as may patients with a human leukocyte antigen
via a different mechanism. (HLA) DR15 phenotype.459
Lenalidomide is generally well tolerated, especially compared to Retrospective analysis of data from the International Myelodys-
thalidomide. The most common side effects are neutropenia and plasia Risk Assessment Workshop showed that treatment with
thrombocytopenia, which are most common in the first month of immunosuppression was associated with an improvement in overall
treatment and which have some correlation to treatment response.447 survival (8.1 vs. 5.2 years, p < .001) and a lower risk of leukemic
Pretreatment thrombocytopenia, a feature atypical of pure del(5q), transformation, but these results could have been subject to selection
appears to negatively correlate with response and perhaps serves as a bias since fewer patients with higher-risk disease are treated upfront
phenotypic proxy for cooperating genetic events that blunt the with immunosuppression.460 On the other hand, a randomized study
molecular sensitivity to the drug.149 Significant thromboembolism comparing ATG plus cyclosporine to best supportive care suggested
has been observed in studies of patients with myeloma who receive a slight improvement in hematologic response in the ATG + cyclo-
lenalidomide, an effect that largely seems to be confined to sporine A group, but there was no difference in disease-free or overall
co-administration of lenalidomide and dexamethasone;448 the manu- survival.461 Subgroup analysis suggested that patients with hypocel-
facturer thus recommends thromboprophylaxis in del(5q) patients as lular marrows were more likely to respond to immunosuppression,
well, though it is not clear that the thrombotic risk is the same in again hinting at biologic similarities to aplastic anemia. In studies
these populations, and thrombocytopenia may limit the safety of this making careful distinction between MDS and aplastic anemia,
approach. however, mortality with ATG treatment was higher in MDS than in
aplastic anemia.345
Alemtuzumab, a monoclonal antibody against anti-CD52, has
Low-Dose Cytarabine activity in highly selected subsets of MDS patients. In particular, one
small phase I/II study of patients yielded a 77% overall response rate
Cytarabine, also known as cytosine arabinoside or Ara-c, is a pyrimi- among patients with IPSS intermediate-1 disease.462 Some patients
dine analog that is widely used in the treatment of a number of with cytogenetic abnormalities normalized their karyotype, and the
hematologic malignancies. In MDS, its use dates to the 1980s.449,450 rate of complete response appeared to increase with time in those
The typical dosing schedule is 5–20 mg/m2 per day via subcutaneous patients who were not lost to follow up. However, alemtuzumab is
injection (daily or twice daily) or continuous IV infusion. Although severely immunosuppressive and the study was hampered by a high
10% to 20% of patients with MDS have some degree of pathologic rate of attrition and careful selection of patients, and the results have
response during Ara-c therapy, these tend to last only a few months, not yet been repeated. The biologic basis for the high response rate
and treatment has not been shown to improve survival or reduce the in this single study remains unclear.
rate of transformation to AML.
One of the drawbacks to treatment with cytarabine or any other
conventional chemotherapeutic agent is that they cause cytopenias as
a primary side effect, and since cytopenias are a central problem in Manipulating the Microenvironment: Vascular
most MDS patients, the agents rapidly become limited in utility.451 Endothelial Growth Factor and Other Targets
The worsening of cytopenias is likely one of the primary reasons for
the lack of survival benefit for cytarabine in MDS. Observations of increased microvessel density in the bone marrow of
MDS463 patients have led to some enthusiasm for targeting vascular
growth factors and other molecules associated with the marrow
Antitumor Necrosis Factor Therapy microenvironment, but results so far have been somewhat disappoint-
ing. For instance, a phase II trial of bevacizumab, a monoclonal
The observation that MDS patients often have elevated levels of antibody directed against vascular endothelial growth factor (VEGF),
inflammatory cytokines and increased rates of apoptosis led to the showed a decrease in VEGF levels and a significant reduction in
postulate that blockade of these pathways might ameliorate the inef- microvascular density, but only one of 21 patients had any type of
fective hematopoiesis found in many patients with MDS. Etanercept, hematologic response.464 Similarly, a phase II study of vatalanib, an
a soluble TNF-α inhibitor, has been analyzed in two small pilot oral VEGF receptor inhibitor, showed hematologic responses in only
studies of MDS patients, with mixed results: cytopenias improved in 5% of patients, though many patients had to withdraw from the
one trial,452 whereas no effect was seen in the other.453 The drug has study because of side effects.465
also been combined with both ATG454 and azacitidine,454 but in Other therapies directed at the microenvironment have similarly
neither case was it clear that the addition made a difference. A separate shown responses in only a minority of patients. For instance, the
case report described sustained erythroid responses in two patients combination of pentoxifylline, a phosphodiesterase inhibitor, with
treated with infliximab, a monoclonal anti-TNF antibody.455 TNF-α dexamethasone and ciprofloxacin (PCD) has been shown to induce
and other inflammatory mediators may play a greater role in some hematologic responses in some patients, but most of these are
subsets of MDS than others,456 and better delineation of these groups transient and probably represent neutrophil demargination from the
may make future study of anti-TNF therapy reasonable in selected corticosteroid.231 A more recent study combining PCD with ami-
patients. fostine, an antiangiogenic agent usually used as a chemoprotectant,
showed a hematologic response rate of 66%, but long-term results
have not been reported.466 Lenalidomide and thalidomide were both
Immunosuppressive Therapy previously thought to act via effects on the microenvironment,467 but
with the discovery of lenalidomide’s role as a catalyst for cereblon-
Corticosteroid therapy was once a mainstay of treatment for MDS, mediated ubiquitination, as described earlier, the extent to which it
but despite reported response rates in the range of 10% to 20%,457 also affects the microenvironment is unclear.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
966 Part VII  Hematologic Malignancies

the promoters of numerous tumor suppressor genes, including


Signal Transduction Inhibitors CDKN2A, CDKN2B,484 FHIT,485 SOCS1,486 CTNNA1,487 and others.
Later, with the advent of genome-wide methylation profiling, it was
Activating mutations in receptor tyrosine kinases like NRAS and further observed that this aberrant methylation is often a global
FLT3 are relatively uncommon in MDS, not because they never occur phenomenon in MDS and that global hypermethylation, in particu-
in this population, but because they usually act as catalysts for pro- lar, appears to confer a poor prognosis.488
gression to secondary AML.238 The consideration for use of FLT3 This characterization of aberrant methylation in MDS soon led
inhibitors in MDS is similar to those in s-AML, and data are limited. to the identification of hypomethylating agents (HMA) as a candidate
A more thorough discussion of these agents may be found in Chapter class of MDS drugs. The prototypical hypomethylator, 5-azacytidine
59 (AML). (azacitidine), was synthesized in the 1960s and its hypomethylating
activity first clearly characterized in 1980.489 It is an azo-substituted
pyrimidine analogue that incorporates into RNA and is from there
Other Agents converted to a deoxynucleotide and incorporated into DNA, where
it binds and irreversibly inhibits DNMT1. The in vitro result is a
A number of other agents have been used in MDS and reported either global decrease in cytosine methylation, which leads to differentiation
in anecdotes or small, nonrandomized series of patients. Historically, in some leukemia cell lines. Of note, azacitidine also appears to have
many patients with RARS were started on vitamin B6 because of its nonepigenetic mechanisms of action, including some degree of direct
efficacy in familial sideroblastic anemia, but it has shown little efficacy cytotoxicity and immune stimulation.490 Both azacitidine and the
in adults.468 Other dietary supplements, include retinoids (vitamin A related compound decitabine (5-aza-2’-deoxycytidine) are now fre-
analogues), vitamin D,469 and vitamin K2,470 have all been tried quently administered in both MDS and AML.
because of favorable effects on differentiation in vitro, but have not Standard dosing of azacitidine is 75 mg/m2 administered subcu-
been effective in humans. A number of other drugs have also been taneously once per day for 7 consecutive days each month.491 This
trialed in humans based on similar observations of prodifferentiation was the dosing schedule used in the first major study of hypometh-
effects in preclinical experiments, all without comparable efficacy in ylating agents in MDS, CALGB 9221, in which azacitidine was
human patients. These include amifostine as a single agent,471 the found to be superior to best supportive care both in terms of quality
solvent hexamethyl bisacetamide,472 and alpha and gamma interferon. of life and risk of progression to AML.492 In this study, about 60%
Interferon, in particular, has recently been shown to cause normal of patients had some degree of response to azacitidine, although
HSCs to exit quiescence in mouse models,473,474 but whether it can consensus response criteria were not used. Most of these responses
be used for similar purposes to “prime” MDS stem cells for subse- were minor hematologic improvement, but about 15% of patients
quent treatment with cytotoxic therapy has not yet been explored. had a pathologic complete response. Most of those who responded
Given their efficacy in promyelocytic leukemia, both all-trans reti- did so within 6 months. There was a nonsignificant trend toward an
noic acid (ATRA) and ATO have been trialed in small numbers of improvement in overall survival with azacitidine, and side effects
MDS patients in a similar attempt to induce differentiation. ATO tended to be mild, including nausea and vomiting and transient
has shown responses in up to 20% of MDS patients in several small myelosuppression.
series,475–478 but the molecular basis for this action and how to predict Based on these data and a large increase in requests for compas-
who will respond are unclear. ATRA by itself has largely been inef- sionate use of the drug after CALGB 9221 was first presented in
fective, likely caused by the absence of the PML-RARA fusion protein 2002, the Food and Drugs Administration (FDA) granted approval
that is the basis for its efficacy in acute promyelocytic leukemia to azacitidine for MDS in 2004. A subsequent study, AZA-001
(APML).479,480 In vitro data suggest that in some AML models, comparing azacitidine 1 : 1 to the patient and doctor choice of either
downstream targets of the retinoic acid receptor may be epigenetically standard induction, low-dose cytarabine, and best supportive care in
silenced by abnormal induction of histone demethylases such as 358 patients with higher-risk MDS or CMML showed a median
LSD1, which can be inhibited by tranylcypromine, a monoamine 9-month improvement in overall survival for those treated with
oxidase inhibitor (MAOI) approved in Europe to treat depression.481 azacitidine (24 versus 15 months).493 This represents the only major
In vitro, the combination of ATRA with tranylcypromine in non- study to date in which a survival benefit has been shown for a drug
APML leukemia derepresses targets of the retinoic acid receptor and in MDS. Other studies have shown that a more convenient 5-day
significantly impairs the ability of leukemia stem cells to engraft in azacitidine dosing schedule appears to be effective, but this has not
immunodeficient mice. Whether this pathway can be similarly been directly compared to the 7-day schedule, which remains the
reactivated in MDS, and whether this is a viable therapeutic combi- standard. One recent study suggested an improvement in response
nation in patients, also remain as yet unexplored. from prolonged administration of azacitidine, though all patients in
A number of other novel agents remain in clinical trials for lower- this study had higher rates of hematologic normalization compared
risk MDS at the time of this writing.482 These include inhibitors of to historical controls, for unclear reasons.494
the transforming growth factor-beta receptor,483 inhibitors of toll-like The other major hypomethylating agent, decitabine (5-aza-2′-
receptors, chimeric antibodies to activin, mitochondrial modulators, deoxycytidine), is also FDA-approved for treatment in all subclasses
RAS pathway inhibitors, and anti-CD33 molecules. In addition, of MDS. Although structurally similar to azacitidine, decitabine has
there are ongoing trials evaluating combinations of existing therapies, a deoxyribose backbone that permits it to be directly incorporated
including hypomethylating agents, histone deacetylase (HDAC) into DNA. At higher doses it introduces DNA cross-linking and
inhibitors, lenalidomide, and hematopoietic growth factors. While cell-cycle arrest, a mechanism more akin to a cytotoxic agent, but at
some of these trials may identify novel therapies or combinations with lower doses it appears to primarily act as a hypomethylating agent.495
efficacy for subsets of MDS patients, whether any of them will have Similar to azacitidine, decitabine has been shown to improve
substantial impact on the treatment landscape for low-risk MDS outcomes for patients with MDS, including a reduced transfusion
remains to be seen. requirement and slower transformation to AML. It may induce
responses more quickly than azacitidine; for instance, in the multi-
center ADOPT (alternative dosing of decitabine for outpatient
Higher-Risk Disease therapy) study, 90% of patients responded after four cycles, rather
than the six cycles seen in the largest azacitidine trials.496
Hypomethylating Agents In contrast to azacitidine, however, decitabine has never been shown
to confer an overall survival benefit. Some of the difference in out-
MDS genomes frequently display aberrant methylation patterns rela- comes may be explained by difficulties establishing the optimal dose
tive to normal hematopoietic cells. This observation was initially and schedule; in the earliest trials it was administered at a dose of
made in the 1990s, when investigators noted hypermethylation in 15 mg/m2 given IV every 8 hours for 3 days,497 but subsequent

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 967

studies showed better hypomethylation and an improved overall remission. Conventional chemotherapy alone is rarely curative in
response rate when it was given at a dose of 20 mg/m2 once daily for MDS, and even those younger patients who achieve a complete
5 consecutive days,498 the dosing strategy used in ADOPT. Some remission will need further consolidation, usually with an allogeneic
recent small studies have suggested an even different dosing strategy, stem cell transplant.
using frequent administration of low doses of decitabine (0.1–0.2 mg/
kg/day) in an attempt to minimize the cytotoxic effects of the drug
while catching more cells in S phase, when the demethylating activity Allogeneic Stem Cell Transplantation
of the drug would be most effective.499
Despite the widespread use of hypomethylating agents in MDS, Allogeneic HSCT is the only known curative therapy for MDS.513
a number of key questions persist. Although studies have shown that This is thought to arise from the fact that MDS progenitor cells,
each can demethylate the promoters of specific tumor suppressors being transformed stem cells, are mostly quiescent and are thus rela-
(e.g., P15INK4B500 for decitabine and PLCB1501 for azacitidine), it is tively resistant to most chemotherapy. Allogeneic HSCT, on the other
not clear that this is the dominant in vivo mechanism. Some studies hand, leverages both the chemotherapy of the conditioning regimen
have shown increased demethylation in patients who respond to and the antitumor effect of the newly transplanted stem cells to
hypomethylating agents,502 but this has not been a universal observa- eliminate all vestiges of the prior hematopoietic system. This is, of
tion, and neither pretreatment methylation levels (either global or at course, a simplified description of the best-case outcome for HSCT.
specific promoters) nor the net change in methylation with treatment The reality for most MDS patients is significantly more complicated,
have been consistently predictive of treatment responsiveness.488,503 although outcomes have steadily improved in recent years as centers
Recent studies have also interrogated the impact of somatic mutations have gained more experience in transplanting older patients using
on hypomethylating agent sensitivity, and TET2 mutations in unrelated and even unmatched donors.514
particular have been shown to increase the likelihood of HMA An exhaustive review of stem cell transplantation is beyond the
response,103 but the overall impact of these findings in the long term scope of this chapter, but a few points specific to MDS bear mention.
remains unclear. Identification of patients for whom transplant might be a reasonable
option is an important first step. This group typically includes
patients younger than 60 years, who are often in otherwise good
“Histone” Deacetylase Inhibitors health and would otherwise be predicted to have a long life expec-
tancy. Older patients who are in otherwise excellent health may be
The observation that MDS frequently involves epigenomic abnor- considered for transplantation as well, usually with a reduced-intensity
malities has provoked interest in other modalities beyond the hypo- conditioning regimen. Although there is no official upper age limit
methylating agents. To date, the next most-studied class of agent is to eligibility, most centers are hesitant to transplant patients older
the histone deacetylase inhibitors, which as their name implies block than 75.515
the deacetylation of histone residues as well as deacetylation of other The stage of disease is also important: in older patients, transplant
cellular protein. In vitro, application of these drugs leads to large-scale is usually reserved for those with excess blasts or other forms of
epigenetic modifications, but in vivo, the drugs have so far not proven higher-risk disease, but it is important to proceed to transplant before
as effective as hypomethylating agents, at least when used alone.504 the disease has evolved into acute leukemia. On the other hand, some
Several agents, however, remain in trials, including valproic acid,505 centers occasionally offer transplantation to younger patients with
vorinostat,506 panobinostat,507 and belinostat.508 Many of these trials lower-risk categories of disease given that they have a longer latency
combine the HDAC inhibitor with some other therapy. One coop- period in which their disease could progress or evolve into leukemia.
erative trial of azacitidine with or without entinostat, E1905, showed This decision, however, is not completely uniform, and some data
no improvement in hematologic response in the entinostat arm, suggest that delayed stem cell transplantation is associated with better
which in fact displayed less demethylation than azacitidine alone, overall survival in patients with low and intermediate-1 IPSS scores
perhaps suggesting some degree of pharmacologic antagonism.494 regardless of age.516
However, in vitro data have also shown significant pharmacologic Once a decision has been made to proceed to transplant, other
differences between the different HDAC inhibitors, suggesting that decisions must follow. The first involves the source of the stem cells.
results for one drug may not be generalizable to the class as a whole. If a healthy, HLA-matched sibling is available, this is often prefera-
Similar negative results were recently reported in the U.S./Canadian ble;517 however, reflective of the average age of MDS patients, most
Intergroup S1117, a large three-arm trial comparing azacitidine alone siblings are older and often have comorbidities, such as cancer or
to azacitidine plus vorinostat to azacitidine plus lenalidomide, as well other illnesses, that preclude them from consideration as donors. If
as a study of azacitidine with or without pracinostat.509 a related donor is unavailable, the next best option is a search of the
national donor registry for an HLA-matched unrelated donor.518
Compared to related donors, transplant from unrelated donors carries
Induction Chemotherapy a higher rate of graft-versus-host disease as a consequence of minor
antigen mismatch. If a matched unrelated donor cannot be found,
AML-like induction regimens (e.g., cytarabine plus an anthracycline) options then include an unrelated donor transplant mismatched at
are not effective for most patients with MDS, with available studies one (or occasionally more) HLA loci, or an “alternative donor source,”
suggesting a remission rate of less than 20%. Hematologic recovery which includes umbilical cord blood519 and haploidentical donors.520
is often incomplete, and in many cases the chemotherapy mainly In the case of MDS patients, this latter type of transplant is often
serves to select for the malignant clone. In fact, a randomized trial obtained from one of the patient’s children.
comparing azacitidine to daunorubicin plus cytarabine in elderly The specific choice of conditioning regimen is likely less important
patients with higher-risk MDS showed that the patients treated with in MDS than in other hematologic diseases given the disease’s intrin-
azacitidine had better outcomes, although only small numbers of sic chemoresistance, as described earlier. Options include fludarabine
patients were treated with induction.493 plus busulfan, fludarabine plus melphalan, busulfan plus cyclophos-
Induction chemotherapy, however, may be reasonable in selected phamide, or cyclophosphamide plus irradiation. Many older patients
younger patients; some studies have suggested remission rates of up with MDS are unlikely to tolerate myeloablative doses of condition-
to 50% in patients younger than 60, though these may have contained ing agents and are instead given reduced doses of conditioning agents.
some patients with de novo AML,510 and the studies were performed These “reduced intensity conditioning” transplants rely more heavily
before the widespread availability of deep sequencing techniques that on the graft-versus-tumor effect of the transplant to eliminate the
might have shown persistence of clonal mutations even in the ablated malignant clone.521,522 Given the intrinsic chemoresistance of MDS
marrows.148 Adding other agents, such as liposomal doxorubicin,511 stem cells, however, this is usually a reasonable tradeoff. Following
topotecan,512 and thalidomide, do not seem to improve the rates of transplant, patients must remain on immunosuppressive medications

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
968 Part VII  Hematologic Malignancies

to prevent the emergence of graft-versus-host disease. A full discus- (PIM kinase inhibitors, B-catenin degraders537), and radioisotope-
sion of posttransplant considerations is outside the scope of this conjugated monoclonal antibodies. As mentioned earlier, whether
chapter, but most aspects of this area are less specific to MDS than any of these drugs will significantly change the landscape of MDS
those discussed previously. therapy is unclear.
As mentioned, despite being potentially curative therapy, trans-
plant in MDS is associated with significant morbidity and mortality.
One study has suggested that 50% to 60% of lower-risk patients and FUTURE DIRECTIONS
20% to 40% with higher-risk disease can expect long-term disease-
free survival following transplant,523 but the alternative view of these While the last ten years have witnessed substantial improvements in
data is that a substantial proportion of patients (40%–50% of low our understanding of the molecular pathophysiology underlying
risk and 60%–80% of high risk) either relapse or die of nonrelapse MDS, similar strides have not been made in its treatment: at the time
complications of transplant. of this writing, it has been almost 10 years since the FDA last
Numerous studies have attempted to dissect patient and disease approved a new drug for treatment of MDS (decitabine in 2006).
attributes that may predict outcomes following transplant. Poor-risk There are few therapies in clinical trials that offer hope of broad or
cytogenetics, including monosomy 7 and complex karyotypes,524 deep efficacy, and much of the ongoing clinical effort is directed at
have been shown to predict a high rate of relapse, and recent data incremental changes to current practice, such as adjusting dosing
suggest that pathogenic TP53, DNMT3A, and TET2 mutations are schedules of currently approved therapies or using modestly effective
predictors of poor outcomes following transplantation as well.104 So drugs in combination. The lack of progress is to a great extent caused
far, no cytogenetic or genetic profiles have been clearly associated with by unfortunate facts of MDS biology, including its origin in quiescent
a favorable response to transplant. Preexisting neutropenia has been stem cells, a paucity of targetable activating mutations, and enrich-
shown to predict an increased risk of serious infections in the post- ment for poorly targetable mutations with much more subtle effects
transplant setting.525 On the other hand, a low blast count (fewer on growth and differentiation. Efforts are further complicated by the
than 5%) is associated with better outcomes, and other data show heterogeneity of biology both between and within individual patients,
that patients who achieved a remission or significant response to as well as the advanced age and poor overall health of many patients
pretransplant chemotherapy have better outcomes as well.526 with MDS.
Despite these significant challenges, there is nevertheless great
opportunity for improvements in care. Advances in stem cell trans-
Other Therapies for Higher-Risk Disease plant techniques, including the availability of alternative donor
sources, refinements of posttransplant immunosuppression, and
Autologous Stem Cell Infusion augmentation of the graft-versus-tumor effect, should continue to
improve the outcomes for MDS patients able to undergo transplant.
Though rarely used in current practice, infusion of autologous stem An equally difficult challenge lies in treatment of those patients ineli-
cells has been attempted in the past, primarily for patients with oli- gible for transplant, and it is in these patients that effective unification
goblastic disease.527,528 There are a number of reasons why this of our improving biologic understanding of MDS with clinical care
approach is suboptimal for MDS patients. The most obvious draw- is most essential.
back is that MDS is by definition a disorder of stem cells, such that
any autologous infusion will ultimately lead to repopulation of the
marrow with the original malignant clone, but without the graft-
versus-leukemia effect imparted by an allogeneic transplant. Moreover, SUGGESTED READINGS
autologous stem cell infusion is really meant to serve as a rescue after
high-dose chemotherapy, and is thus most successful when given for Abel GA, Klaassen R, Lee SJ, et al: Patient-reported outcomes for the
a chemotherapy-sensitive disease, which MDS is not. Finally, given myelodysplastic syndromes: a new MDS-specific measure of quality of
the ineffective hematopoiesis many patients experience before treat- life. Blood 123(3):451–452, 2014.
ment, hematopoietic recovery following high-dose chemotherapy is Bejar R, Stevenson K, Abdel-Wahab O, et al: Clinical effect of point muta-
often poor. Since most patients who would be candidates for autolo- tions in myelodysplastic syndromes. N Engl J Med 364(26):2496–2506,
gous stem cell rescue would also be eligible for other, more effective 2011.
modalities of therapy, there are usually few reasons to pursue this Bejar R, Stevenson KE, Caughey B, et al: Somatic mutations predict poor
avenue. outcome in patients with myelodysplastic syndrome after hematopoietic
stem-cell transplantation. J Clin Oncol 32(25):2691–2698, 2014.
Bejar R, Stevenson KE, Caughey BA, et al: Validation of a prognostic model
Other Drugs and the impact of mutations in patients with lower-risk myelodysplastic
syndromes. J Clin Oncol 30(27):3376–3382, 2012.
Most drugs known to have activity in AML have been tried for MDS Cogle CR, Iannacone MR, Yu D, et al: High rate of uncaptured myelodys-
as well, usually with suboptimal results. These include etoposide,529 plastic syndrome cases and an improved method of case ascertainment.
irinotecan, gemcitabine,530 low-dose melphalan,531 idarubicin,510 and Leuk Res 38(1):71–75, 2014.
clofarabine.532 Both hydroxyurea and low-dose oral etoposide can be Damm F, Fontenay M, Bernard OA: Point mutations in myelodysplastic
used to control proliferation in patients with excess blasts, but they syndromes. N Engl J Med 365(12):1154–1155, author reply 155,
do not target the underlying disease. Clofarabine is sometimes effec- 2011.
tive as a bridge to transplant therapy in patients with excess blasts Della Porta MG, Tuechler H, Malcovati L, et al: Validation of WHO
despite hypomethylating agent therapy, but renal and hepatic toxicity classification-based Prognostic Scoring System (WPSS) for myelodysplas-
and severe skin rash are limiting.533 tic syndromes and comparison with the revised International Prognostic
As with lower-risk disease, there are a number of ongoing trials Scoring System (IPSS-R). A study of the International Working Group
for patients with high-risk MDS as of this writing. Reflective of the for Prognosis in Myelodysplasia (IWG-PM). Leukemia 2015.
poorer prognosis of these patients compared to those with lower-risk Genovese G, Kähler AK, Handsaker RE, et al: Clonal hematopoiesis and
disease, many of these trials either examine different combinations blood-cancer risk inferred from blood DNA sequence. N Engl J Med
of high-dose chemotherapy or modification of protocols for alloge- 371(26):2477–2487, 2014.
neic stem cell transplant, and are often open to both MDS and Gerds AT, Gooley TA, Estey EH, et al: Pretransplantation therapy with
AML patients. However, there are a number of novel or repurposed azacitidine vs induction chemotherapy and posttransplantation outcome
agents in trials as well, including immunotherapies (ipilimumab,534 in patients with MDS. Biol Blood Marrow Transplant 18(8):1211–1218,
adoptive T-cell transfer,535 and NK cell therapy536), small molecules 2012.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 969

Greenberg PL, Tuechler H, Schanz J, et al: Revised international prognostic Papaemmanuil E, Gerstung M, Malcovati L, et al: Clinical and biological
scoring system for myelodysplastic syndromes. Blood 120(12):2454–2465, implications of driver mutations in myelodysplastic syndromes. Blood
2012. 122(22):3616–3627, quiz 3699, 2013.
Haferlach T, Nagata Y, Grossmann V, et al: Landscape of genetic lesions in Pascal L, Beyne-Rauzy O, Brechignac S, et al: Cardiac iron overload
944 patients with myelodysplastic syndromes. Leukemia 28(2):241–247, assessed by T2* magnetic resonance imaging and cardiac function in
2014. regularly transfused myelodysplastic syndrome patients. Br J Haematol
Hahn CN, Chong C-E, Carmichael CL, et al: Heritable GATA2 mutations 162(3):413–415, 2013.
associated with familial myelodysplastic syndrome and acute myeloid Saunthararajah Y, Sekeres M, Advani A, et al: Evaluation of noncytotoxic
leukemia. Nat Genet 43(10):1012–1017, 2011. DNMT1-depleting therapy in patients with myelodysplastic syndromes.
Hahn T, McCarthy PL, Hassebroek A, et al: Significant improvement in J Clin Invest 2015.
survival after allogeneic hematopoietic cell transplantation during a period Schanz J, Tüchler H, Solé F, et al: New comprehensive cytogenetic scoring
of significantly increased use, older recipient age, and use of unrelated system for primary myelodysplastic syndromes (MDS) and oligoblastic
donors. J Clin Oncol 31(19):2437–2449, 2013. acute myeloid leukemia after MDS derived from an international database
Jaiswal S, Fontanillas P, Flannick J, et al: Age-related clonal hematopoiesis merge. J Clin Oncol 30(8):820–829, 2012.
associated with adverse outcomes. N Engl J Med 371(26):2488–2498, Schneider RK, Ademà V, Heckl D, et al: Role of casein kinase 1A1 in the
2014. biology and targeted therapy of del(5q) MDS. Cancer Cell 26(4):509–520,
Kon A, Shih L-Y, Minamino M, et al: Recurrent mutations in multiple 2014.
components of the cohesin complex in myeloid neoplasms. Nat Genet Steensma DP: Dysplasia has A differential diagnosis: distinguishing genuine
45(10):1232–1237, 2013. myelodysplastic syndromes (MDS) from mimics, imitators, copycats and
Kuter DJ, Begley CG: Recombinant human thrombopoietin: basic biology impostors. Curr Hematol Malig Rep 7(4):310–320, 2012.
and evaluation of clinical studies. Blood 100(10):3457–3469, 2002. Steensma DP: Historical perspectives on myelodysplastic syndromes. Leuk
List A, Kurtin S, Roe DJ, et al: Efficacy of lenalidomide in myelodysplastic Res 36:1441, 2012.
syndromes. N Engl J Med 352(6):549–557, 2005. Walter MJ, Shen D, Ding L, et al: Clonal architecture of secondary acute
Lyons RM, Marek BJ, Paley C, et al: Comparison of 24-month outcomes myeloid leukemia. N Engl J Med 366(12):1090–1098, 2012.
in chelated and non-chelated lower-risk patients with myelodysplas- Yoshida K, Sanada M, Shiraishi Y, et al: Frequent pathway mutations of
tic syndromes in a prospective registry. Leuk Res 38(2):149–154, splicing machinery in myelodysplasia. Nature 478(7367):64–69, 2011.
2014. Yoshizato T, Dumitriu B, Hosokawa K, et al: Somatic mutations and clonal
Nikoloski G, Langemeijer SMC, Kuiper RP, et al: Somatic mutations of the hematopoiesis in aplastic anemia. N Engl J Med 373(1):35–47, 2015.
histone methyltransferase gene EZH2 in myelodysplastic syndromes. Nat
Genet 42(8):665–667, 2010.
Papaemmanuil E, Cazzola M, Boultwood J, et al: Somatic SF3B1 mutation in REFERENCES
myelodysplasia with ring sideroblasts. N Engl J Med 365(15):1384–1395,
2011. For the complete list of references, log on to www.expertconsult.com.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 969.e1

REFERENCES 25. Ma X, Does M, Raza A, et al: Myelodysplastic syndromes: incidence


and survival in the United States. Cancer 109(8):1536–1542, 2007.
1. Steensma DP: Historical perspectives on myelodysplastic syndromes. 26. Cogle CR, Iannacone MR, Yu D, et al: High rate of uncaptured
Leuk Res 36:1441, 2012. myelodysplastic syndrome cases and an improved method of case
2. Luzzatto A: Sull’ anemia grave megaloblastica senza reporto ematologico ascertainment. Leuk Res 38(1):71–75, 2014.
corrispondente (anemia pseudoaplastica). Riv veneta di sc med Venezia 27. Cogle CR, Craig BM, Rollison DE, et al: Incidence of the myelodys-
47:193–212, 1907. plastic syndromes using a novel claims-based algorithm: high number of
3. Guglielmo G: di Eritremie acute. BollettinoSocieta Medico Chir uncaptured cases by cancer registries. Blood 117(26):7121–7125, 2011.
1:665–673, 1926. 28. Pavlu J, Emmerson J, Marks AJ, et al: Idiopathic cytopenia of
4. Rhoads C, Barker W: Refractory anemia: analysis of 100 cases. JAMA undetermined significance and the minimal criteria for a diagnosis of
110:794–796, 1938. myelodysplastic syndrome. Leuk Lymphoma 52(3):515–516, 2011.
5. Rosati S, Mick R, Xu F, et al: Refractory cytopenia with multilineage 29. Sekeres MA, Schoonen WM, Kantarjian H, et al: Characteristics of US
dysplasia: further characterization of an “unclassifiable” myelodysplastic patients with myelodysplastic syndromes: results of six cross-sectional
syndrome. Leukemia 10(1):20–26, 1996. physician surveys. J Natl Cancer Inst 100(21):1542–1551, 2008.
6. Chevallier P: Sur la terminologie des leucoses et les affections-frontieres: 30. Marisavljevic D, Savic A, Zeremski V, et al: Myelodysplastic syndromes
les odoleucoses. Sangre (Barc) 15:587–593, 1942. in adults aged less than 50 years: incidence and clinicopathological data.
7. Hamilton-Paterson J: Pre-leukaemic anaemia. Acta Haematol 2:309– J BUON 19(4):999–1005, 2014.
316, 1949. 31. Jaiswal S, Fontanillas P, Flannick J, et al: Age-related clonal hematopoiesis
8. Block M, Jacobson LO, Bethard WF: Preleukemic acute human associated with adverse outcomes. N Engl J Med 371(26):2488–2498,
leukemia. J Am Med Assoc 152(11):1018–1028, 1953. 2014.
9. Saarni MI, Linman JW: Preleukemia. The hematologic syndrome 32. Genovese G, Kähler AK, Handsaker RE, et al: Clonal Hematopoiesis
preceding acute leukemia. Am J Med 55(1):38–48, 1973. and Blood-Cancer Risk Inferred from Blood DNA Sequence. N Engl J
10. Dreyfus B, Rochant H, Sultan C, et al: Refractory anemia with Med 371(26):2477–2487, 2014.
excess myeloblasts in the bone marrow. Study of 11 cases]. Presse Med 33. Stary J, Baumann I, Creutzig U, et al: Getting the numbers straight
78(8):359–364, 1970. in pediatric MDS: distribution of subtypes after exclusion of down
11. Dreyfus B: Preleukemic states. I. Definition and classification. II. syndrome. Pediatr Blood Cancer 50(2):435–436, 2008.
Refractory anemia with an excess of myeloblasts in the bone marrow 34. Germing U, Aul C, Niemeyer CM, et al: Epidemiology, classification
(smoldering acute leukemia). Nouv Rev Fr Hematol Blood Cells and prognosis of adults and children with myelodysplastic syndromes.
17(1–2):33–55, 1976. Ann Hematol 87(9):691–699, 2008.
12. Bennett JM, Catovsky D, Daniel MT, et al: Proposals for the clas- 35. Lange BJ, Kobrinsky N, Barnard DR, et al: Distinctive demography,
sification of the acute leukaemias. French-American-British (FAB) biology, and outcome of acute myeloid leukemia and myelodysplastic
co-operative group. Br J Haematol 33(4):451–458, 1976. syndrome in children with Down syndrome: Children’s Cancer Group
13. Galton D, Dacie J: Classification of the acute leukaemias. Blood Cells Studies 2861 and 2891. Blood 91(2):608–615, 1998.
1:17–24, 1975. 36. Butturini A, Gale RP, Verlander PC, et al: Hematologic abnormalities
14. Linman JW: Myelomonocytic leukemia and its preleukemic phase. J in Fanconi anemia: an International Fanconi Anemia Registry study.
Chronic Dis 22(11):713–716, 1970. Blood 84(5):1650–1655, 1994.
15. Bennett JM, Catovsky D, Daniel MT, et al: Proposals for the classifica- 37. Hahn CN, Chong C-E, Carmichael CL, et al: Heritable GATA2
tion of the myelodysplastic syndromes. Br J Haematol 51(2):189–199, mutations associated with familial myelodysplastic syndrome and acute
1982. myeloid leukemia. Nat Genet 43(10):1012–1017, 2011.
16. Cabello AI, Collado R, Ruiz MA, et al: A retrospective analysis of 38. Owen CJ, Toze CL, Koochin A, et al: Five new pedigrees with inherited
myelodysplastic syndromes with thrombocytosis: reclassification of the RUNX1 mutations causing familial platelet disorder with propensity to
cases by WHO proposals. Leuk Res 29(4):365–370, 2005. myeloid malignancy. Blood 112(12):4639–4645, 2008.
17. Voglová J, Chrobák L, Neuwirtová R, et al: Myelodysplastic and 39. Zhang MY, Churpek JE, Keel SB, et al: Germline ETV6 mutations
myeloproliferative type of chronic myelomonocytic leukemia–distinct in familial thrombocytopenia and hematologic malignancy. Nat Genet
subgroups or two stages of the same disease? Leuk Res 25(6):493–499, 47(2):180–185, 2015.
2001. 40. Yamaguchi H, Baerlocher GM, Lansdorp PM, et al: Mutations of the
18. Giagounidis AA, Germing U, Haase S, et al: Clinical, morphological, human telomerase RNA gene (TERC) in aplastic anemia and myelo-
cytogenetic, and prognostic features of patients with myelodysplastic dysplastic syndrome. Blood 102(3):916–918, 2003.
syndromes and del(5q) including band q31. Leukemia 18(1):113–119, 41. Surveillance, Epidemiology, and End Results Program.html [Internet].
2004. [cited 2015 Feb 25];Available from: <http://seer.cancer.gov/>.
19. Hasserjian RP, Campigotto F, Klepeis V, et al: De novo acute myeloid 42. Komrokji RS, Padron E, Ebert BL, et al: Deletion 5q MDS: molecular
leukemia with 20-29% blasts is less aggressive than acute myeloid and therapeutic implications. Best Pract Res Clin Haematol 26(4):365–
leukemia with ≥30% blasts in older adults: a Bone Marrow Pathology 375, 2013.
Group study. Am J Hematol 89(11):E193–E199, 2014. 43. Yin SN, Hayes RB, Linet MS, et al: A cohort study of cancer among
20. Valent P, Bain BJ, Bennett JM, et al: Idiopathic cytopenia of unde- benzene-exposed workers in China: overall results. Am J Ind Med
termined significance (ICUS) and idiopathic dysplasia of uncertain 29(3):227–235, 1996.
significance (IDUS), and their distinction from low risk MDS. Leuk 44. Tong H, Hu C, Yin X, et al: A Meta-analysis of the relationship between
Res 36(1):1–5, 2012. cigarette smoking and incidence of myelodysplastic syndromes. PLoS
21. Riccardi A, Giordano M, Girino M, et al: Refractory cytopenias: ONE 8(6):e67537, 2013.
clinical course according to bone marrow cytology and cellularity. Blut 45. Chen B, Zhao W-L, Jin J, et al: Clinical and cytogenetic features of
54(3):153–163, 1987. 508 Chinese patients with myelodysplastic syndrome and comparison
22. Della Porta MG, Travaglino E, Boveri E, et al: Minimal morpho- with those in Western countries. Leukemia 19(5):767–775, 2005.
logical criteria for defining bone marrow dysplasia: a basis for clinical 46. Gologan R, Georgescu D, Tatic A, et al: Epidemiological data from the
implementation of WHO classification of myelodysplastic syndromes. registry of patients with myelodysplastic syndrome in a single hospital
Leukemia 29(1):66–75, 2015. center of Romania. Leuk Res 33(11):1556–1561, 2009.
23. Sekeres MA: The epidemiology of myelodysplastic syndromes. Hematol 47. Matsuda A, Germing U, Jinnai I, et al: Difference in clinical features
Oncol Clin North Am 24(2):287–294, 2010. between Japanese and German patients with refractory anemia in
24. Craig BM, Rollison DE, List AF, et al: Underreporting of myeloid myelodysplastic syndromes. Blood 106(8):2633–2640, 2005.
malignancies by United States cancer registries. Cancer Epidemiol 48. Iwanaga M, Hsu W-L, Soda M, et al: Risk of myelodysplastic syn-
Biomark Prev 21(3):474–481, 2012. dromes in people exposed to ionizing radiation: a retrospective cohort

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
969.e2 Part VII  Hematologic Malignancies

study of Nagasaki atomic bomb survivors. J Clin Oncol 29(4):428–434, 71. Kelly L, Clark J, Gilliland DG: Comprehensive genotypic analysis
2011. of leukemia: clinical and therapeutic implications. Curr Opin Oncol
49. Ben-Yehuda D, Krichevsky S, Caspi O, et al: Microsatellite instabil- 14(1):10–18, 2002.
ity and p53 mutations in therapy-related leukemia suggest mutator 72. Anastasi J, Feng J, Le Beau MM, et al: Cytogenetic clonality in myelo-
phenotype. Blood 88(11):4296–4303, 1996. dysplastic syndromes studied with fluorescence in situ hybridization:
50. Pedersen-Bjergaard J, Philip P, Larsen SO, et al: Chromosome aberra- lineage, response to growth factor therapy, and clone expansion. Blood
tions and prognostic factors in therapy-related myelodysplasia and acute 81(6):1580–1585, 1993.
nonlymphocytic leukemia. Blood 76(6):1083–1091, 1990. 73. Gondek LP, Tiu R, O’Keefe CL, et al: Chromosomal lesions and
51. Karp JE, Sarkodee-Adoo CB: Therapy-related acute leukemia. Clin Lab uniparental disomy detected by SNP arrays in MDS, MDS/MPD,
Med 20(1):71–81, ix, 2000. and MDS-derived AML. Blood 111(3):1534–1542, 2008.
52. Curtis RE, Boice JD, Stovall M, et al: Risk of leukemia after che- 74. Damm F, Fontenay M, Bernard OA: Point mutations in myelodysplas-
motherapy and radiation treatment for breast cancer. N Engl J Med tic syndromes. N Engl J Med 365(12):1154–1155, author reply 155,
326(26):1745–1751, 1992. 2011.
53. Christiansen DH, Andersen MK, Pedersen-Bjergaard J: Mutations with 75. Bravo GM, Lee E, Merchan B, et al: Integrating genetics and epi-
loss of heterozygosity of p53 are common in therapy-related myelodys- genetics in myelodysplastic syndromes: advances in pathogenesis and
plasia and acute myeloid leukemia after exposure to alkylating agents disease evolution. Br J Haematol 166(5):646–659, 2014.
and significantly associated with deletion or loss of 5q, a complex 76. Welch JS, Ley TJ, Link DC, et al: The origin and evolution of muta-
karyotype, and a poor prognosis. J Clin Oncol 19(5):1405–1413, 2001. tions in acute myeloid leukemia. Cell 150(2):264–278, 2012.
54. Super HJ, McCabe NR, Thirman MJ, et al: Rearrangements of the 77. Vogelstein B, Papadopoulos N, Velculescu VE, et al: Cancer genome
MLL gene in therapy-related acute myeloid leukemia in patients landscapes. Science 339(6127):1546–1558, 2013.
previously treated with agents targeting DNA-topoisomerase II. Blood 78. Papaemmanuil E, Gerstung M, Malcovati L, et al: Clinical and bio-
82(12):3705–3711, 1993. logical implications of driver mutations in myelodysplastic syndromes.
55. Nimer SD: MDS: a stem cell disorder—but what exactly is wrong with Blood 122(22):3616–3627, quiz 3699, 2013.
the primitive hematopoietic cells in this disease? Hematology Am Soc 79. Bejar R, Stevenson K, Abdel-Wahab O, et al: Clinical effect of point
Hematol Educ Program 2008(1):43–51, 2008. mutations in myelodysplastic syndromes. N Engl J Med 364(26):2496–
56. Pang WW, Pluvinage JV, Price EA, et al: Hematopoietic stem cell and 2506, 2011.
progenitor cell mechanisms in myelodysplastic syndromes. Proc Natl 80. Haferlach T, Nagata Y, Grossmann V, et al: Landscape of genetic
Acad Sci USA 110(8):3011–3016, 2013. lesions in 944 patients with myelodysplastic syndromes. Leukemia
57. Janssen JW, Buschle M, Layton M, et al: Clonal analysis of myelo- 28(2):241–247, 2014.
dysplastic syndromes: evidence of multipotent stem cell origin. Blood 81. Malcovati L, Papaemmanuil E, Ambaglio I: Driver somatic mutations
73(1):248–254, 1989. identify distinct disease entities within myeloid neoplasms with myelo-
58. Walter MJ, Shen D, Ding L, et al: Clonal architecture of secondary dysplasia. Blood 124:1513–1521, 2014.
acute myeloid leukemia. N Engl J Med 366(12):1090–1098, 2012. 82. Lindsley RC, Ebert BL: The biology and clinical impact of genetic
59. Magee JA, Piskounova E, Morrison SJ: Cancer stem cells: impact, lesions in myeloid malignancies. Blood 122(23):3741–3748, 2013.
heterogeneity, and uncertainty. Cancer Cell 21(3):283–296, 2012. 83. Yoshida K, Sanada M, Shiraishi Y, et al: Frequent pathway mutations
60. Clevers H: The cancer stem cell: premises, promises and challenges. Nat of splicing machinery in myelodysplasia. Nature 478(7367):64–69,
Med 313–319, 2011. 2011.
61. Thanopoulou E: Engraftment of NOD/SCID- 2 microglobulin null 84. Je EM, Yoo NJ, Kim YJ, et al: Mutational analysis of splicing machinery
mice with multilineage neoplastic cells from patients with myelodys- genes SF3B1, U2AF1 and SRSF2 in myelodysplasia and other common
plastic syndrome. Blood 103(11):4285–4293, 2004. tumors. Int J Cancer 133(1):260–265, 2013.
62. Gerritsen WR, Donohue J, Bauman J, et al: Clonal analysis of myelo- 85. Boultwood J, Dolatshad H, Varanasi SS, et al: The role of splicing
dysplastic syndrome: monosomy 7 is expressed in the myeloid lineage, factor mutations in the pathogenesis of the myelodysplastic syndromes.
but not in the lymphoid lineage as detected by fluorescent in situ Adv Biol Regul 54:153–161, 2014.
hybridization. Blood 80(1):217–224, 1992. 86. Visconte V, Rogers HJ, Singh J, et al: SF3B1 haploinsufficiency leads
63. White N, Nacheva E, Asimakopoulous F, et al: Deletion of chromo- to formation of ring sideroblasts in myelodysplastic syndromes. Blood
some 20q can occur in a multipotent precursor of both myeloid cells 120(16):3173–3186, 2012.
and B cells. Blood 83(10):2809–2816, 1994. 87. Malcovati L, Karimi M, Papaemmanuil E, et al: SF3B1 mutation
64. Woll PS, Kjällquist U, Chowdhury O, et al: Myelodysplastic syndromes identifies a distinct subset of myelodysplastic syndrome with ring
are propagated by rare and distinct human cancer stem cells in vivo. sideroblasts. Blood 126(2):233–241, 2015.
Cancer Cell 25(6):794–808, 2014. 88. Papaemmanuil E, Cazzola M, Boultwood J, et al: Somatic SF3B1
65. Will B, Zhou L, Vogler TO, et al: Stem and progenitor cells in myelo- mutation in myelodysplasia with ring sideroblasts. N Engl J Med
dysplastic syndromes show aberrant stage-specific expansion and harbor 365(15):1384–1395, 2011.
genetic and epigenetic alterations. Blood 120(10):2076–2086, 2012. 89. Damm F, Thol F, Kosmider O, et al: SF3B1 mutations in myelodys-
66. Jaiswal S, Ebert BL: MDS is a stem cell disorder after all. Cancer Cell plastic syndromes: clinical associations and prognostic implications.
25(6):713–714, 2014. Leukemia 26(5):1137–1140, 2012.
67. Corces-Zimmerman MR, Hong W-J, Weissman IL, et al: Preleukemic 90. Malcovati L, Papaemmanuil E, Bowen DT, et al: Clinical significance of
mutations in human acute myeloid leukemia affect epigenetic regulators SF3B1 mutations in myelodysplastic syndromes and myelodysplastic/
and persist in remission. Proc Natl Acad Sci USA 111(7):2548–2553, myeloproliferative neoplasms. Blood 118(24):6239–6246, 2011.
2014. 91. Thol F, Kade S, Schlarmann C, et al: Frequency and prognostic impact
68. Chao MP, Seita J, Weissman IL: Establishment of a normal hematopoi- of mutations in SRSF2, U2AF1, and ZRSR2 in patients with myelo-
etic and leukemia stem cell hierarchy [Internet]. In Cold Spring Harbor dysplastic syndromes. Blood 119(15):3578–3584, 2012.
symposia on quantitative biology, 2008, Cold Spring Harbor Laboratory 92. Meggendorfer M, Roller A, Haferlach T, et al: SRSF2 mutations in
Press [cited 2014 Dec 31]. p. sqb – 2008. Available from:: <http:// 275 cases with chronic myelomonocytic leukemia (CMML). Blood
symposium.cshlp.org/content/early/2008/11/06/sqb.2008.73.031 120(15):3080–3088, 2012.
.short>. 93. Wu S-J, Kuo Y-Y, Hou H-A, et al: The clinical implication of SRSF2
69. Bejar R, Levine R, Ebert BL: Unraveling the molecular pathophysiology mutation in patients with myelodysplastic syndrome and its stability
of myelodysplastic syndromes. J Clin Oncol 29(5):504–515, 2011. during disease evolution. Blood 120(15):3106–3111, 2012.
70. Jacobs RH, Cornbleet MA, Vardiman JW, et al: Prognostic implications 94. Graubert T, Shen D, Ding L: Recurrent mutations in the U2AF1
of morphology and karyotype in primary myelodysplastic syndromes. splicing factor in myelodysplastic syndromes. Nat Genet 44(1):53–U77,
Blood 67(6):1765–1772, 1986. 2012.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 969.e3

95. Okeyo-Owuor T, White BS, Chatrikhi R, et al: U2AF1 mutations 120. Wang J, Ai X, Gale RP, et al: TET2, ASXL1 and EZH2 mutations in
alter sequence specificity of pre-mRNA binding and splicing. Leukemia Chinese with myelodysplastic syndromes. Leuk Res 37(3):305–311,
2014. 2013.
96. Wu S-J, Tang J-L, Lin C-T, et al: Clinical implications of U2AF1 121. Ezhkova E, Pasolli HA, Parker JS, et al: Ezh2 orchestrates gene expres-
mutation in patients with myelodysplastic syndrome and its stability sion for the stepwise differentiation of tissue-specific stem cells. Cell
during disease progression. Am J Hematol 88(11):E277–E282, 2013. 136(6):1122–1135, 2009.
97. Langemeijer SMC, Kuiper RP, Berends M, et al: Acquired mutations 122. Carey BW, Finley LWS, Cross JR, et al: Intracellular α-ketoglutarate
in TET2 are common in myelodysplastic syndromes. Nat Genet maintains the pluripotency of embryonic stem cells. Nature 518:413–
41(7):838–842, 2009. 416, 2015.
98. Kosmider O, Gelsi-Boyer V, Ciudad M, et al: TET2 gene mutation 123. Wang X, Dai H, Wang Q, et al: EZH2 mutations are related to low
is a frequent and adverse event in chronic myelomonocytic leukemia. blast percentage in bone marrow and -7/del(7q) in de novo acute
Haematologica 94(12):1676–1681, 2009. myeloid leukemia. PLoS ONE 8(4):e61341, 2013.
99. Nakajima H, Kunimoto H: TET2 as an epigenetic master regulator for 124. Chen C, Liu Y, Rappaport AR, et al: MLL3 is a haploinsufficient 7q
normal and malignant hematopoiesis. Cancer Sci 105(9):1093–1099, tumor suppressor in acute myeloid leukemia. Cancer Cell 25(5):652–
2014. 665, 2014.
100. Wu H, Zhang Y: Mechanisms and functions of Tet protein-mediated 125. Jankowska AM, Makishima H, Tiu RV, et al: Mutational spectrum
5-methylcytosine oxidation. Genes Dev 25(23):2436–2452, 2011. analysis of chronic myelomonocytic leukemia includes genes associ-
101. Jankowska AM, Szpurka H, Tiu RV, et al: Loss of heterozygosity 4q24 ated with epigenetic regulation: UTX, EZH2, and DNMT3A. Blood
and TET2 mutations associated with myelodysplastic/myeloproliferative 118(14):3932–3941, 2011.
neoplasms. Blood 113(25):6403–6410, 2009. 126. Ernst T, Chase AJ, Score J, et al: Inactivating mutations of the
102. Yamazaki J, Taby R, Vasanthakumar A, et al: Effects of TET2 mutations histone methyltransferase gene EZH2 in myeloid disorders. Nat Genet
on DNA methylation in chronic myelomonocytic leukemia. Epigenetics 42(8):722–726, 2010.
7(2):201–207, 2012. 127. Nikoloski G, Langemeijer SMC, Kuiper RP, et al: Somatic mutations
103. Bejar R, Lord A, Stevenson K, et al: TET2 mutations predict response of the histone methyltransferase gene EZH2 in myelodysplastic syn-
to hypomethylating agents in myelodysplastic syndrome patients. Blood dromes. Nat Genet 42(8):665–667, 2010.
124(17):2705–2712, 2014. 128. Losman J-A, Looper RE, Koivunen P, et al: R)-2-hydroxyglutarate
104. Bejar R, Stevenson KE, Caughey B, et al: Somatic mutations predict is sufficient to promote leukemogenesis and its effects are reversible.
poor outcome in patients with myelodysplastic syndrome after hema- Science 339(6127):1621–1625, 2013.
topoietic stem-cell transplantation. J Clin Oncol 32(25):2691–2698, 129. Figueroa ME, Abdel-Wahab O, Lu C, et al: Leukemic IDH1 and
2014. IDH2 mutations result in a hypermethylation phenotype, disrupt
105. Yang L, Rau R, Goodell MA: DNMT3A in haematological malignan- TET2 function, and impair hematopoietic differentiation. Cancer Cell
cies. Nat Rev Cancer 15(3):152–165, 2015. 18(6):553–567, 2010.
106. Ley TJ, Ding L, Walter MJ, et al: DNMT3A mutations in acute 130. Ito Y, Bae S-C, Chuang LSH: The RUNX family: developmental
myeloid leukemia. N Engl J Med 363(25):2424–2433, 2010. regulators in cancer. Nat Rev Cancer 15(2):81–95, 2015.
107. Russler-Germain DA, Spencer DH, Young MA, et al: The R882H 131. Miyoshi H, Shimizu K, Kozu T, et al: t(8;21) breakpoints on chromo-
DNMT3A mutation associated with AML dominantly inhibits wild- some 21 in acute myeloid leukemia are clustered within a limited region
type DNMT3A by blocking its ability to form active tetramers. Cancer of a single gene, AML1. Proc Natl Acad Sci USA 88(23):10431–10434,
Cell 25(4):442–454, 2014. 1991.
108. Challen GA, Sun D, Jeong M, et al: Dnmt3a is essential for hemato- 132. Golub TR, Barker GF, Bohlander SK, et al: Fusion of the TEL gene on
poietic stem cell differentiation. Nat Genet 44(1):23–31, 2011. 12p13 to the AML1 gene on 21q22 in acute lymphoblastic leukemia.
109. Mayle A, Yang L, Rodriguez B, et al: Dnmt3a loss predisposes murine Proc Natl Acad Sci USA 92(11):4917–4921, 1995.
hematopoietic stem cells to malignant transformation. Blood 2014. 133. Harada H, Harada Y, Niimi H, et al: High incidence of somatic
110. Walter MJ, Ding L, Shen D, et al: Recurrent DNMT3A mutations in mutations in the AML1/RUNX1 gene in myelodysplastic syndrome
patients with myelodysplastic syndromes. Leukemia 25(7):1153–1158, and low blast percentage myeloid leukemia with myelodysplasia. Blood
2011. 103(6):2316–2324, 2004.
111. Thol F, Winschel C, Lüdeking A, et al: Rare occurrence of DNMT3A 134. Harada Y, Harada H: Molecular pathways mediating MDS/AML with
mutations in myelodysplastic syndromes. Haematologica 96(12):1870– focus on AML1/RUNX1 point mutations. J Cell Physiol 220(1):16–20,
1873, 2011. 2009.
112. Bejar R, Stevenson KE, Caughey BA, et al: Validation of a prognostic 135. Matsuura S, Komeno Y, Stevenson KE, et al: Expression of the runt
model and the impact of mutations in patients with lower-risk myelo- homology domain of RUNX1 disrupts homeostasis of hematopoietic
dysplastic syndromes. J Clin Oncol 30(27):3376–3382, 2012. stem cells and induces progression to myelodysplastic syndrome. Blood
113. Bejar R: Clinical and genetic predictors of prognosis in myelodysplastic 120(19):4028–4037, 2012.
syndromes. Haematologica 99(6):956–964, 2014. 136. Huang G, Zhao X, Wang L, et al: The ability of MLL to bind RUNX1
114. Gelsi-Boyer V, Trouplin V, Adélaïde J, et al: Mutations of polycomb- and methylate H3K4 at PU. 1 regulatory regions is impaired by MDS/
associated gene ASXL1 in myelodysplastic syndromes and chronic AML-associated RUNX1/AML1 mutations. Blood 118(25):6544–6552,
myelomonocytic leukaemia. Br J Haematol 145(6):788–800, 2009. 2011.
115. Fisher CL, Pineault N, Brookes C, et al: Loss-of-function additional sex 137. Fears S, Gavin M, Zhang DE, et al: Functional characterization of
combs like 1 mutations disrupt hematopoiesis but do not cause severe ETV6 and ETV6/CBFA2 in the regulation of the MCSFR proximal
myelodysplasia or leukemia. Blood 115(1):38–46, 2010. promoter. Proc Natl Acad Sci USA 94(5):1949–1954, 1997.
116. Abdel-Wahab O, Adli M, LaFave LM, et al: ASXL1 mutations promote 138. Peeters P, Wlodarska I, Baens M, et al: Fusion of ETV6 to MDS1/EVI1
myeloid transformation through loss of PRC2-mediated gene repres- as a result of t(3;12)(q26;p13) in myeloproliferative disorders. Cancer
sion. Cancer Cell 22(2):180–193, 2012. Res 57(4):564–569, 1997.
117. Abdel-Wahab O, Gao J, Adli M, et al: Deletion of Asxl1 results in 139. Wlodarska I, Selleri L, La Starza R, et al: Molecular cytogenetics local-
myelodysplasia and severe developmental defects in vivo. J Exp Med izes two new breakpoints on 11q23.3 and 21q11.2 in myelodysplastic
210(12):2641–2659, 2013. syndrome with t(11;21) translocation. Genes Chromosomes Cancer
118. Boultwood J, Perry J, Pellagatti A, et al: Frequent mutation of the 24(3):199–206, 1999.
polycomb-associated gene ASXL1 in the myelodysplastic syndromes 140. Padron E, Yoder S, Kunigal S, et al: ETV6 and signaling gene muta-
and in acute myeloid leukemia. Leukemia 24(5):1062–1065, 2010. tions are associated with secondary transformation of myelodysplastic
119. Carbuccia N, Murati A, Trouplin V, et al: Mutations of ASXL1 gene syndromes to chronic myelomonocytic leukemia. Blood 123(23):3675–
in myeloproliferative neoplasms. Leukemia 23(11):2183–2186, 2009. 3677, 2014.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
969.e4 Part VII  Hematologic Malignancies

141. Hyde RK, Liu PP: GATA2 mutations lead to MDS and AML. Nat 163. Christiansen DH, Andersen MK, Desta F, et al: Mutations of genes
Genet 43(10):926–927, 2011. in the receptor tyrosine kinase (RTK)/RAS-BRAF signal transduction
142. Ostergaard P, Simpson MA, Connell FC, et al: Mutations in GATA2 pathway in therapy-related myelodysplasia and acute myeloid leukemia.
cause primary lymphedema associated with a predisposition to acute Leukemia 19(12):2232–2240, 2005.
myeloid leukemia (Emberger syndrome). Nat Genet 43(10):929–931, 164. Score J, Hidalgo-Curtis C, Jones AV, et al: Inactivation of poly-
2011. comb repressive complex 2 components in myeloproliferative and
143. Hsu AP, Sampaio EP, Khan J, et al: Mutations in GATA2 are associated myelodysplastic/myeloproliferative neoplasms. Blood 119(5):1208–
with the autosomal dominant and sporadic monocytopenia and myco- 1213, 2012.
bacterial infection (MonoMAC) syndrome. Blood 118(10):2653–2655, 165. Makishima H, Cazzolli H, Szpurka H, et al: Mutations of e3 ubiquitin
2011. ligase cbl family members constitute a novel common pathogenic lesion
144. Dickinson RE, Griffin H, Bigley V, et al: Exome sequencing identifies in myeloid malignancies. J Clin Oncol 27(36):6109–6116, 2009.
GATA-2 mutation as the cause of dendritic cell, monocyte, B and NK 166. Di Giacomo D, Lema Fernandez AG, Pierini T, et al: The GNAS1 gene
lymphoid deficiency. Blood 118(10):2656–2658, 2011. in myelodysplastic syndromes (MDS). Leuk Res 38(7):804–807, 2014.
145. Dickinson RE, Milne P, Jardine L, et al: The evolution of cellular 167. Yoda A, Adelmant G, Tamburini J, et al: Mutations in G protein β
deficiency in GATA2 mutation. Blood 123(6):863–874, 2014. subunits promote transformation and kinase inhibitor resistance. Nat
146. Micol J-B, Abdel-Wahab O: Collaborating constitutive and somatic Med 21(1):71–75, 2014.
genetic events in myeloid malignancies: ASXL1 mutations in patients 168. Jankowska AM, Gondek LP, Szpurka H, et al: Base excision repair
with germline GATA2 mutations. Haematologica 99(2):201–203, 2014. dysfunction in a subgroup of patients with myelodysplastic syndrome.
147. Kaneko H, Misawa S, Horiike S, et al: TP53 mutations emerge at early Leukemia 22(3):551–558, 2008.
phase of myelodysplastic syndrome and are associated with complex 169. Kawankar N, Vundinti BR: Cytogenetic abnormalities in myelodysplas-
chromosomal abnormalities. Blood 85(8):2189–2193, 1995. tic syndrome: an overview. Hematology 16(3):131–138, 2011.
148. Lindsley RC, Mar BG, Mazzola E, et al: Acute myeloid leukemia ontog- 170. Swerdlow S, Campo E, Harris N, et al WHO Classification of Tumours
eny is defined by distinct somatic mutations. Blood 125:1367–1376, of Haematopoietic and Lymphoid Tissue (IARC WHO Classification of
2015. Tumours). 4th ed. 2008.
149. Jadersten M, Saft L, Smith A: TP53 mutations in low-risk myelodys- 171. Ebert BL: Molecular dissection of the 5q deletion in myelodysplastic
plastic syndromes with del(5q) predict disease progression. J Clin Oncol syndrome. Semin Oncol 38(5):621–626, 2011.
29(15):1971–1979, 2011. 172. Pedersen B: Anatomy of the 5q- deletion: different sex ratios and
150. Wong TN, Ramsingh G, Young AL, et al: Role of TP53 mutations in deleted 5q bands in MDS and AML. Leukemia 10(12):1883–1890,
the origin and evolution of therapy-related acute myeloid leukaemia. 1996.
Nature 518:552–555, 2014. 173. List A, Dewald G, Bennett J, et al: Lenalidomide in the myelodysplastic
151. Kottaridis PD, Gale RE, Frew ME, et al: The presence of a FLT3 syndrome with chromosome 5q deletion. N Engl J Med 355(14):1456–
internal tandem duplication in patients with acute myeloid leukemia 1465, 2006.
(AML) adds important prognostic information to cytogenetic risk 174. Boultwood J, Pellagatti A, Wainscoat JS: Haploinsufficiency of
group and response to the first cycle of chemotherapy: analysis of 854 ribosomal proteins and p53 activation in anemia: Diamond-Blackfan
patients from the United Kingdom Medical Research Council AML 10 anemia and the 5q- syndrome. Adv Biol Regul 52(1):196–203, 2012.
and 12 trials. Blood 98(6):1752–1759, 2001. 175. Ebert BL, Pretz J, Bosco J, et al: Identification of RPS14 as a 5q- syn-
152. Kralovics R, Passamonti F, Buser AS, et al: A gain-of-function mutation drome gene by RNA interference screen. Nature 451(7176):335–339,
of JAK2 in myeloproliferative disorders. N Engl J Med 352(17):1779– 2008.
1790, 2005. 176. Kumar MS, Narla A, Nonami A, et al: Coordinate loss of a microRNA
153. Pardanani AD, Levine RL, Lasho T, et al: MPL515 mutations in myelo- and protein-coding gene cooperate in the pathogenesis of 5q- syndrome.
proliferative and other myeloid disorders: a study of 1182 patients. Blood 118(17):4666–4673, 2011.
Blood 108(10):3472–3476, 2006. 177. Schneider RK, Ademà V, Heckl D, et al: Role of casein kinase 1A1
154. Furitsu T, Tsujimura T, Tono T, et al: Identification of mutations in in the biology and targeted therapy of del(5q) MDS. Cancer Cell
the coding sequence of the proto-oncogene c-kit in a human mast 26(4):509–520, 2014.
cell leukemia cell line causing ligand-independent activation of c-kit 178. Stoddart A, Fernald AA, Wang J, et al: Haploinsufficiency of del(5q)
product. J Clin Invest 92(4):1736–1744, 1993. genes, Egr1 and Apc, cooperate with Tp53 loss to induce acute myeloid
155. Bains A, Luthra R, Medeiros LJ, et al: FLT3 and NPM1 mutations leukemia in mice. Blood 123(7):1069–1078, 2014.
in myelodysplastic syndromes: Frequency and potential value for 179. Chen TH-P, Kambal A, Krysiak K, et al: Knockdown of Hspa9, a
predicting progression to acute myeloid leukemia. Am J Clin Pathol del(5q31.2) gene, results in a decrease in hematopoietic progenitors in
135(1):62–69, 2011. mice. Blood 117(5):1530–1539, 2011.
156. Gallagher A, Darley R, Padua RA: RAS and the myelodysplastic 180. Sportoletti P, Grisendi S, Majid SM, et al: Npm1 is a haploinsufficient
syndromes. Pathol Biol (Paris) 45(7):561–568, 1997. suppressor of myeloid and lymphoid malignancies in the mouse. Blood
157. Side LE, Curtiss NP, Teel K, et al: RAS, FLT3, and TP53 mutations 111(7):3859–3862, 2008.
in therapy-related myeloid malignancies with abnormalities of chromo- 181. Horrigan SK, Arbieva ZH, Xie HY, et al: Delineation of a minimal
somes 5 and 7. Genes Chromosomes Cancer 39(3):217–223, 2004. interval and identification of 9 candidates for a tumor suppressor gene
158. Steensma DP, Dewald GW, Lasho TL, et al: The JAK2 V617F activat- in malignant myeloid disorders on 5q31. Blood 95(7):2372–2377,
ing tyrosine kinase mutation is an infrequent event in both “atypical” 2000.
myeloproliferative disorders and myelodysplastic syndromes. Blood 182. Ebert BL: Genetic deletions in AML and MDS. Best Pract Res Clin
106(4):1207–1209, 2005. Haematol 23(4):457–461, 2010.
159. Hellstrom-Lindberg E, Cazzola M: The role of JAK2 mutations in 183. Kere J, Ruutu T, la Chapelle A: de. Monosomy 7 in granulocytes and
RARS and other MDS. Hematology Am Soc Hematol Educ Program monocytes in myelodysplastic syndrome. N Engl J Med 316(9):499–503,
52–59, 2008. 1987.
160. Kon A, Shih L-Y, Minamino M, et al: Recurrent mutations in multiple 184. Wong CC, Martincorena I, Rust AG, et al: Inactivating CUX1 muta-
components of the cohesin complex in myeloid neoplasms. Nat Genet tions promote tumorigenesis. Nat Genet 46(1):33–38, 2014.
45(10):1232–1237, 2013. 185. Le Beau M, Espinosa R, 3rd, Davis E, et al: Cytogenetic and molecular
161. Thota S, Viny A, Makishima H: Genetic alterations of the cohesin delineation of a region of chromosome 7 commonly deleted in malig-
complex genes in myeloid malignancies. Blood 8:2014–2024, 2014. nant myeloid diseases. Blood 88:1930–1935, 1996.
162. Tartaglia M, Niemeyer CM, Fragale A, et al: Somatic mutations in 186. Cordoba I, González-Porras JR, Nomdedeu B, et al: Better prognosis
PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syn- for patients with del(7q) than for patients with monosomy 7 in myelo-
dromes and acute myeloid leukemia. Nat Genet 34(2):148–150, 2003. dysplastic syndrome. Cancer 118(1):127–133, 2012.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 969.e5

187. Saumell S, Florensa L, Luño E, et al: Prognostic value of trisomy 8 as a 206. Rubin CM, Larson RA, Anastasi J, et al: t(3;21)(q26;q22): a recurring
single anomaly and the influence of additional cytogenetic aberrations chromosomal abnormality in therapy-related myelodysplastic syndrome
in primary myelodysplastic syndromes. Br J Haematol 159(3):311–321, and acute myeloid leukemia. Blood 76(12):2594–2598, 1990.
2012. 207. Testoni N, Borsaru G, Martinelli G, et al: 3q21 and 3q26 cytogenetic
188. Sloand EM, Mainwaring L, Fuhrer M, et al: Preferential suppression abnormalities in acute myeloblastic leukemia: biological and clinical
of trisomy 8 compared with normal hematopoietic cell growth by features. Haematologica 84(8):690–694, 1999.
autologous lymphocytes in patients with trisomy 8 myelodysplastic 208. Gupta M, Ashok Kumar J, Sitaram U, et al: The t(6;9)(p22;q34) in
syndrome. Blood 106(3):841–851, 2005. myeloid neoplasms: a retrospective study of 16 cases. Cancer Genet
189. Nilsson L, Astrand-Grundström I, Anderson K, et al: Involvement and Cytogenet 203(2):297–302, 2010.
functional impairment of the CD34(+)CD38(-)Thy-1(+) hematopoi- 209. Boulais PE, Frenette PS: Making sense of hematopoietic stem cell
etic stem cell pool in myelodysplastic syndromes with trisomy 8. Blood niches. Blood 2015.
100(1):259–267, 2002. 210. Kfoury Y, Scadden DT: Mesenchymal Cell Contributions to the Stem
190. Sloand EM: Preferential suppression of trisomy 8 compared with normal Cell Niche. Cell Stem Cell 16(3):239–253, 2015.
hematopoietic cell growth by autologous lymphocytes in patients with 211. Verhoef GE, De Schouwer P, Ceuppens JL, et al: Measurement of
trisomy 8 myelodysplastic syndrome. Blood 106(3):841–851, 2005. serum cytokine levels in patients with myelodysplastic syndromes.
191. Toyonaga T, Nakase H, Matsuura M, et al: Refractoriness of intestinal Leukemia 6(12):1268–1272, 1992.
Behçet’s disease with myelodysplastic syndrome involving trisomy 8 212. Schipperus MR, Sonneveld P, Lindemans J, et al: The combined effects
to medical therapies - our case experience and review of the literature. of Il-3, GM-CSF and G-CSF on the in vitro growth of myelodysplastic
Digestion 88(4):217–221, 2013. myeloid progenitor cells. Leuk Res 14(11–12):1019–1025, 1990.
192. Haase D, Germing U, Schanz J, et al: New insights into the prognostic 213. Vas V, Senger K, Dörr K, et al: Aging of the microenvironment influ-
impact of the karyotype in MDS and correlation with subtypes: evi- ences clonality in hematopoiesis. PLoS ONE 7(8):e42080, 2012.
dence from a core dataset of 2124 patients. Blood 110(13):4385–4395, 214. Raaijmakers MHGP, Mukherjee S, Guo S, et al: Bone progenitor
2007. dysfunction induces myelodysplasia and secondary leukaemia. Nature
193. Bacher U, Haferlach T, Schnittger S, et al: Investigation of 305 patients 464(7290):852–857, 2010.
with myelodysplastic syndromes and 20q deletion for associated cyto- 215. Santamaria C, Muntion S, Roson B, et al: Impaired expression of
genetic and molecular genetic lesions and their prognostic impact. Br DICER, DROSHA, SBDS and some microRNAs in mesenchymal
J Haematol 164(6):822–833, 2014. stromal cells from myelodysplastic syndrome patients. Haematologica
194. Braun T, de Botton S, Taksin A-L, et al: Characteristics and outcome of 97(8):1218–1224, 2012.
myelodysplastic syndromes (MDS) with isolated 20q deletion: a report 216. Kode A, Manavalan JS, Mosialou I, et al: Leukaemogenesis induced by
on 62 cases. Leuk Res 35(7):863–867, 2011. an activating β-catenin mutation in osteoblasts. Nature 506(7487):240–
195. Clarke M, Dumon S, Ward C, et al: MYBL2 haploinsufficiency 244, 2014.
increases susceptibility to age-related haematopoietic neoplasia. Leuke- 217. Warlick ED, Miller JS: Myelodysplastic syndromes: the role of the
mia 27(3):661–670, 2013. immune system in pathogenesis. Leuk Lymphoma 52(11):2045–2049,
196. Heinrichs S, Conover LF, Bueso-Ramos CE, et al: MYBL2 is a sub- 2011.
haploinsufficient tumor suppressor gene in myeloid malignancy. Elife 218. Wei Y, Dimicoli S, Bueso-Ramos C, et al: Toll-like receptor alterations
2:e00825, 2013. in myelodysplastic syndrome. Leukemia 27(9):1832–1840, 2013.
197. Wang PW, Eisenbart JD, Espinosa R, et al: Refinement of the smallest 219. Maratheftis CI, Andreakos E, Moutsopoulos HM, et al: Toll-like
commonly deleted segment of chromosome 20 in malignant myeloid receptor-4 is up-regulated in hematopoietic progenitor cells and
diseases and development of a PAC-based physical and transcription contributes to increased apoptosis in myelodysplastic syndromes. Clin
map. Genomics 67(1):28–39, 2000. Cancer Res 13(4):1154–1160, 2007.
198. Soenen V, Preudhomme C, Roumier C, et al: 17p Deletion in 220. Starczynowski DT, Kuchenbauer F, Argiropoulos B, et al: Identifica-
acute myeloid leukemia and myelodysplastic syndrome. Analysis tion of miR-145 and miR-146a as mediators of the 5q- syndrome
of breakpoints and deleted segments by fluorescence in situ. Blood phenotype. Nat Med 16(1):49–58, 2010.
91(3):1008–1015, 1998. 221. Keerthivasan G, Mei Y, Zhao B, et al: Aberrant overexpression of
199. Jary L, Mossafa H, Fourcade C, et al: The 17p-syndrome: a distinct CD14 on granulocytes sensitizes the innate immune response in mDia1
myelodysplastic syndrome entity? Leuk Lymphoma 25(1–2):163–168, heterozygous del(5q) MDS. Blood 124(5):780–790, 2014.
1997. 222. Chen X, Eksioglu EA, Zhou J, et al: Induction of myelodysplasia by
200. Patnaik MM, Hanson CA, Hodnefield JM, et al: Monosomal karyotype myeloid-derived suppressor cells. J Clin Invest 123(11):4595–4611,
in myelodysplastic syndromes, with or without monosomy 7 or 5, is 2013.
prognostically worse than an otherwise complex karyotype. Leukemia 223. Kristinsson SY, Björkholm M, Hultcrantz M, et al: Chronic immune
25(2):266–270, 2011. stimulation might act as a trigger for the development of acute myeloid
201. Valcarcel D, Adema V, Sole F: Complex, not monosomal, karyotype is leukemia or myelodysplastic syndromes. J Clin Oncol 29(21):2897–
the cytogenetic marker of poorest prognosis in patients with primary 2903, 2011.
myelodysplastic syndrome. J Clin Oncol 31(7):916–922, 2013. 224. Nand S, Godwin JE: Hypoplastic myelodysplastic syndrome. Cancer
202. Wong AK, Fang B, Zhang L, et al: Loss of the Y chromosome: an 62(5):958–964, 1988.
age-related or clonal phenomenon in acute myelogenous leukemia/ 225. Epperson DE, Nakamura R, Saunthararajah Y, et al: Oligoclonal T cell
myelodysplastic syndrome? Arch Pathol Lab Med 132(8):1329–1332, expansion in myelodysplastic syndrome: evidence for an autoimmune
2008. process. Leuk Res 25(12):1075–1083, 2001.
203. Pedersen-Bjergaard J, Pedersen M, Roulston D, et al: Different genetic 226. Anderson RW, Volsky DJ, Greenberg B, et al: Lymphocyte abnormali-
pathways in leukemogenesis for patients presenting with therapy-related ties in preleukemia–I. Decreased NK activity, anomalous immunoregu-
myelodysplasia and therapy-related acute myeloid leukemia. Blood latory cell subsets and deficient EBV receptors. Leuk Res 7(3):389–395,
86(9):3542–3552, 1995. 1983.
204. Wlodarska I, Marynen P, La Starza R, et al: The ETV6, CDKN1B and 227. Knox SJ, Greenberg BR, Anderson RW, et al: Studies of T-lymphocytes
D12S178 loci are involved in a segment commonly deleted in various in preleukemic disorders and acute nonlymphocytic leukemia: in vitro
12p aberration in different hematological malignancies. Cytogenet Cell radiosensitivity, mitogenic responsiveness, colony formation, and
Genet 72(2–3):229–235, 1996. enumeration of lymphocytic subpopulations. Blood 61(3):449–455,
205. Pedersen-Bjergaard J, Philip P: Balanced translocations involving 1983.
chromosome bands 11q23 and 21q22 are highly characteristic of 228. Volsky DJ, Anderson RW: Deficiency in Epstein-Barr virus receptors on
myelodysplasia and leukemia following therapy with cytostatic agents B-lymphocytes of preleukemia patients. Cancer Res 43(8):3923–3926,
targeting at DNA-topoisomerase II. Blood 78(4):1147–1148, 1991. 1983.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
969.e6 Part VII  Hematologic Malignancies

229. Parker JE, Fishlock KL, Mijovic A, et al: Low-risk” myelodysplastic 251. Chanarin I: Megaloblastic anaemia, cobalamin, and folate. J Clin Pathol
syndrome is associated with excessive apoptosis and an increased ratio 40(9):978–984, 1987.
of pro- versus anti-apoptotic bcl-2-related proteins. Br J Haematol 252. Aitelli C, Wasson L, Page R: Pernicious anemia: presentations mimick-
103(4):1075–1082, 1998. ing acute leukemia. South Med J 97(3):295–297, 2004.
230. Letai A: Restoring cancer’s death sentence. Cancer Cell 10(5):343–345, 253. Stabler SP: Clinical practice. Vitamin B12 deficiency. N Engl J Med
2006. 368(2):149–160, 2013.
231. Reza S, Shetty V, Dar S, et al: Tumor necrosis factor-alpha levels 254. Balsa JA, Botella-Carretero JI, Gómez-Martín JM, et al: Copper and
decrease with anticytokine therapy in patients with myelodysplastic zinc serum levels after derivative bariatric surgery: differences between
syndromes. J Interferon Cytokine Res 18(10):871–877, 1998. Roux-en-Y Gastric bypass and biliopancreatic diversion. Obes Surg
232. Shetty V, Mundle S, Alvi S, et al: Measurement of apoptosis, prolifera- 21(6):744–750, 2011.
tion and three cytokines in 46 patients with myelodysplastic syndromes. 255. Fiske DN, McCoy HE, Kitchens CS: Zinc-induced sideroblastic
Leuk Res 20(11–12):891–900, 1996. anemia: report of a case, review of the literature, and description of
233. Letai AG: Diagnosing and exploiting cancer’s addiction to blocks in the hematologic syndrome. Am J Hematol 46(2):147–150, 1994.
apoptosis. Nat Rev Cancer 8(2):121–132, 2008. 256. Halfdanarson TR, Kumar N, Li C-Y, et al: Hematological manifes-
234. Parker JE, Mufti GJ, Rasool F, et al: The role of apoptosis, proliferation, tations of copper deficiency: a retrospective review. Eur J Haematol
and the Bcl-2-related proteins in the myelodysplastic syndromes and 80(6):523–531, 2008.
acute myeloid leukemia secondary to MDS. Blood 96(12):3932–3938, 257. Imataki O, Ohnishi H, Kitanaka A, et al: Pancytopenia complicated
2000. with peripheral neuropathy due to copper deficiency: clinical diagnostic
235. Slape CI, Saw J, Jowett JBM, et al: Inhibition of apoptosis by BCL2 review. Intern Med 47(23):2063–2065, 2008.
prevents leukemic transformation of a murine myelodysplastic syn- 258. Huff JD, Keung Y-K, Thakuri M, et al: Copper deficiency causes revers-
drome. Blood 120(12):2475–2483, 2012. ible myelodysplasia. Am J Hematol 82(7):625–630, 2007.
236. Beurlet S, Omidvar N, Gorombei P, et al: BCL-2 inhibition with 259. Chanarin I: Alcohol and haemopoiesis. Br J Haematol 17(6):515–516,
ABT-737 prolongs survival in an NRAS/BCL-2 mouse model of AML 1969.
by targeting primitive LSK and progenitor cells. Blood 122(16):2864– 260. Scott JM, Weir DG: Drug-induced megaloblastic change. Clin Haema-
2876, 2013. tol 9(3):587–606, 1980.
237. Bogenberger JM, Kornblau SM, Pierceall WE, et al: BCL-2 family pro- 261. Hongeng S, May W, Crist WM: Transient myeloid dysplasia due to
teins as 5-Azacytidine-sensitizing targets and determinants of response valproic acid. Clin Pediatr (Phila) 36(6):361–364, 1997.
in myeloid malignancies. Leukemia 28(8):1657–1665, 2014. 262. Kennedy GA, Kay TD, Johnson DW, et al: Neutrophil dysplasia char-
238. Takahashi K, Jabbour E, Wang X, et al: Dynamic acquisition of FLT3 acterised by a pseudo-Pelger-Huet anomaly occurring with the use of
or RAS alterations drive a subset of patients with lower risk MDS to mycophenolate mofetil and ganciclovir following renal transplantation:
secondary AML. Leukemia 27(10):2081–2083, 2013. a report of five cases. Pathology (Phila) 34(3):263–266, 2002.
239. Murphy DM, Bejar R, Stevenson K, et al: NRAS mutations with low 263. Taegtmeyer AB, Halil O, Bell AD, et al: Neutrophil dysplasia (acquired
allele burden have independent prognostic significance for patients with pseudo-pelger anomaly) caused by ganciclovir. Transplantation 80(1):
lower risk myelodysplastic syndromes. Leukemia 27(10):2077–2081, 127–130, 2005.
2013. 264. Gibbs SDJ, Westerman DA, McCormack C, et al: Severe and pro-
240. Lorenzo F, Nishii K, Monma F, et al: Mutational analysis of the KIT longed myeloid haematopoietic toxicity with myelodysplastic features
gene in myelodysplastic syndrome (MDS) and MDS-derived leukemia. following alemtuzumab therapy in patients with peripheral T-cell
Leuk Res 30(10):1235–1239, 2006. lymphoproliferative disorders. Br J Haematol 130(1):87–91, 2005.
241. Kaeferstein A, Krug U, Tiesmeier J, et al: The emergence of a C/ 265. Frewin RJ, Provan D, Smith AG: Myelodysplasia occurring after
EBPalpha mutation in the clonal evolution of MDS towards secondary fludarabine treatment for chronic lymphocytic leukaemia. Clin Lab
AML. Leukemia 17(2):343–349, 2003. Haematol 19(2):151–152, 1997.
242. Pagano L, Pulsoni A, Vignetti M, et al: Secondary acute myeloid 266. Buchbinder R, Hall S, Ryan PF, et al: Severe bone marrow failure due
leukaemia: results of conventional treatments. Experience of GIMEMA to low dose oral methotrexate. J Rheumatol 15(10):1586–1588, 1988.
trials. Ann Oncol 16(2):228–233, 2005. 267. Piso RJ, Kriz K, Desax M-C: Severe isoniazid related sideroblastic
243. Leone G, Mele L, Pulsoni A, et al: The incidence of secondary leuke- anemia. Hematol Rep 3(1):e2, 2011.
mias. Haematologica 84(10):937–945, 1999. 268. Beck EA, Ziegler G, Schmid R, et al: Reversible sideroblastic anemia
244. Gerstung M, Pellagatti A, Malcovati L, et al: Combining gene mutation caused by chloramphenicol. Acta Haematol 38(1):1–10, 1967.
with gene expression data improves outcome prediction in myelodys- 269. Verwilghen R, Reybrouck G, Callens L, et al: Antituberculous drugs
plastic syndromes. Nat Commun 6:5901, 2015. and sideroblastic anaemia. Br J Haematol 11:92–98, 1965.
245. Kobayashi Y, Nakayama M, Uemura N, et al: Analysis of myelo- 270. Colucci G, Silzle T, Solenthaler M: Pyrazinamide-induced sideroblastic
dysplastic syndrome clones arising after multiple myeloma: a case anemia. Am J Hematol 87(3):305, 2012.
study by correlative interphase cytogenetic analysis. Jpn J Clin Oncol 271. Haden HT: Pyridoxine-responsive sideroblastic anemia due to antitu-
29(8):374–377, 1999. berculous drugs. Arch Intern Med 120(5):602–606, 1967.
246. Lorand-Metze I, Lima CS, Cardinalli IA, et al: Association of a 272. Scott J, Finegold S, Belkin G, et al: A controlled double-blind
myelodysplastic syndrome with hairy cell leukaemia. Eur J Haematol study of the hematologic toxicity of chloramphenicol. N Engl J Med
55(5):341–343, 1995. 272:1137–1142, 1965.
247. Mitterbauer G, Schwarzmeier J, Mitterbauer M, et al: Myelodysplastic 273. Karcher DS, Frost AR: The bone marrow in human immunodeficiency
syndrome/acute myeloid leukemia supervening previously untreated virus (HIV)-related disease. Morphology and clinical correlation. Am J
chronic B-lymphocytic leukemia: demonstration of the concomitant Clin Pathol 95(1):63–71, 1991.
presence of two different malignant clones by immunologic and 274. Scadden DT, Zon LI, Groopman JE: Pathophysiology and management
molecular analysis. Ann Hematol 74(4):193–197, 1997. of HIV-associated hematologic disorders. Blood 74(5):1455–1463,
248. Wells DA, Hall MC, Shulman HM, et al: Occult B cell malignan- 1989.
cies can be detected by three-color flow cytometry in patients with 275. Recommendation Summary - US Preventive Services Task Force
cytopenias. Leukemia 12(12):2015–2023, 1998. [Internet]. [cited 2015 Mar 17];Available from: <http://www
249. Saunthararajah Y, Molldrem JL, Rivera M, et al: Coincident myelodys- .uspreventiveservicestaskforce.org/Page/Topic/recommendation-summary/
plastic syndrome and T-cell large granular lymphocytic disease: clinical human-immunodeficiency-virus-hiv-infection-screening>.
and pathophysiological features. Br J Haematol 112(1):195–200, 2001. 276. Kasahara S, Hara T, Itoh H, et al: Hypoplastic myelodysplastic syn-
250. Steensma DP: Dysplasia has A differential diagnosis: distinguishing dromes can be distinguished from acquired aplastic anaemia by bone
genuine myelodysplastic syndromes (MDS) from mimics, imitators, marrow stem cell expression of the tumour necrosis factor receptor. Br
copycats and impostors. Curr Hematol Malig Rep 7(4):310–320, 2012. J Haematol 118(1):181–188, 2002.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 969.e7

277. Pagliuca A, Layton DM, Manoharan A, et al: Myelofibrosis in primary 302. Kornberg A, Goldfarb A: Preleukemia manifested by hemolytic anemia
myelodysplastic syndromes: a clinico-morphological study of 10 cases. with pyruvate-kinase deficiency. Arch Intern Med 146(4):785–786,
Br J Haematol 71(4):499–504, 1989. 1986.
278. Nangalia J, Green TR: The evolving genomic landscape of myelo- 303. Hast R: Sideroblasts in myelodysplasia: their nature and clinical signifi-
proliferative neoplasms. Hematology Am Soc Hematol Educ Program cance. Scand J Haematol Suppl 45:53–55, 1986.
2014(1):287–296, 2014. 304. Steensma DP, Higgs DR, Fisher CA, et al: Acquired somatic ATRX
279. Steensma DP, Heptinstall KV, Johnson VM, et al: Common trouble- mutations in myelodysplastic syndrome associated with alpha thalas-
some symptoms and their impact on quality of life in patients with semia (ATMDS) convey a more severe hematologic phenotype than
myelodysplastic syndromes (MDS): results of a large internet-based germline ATRX mutations. Blood 103(6):2019–2026, 2004.
survey. Leuk Res 32(5):691–698, 2008. 305. Annino L, Di Giovanni S, Tentorì L, et al: Acquired hemoglobin H
280. Pomeroy C, Oken MM, Rydell RE, et al: Infection in the myelodys- disease in a case of refractory anemia with excess of blasts (RAEB) evolv-
plastic syndromes. Am J Med 90(3):338–344, 1991. ing into acute nonlymphoid leukemia. Acta Haematol 72(1):41–44,
281. Raman B, Van Slyck E, Riddle J, et al: Platelet function and structure 1984.
in myeloproliferative disease, myelodysplastic syndrome, and secondary 306. French Registry of Acute Leukemia and Myelodysplastic Syndromes:
thrombocytosis. Am J Clin Pathol 647:91, 1989. Age distribution and hemogram analysis of the 4496 cases recorded
282. Avivi I, Rosenbaum H, Levy Y, et al: Myelodysplastic syndrome and during and classified according to criteria FAB, Groupe Francais de
associated skin lesions: a review of the literature. Leuk Res 23(4):323– Morphologie Hematologique. Cancer 60:1982–1983, 1987. 1385.
330, 1999. 307. Cech P, Markert M, Perrin LH: Partial myeloperoxidase deficiency in
283. Rochet NM, Chavan RN, Cappel MA, et al: Sweet syndrome: clinical preleukemia. Blut 47(1):21–30, 1983.
presentation, associations, and response to treatment in 77 patients. 308. Prodan M, Tulissi P, Perticarari S, et al: Flow cytometric assay for the
J Am Acad Dermatol 69(4):557–564, 2013. evaluation of phagocytosis and oxidative burst of polymorphonuclear
284. Ten Oever J, Kuijper PHM, Kuijpers ALA, et al: Complete remission leukocytes and monocytes in myelodysplastic disorders. Haematologica
of MDS RAEB following immunosuppressive treatment in a patient 80(3):212–218, 1995.
with Sweet’s syndrome. Neth J Med 67(8):347–350, 2009. 309. Ruutu P: Granulocyte function in myelodysplastic syndromes. Scand J
285. Jo T, Horio K, Migita K: Sweet’s syndrome in patients with MDS and Haematol Suppl 45:66–70, 1986.
MEFV mutations. N Engl J Med 372(7):686–688, 2015. 310. Schanz J, Tüchler H, Solé F, et al: New comprehensive cytogenetic
286. Srivastava M, Rencic A, Nousari HC: A rapidly expanding ulcer. scoring system for primary myelodysplastic syndromes (MDS) and
Myelodysplastic syndrome-associated (paraneoplastic) pyoderma gan- oligoblastic acute myeloid leukemia after MDS derived from an inter-
grenosum. Arch Dermatol 139(4):531–536, 2003. national database merge. J Clin Oncol 30(8):820–829, 2012.
287. Duguid JK, Mackie MJ, McVerry BA: Skin infiltration associated with 311. Tricot G, De Wolf-Peeters C, Vlietinck R, et al: Bone marrow histology
chronic myelomonocytic leukaemia. Br J Haematol 53(2):257–264, in myelodysplastic syndromes. II. Prognostic value of abnormal localiza-
1983. tion of immature precursors in MDS. Br J Haematol 58(2):217–225,
288. Enright H, Miller W: Autoimmune phenomena in patients with 1984.
myelodysplastic syndromes. Leuk Lymphoma 24(5–6):483–489, 1997. 312. Della Porta MG, Malcovati L, Boveri E, et al: Clinical relevance of
289. George S, Newman E: Seronegative inflammatory arthritis in the bone marrow fibrosis and CD34-positive cell clusters in primary
myelodysplastic syndromes. Semin Arthritis Rheum 21:345–354, myelodysplastic syndromes. J Clin Oncol 27(5):754–762, 2009.
1992. 313. Williamson PJ, Oscier DG, Mufti GJ, et al: Pyogenic abscesses in the
290. Green AR, Shuttleworth D, Bowen DT, et al: Cutaneous vasculitis myelodysplastic syndrome. BMJ 299(6695):375–376, 1989.
in patients with myelodysplasia. Br J Haematol 74(3):364–365, 314. Rasi V, Lintula R: Platelet function in the myelodysplastic syndromes.
1990. Scand J Haematol Suppl 45:71–73, 1986.
291. Tsuji G, Maekawa S, Saigo K, et al: Dermatomyositis and myelo- 315. Girtovitis FI, Ntaios G, Papadopoulos A, et al: Defective platelet aggre-
dysplastic syndrome with myelofibrosis responding to methotrexate gation in myelodysplastic syndromes. Acta Haematol 118(2):117–122,
therapy. Am J Hematol 74(3):175–178, 2003. 2007.
292. Enright H, Jacob HS, Vercellotti G, et al: Paraneoplastic autoimmune 316. Zeidman A, Sokolover N, Fradin Z, et al: Platelet function and its
phenomena in patients with myelodysplastic syndromes: response to clinical significance in the myelodysplastic syndromes. Hematol J
immunosuppressive therapy. Br J Haematol 91(2):403–408, 1995. 5(3):234–238, 2004.
293. Heo S-W, Cho K-H, Ryu J-I, et al: A case of relapsing polychondritis 317. Szpurka H, Tiu R, Murugesan G, et al: Refractory anemia with ringed
associated with myelodysplastic syndrome with erythroid hypoplasia/ sideroblasts associated with marked thrombocytosis (RARS-T), another
aplasia. Korean J Intern Med 18(4):251–254, 2003. myeloproliferative condition characterized by JAK2 V617F mutation.
294. Steurer M, Fritsche G, Tzankov A, et al: Large-vessel arteritis and Blood 108(7):2173–2181, 2006.
myelodysplastic syndrome: report of two cases. Eur J Haematol 318. Kuriyama K, Tomonaga M, Matsuo T, et al: Diagnostic significance of
73(2):128–133, 2004. detecting pseudo-Pelger-Huët anomalies and micro-megakaryocytes in
295. Saitoh T, Murakami H, Uchiumi H, et al: Myelodysplastic syndromes myelodysplastic syndrome. Br J Haematol 63(4):665–669, 1986.
with nephrotic syndrome. Am J Hematol 60(3):200–204, 1999. 319. Queisser W, Queisser U, Ansmann M, et al: Megakaryocyte poly-
296. Linman JW, Bagby GC: The preleukemic syndrome (hemopoietic ploidization in acute leukaemia and preleukaemia. Br J Haematol
dysplasia). Cancer 42(2 Suppl):854–864, 1978. 28(2):261–270, 1974.
297. Noel P, Solberg L, Jr: Myelodysplastic syndromes. Pathog Diagn Treat 320. Takagi S, Kitagawa S, Takeda A, et al: Natural killer-interferon system
Crit Rev Oncol 12(3):193–215, 1992. in patients with preleukaemic states. Br J Haematol 58(1):71–81, 1984.
298. Bowen DT, Jacobs A, Cotes PM, et al: Serum erythropoietin and eryth- 321. Kerndrup G, Meyer K, Ellegaard J, et al: Natural killer (NK)-cell activ-
ropoiesis in patients with myelodysplastic syndromes. Eur J Haematol ity and antibody-dependent cellular cytotoxicity (ADCC) in primary
44(1):30–32, 1990. preleukemic syndrome. Leuk Res 8(2):239–247, 1984.
299. Kushner J, Porter J, Olivieri N: Secondary iron overload. Hematology 322. Britanova OV, Putintseva EV, Shugay M, et al: Age-related decrease in
Am Soc Hematol Educ Program 47, 2001. TCR repertoire diversity measured with deep and normalized sequence
300. Choi JW, Kim Y, Fujino M, et al: Significance of fetal hemoglobin- profiling. J Immunol 1950 192(6):2689–2698, 2014.
containing erythroblasts (F blasts) and the F blast/F cell ratio in 323. Gibson KL, Wu Y-C, Barnett Y, et al: B-cell diversity decreases in old
myelodysplastic syndromes. Leukemia 16(8):1478–1483, 2002. age and is correlated with poor health status. Aging Cell 8(1):18–25,
301. Lopez M, Bonnet-Gajdos M, Reviron M, et al: An acute leukaemia 2009.
augured before clinical signs by blood group antigen abnormalities and 324. Wells RA, Leber B, Buckstein R, et al: Iron overload in myelodysplastic
low levels of A and H blood group transferase activities in erythrocytes. syndromes: a Canadian consensus guideline. Leuk Res 32(9):1338–1353,
Br J Haematol 63(3):535–539, 1986. 2008.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
969.e8 Part VII  Hematologic Malignancies

325. Wimazal F, Sperr WR, Kundi M, et al: Prognostic value of lactate 345. Kulasekararaj AG, Jiang J, Smith AE, et al: Somatic mutations identify
dehydrogenase activity in myelodysplastic syndromes. Leuk Res 25(4): a subgroup of aplastic anemia patients who progress to myelodysplastic
287–294, 2001. syndrome. Blood 124(17):2698–2704, 2014.
326. Economopoulos T, Economidou J, Giannopoulos G, et al: Immune 346. Iwanaga M, Furukawa K, Amenomori T, et al: Paroxysmal nocturnal
abnormalities in myelodysplastic syndromes. J Clin Pathol 38(8):908– haemoglobinuria clones in patients with myelodysplastic syndromes. Br
911, 1985. J Haematol 102(2):465–474, 1998.
327. Mufti GJ, Figes A, Hamblin TJ, et al: Immunological abnormalities in 347. Sokol L, Loughran TP: Large granular lymphocyte leukemia. Oncologist
myelodysplastic syndromes. I. Serum immunoglobulins and autoanti- 11(3):263–273, 2006.
bodies. Br J Haematol 63(1):143–147, 1986. 348. Lane AA, Odejide O, Kopp N, et al: Low frequency clonal muta-
328. Senent L, Arenillas L, Luño E, et al: Reproducibility of the World tions recoverable by deep sequencing in patients with aplastic anemia.
Health Organization 2008 criteria for myelodysplastic syndromes. Leukemia 27(4):968–971, 2013.
Haematologica 98(4):568–575, 2013. 349. Yoshizato T, Dumitriu B, Hosokawa K, et al: Somatic mutations and
329. Font P, Loscertales J, Benavente C, et al: Inter-observer variance with clonal hematopoiesis in aplastic anemia. N Engl J Med 373(1):35–47,
the diagnosis of myelodysplastic syndromes (MDS) following the 2008 2015.
WHO classification. Ann Hematol 92(1):19–24, 2013. 350. Koskela HLM, Eldfors S, Ellonen P, et al: Somatic STAT3 mutations in
330. Greenberg P, Cox C, LeBeau MM, et al: International scoring large granular lymphocytic leukemia. N Engl J Med 366(20):1905–1913,
system for evaluating prognosis in myelodysplastic syndromes. Blood 2012.
89(6):2079–2088, 1997. 351. Ohyashiki K, Sasao I, Ohyashiki JH, et al: Clinical and cytogenetic
331. Greenberg PL, Tuechler H, Schanz J, et al: Revised international characteristics of myelodysplastic syndromes developing myelofibrosis.
prognostic scoring system for myelodysplastic syndromes. Blood Cancer 68(1):178–183, 1991.
120(12):2454–2465, 2012. 352. Rumi E, Pietra D, Pascutto C, et al: Clinical effect of driver muta-
332. Kantarjian H, O’Brien S, Ravandi F, et al: Proposal for a new risk tions of JAK2, CALR, or MPL in primary myelofibrosis. Blood
model in myelodysplastic syndrome that accounts for events not con- 124(7):1062–1069, 2014.
sidered in the original International Prognostic Scoring System. Cancer 353. Zhang S-J, Rampal R, Manshouri T, et al: Genetic analysis of patients
113(6):1351–1361, 2008. with leukemic transformation of myeloproliferative neoplasms shows
333. Garcia-Manero G, Shan J, Faderl S, et al: A prognostic score for patients recurrent SRSF2 mutations that are associated with adverse outcome.
with lower risk myelodysplastic syndrome. Leukemia 22(3):538–543, Blood 119(19):4480–4485, 2012.
2008. 354. Kantarjian HM, Keating MJ, Walters RS, et al: Therapy-related
334. Malcovati L, Della Porta MG, Strupp C, et al: Impact of the degree of leukemia and myelodysplastic syndrome: clinical, cytogenetic, and
anemia on the outcome of patients with myelodysplastic syndrome and prognostic features. J Clin Oncol 4(12):1748–1757, 1986.
its integration into the WHO classification-based Prognostic Scoring 355. Tallman MS, Gray R, Bennett JM, et al: Leukemogenic potential of adju-
System (WPSS). Haematologica 96(10):1433–1440, 2011. vant chemotherapy for early-stage breast cancer: the Eastern Cooperative
335. Voso MT, Fenu S, Latagliata R, et al: Revised International Prognostic Oncology Group experience. J Clin Oncol 13(7):1557–1563, 1995.
Scoring System (IPSS) predicts survival and leukemic evolution of 356. Smith RE, Bryant J, DeCillis A, et al: Acute myeloid leukemia and
myelodysplastic syndromes significantly better than IPSS and WHO myelodysplastic syndrome after doxorubicin-cyclophosphamide adju-
Prognostic Scoring System: validation by the Gruppo Romano Mielo- vant therapy for operable breast cancer: the National Surgical Adjuvant
displasie Italian Regional Database. J Clin Oncol 31(21):2671–2677, Breast and Bowel Project Experience. J Clin Oncol 21(7):1195–1204,
2013. 2003.
336. Neukirchen J, Lauseker M, Blum S, et al: Validation of the revised 357. Darrington DL, Vose JM, Anderson JR, et al: Incidence and character-
international prognostic scoring system (IPSS-R) in patients with ization of secondary myelodysplastic syndrome and acute myelogenous
myelodysplastic syndrome: a multicenter study. Leuk Res 38(1):57–64, leukemia following high-dose chemoradiotherapy and autologous stem-
2014. cell transplantation for lymphoid malignancies. J Clin Oncol 12(12):
337. Zeidan AM, Lee J-W, Prebet T, et al: Comparison of the prognostic 2527–2534, 1994.
utility of the revised International Prognostic Scoring System and the 358. Friedberg JW, Neuberg D, Stone RM, et al: Outcome in patients with
French Prognostic Scoring System in azacitidine-treated patients with myelodysplastic syndrome after autologous bone marrow transplanta-
myelodysplastic syndromes. Br J Haematol 166(3):352–359, 2014. tion for non-Hodgkin’s lymphoma. J Clin Oncol 17(10):3128–3135,
338. Della Porta MG, Alessandrino EP, Bacigalupo A, et al: Predictive factors 1999.
for the outcome of allogeneic transplantation in patients with MDS 359. Palumbo A, Bringhen S, Kumar SK, et al: Second primary malignancies
stratified according to the revised IPSS-R. Blood 123(15):2333–2342, with lenalidomide therapy for newly diagnosed myeloma: a meta-
2014. analysis of individual patient data. Lancet Oncol 15(3):333–342, 2014.
339. Komrokji RS, Corrales-Yepez M, Al Ali N, et al: Validation of the MD 360. Andersen MK, Christiansen DH, Pedersen-Bjergaard J: Centromeric
Anderson Prognostic Risk Model for patients with myelodysplastic breakage and highly rearranged chromosome derivatives associated
syndrome. Cancer 118(10):2659–2664, 2012. with mutations of TP53 are common in therapy-related MDS and
340. van Spronsen MF, Ossenkoppele GJ, Holman R, et al: Improved risk AML after therapy with alkylating agents: an M-FISH study. Genes
stratification by the integration of the revised international prognostic Chromosomes Cancer 42(4):358–371, 2005.
scoring system with the myelodysplastic syndromes comorbidity index. 361. Pedersen-Bjergaard J: Radiotherapy- and chemotherapy-induced myelo-
Eur J Cancer 1990 50(18):3198–3205, 2014. dysplasia and acute myeloid leukemia. A review. Leuk Res 16(1):61–65,
341. Malcovati L, Germing U, Kuendgen A, et al: Time-dependent prog- 1992.
nostic scoring system for predicting survival and leukemic evolution in 362. Mistry AR, Felix CA, Whitmarsh RJ, et al: DNA topoisomerase II in
myelodysplastic syndromes. J Clin Oncol 25(23):3503–3510, 2007. therapy-related acute promyelocytic leukemia. N Engl J Med 352(15):
342. Della Porta MG, Tuechler H, Malcovati L, et al: Validation of WHO 1529–1538, 2005.
classification-based Prognostic Scoring System (WPSS) for myelodys- 363. Smith SM, Le Beau MM, Huo D, et al: Clinical-cytogenetic associa-
plastic syndromes and comparison with the revised International Prog- tions in 306 patients with therapy-related myelodysplasia and myeloid
nostic Scoring System (IPSS-R). A study of the International Working leukemia: the University of Chicago series. Blood 102(1):43–52, 2003.
Group for Prognosis in Myelodysplasia (IWG-PM). Leukemia 2015. 364. Fukumoto JS, Greenberg PL: Management of patients with higher risk
343. Van den Berghe H, Cassiman JJ, David G, et al: Distinct haematologi- myelodysplastic syndromes. Crit Rev Oncol Hematol 56(2):179–192,
cal disorder with deletion of long arm of no. 5 chromosome. Nature 2005.
251(5474):437–438, 1974. 365. Abruzzese E, Radford JE, Miller JS, et al: Detection of abnormal pre-
344. Sokal G, Michaux JL, Van Den Berghe H, et al: A new hematologic transplant clones in progenitor cells of patients who developed myelo-
syndrome with a distinct karyotype: the 5 q–chromosome. Blood dysplasia after autologous transplantation. Blood 94(5):1814–1819,
46(4):519–533, 1975. 1999.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 969.e9

366. Broseus J, Alpermann T, Wulfert M, et al: Age, JAK2(V617F) and 388. Schouten HC, Vellenga E, van Rhenen DJ, et al: Recombinant human
SF3B1 mutations are the main predicting factors for survival in erythropoietin in patients with myelodysplastic syndromes. Leukemia
refractory anaemia with ring sideroblasts and marked thrombocytosis. 5(5):432–436, 1991.
Leukemia 27(9):1826–1831, 2013. 389. Hellstrom-Lindberg E, Gulbrandsen N, Lindberg G, et al: A validated
367. Ceesay MM, Lea NC, Ingram W, et al: The JAK2 V617F mutation is decision model for treating the anaemia of myelodysplastic syndromes
rare in RARS but common in RARS-T. Leukemia 20(11):2060–2061, with erythropoietin + granulocyte colony-stimulating factor: significant
2006. effects on quality of life. Br J Haematol 120(6):1037–1046, 2003.
368. Remacha A, Puget G, Nomdedéu JF, et al: Occurrence of the JAK2 390. Park S, Grabar S, Kelaidi C, et al: Predictive factors of response and
V617F mutation in the WHO provisional entity: Myelodysplastic/ survival in myelodysplastic syndrome treated with erythropoietin and
myeloproliferative disease, unclassifiable-refractory anemia with ringed G-CSF: the GFM experience. Blood 111(2):574–582, 2008.
sideroblasts associated with marked thrombocytosis. Haematologica 391. Rigolin GM, Porta MD, Ciccone M, et al: In patients with myelodys-
91:719–720, 2006. plastic syndromes response to rHuEPO and G-CSF treatment is related
369. Cheson BD, Bennett JM, Kantarjian H, et al: Report of an international to an increase of cytogenetically normal CD34 cells. Br J Haematol
working group to standardize response criteria for myelodysplastic 126(4):501–507, 2004.
syndromes. Blood 96(12):3671–3674, 2000. 392. Musto P, Matera R, Minervini MM, et al: Low serum levels of tumor
370. Jansen AJG, Essink-Bot M-L, Beckers EAM, et al: Quality of life necrosis factor and interleukin-1 beta in myelodysplastic syndromes
measurement in patients with transfusion-dependent myelodysplastic responsive to recombinant erythropoietin. Haematologica 79(3):265–
syndromes. Br J Haematol 121(2):270–274, 2003. 268, 1994.
371. Abel GA, Klaassen R, Lee SJ, et al: Patient-reported outcomes for the 393. Goy A, Belanger C, Casadevall N, et al: High doses of intravenous
myelodysplastic syndromes: a new MDS-specific measure of quality of recombinant erythropoietin for the treatment of anaemia in myelodys-
life. Blood 123(3):451–452, 2014. plastic syndrome. Br J Haematol 84(2):232–237, 1993.
372. Hellstrom-Lindberg E, Malcovati L: Supportive care and use of hema- 394. Stasi R, Abruzzese E, Lanzetta G, et al: Darbepoetin alfa for the treat-
topoietic growth factors in myelodysplastic syndromes. Semin Hematol ment of anemic patients with low- and intermediate-1-risk myelodys-
45(1):14–22, 2008. plastic syndromes. Ann Oncol 16(12):1921–1927, 2005.
373. Cazzola M, Malcovati L: Myelodysplastic syndromes–coping with 395. Moyo V, Lefebvre P, Duh MS, et al: Erythropoiesis-stimulating agents
ineffective hematopoiesis. N Engl J Med 352(6):536–538, 2005. in the treatment of anemia in myelodysplastic syndromes: a meta-
374. Hebert P, Wells G, Blajchman M, et al: A multicenter, randomized, con- analysis. Ann Hematol 87(7):527–536, 2008.
trolled clinical trial of transfusion requirements in critical care. Transfus 396. Terpos E, Mougiou A, Kouraklis A, et al: Prolonged administration of
Requir Crit Care Care Trials Group. N Engl J Med 340(6):409–417, erythropoietin increases erythroid response rate in myelodysplastic syn-
1999. dromes: a phase II trial in 281 patients. Br J Haematol 118(1):174–180,
375. Hajjar LA, Vincent J-L, Galas FRBG, et al: Transfusion requirements 2002.
after cardiac surgery: the TRACS randomized controlled trial. JAMA 397. Verhoef GE, Zachée P, Ferrant A, et al: Recombinant human erythro-
304(14):1559–1567, 2010. poietin for the treatment of anemia in the myelodysplastic syndromes:
376. Fakhry S, Fata P: How low is too low? Cardiac risks with anemia. Crit a clinical and erythrokinetic assessment. Ann Hematol 64(1):16–21,
Care 8(Suppl 2):S11–S14, 2004. 1992.
377. Oliva EN, Schey C, Hutchings AS: A review of anemia as a cardiovas- 398. Kurzrock R, Talpaz M, Estey E, et al: Erythropoietin treatment in
cular risk factor in patients with myelodysplastic syndromes. Am J Blood patients with myelodysplastic syndrome and anemia. Leukemia
Res 1(2):160–166, 2011. 5(11):985–990, 1991.
378. Anderson KC, Weinstein HJ: Transfusion-associated graft-versus-host 399. Spiriti MAA, Latagliata R, Niscola P, et al: Impact of a new dosing
disease. N Engl J Med 323(5):315–321, 1990. regimen of epoetin alfa on quality of life and anemia in patients with
379. Chacko J, Pennell DJ, Tanner MA, et al: Myocardial iron loading by low-risk myelodysplastic syndrome. Ann Hematol 84(3):167–176,
magnetic resonance imaging T2* in good prognostic myelodysplastic 2005.
syndrome patients on long-term blood transfusions. Br J Haematol 400. Jadersten M, Malcovati L, Dybedal I, et al: Erythropoietin and
138(5):587–593, 2007. granulocyte-colony stimulating factor treatment associated with
380. Pascal L, Beyne-Rauzy O, Brechignac S, et al: Cardiac iron overload improved survival in myelodysplastic syndrome. J Clin Oncol
assessed by T2* magnetic resonance imaging and cardiac function in 26(21):3607–3613, 2008.
regularly transfused myelodysplastic syndrome patients. Br J Haematol 401. Hellström-Lindberg E, Negrin R, Stein R, et al: Erythroid response to
162(3):413–415, 2013. treatment with G-CSF plus erythropoietin for the anaemia of patients
381. Malcovati L: Impact of transfusion dependency and secondary iron with myelodysplastic syndromes: proposal for a predictive model. Br J
overload on the survival of patients with myelodysplastic syndromes. Haematol 99(2):344–351, 1997.
Leuk Res 31(Suppl 3):S2–S6, 2007. 402. Rizzo JD, Brouwers M, Hurley P, et al: American Society of Hematology/
382. Chee CE, Steensma DP, Wu W, et al: Neither serum ferritin nor the American Society of Clinical Oncology clinical practice guideline
number of red blood cell transfusions affect overall survival in refrac- update on the use of epoetin and darbepoetin in adult patients with
tory anemia with ringed sideroblasts. Am J Hematol 83(8):611–613, cancer. Blood 116(20):4045–4059, 2010.
2008. 403. Phrommintikul A, Haas SJ, Elsik M, et al: Mortality and target haemo-
383. Rose EH, Abels RI, Nelson RA, et al: The use of r-HuEpo in the globin concentrations in anaemic patients with chronic kidney disease
treatment of anaemia related to myelodysplasia (MDS). Br J Haematol treated with erythropoietin: a meta-analysis. Lancet 369(9559):381–388,
89(4):831–837, 1995. 2007.
384. Bessho M, Jinnai I, Matsuda A, et al: Improvement of anemia by 404. NCCN practice guidelines for the myelodysplastic syndromes. Natl
recombinant erythropoietin in patients with myelodysplastic syndromes Compr Cancer Netw Oncol Williston Park 12:53, 1998.
and aplastic anemia. Int J Cell Cloning 8(6):445–458, 1990. 405. Vadhan-Raj S, Keating M, LeMaistre A, et al: Effects of recombinant
385. Stebler C, Tichelli A, Dazzi H, et al: High-dose recombinant human human granulocyte-macrophage colony-stimulating factor in patients
erythropoietin for treatment of anemia in myelodysplastic syndromes with myelodysplastic syndromes. N Engl J Med 317(25):1545–1552,
and paroxysmal nocturnal hemoglobinuria: a pilot study. Exp Hematol 1987.
18(11):1204–1208, 1990. 406. Antin JH, Smith BR, Holmes W, et al: Phase I/II study of recom-
386. Bowen D, Culligan D, Jacobs A: The treatment of anaemia in the binant human granulocyte-macrophage colony-stimulating factor in
myelodysplastic syndromes with recombinant human erythropoietin. aplastic anemia and myelodysplastic syndrome. Blood 72(2):705–713,
Br J Haematol 77(3):419–423, 1991. 1988.
387. Hellstrom E, Birgegard G, Lockner D, et al: Treatment of myelo- 407. Negrin RS, Haeuber DH, Nagler A, et al: Treatment of myelodysplastic
dysplastic syndromes with recombinant human erythropoietin. Eur J syndromes with recombinant human granulocyte colony-stimulating
Haematol 47(5):355–360, 1991. factor. A phase I-II trial. Ann Intern Med 110(12):976–984, 1989.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
969.e10 Part VII  Hematologic Malignancies

408. Jadersten M, Montgomery S, Dybedal I, et al: Long-term outcome of 430. Neukirchen J, Fox F, Kündgen A, et al: Improved survival in MDS
treatment of anemia in MDS with erythropoietin and G-CSF. Blood patients receiving iron chelation therapy - a matched pair analysis of 188
106(3):803–811, 2005. patients from the Düsseldorf MDS registry. Leuk Res 36(8):1067–1070,
409. Engelfriet CP, Reesink HW, Klein HG, et al: International forum: 2012.
granulocyte transfusions. Vox Sang 79(1):59–66, 2000. 431. Malcovati L, Porta MGD, Pascutto C, et al: Prognostic factors and
410. Freifeld AG, Bow EJ, Sepkowitz KA, et al: Clinical practice guideline life expectancy in myelodysplastic syndromes classified according
for the use of antimicrobial agents in neutropenic patients with cancer: to WHO criteria: a basis for clinical decision making. J Clin Oncol
2010 Update by the Infectious Diseases Society of America. Clin Infect 23(30):7594–7603, 2005.
Dis 52(4):427–431, 2011. 432. Takatoku M, Uchiyama T, Okamoto S, et al: Retrospective nationwide
411. Cornely OA, Maertens J, Winston DJ, et al: Posaconazole vs. flucon- survey of Japanese patients with transfusion-dependent MDS and
azole or itraconazole prophylaxis in patients with neutropenia. N Engl aplastic anemia highlights the negative impact of iron overload on
J Med 356(4):348–359, 2007. morbidity/mortality. Eur J Haematol 78(6):487–494, 2007.
412. Bergmann OJ, Mogensen SC, Ellermann-Eriksen S, et al: Acyclovir 433. Jensen PD, Heickendorff L, Pedersen B, et al: The effect of iron chela-
prophylaxis and fever during remission-induction therapy of patients tion on haemopoiesis in MDS patients with transfusional iron overload.
with acute myeloid leukemia: a randomized, double-blind, placebo- Br J Haematol 94(2):288–299, 1996.
controlled trial. J Clin Oncol 15(6):2269–2274, 1997. 434. Guariglia R, Martorelli MC, Villani O, et al: Positive effects on hemato-
413. Kantarjian H, Giles F, List A, et al: The incidence and impact of throm- poiesis in patients with myelodysplastic syndrome receiving deferasirox
bocytopenia in myelodysplastic syndromes. Cancer 109(9):1705–1714, as oral iron chelation therapy: a brief review. Leuk Res 35(5):566–570,
2007. 2011.
414. Neukirchen J, Blum S, Kuendgen A, et al: Platelet counts and haem- 435. Leitch HA, Chan C, Leger CS, et al: Improved survival with iron chela-
orrhagic diathesis in patients with myelodysplastic syndromes. Eur J tion therapy for red blood cell transfusion dependent lower IPSS risk
Haematol 83(5):477–482, 2009. MDS may be more significant in patients with a non-RARS diagnosis.
415. Gonzalez-Porras JR, Cordoba I, Such E, et al: Prognostic impact of Leuk Res 36(11):1380–1386, 2012.
severe thrombocytopenia in low-risk myelodysplastic syndrome. Cancer 436. Rose C, Brechignac S, Vassilief D, et al: Does iron chelation therapy
117(24):5529–5537, 2011. improve survival in regularly transfused lower risk MDS patients? A
416. Rebulla P, Finazzi G, Marangoni F, et al: The threshold for prophylactic multicenter study by the GFM (Groupe Francophone des Myélodys-
platelet transfusions in adults with acute myeloid leukemia. Gruppo plasies). Leuk Res 34(7):864–870, 2010.
Italiano Malattie Ematologiche Maligne dell’Adulto. N Engl J Med 437. Mainous AG, Tanner RJ, Hulihan MM, et al: The impact of chelation
337(26):1870–1875, 1997. therapy on survival in transfusional iron overload: a meta-analysis of
417. Wandt H, Schaefer-Eckart K, Wendelin K, et al: Therapeutic platelet myelodysplastic syndrome. Br J Haematol 167(5):720–723, 2014.
transfusion versus routine prophylactic transfusion in patients with 438. Bennett JM, MDS Foundation’s Working Group on Transfusional Iron
haematological malignancies: an open-label, multicentre, randomised Overload: Consensus statement on iron overload in myelodysplastic
study. Lancet 380(9850):1309–1316, 2012. syndromes. Am J Hematol 83(11):858–861, 2008.
418. Bussel JB, Cheng G, Saleh MN, et al: Eltrombopag for the treat- 439. Armand P, Kim HT, Cutler CS, et al: Prognostic impact of elevated
ment of chronic idiopathic thrombocytopenic purpura. N Engl J Med pretransplantation serum ferritin in patients undergoing myeloablative
357(22):2237–2247, 2007. stem cell transplantation. Blood 109(10):4586–4588, 2007.
419. Kuter DJ, Begley CG: Recombinant human thrombopoietin: basic 440. Di Tucci AA, Matta G, Deplano S, et al: Myocardial iron overload
biology and evaluation of clinical studies. Blood 100(10):3457–3469, assessment by T2* magnetic resonance imaging in adult transfusion
2002. dependent patients with acquired anemias. Haematologica 93(9):1385–
420. Kantarjian H, Fenaux P, Sekeres MA, et al: Safety and efficacy of 1388, 2008.
romiplostim in patients with lower-risk myelodysplastic syndrome and 441. List AF: New approaches to the treatment of myelodysplasia. Oncologist
thrombocytopenia. J Clin Oncol 28(3):437–444, 2010. 7(Suppl 1):39–49, 2002.
421. Kantarjian HM, Giles FJ, Greenberg PL, et al: Phase 2 study of romiplos- 442. Krönke J, Fink EC, Hollenbach PW, et al: Lenalidomide induces
tim in patients with low- or intermediate-risk myelodysplastic syndrome ubiquitination and degradation of CK1α in del(5q) MDS. Nature
receiving azacitidine therapy. Blood 116(17):3163–3170, 2010. 523(7559):183–188, 2015.
422. Sekeres MA, Kantarjian H, Fenaux P, et al: Subcutaneous or intrave- 443. Fink EC, Krönke J, Hurst SN, et al: Lenalidomide Induces Ubiquitina-
nous administration of romiplostim in thrombocytopenic patients with tion and Degradation of CSNK1A1 in MDS with Del(5q). Blood
lower risk myelodysplastic syndromes. Cancer 117(5):992–1000, 2011. 124:4, 2014.
423. Greenberg PL, Garcia-Manero G, Moore M, et al: A randomized 444. List A, Kurtin S, Roe DJ, et al: Efficacy of lenalidomide in myelodys-
controlled trial of romiplostim in patients with low- or intermediate- plastic syndromes. N Engl J Med 352(6):549–557, 2005.
risk myelodysplastic syndrome receiving decitabine. Leuk Lymphoma 445. Raza A, Reeves JA, Feldman EJ, et al: Phase 2 study of lenalidomide
54(2):321–328, 2013. in transfusion-dependent, low-risk, and intermediate-1 risk myelo-
424. Svensson T, Chowdhury O, Garelius H, et al: A pilot phase I dose finding dysplastic syndromes with karyotypes other than deletion 5q. Blood
safety study of the thrombopoietin-receptor agonist, eltrombopag, in 111(1):86–93, 2008.
patients with myelodysplastic syndrome treated with azacitidine. Eur J 446. Santini V, Prebet T, Fenaux P, et al: Minimizing risk of hypomethylating
Haematol 93(5):439–445, 2014. agent failure in patients with higher-risk MDS and practical manage-
425. Kuter DJ, Mufti GJ, Bain BJ, et al: Evaluation of bone marrow reticulin ment recommendations. Leuk Res 38(12):1381–1391, 2014.
formation in chronic immune thrombocytopenia patients treated with 447. Sekeres MA, Maciejewski JP, Giagounidis AAN, et al: Relationship
romiplostim. Blood 114(18):3748–3756, 2009. of treatment-related cytopenias and response to lenalidomide in
426. Brierley CK, Steensma DP: Thrombopoiesis-stimulating agents and patients with lower-risk myelodysplastic syndromes. J Clin Oncol
myelodysplastic syndromes. Br J Haematol 2015. 26(36):5943–5949, 2008.
427. Giagounidis A, Mufti GJ, Fenaux P, et al: Results of a randomized, 448. Richardson P, Jagannath S, Hussein M, et al: Safety and efficacy of
double-blind study of romiplostim versus placebo in patients with low/ single-agent lenalidomide in patients with relapsed and refractory
intermediate-1-risk myelodysplastic syndrome and thrombocytopenia. multiple myeloma. Blood 114(4):772–778, 2009.
Cancer 120(12):1838–1846, 2014. 449. Wisch JS, Griffin JD, Kufe DW: Response of preleukemic syndromes
428. Giardina PJ, Grady RW: Chelation therapy in beta-thalassemia: an to continuous infusion of low-dose cytarabine. N Engl J Med
optimistic update. Semin Hematol 38(4):360–366, 2001. 309(26):1599–1602, 1983.
429. Lyons RM, Marek BJ, Paley C, et al: Comparison of 24-month outcomes 450. Mufti GJ, Oscier DG, Hamblin TJ, et al: Low doses of cytarabine in the
in chelated and non-chelated lower-risk patients with myelodysplastic treatment of myelodysplastic syndrome and acute myeloid leukemia. N
syndromes in a prospective registry. Leuk Res 38(2):149–154, 2014. Engl J Med 309(26):1653–1654, 1983.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 969.e11

451. Manoharan A: Low-dose cytarabine therapy in hypoplastic acute phase II clinical trial with a differentiation-inducing agent. Blood
leukemia. N Engl J Med 309(26):1652–1653, 1983. 80(10):2604–2609, 1992.
452. Deeg HJ, Gotlib J, Beckham C, et al: Soluble TNF receptor fusion 473. Essers MAG, Offner S, Blanco-Bose WE, et al: IFNalpha activates
protein (etanercept) for the treatment of myelodysplastic syndrome: a dormant haematopoietic stem cells in vivo. Nature 458(7240):904–908,
pilot study. Leukemia 16(2):162–164, 2002. 2009.
453. Rosenfeld C, Bedell C: Pilot study of recombinant human soluble 474. Walter D, Lier A, Geiselhart A, et al: Exit from dormancy provokes
tumor necrosis factor receptor (TNFR:Fc) in patients with low risk DNA-damage-induced attrition in haematopoietic stem cells. Nature
myelodysplastic syndrome. Leuk Res 26(8):721–724, 2002. 520:549–552, 2015.
454. Scott BL, Ramakrishnan A, Fosdal M, et al: Anti-thymocyte globulin 475. List A, Beran M, DiPersio J, et al: Opportunities for Trisenox (arsenic
plus etanercept as therapy for myelodysplastic syndromes (MDS): a trioxide) in the treatment of myelodysplastic syndromes. Leukemia
phase II study. Br J Haematol 149(5):706–710, 2010. 17(8):1499–1507, 2003.
455. Stasi R, Amadori S: Infliximab chimaeric anti-tumour necrosis factor 476. Schiller GJ, Slack J, Hainsworth JD, et al: Phase II multicenter study
alpha monoclonal antibody treatment for patients with myelodysplastic of arsenic trioxide in patients with myelodysplastic syndromes. J Clin
syndromes. Br J Haematol 116(2):334–337, 2002. Oncol 24(16):2456–2464, 2006.
456. Braun T, Fenaux P: Myelodysplastic syndromes (MDS) and auto- 477. Bejanyan N, Tiu R, Raza A: A phase 2 trial of combination therapy
immune disorders (AD): cause or consequence? Best Pract Res Clin with thalidomide, arsenic trioxide, dexamethasone, and ascorbic acid
Haematol 26(4):327–336, 2013. (TADA) in patients with overlap myelodysplastic/myeloproliferative
457. Biesma DH, van den Tweel JG, Verdonck LF: Immunosuppressive neoplasms (MDS/MPN) or primary myelofibrosis (PMF). Cancer
therapy for hypoplastic myelodysplastic syndrome. Cancer 79(8):1548– 118:3968–3976, 2012.
1551, 1997. 478. Sekeres MA, Maciejewski JP, Erba HP, et al: A Phase 2 study of com-
458. Molldrem JJ, Leifer E, Bahceci E, et al: Antithymocyte globulin for bination therapy with arsenic trioxide and gemtuzumab ozogamicin in
treatment of the bone marrow failure associated with myelodysplastic patients with myelodysplastic syndromes or secondary acute myeloid
syndromes. Ann Intern Med 137(3):156–163, 2002. leukemia. Cancer 117(6):1253–1261, 2011.
459. Saunthararajah Y, Nakamura R, Nam J-M, et al: HLA-DR15 (DR2) 479. Hast R, Axdorph S, Laurén L, et al: Absent clinical effects of retinoic
is overrepresented in myelodysplastic syndrome and aplastic anemia acid and isoretinoin treatment in the myelodysplastic syndrome.
and predicts a response to immunosuppression in myelodysplastic Hematol Oncol 7(4):297–301, 1989.
syndrome. Blood 100(5):1570–1574, 2002. 480. Ohno R, Naoe T, Hirano M, et al: Treatment of myelodysplastic
460. Sloand EM, Wu CO, Greenberg P, et al: Factors affecting response and syndromes with all-trans retinoic acid. Leukemia Study Group of the
survival in patients with myelodysplasia treated with immunosuppres- Ministry of Health and Welfare. Blood 81(5):1152–1154, 1993.
sive therapy. J Clin Oncol 26(15):2505–2511, 2008. 481. Schenk T, Chen WC, Göllner S, et al: Inhibition of the LSD1
461. Passweg JR, Giagounidis AAN, Simcock M, et al: Immunosuppressive (KDM1A) demethylase reactivates the all-trans-retinoic acid differen-
therapy for patients with myelodysplastic syndrome: a prospective ran- tiation pathway in acute myeloid leukemia. Nat Med 18(4):605–611,
domized multicenter phase III trial comparing antithymocyte globulin 2012.
plus cyclosporine with best supportive care–SAKK 33/99. J Clin Oncol 482. Search of: MDS AND (“low” or “intermediate”) NOT (“high-risk” or
29(3):303–309, 2011. “AML” or “acute myeloid leukemia”) | Open Studies | Exclude Unknown |
462. Sloand EM, Olnes MJ, Shenoy A, et al: Alemtuzumab treatment of Adult | Phase 1, 2, 3 - List Results - ClinicalTrials.gov [Internet].
intermediate-1 myelodysplasia patients is associated with sustained 483. Zhou L, Nguyen AN, Sohal D, et al: Inhibition of the TGF-beta recep-
improvement in blood counts and cytogenetic remissions. J Clin Oncol tor I kinase promotes hematopoiesis in MDS. Blood 112(8):3434–3443,
28(35):5166–5173, 2010. 2008.
463. Keyhani A, Jendiroba DB, Freireich EJ: Angiogenesis and leukemia. 484. Quesnel B, Guillerm G, Vereecque R, et al: Methylation of the
Leuk Res 25(8):639–645, 2001. p15(INK4b) gene in myelodysplastic syndromes is frequent and
464. Legros L, Slama B, Karsenti J-M, et al: Treatment of myelodysplastic acquired during disease progression. Blood 91(8):2985–2990,
syndromes with excess of blasts by bevacizumab is well tolerated and 1998.
is associated with a decrease of VEGF plasma level. Ann Hematol 485. Lin J, Yao D, Qian J, et al: Methylation status of fragile histidine triad
91(1):39–46, 2012. (FHIT) gene and its clinical impact on prognosis of patients with
465. Gupta P, Mulkey F, Hasserjian RP, et al: A phase II study of the myelodysplastic syndrome. Leuk Res 32(10):1541–1545, 2008.
oral VEGF receptor tyrosine kinase inhibitor vatalanib (PTK787/ 486. Brakensiek K, Länger F, Schlegelberger B, et al: Hypermethylation of
ZK222584) in myelodysplastic syndrome: Cancer and Leukemia the suppressor of cytokine signalling-1 (SOCS-1) in myelodysplastic
Group B study 10105 (Alliance). Invest New Drugs 31(5):1311–1320, syndrome. Br J Haematol 130(2):209–217, 2005.
2013. 487. Liu TX, Becker MW, Jelinek J, et al: Chromosome 5q deletion and
466. Erikci AA, Ozturk A, Karagoz B, et al: Results of combination therapy epigenetic suppression of the gene encoding alpha-catenin (CTNNA1)
with amifostine, pentoxifylline, ciprofloxacin and dexamethasone in in myeloid cell transformation. Nat Med 13(1):78–83, 2007.
patients with myelodysplastic syndrome and acute myeloid leukemia. 488. Shen L, Kantarjian H, Guo Y, et al: DNA methylation predicts survival
Hematology 13(5):289–292, 2008. and response to therapy in patients with myelodysplastic syndromes. J
467. List AF: Emerging data on IMiDs in the treatment of myelodysplastic Clin Oncol 28(4):605–613, 2010.
syndromes (MDS). Semin Oncol 32(4 Suppl 5):S31–S35, 2005. 489. Jones PA, Taylor SM: Cellular differentiation, cytidine analogs and
468. Vuylsteke J, Verloop MC, Drogendijk AC: Favourable effect of pyridox- DNA methylation. Cell 20(1):85–93, 1980.
ine and ascorbic acid in a patient with refractory sideroblastic anaemia 490. de Vos D: Epigenetic drugs: a longstanding story. Semin Oncol
and haemochromatosis. Acta Med Scand 169:113–123, 1961. 32(5):437–442, 2005.
469. Nagler A, Rikilis I, Tatarsky I, et al: Effect of 1,25-dihydroxyvitamin 491. Lyons RM, Cosgriff TM, Modi SS, et al: Hematologic response to three
D3 and 13-cis-retinoic acid on in vitro hematopoiesis in the myelodys- alternative dosing schedules of azacitidine in patients with myelodys-
plastic syndromes. J Lab Clin Med 110(2):237–244, 1987. plastic syndromes. J Clin Oncol 27(11):1850–1856, 2009.
470. Takami A, Nakao S, Ontachi Y, et al: Successful therapy of myelo- 492. Silverman LR, Demakos EP, Peterson BL, et al: Randomized controlled
dysplastic syndrome with menatetrenone, a vitamin K2 analog. Int J trial of azacitidine in patients with the myelodysplastic syndrome: a study
Hematol 69(1):24–26, 1999. of the cancer and leukemia group B. J Clin Oncol 20(10):2429–2440,
471. Tefferi A, Elliott MA, Steensma DP, et al: Amifostine alone and in 2002.
combination with erythropoietin for the treatment of favorable myelo- 493. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al: Efficacy of azacitidine
dysplastic syndrome. Leuk Res 25(2):183–185, 2001. compared with that of conventional care regimens in the treatment
472. Andreeff M, Stone R, Michaeli J, et al: Hexamethylene bisacetamide of higher-risk myelodysplastic syndromes: a randomised, open-label,
in myelodysplastic syndrome and acute myelogenous leukemia: a phase III study. Lancet Oncol 10(3):223–232, 2009.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
969.e12 Part VII  Hematologic Malignancies

494. Prebet T, Sun Z, Figueroa ME, et al: Prolonged administration of of myelodysplastic syndromes: an evidence-based review. Biol Blood
azacitidine with or without entinostat for myelodysplastic syndrome Marrow Transplant 15(2):137–172, 2009.
and acute myeloid leukemia with myelodysplasia-related changes: 514. Hahn T, McCarthy PL, Hassebroek A, et al: Significant improvement
results of the US Leukemia Intergroup trial E1905. J Clin Oncol in survival after allogeneic hematopoietic cell transplantation during
32(12):1242–1248, 2014. a period of significantly increased use, older recipient age, and use of
495. Qin T, Castoro R, El Ahdab S, et al: Mechanisms of resistance to unrelated donors. J Clin Oncol 31(19):2437–2449, 2013.
decitabine in the myelodysplastic syndrome. PLoS ONE 6(8):e23372, 515. Brunner AM, Kim HT, Coughlin E, et al: Outcomes in patients age
2011. 70 or older undergoing allogeneic hematopoietic stem cell transplanta-
496. Steensma DP, Baer MR, Slack JL, et al: Multicenter study of decitabine tion for hematologic malignancies. Biol Blood Marrow Transplant
administered daily for 5 days every 4 weeks to adults with myelo- 19(9):1374–1380, 2013.
dysplastic syndromes: the alternative dosing for outpatient treatment 516. Cutler CS, Lee SJ, Greenberg P, et al: A decision analysis of allogeneic
(ADOPT) trial. J Clin Oncol 27(23):3842–3848, 2009. bone marrow transplantation for the myelodysplastic syndromes:
497. Kantarjian H, Issa J-PJ, Rosenfeld CS, et al: Decitabine improves delayed transplantation for low-risk myelodysplasia is associated with
patient outcomes in myelodysplastic syndromes: results of a phase III improved outcome. Blood 104(2):579–585, 2004.
randomized study. Cancer 106(8):1794–1803, 2006. 517. Sierra J, Pérez WS, Rozman C, et al: Bone marrow transplantation
498. Kantarjian H, Oki Y, Garcia-Manero G, et al: Results of a randomized from HLA-identical siblings as treatment for myelodysplasia. Blood
study of 3 schedules of low-dose decitabine in higher-risk myelo- 100(6):1997–2004, 2002.
dysplastic syndrome and chronic myelomonocytic leukemia. Blood 518. Castro-Malaspina H, Harris RE, Gajewski J, et al: Unrelated donor
109(1):52–57, 2007. marrow transplantation for myelodysplastic syndromes: outcome
499. Saunthararajah Y, Sekeres M, Advani A, et al: Evaluation of noncy- analysis in 510 transplants facilitated by the National Marrow Donor
totoxic DNMT1-depleting therapy in patients with myelodysplastic Program. Blood 99(6):1943–1951, 2002.
syndromes. J Clin Invest 2015. 519. Majhail NS, Brunstein CG, Shanley R, et al: Reduced-intensity
500. Quesnel B, Fenaux P: P15INK4b gene methylation and myelodysplas- hematopoietic cell transplantation in older patients with AML/MDS:
tic syndromes. Leuk Lymphoma 35(5–6):437–443, 1999. umbilical cord blood is a feasible option for patients without HLA-
501. Follo MY, Russo D, Finelli C, et al: Epigenetic regulation of nuclear matched sibling donors. Bone Marrow Transplant 47(4):494–498, 2012.
PI-PLCbeta1 signaling pathway in low-risk MDS patients during 520. Di Stasi A, Milton DR, Poon LM, et al: Similar transplantation
azacitidine treatment. Leukemia 26(5):943–950, 2012. outcomes for acute myeloid leukemia and myelodysplastic syndrome
502. Figueroa ME, Skrabanek L, Li Y, et al: MDS and secondary AML patients with haploidentical versus 10/10 human leukocyte antigen-
display unique patterns and abundance of aberrant DNA methylation. matched unrelated and related donors. Biol Blood Marrow Transplant
Blood 114(16):3448–3458, 2009. 20(12):1975–1981, 2014.
503. Fandy TE, Herman JG, Kerns P, et al: Early epigenetic changes and 521. Koreth J, Pidala J, Perez WS, et al: Role of reduced-intensity condition-
DNA damage do not predict clinical response in an overlapping ing allogeneic hematopoietic stem-cell transplantation in older patients
schedule of 5-azacytidine and entinostat in patients with myeloid with de novo myelodysplastic syndromes: an international collaborative
malignancies. Blood 114(13):2764–2773, 2009. decision analysis. J Clin Oncol 31(21):2662–2670, 2013.
504. Quintas-Cardama A, Santos F, Garcia-Manero G: Histone deacetylase 522. Alyea EP, Kim HT, Ho V, et al: Impact of conditioning regimen inten-
inhibitors for the treatment of myelodysplastic syndrome and acute sity on outcome of allogeneic hematopoietic cell transplantation for
myeloid leukemia. Leukemia 25(2):226–235, 2011. advanced acute myelogenous leukemia and myelodysplastic syndrome.
505. Kuendgen A, Strupp C, Aivado M, et al: Treatment of myelodysplastic Biol Blood Marrow Transplant 12(10):1047–1055, 2006.
syndromes with valproic acid alone or in combination with all-trans 523. Appelbaum FR, Anderson J: Allogeneic bone marrow transplantation
retinoic acid. Blood 104(5):1266–1269, 2004. for myelodysplastic syndrome: outcomes analysis according to IPSS
506. Prebet T, Vey N: Vorinostat in acute myeloid leukemia and myelo- score. Leukemia 12(Suppl 1):S25–S29, 1998.
dysplastic syndromes. Expert Opin Investig Drugs 20(2):287–295, 524. Armand P, Kim HT, DeAngelo DJ, et al: Impact of cytogenetics on
2011. outcome of de novo and therapy-related AML and MDS after allo-
507. Ottmann O, DeAngelo D, Garcia-Manero G, et al: Determination geneic transplantation. Biol Blood Marrow Transplant 13(6):655–664,
of a phase II dose of panobinostat in combination with 5-azacitidine 2007.
in patients with myelodysplastic syndromes, chronic myelomonocytic 525. Scott BL, Park JY, Deeg HJ, et al: Pretransplant neutropenia is associ-
leukemia, or acute myeloid leukemia. Blood Meet Extr 459:118, ated with poor-risk cytogenetic features and increased infection-related
2011. mortality in patients with myelodysplastic syndromes. Biol Blood
508. Cashen A, Juckett M, Jumonville A, et al: Phase II study of the histone Marrow Transplant 14(7):799–806, 2008.
deacetylase inhibitor belinostat (PXD101) for the treatment of myelo- 526. Gerds AT, Gooley TA, Estey EH, et al: Pretransplantation therapy with
dysplastic syndrome (MDS). Ann Hematol 91(1):33–38, 2012. azacitidine vs induction chemotherapy and posttransplantation outcome
509. Sekeres MA, Othus M, List AF, et al: A randomized phase II study of in patients with MDS. Biol Blood Marrow Transplant 18(8):1211–1218,
azacitidine combined with lenalidomide or with vorinostat vs. azaciti- 2012.
dine monotherapy in higher-risk myelodysplastic syndromes (MDS) 527. De Witte T, Van Biezen A, Hermans J, et al: Autologous bone marrow
and chronic myelomonocytic leukemia (CMML): North American transplantation for patients with myelodysplastic syndrome (MDS) or
Intergroup Study SWOG S1117. Blood 124:LBA–5, 2014. acute myeloid leukemia following MDS. Chronic and Acute Leukemia
510. Beran M, Shen Y, Kantarjian H, et al: High-dose chemotherapy in Working Parties of the European Group for Blood and Marrow Trans-
high-risk myelodysplastic syndrome: covariate-adjusted comparison of plantation. Blood 90(10):3853–3857, 1997.
five regimens. Cancer 92(8):1999–2015, 2001. 528. de Witte T, Suciu S, Brand R, et al: Autologous stem cell transplanta-
511. Cortes J, Kantarjian H, Albitar M, et al: A randomized trial of liposo- tion in myelodysplastic syndromes. Semin Hematol 44(4):274–277,
mal daunorubicin and cytarabine versus liposomal daunorubicin and 2007.
topotecan with or without thalidomide as initial therapy for patients 529. Ogata K, Nomura T: Application of low-dose etoposide therapy for
with poor prognosis acute myelogenous leukemia or myelodysplastic myelodysplastic syndromes. Leuk Lymphoma 12(1–2):35–39, 1993.
syndrome. Cancer 97(5):1234–1241, 2003. 530. Mario AD, Pagano L, Mele L, et al: Use of gemcitabine (GEM) in
512. Beran M, Estey E, O’Brien S, et al: Topotecan and cytarabine is an advanced myelodysplastic syndromes. Ann Oncol 12(10):1494, 2001.
active combination regimen in myelodysplastic syndromes and chronic 531. Denzlinger C, Bowen D, Benz D, et al: Low-dose melphalan induces
myelomonocytic leukemia. J Clin Oncol 17(9):2819–2830, 1999. favourable responses in elderly patients with high-risk myelodysplastic
513. Oliansky DM, Antin JH, Bennett JM, et al: The role of cytotoxic syndromes or secondary acute myeloid leukaemia. Br J Haematol
therapy with hematopoietic stem cell transplantation in the therapy 108(1):93–95, 2000.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.
Chapter 60  Myelodysplastic Syndromes 969.e13

532. Bryan J, Kantarjian H, Prescott H, et al: Clofarabine in the treatment of 535. Clinical Trials (PDQ®) - National Cancer Institute [Internet]. [cited
myelodysplastic syndromes. Expert Opin Investig Drugs 23(2):255–263, 2015 Mar 20];Available from: <http://www.cancer.gov/clinicaltrials/
2014. search/view?cdrid=737176&version=Patient>.
533. Roberts DA, Wadleigh M, McDonnell AM, et al: Low efficacy and high 536. Gleason MK, Ross JA, Warlick ED, et al: CD16xCD33 bispecific
mortality associated with clofarabine treatment of relapsed/refractory killer cell engager (BiKE) activates NK cells against primary MDS and
acute myeloid leukemia and myelodysplastic syndromes. Leuk Res MDSC CD33+ targets. Blood 123(19):3016–3026, 2014.
39(2):204–210, 2015. 537. Grant S: New agents for AML and MDS. Best Pract Res Clin Haematol
534. Ipilimumab in treating patients with relapsed or refractory high-risk 22(4):501–507, 2009.
myelodysplastic syndrome or acute myeloid leukemia—Full Text
View—ClinicalTrials.gov [Internet]. [cited 2015 Mar 20]; Available
from: <https://clinicaltrials.gov/ct2/show/NCT01757639>.

Downloaded for asmaa mohammad (asmaa.mohamed@kasralainy.edu.eg) at EKB Data Centre from ClinicalKey.com by Elsevier on January 22, 2018.
For personal use only. No other uses without permission. Copyright ©2018. Elsevier Inc. All rights reserved.

Das könnte Ihnen auch gefallen