Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

SYMPOSIUM ON NEOPLASTIC HEMATOLOGY AND MEDICAL ONCOLOGY

Myelodysplastic Syndromes: Diagnosis and


Treatment
David P. Steensma, MD

CME Activity
Target Audience: The target audience for Mayo Clinic Proceedings is primar- Safeguards against commercial bias have been put in place. Faculty also will
ily internal medicine physicians and other clinicians who wish to advance disclose any off-label and/or investigational use of pharmaceuticals or instru-
For editorial
their current knowledge of clinical medicine and who wish to stay abreast ments discussed in their presentation. Disclosure of this information will be comment, see
of advances in medical research. published in course materials so that those participants in the activity may page 848
Statement of Need: General internists and primary care physicians must formulate their own judgments regarding the presentation.
maintain an extensive knowledge base on a wide variety of topics covering In their editorial and administrative roles, William L. Lanier, Jr, MD, Terry L.
all body systems as well as common and uncommon disorders. Mayo Clinic Jopke, Kimberly D. Sankey, and Nicki M. Smith, MPA, have control of the con- Dana-Farber Cancer Insti-
Proceedings aims to leverage the expertise of its authors to help physicians tent of this program but have no relevant financial relationship(s) with industry. tute and Harvard Medical
understand best practices in diagnosis and management of conditions Dr Steensma is a consultant for Celgene, MEI Pharma, Astex, and H3/Eisai and School, Boston, MA.
encountered in the clinical setting. serves on the Data Safety Monitoring Committee for Amgen and Novartis.
Accreditation: Mayo Clinic College of Medicine is accredited by the Accred- Only azacitidine, decitabine, and lenalidomide are FDA approved for MDS.
itation Council for Continuing Medical Education to provide continuing med- All other agents and uses are off label or experimental.
ical education for physicians. Method of Participation: In order to claim credit, participants must com-
Credit Statement: Mayo Clinic College of Medicine designates this journal- plete the following:
based CME activity for a maximum of 1.0 AMA PRA Category 1 Credit(s). 1. Read the activity.
Physicians should claim only the credit commensurate with the extent of 2. Complete the online CME Test and Evaluation. Participants must achieve
their participation in the activity. a score of 80% on the CME Test. One retake is allowed.
Learning Objectives: On completion of this article, you should be able to (1) Visit www.mayoclinicproceedings.com, select CME, and then select CME ar-
diagnose myelodysplastic syndrome in a patient with unexplained cytopenias; ticles to locate this article online to access the online process. On successful
(2) describe the role of molecular testing in evaluation of patients with sus- completion of the online test and evaluation, you can instantly download and
pected myelodysplastic syndromes; and (3) apply risk stratification tools to print your certificate of credit.
choose appropriate initial therapy for patients with myelodysplastic syndromes. Estimated Time: The estimated time to complete each article is approxi-
Disclosures: As a provider accredited by ACCME, Mayo Clinic College of mately 1 hour.
Medicine (Mayo School of Continuous Professional Development) must Hardware/Software: PC or MAC with Internet access.
ensure balance, independence, objectivity, and scientific rigor in its educa- Date of Release: 7/1/2015
tional activities. Course Director(s), Planning Committee members, Faculty, Expiration Date: 6/30/2017 (Credit can no longer be offered after it has
and all others who are in a position to control the content of this educational passed the expiration date.)
activity are required to disclose all relevant financial relationships with any Privacy Policy: http://www.mayoclinic.org/global/privacy.html
commercial interest related to the subject matter of the educational activity. Questions? Contact dletcsupport@mayo.edu.

Abstract

In the past few years, new biological insights into the myelodysplastic syndromes (MDS) resulting from
molecular genetic analysis have improved pathologic understanding, but treatment advances have not kept
pace. More than 40 genes are now known to be recurrently mutated in MDS. However, because most of
these genes encode spliceosome components, chromatic remodeling factors, epigenetic pattern modula-
tors, or transcription factors rather than more easily inhibited activated tyrosine kinases, there are as of yet
few narrowly targeted therapies available for MDS. Three drugsdazacitidine, decitabine, and lenalido-
midedwere approved by the US Food and Drug Administration for MDS indications a decade ago, and
these agents can improve hematopoiesis, delay disease progression, and improve survival and quality of
life for a subset of patients. However, only a few patients with MDS respond to these agents, and their
benefit is temporary. The only potentially curative therapy for MDS is allogeneic hematopoietic stem cell
transplant, but owing to the advanced age of many patients with MDS and the frequency of serious co-
morbid conditions, less than 10% of patients currently undergo stem cell transplant. This narrative review
summarizes the current understanding of MDS and treatment options for these challenging disorders.
ª 2015 Mayo Foundation for Medical Education and Research n Mayo Clin Proc. 2015;90(7):969-983

T
he myelodysplastic syndromes (MDS) myeloid neoplasms characterized by ineffective
are the most commonly diagnosed ac- clonal hematopoiesis and abnormal “dysplastic”
quired bone marrow failure syndromes cell morphology, resulting in peripheral blood
in adults.1 The umbrella term MDS is used to cytopenias and functional blood cell abnormal-
describe a heterogeneous group of chronic ities.2,3 The MDS are inherently unstable and

Mayo Clin Proc. n July 2015;90(7):969-983 n http://dx.doi.org/10.1016/j.mayocp.2015.04.001 969


www.mayoclinicproceedings.org n ª 2015 Mayo Foundation for Medical Education and Research
MAYO CLINIC PROCEEDINGS

may progress over time, including evolution to 70 years, and the incidence increases steadily
acute myeloid leukemia (AML). Acute myeloid with age. In Eastern Europe and parts of Asia,
leukemia is currently defined by the World the median age at diagnosis is younger than in
Health Organization (WHO) as at least 20% the West, for unclear reasons. Although MDS
blasts in the marrow or blood or the presence have a modest male predominance, a specific
of either myeloid sarcomas or certain AML- MDS subtypedMDS associated with isolated
defining karyotypes, such as t(15;17) and deletion of the long arm of chromosome 5, hypo-
t(8;21). Therefore, all patients with MDS have lobated megakaryocytes, and erythroid hypopla-
less than 20% marrow blasts, by definition.4 sia (so-called 5qe syndrome)dis more common
in women.11
EPIDEMIOLOGIC PROFILE Approximately 85% to 90% of MDS cases
Owing to the use of ambiguous diagnostic ter- are idiopathic and result from aging-related he-
minology, the exclusion of MDS cases from matopoietic stem cell injury. Secondary or
most cancer registries, and the incomplete eval- therapy-related MDS (t-MDS) represents 10%
uation of many older patients with cytopenias, to 15% of cases.12 Myelodysplastic syndrome
the incidence and prevalence of MDS have can be induced by exogenous DNA-damaging
been difficult to estimate accurately.5 Data agents, including DNA alkylating drugs (eg,
from the US National Cancer Institute’s Surveil- cyclophosphamide and melphalan), inhibitors
lance, Epidemiology, and End Results Program of topoisomerase II (eg, etoposide), ionizing
(which has captured MDS cases since 2001) sug- radiation, and volatile hydrocarbons (eg, ben-
gest that 10,000 to 12,000 new cases are diag- zene). Although it can be difficult to prove a
nosed in the United States each year (ie, w3-4 direct link between an exposure and subsequent
cases per 100,000 persons per year).6 However, MDS development, features supporting t-MDS
analysis of Medicare claims in conjunction with rather than de novo MDS include complex kar-
Surveillance, Epidemiology, and End Results yotype (defined as 3 acquired chromosome
data indicates that the actual incidence of MDS abnormalities), abnormalities of chromosomes
in the United States may be closer to 30,000 to 5 and 7, and TP53 mutation.
40,000 new cases per yeardseveral times higher Pediatric MDS, which are rare, are frequently
than the incidence of AML.7 associated with inborn disorders such as Fanconi
Aging is the most important risk factor for anemia, Down syndrome, and congenital neutro-
the development of MDS. Owing to errors in penia.13 Germline mutations in RUNX1 and
DNA replication and spontaneous mutations GATA2 transcription factors also predispose to
from normal metabolic by-products (eg, conver- MDS. Germline RUNX1 mutations are associated
sion of cytosine to thymidine by oxidative deam- with a prodrome of thrombocytopenia that is
ination from reactive oxygen species), coding often mistaken for immune thrombocytopenia,
mutations accumulate in hematopoietic stem and GATA2 mutations may be associated with
cells at a mean  SD rate of 0.130.02 exonic monocytopenia, mycobacterial infections, and
mutations per year of life.8 When an acquired lymphedema (MonoMAC syndrome).14,15
DNA mutation or combination of mutations pro-
motes growth or generates a survival advantage CLASSIFICATION
to a hematopoietic stem or progenitor cell, clonal The fourth edition of the WHO Classification of
hematopoiesis emerges. Approximately 10% of Tumours of Hematopoietic and Lymphoid Tissues,
individuals older than 70 years have clonal mu- published in 2008, is currently the most widely
tations in genes associated with myeloid used MDS classification (Table 1).4 The 2008
neoplasia, such as DNMT3A, TET2, and SF3B1, WHO MDS classification is similar to the
and these persons have a 0.5% to 1% chance earliest formal MDS classification, the 1982
per year of acquiring additional mutations that MDS classification of the French-American-
lead to progression to MDS or another hemato- British Cooperative Group. In 2016, a revision
logic neoplasm, similar in magnitude to the of the WHO classification will be published
risk of monoclonal gammopathy of undeter- that will simplify classification and formally
mined significance progressing to myeloma.9,10 address the role of molecular genetic testing
In the United States and Western Europe, the in myeloid neoplasia diagnosis. The current
median age at diagnosis of MDS is approximately WHO classification has limited prognostic
n n
970 Mayo Clin Proc. July 2015;90(7):969-983 http://dx.doi.org/10.1016/j.mayocp.2015.04.001
www.mayoclinicproceedings.org
another condition.
cussed further herein.16

www.mayoclinicproceedings.org
DIAGNOSTIC EVALUATION

Mayo Clin Proc. n July 2015;90(7):969-983


n
been developed, as described further herein.

group that includes chronic myelomonocytic leu-

tion of ATRX, a chromatin remodeling factor


exhibit both MDS and myeloproliferative features
value, so other risk stratification tools have

plete blood cell count is performed to evaluate


with MDS may seek medical attention because

are discovered to have MDS only when a com-


precursors or Auer rods are present, and, to

patients are asymptomatic at diagnosis and


of fatigue, dyspnea, poor exercise tolerance,
that alters alpha-globin expression.18 Patients
less than 100109/L or an absolute neutrophil
g/dL; 80% of patients have a platelet count
level of patients at diagnosis is 9.5 g/dL (to

cytic but rarely can be microcytic owing to


multiple cytopenias. The median hemoglobin
penia in MDS, but most patients present with
AML because outcomes in these patients are
related MDS is grouped with therapy-related
dysplastic cells, whether ring sideroblast erythroid
The 2008 WHO classification considers

toid erythroid maturation, binucleate erythroid


more than 10% of examined cells is considered
Currently, the observation of dysplastic
bruising, bleeding, or an infection, but some
patients have a hemoglobin level less than 11
convert to g/L, multiply by 10.0), and 75% of
MDS/MPN are distinct from those of MDS
and refractory anemia with ring sideroblasts

genetics, biology, and clinical approach to


and marked thrombocytosis (which requires a
such as leukocytosis and thrombocytosisda
poor regardless of the blast count. For cases that

acquired alpha-thalassemia from somatic muta-


ated with MDS is usually macrocytic or normo-
count less than 1.0109/L.17 Anemia associ-
Anemia is the most commonly observed cyto-
cation includes a separate MDS/myeloprolifera-
platelet count >450109/L)dthe WHO classifi-
a limited extent, karyotype results. Therapy-
myeloid cell lineages exhibit greater than 10% of
marrow and blood blast proportion, which

kemia (defined by >1109/L blood monocytes)

without proliferative features and are not dis-


tive neoplasm (MPN) overlap category. The

observed dysplastic features include megaloblas-


important for MDS diagnosis.19 Commonly
blood and marrow cell morphologic features in
TABLE 1. 2008 World Health Organization (WHO) Classification of Myelodysplastic Syndromes (MDS)a,b
Key features WHO-estimated
patients with
Name Abbreviation Peripheral blood Bone marrow MDS (%)
MYELODYSPLASTIC SYNDROMES: DIAGNOSIS AND TREATMENT

RCUD
Refractory anemia RA Anemia; <1% peripheral blood blasts Unilineage erythroid dysplasia (in >10% of cells); <5% blasts 10-20

http://dx.doi.org/10.1016/j.mayocp.2015.04.001
Refractory neutropenia RN Neutropenia; <1% peripheral blood blasts Unilineage granulocytic dysplasia; <5% blasts <1
Refractory thrombocytopenia RT Thrombocytopenia; <1% blasts Unilineage megakaryocytic dysplasia; <5% blasts <1
Refractory anemia with ring sideroblasts RARS Anemia; no blasts Unilineage erythroid dysplasia; 15% of erythroid precursors are
ring sideroblasts; <5% blasts 3-11
Refractory cytopenias with multilineage dysplasia RCMD Cytopenia(s); <1% blasts; no Auer rods Multilineage dysplasia  ring sideroblasts; <5% blasts; no Auer rods 30
Refractory anemia with excess blasts, type 1 RAEB-1 Cytopenia(s); <5% blasts; no Auer rods Unilineage or multilineage dysplasia; 5%-9% blasts; no Auer rods 40c
Refractory anemia with excess blasts, type 2 RAEB-2 Cytopenia(s); 5%-19% blasts;  Auer rods Unilineage or multilineage dysplasia; 10%-19% blasts;  Auer rods
MDS associated with isolated del(5q) Del(5q) Anemia; normal or high platelet count; 1% blasts Isolated 5q31 chromosome deletion; anemia, hypolobated megakaryocytes <5
Childhood MDS, including refractory
cytopenia of childhood (provisional) RCC Pancytopenia <5% marrow blasts for RCC; marrow usually hypocellular w1
MDS, unclassifiable MDS-U Cytopenias; 1% blasts Does not fit other categories; dysplasia; <5% blasts; if no dysplasia, Unknown
MDS-associated karyotype
a
RCUD ¼ refractory cytopenias with unilineage dysplasia.
b
If peripheral blood blasts are 2% to 4%, the diagnosis is RAEB-1 even if marrow blasts are less than 5%. If Auer rods are present, the WHO considers the diagnosis RAEB-2 if the blast proportion is less than 20% (even if <10%) or
acute myeloid leukemia (AML) if at least 20% blasts. Cases of RCUD, RARS, or RCMD where the peripheral blood blasts are exactly 1% should be considered MDS-U according to the WHO. For all subtypes, peripheral blood
monocyte counts must be less than 1109/L. Bicytopenia may be observed in RCUD subtypes, but pancytopenia with unilineage marrow dysplasia should be classified as MDS-U. Therapy-related MDS, whether due to alkylating
agents, topoisomerase II inhibitors, or radiation, is classified together with therapy-related AML in the WHO classification of AML and precursor lesions. The listing in this table excludes MDS/myeloproliferative neoplasm overlap
categories, such as chronic myelomonocytic leukemia, juvenile myelomonocytic leukemia, and RARS with thrombocytosis.
c
This 40% figure represents the proportion of patients with RAEB-1 or RAEB-2, collectively.
Adapted from WHO.4

971
MAYO CLINIC PROCEEDINGS

precursor cells and other nucleation abnormal- and azathioprine), alcohol abuse, human immu-
ities, ring sideroblasts, neutrophil hypolobulation nodeficiency virus infection, and immune-
or hypogranulation, and small megakaryocytes mediated cytopenias need to be excluded.
with abnormally segmented nuclei. The diag- Although the blood smear may suggest MDS,
nostic requirement for morphologic dysplasia marrow aspiration is essential to establish the
may evolve with more routine use of molecular diagnosis. Bone marrow core (trephine) biopsy
genetic testing in diagnostic evaluation. provides complementary information on
Dysplastic morphologic features are often cellularity and architecture, megakaryocyte
accompanied by cellular dysfunction. For morphology, and the presence of fibrosisduseful
example, hypogranular neutrophils have information that may inform therapeutic
impaired bactericidal activity, which increases decisions.
neutropenia-associated infection risk, and Flow cytometric analysis is often used by
platelets in MDS often express abnormally low pathologists to evaluate patients suspected of
levels of procoagulant cell surface markers or having MDS.26 Flow cytometry may detect
lack intracellular granules, which exacerbates aberrant antigen expression by hematopoietic
bleeding from thrombocytopenia.20,21 As a cells or abnormal cell populations, such as
result of such functional defects, infection and increased blasts, but flow cytometry is currently
bleeding risks in MDS correlate poorly with a complementary assay that is best interpreted
the circulating neutrophil and platelet count. in the context of marrow morphology. Because
Although the bone marrow in MDS usually flow cytometric enumeration of marrow blasts
is normocellular or hypercellular for age, 10% is subject to various technical artifacts, it should
to 20% of cases are accompanied by a hypocel- not replace a marrow aspirate manual differen-
lular marrow. These hypoplastic or hypocellu- tial count.
lar MDS cases may be difficult to distinguish In contrast to flow cytometry, cytogenetic
from aplastic anemia and may have an studies of the marrow are essential in evaluation
increased likelihood of response to immuno- of suspected MDS. Abnormal cytogenetic results
suppressive drug therapy.22 Increased reticulin can provide confirmation of the diagnosis when
fibrosis is frequently present in the marrow in the morphologic profile is ambiguous. Specific
MDS; severe fibrosis is associated with poorer karyotypes correlate with prognosis and response
prognosis.23 Occasionally, patients with MDS to treatment (Tables 2 and 3), such as deletion of
present with immune dysregulation, paraneo- chromosome arm 5q and high response rate to
plastic syndromes, or other clonal disorders, lenalidomide.17,27,28 Rarely, fluorescence in situ
such as a small B-cell clone, paroxysmal hybridization (FISH) analysis with probes
nocturnal hemoglobinuria clone, and mono- directed toward common MDS-associated abnor-
clonal gammopathy of undetermined signifi- malities reveals specific chromosomal transloca-
cance. The influence of paroxysmal nocturnal tions and gains or losses of large DNA segments
hemoglobinuria clones on clinical behavior is not detected using standard cytogenetic methods.
unpredictable, but these clones are usually The yield of FISH is low if karyotyping is success-
small, clinically irrelevant, and not associated ful, and the clinical relevance of small clones
with hemolysis.24 Leukocytosis and spleno- detectable only by FISH and not by karyotyping
megaly are rare in MDS, and their presence sug- is unclear.29 Although the WHO classification al-
gests the possibility of chronic myelomonocytic lows labeling patients with cytogenetic abnormal-
leukemia or another MDS/MPN overlap syn- ities but bland morphologic features as having
drome, or an alternative diagnosis. unclassifiable MDS, certain karyotypes (trisomy
Diagnostic evaluation of the patient with sus- 8, loss of the Y chromosome, and deletion 20q)
pected MDS includes medical history and phys- are not specific enough to diagnose unclassifiable
ical examination, complete blood cell count, MDS.4
review of the blood smear, bone marrow aspirate Almost all patients who develop t-MDS
and biopsy, and laboratory tests to rule out other secondary to exposure to mutagenic agents
disorders that mimic MDS.25 Not all that is have chromosomal abnormalities.30 Therapy-
dysplastic is MDS, and nutritional deficiency related MDS that is associated with previous
(iron, vitamin B12, folate, and copper), medication treatment with alkylating agents or exposure
effect (eg, antimetabolites, such as methotrexate to ionizing radiation frequently demonstrates
n n
972 Mayo Clin Proc. July 2015;90(7):969-983 http://dx.doi.org/10.1016/j.mayocp.2015.04.001
www.mayoclinicproceedings.org
MYELODYSPLASTIC SYNDROMES: DIAGNOSIS AND TREATMENT

TABLE 2. The 1997 International Prognostic Scoring System (IPSS) for Myelodysplastic Syndromesa
Category scoreb
Prognostic factor 0 (best) 0.5 1.0 1.5 2.0 (worst)
Marrow blasts (%) <5 5-10 e 11-20 21-30c
Karyotype Good: normal, isolated eY, isolated Intermediate: all karyotypes Poor: abnormal chromosome e e
del(5q), or isolated del(20q) not defined as good or poor 7 or a complex karyotype
(3 anomalies)
Peripheral blood
cytopeniasd 0 or 1 2 or 3 e e e

Median survival (y) Time until 25% of


Total Patients aged Patients aged surviving patients
Risk category score All patients <60 y (n¼205) 60 y (n¼611) developed leukemia (y)
Low 0 5.7 11.8 4.8 9.4
Intermediate-1 0.5 or 1.0 3.5 5.2 2.7 3.3
Intermediate-2 1.5 or 2.0 1.2 1.8 1.1 1.1
High 2.5 0.4 0.3 0.5 0.2
a
Although replaced by the revised IPSS in 2012, numerous clinical trial protocols still use the original IPSS for determination of eligibility.
b
Scoring system: A point value from 0 to 2.0 is determined for each of the 3 prognostic factors, and the 3 values are summed to obtain the total IPSS score.
c
No longer considered myelodysplastic syndrome (redefined as acute myeloid leukemia by the World Health Organization in 2001).
d
The IPSS definition of peripheral blood cytopenias: hemoglobin level less than 10 g/dL; absolute neutrophil count less than 1.8109L; and platelet count less than 100109L.
Adapted from Blood.17

losses involving chromosomes 5 and 7 and MDS-associated genes, the negative predictive
emerges 3 to 7 years after exposure. Patients value of a normal result on an MDS mutation
treated with epipodophyllotoxins may exhibit panel is relatively high, and nonneoplastic
translocations at the chromosome 11q23 locus causes of cytopenias should be carefully
involving the MLL gene; the latency period be- excluded when mutation results are normal.35
tween exposure and diagnosis with this form However, because clonal hematopoiesis is pre-
of t-MDS/AML is typically 1 to 3 years. sent in more than 10% of healthy people older
Increasingly, molecular genetic profiling is than 70 years, clinicians must be cautious in
used to evaluate patients suspected of having interpreting mutations in patients with a
MDS. Molecular genetic profiling can augment normal karyotype and without morphologic
prognostic assessment and predict outcomes changes of dysplasia.
after allogeneic stem cell transplant.31,32 Molec-
ular genetic profiling may also be useful diag- PROGNOSIS
nostically, especially in cases that lack another Risk assessment in MDS is important for therapy
explanation for cytopenias but do not meet selection and for counseling patients. Although
the diagnostic criteria for MDS or another dis- the natural history of MDS includes a risk of pro-
order. The term idiopathic cytopenia(s) of unde- gression to AML in 25% to 30% of patients, most
termined significance (ICUS) has been used for patients with MDS do not develop AML and
such patients, and although some individuals instead die of complications of cytopenias or of
with ICUS will eventually be diagnosed as hav- unrelated conditions that are common in geriatric
ing MDS or AML, others will resolve or be populations, such as cardiovascular disease.36
found to have a nonneoplastic diagnosis.33 This observation led to a change in the name of
Several commercial vendors offer MDS molec- these disorders in the 1970s from the simpler
ular genetic panels, and a homegrown clinically but incomplete designation preleukemia to MDS.
validated 96-gene panel that returns results in Until recently, the most widely used risk strat-
72 hours is now routinely used for evaluating ification tool for MDS was the 1997 International
patients with ICUS or suspected MDS in the au- Prognostic Scoring System (IPSS) (Table 2).17 The
thor’s practice.34 Because more than 80% of pa- IPSS, which is still reflected in drug labels and
tients with MDS have a somatic mutation in 1 used for clinical trial eligibility determination, is
of the more than 40 most commonly mutated based on the sum of 3 subscores: karyotype,

Mayo Clin Proc. n July 2015;90(7):969-983 n http://dx.doi.org/10.1016/j.mayocp.2015.04.001 973


www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

TABLE 3. 2012 Revised International Prognostic Scoring System (IPSS-R)a-c


Updated cytogenetic classification for use in the IPSS-R (n¼7012)
Time until 25% of
Median patients developed Patients in
Risk group Included karyotypes survival (y) AML (y) this group (%)
Very good del(11q), -Y 5.4 Not reached 4
Good Normal, del(20q), del(5q) alone or with 1 other anomaly, del(12p) 4.8 9.4 72
Intermediate þ8, del(7q), i17q, þ19, any other single or double abnormality not 2.7 2.5 13
listed, 2 or more independent clones
Poor Abnormal 3q, -7, double abnormality include -7/del(7q), complex with 1.5 1.7 4
3 abnormalities
Very poor Complex with >3 abnormalities 0.7 0.7 7

IPSS-R
Categories and associated scores
Parameter Very good Good Intermediate Poor Very Poor
Cytogenetic risk group 0 1 2 3 4
Marrow blasts (%) <2 2-<5 5-10 >10
0 1 2 3
Hemoglobin (g/dL) 10 8-<10 <8
0 1 1.5
Absolute neutrophil count (109/L) 0.8 <0.8
0 0.5
Platelet count (109/L) >100 50-100 <50
0 0.5 1

IPSS-R (see: http://www.mds-foundation.org/ipss-r-calculator/)


% patients in this risk group Median survival, Median survival for pts Time until 25% of patients
Risk group Points (n¼7012; AML data on 6485) years under 60 years develop AML, years
Very low 0-1.5 19% 8.8 Not reached Not reached
Low 2.0-3.0 38% 5.3 8.8 10.8
Intermediate 3.5-4.5 20% 3.0 5.2 3.2
High 5.0-6.0 13% 1.5 2.1 1.4
Very high >6.0 10% 0.8 0.9 0.7
a
AML ¼ acute myeloid leukemia.
b
SI conversion factors: To convert hemoglobin values to g/L, multiply by 10.0.
c
Possible range of summed scores: 0-10.
Adapted from Blood.28

number of cytopenias, and proportion of marrow factors such as transfusion dependence, and did
blasts. Patients younger than 60 years with a low not adequately stratify patients with MPN fea-
IPSS score have median survival of more than 5 tures or those with t-MDS. In addition, in each
years, and older patients with a high IPSS score IPSS risk group there are wide variations in pa-
have median survival of only less than 6 months, tient outcomes. Several newer MDS prognostic
if treated with supportive care alone. For each systems have been introduced since 2007 to try
IPSS risk group, outcomes tend to be better for to overcome these limitations. The WHO-based
younger patients than for older patients. Prognostic Scoring System integrates the WHO
The 1997 IPSS had several important limita- classification with karyotyping data, the degree
tions.37 It was validated only for adult patients of anemia, and the presence or absence of marrow
with de novo disease treated with supportive fibrosis; it is used primarily in Europe.38 A general
care or hematopoietic growth factors. It did not risk model proposed by investigators at the MD
weight severity of cytopenias, included a limited Anderson Cancer Center is valid across a broad
repertoire of karyotypes, did not include risk spectrum of patients with MDS, including those
n n
974 Mayo Clin Proc. July 2015;90(7):969-983 http://dx.doi.org/10.1016/j.mayocp.2015.04.001
www.mayoclinicproceedings.org
MYELODYSPLASTIC SYNDROMES: DIAGNOSIS AND TREATMENT

with t-MDS and those who previously have been and chromatin remodeling (Figure 1).35 Cell
treated with a disease-modifying drug.39 A risk culture studies with primary MDS samples have
model for IPSS lower-risk MDS also was devel- found reduced growth of multilineage colony-
oped at the MD Anderson Cancer Center.40 forming unit progenitors.43,44 Experimental
A revised IPSS (IPSS-R) based on analysis of evidence also implicates inhibitory cytokines
more than 7000 patients from more than 10 and increased intramedullary apoptosis as con-
countries was published in 2012 (Table 3).28 tributors to ineffective hematopoiesis in early
The major changes in the IPSS-R from the IPSS MDS.44 The role of the marrow microenviron-
are that the IPSS-R includes a broader range of cy- ment and hematopoietic niche in MDS continues
togenetic abnormalities than those included in the to undergo scrutiny as stromal support of the
1997 IPSS, and the IPSS-R also weights cytoge- growth and maturation of normal hematopoietic
netic findings more heavily than other variables, progenitors is impaired in MDS, and in a murine
such as cytopenias, whereas the IPSS weighted model, mutation of a gene in osteoprogenitor cells
blasts most heavily. In addition, blast cutoff points resulted in an MDS-like phenotype.45
are different in the IPSS-R, which has 5 risk cate-
gories compared with 4 for the IPSS, and the de- TREATMENT
gree of cytopenias is considered in the IPSS-R, Other than allogeneic hematopoietic stem cell
whereas the IPSS considered only the number of transplant (HSCT), which is currently used in
cytopenias. Despite these improvements, the less than 10% of patients and is successful 20%
IPSS-R is still valid only in patients with de to 50% of the time, there is no cure for MDS.46
novo MDS at the time of diagnosis. In addition, Three medications have specific US Food and
other prognostically important variables, such Drug Administration (FDA) approval for
as the presence of comorbid conditions, kinetics MDS-related indications: azacitidine, a DNA
of clonal evolution, and molecular genetic find- methyltransferase inhibitor and nucleoside
ings, are not accounted for by the IPSS-R or by analogue that was approved in 2004; lenalido-
any of the other MDS risk stratification tools. mide, a modulator of cereblon and ubiquitin
In the future, molecular genetic data will ligase activity that was approved in 2005; and
be increasingly used for risk stratification. decitabine, another DNA methyltransferase in-
For example, in 1 study, mutations in EZH2, hibitor that was approved in 2006. Several other
ETV6, ASXL1, RUNX1, and TP53 retained drugs, such as hematopoietic growth factors,
prognostic significance independent of the are commonly used off label in the MDS setting.
IPSS in a multivariable analysis, and the over- Risk stratification systems, such as the IPSS-
all mutation burden was also prognostic, with R, supplemented by molecular genetic testing
a higher risk of AML progression and death in and clinical assessment, can aid treating physi-
those with more detected mutations.41 cians in therapeutic decision making.47,48 The
goals of MDS therapy depend on prognostic
BIOLOGY assessment and the individual patient profile.
A detailed discussion of MDS biology is beyond For lower-risk patients, common goals of therapy
the scope of this review. Cytogenetic and mo- include symptom control and quality of life,
lecular genetic abnormalities indicate that improvement of hematopoiesis, and delay of dis-
MDS are clonal disorders. The neoplastic clone ease progression.49,50 For higher-risk patients,
in MDS includes stem/progenitor cells and extension of survival has been reported with aza-
more differentiated myeloid, erythroid, and citidine therapy.49-51 Figure 2 displays a general
megakaryocytic cells; B cells are also sometimes treatment algorithm for MDS. Experimental com-
part of the clonal process, but T cells are rarely pounds currently undergoing testing in patients
involved, although T-cell clones similar to with MDS were recently reviewed elsewhere.52
those seen in large granular lymphocyte leuke-
mia can be detected in some patients.42 Most Supportive Care: Transfusions and Iron
marrow and blood cells in MDS are clonal, Chelation
even if blast counts are not increased. The proportion of patients with MDS receiving
Recurrent MDS-associated DNA mutations red blood cell transfusion is approximately 40%
cluster in genes that encode biological pathways in lower-risk patients and 60% to 80% in
for messenger RNA splicing, DNA methylation, higher-risk patients.53 Patients receiving regular

Mayo Clin Proc. n July 2015;90(7):969-983 n http://dx.doi.org/10.1016/j.mayocp.2015.04.001 975


www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

MDS mutation landscape

IPSS independent good prognosis


No clear independent effect
Proliferation CDKN2A <1%
IPSS independent poor prognosis
CBL
2% Impaired differentiation
JAK2
BRAF <1% PTPN11 <1%
3%
RUNX1
ETV6 SETBP1
9%
GNAS <1% 3% 7%
KRAS NRAS PTEN <1%
1% 4%
STAG2
NPM1 2% and other
TP53
Epigenetic regulation cohesins -
8%
Other 5%-10%
EZH2
6% ASXL1
DNMT3A 14% SF1 Pre-mRNA splicing SF3A1
8% 1%
1%

U2AF1 PRPF40B
IDH1/2 1%
2% 8%
TET2 SF3B1
21% UTX ATRX 22%
SRSF2
1% <1% U2AF65
ZRSR2 11%
5% <1%

FIGURE 1. Recurrent somatic mutations in myelodysplastic syndromes (MDS), including approximate frequency and prognostic
significance. Some mutations influence the phenotype and are, therefore, more common in specific subtypes of MDS; for example,
SF3B1 mutations are found in more than 50% of patients with refractory anemia with ring sideroblasts, ATRX mutations are most
common in MDS associated with acquired alpha-thalassemia, and SRSF2 mutations are more common in myeloproliferative
neoplasm/MDS overlap syndromes, such as chronic myelomonocytic leukemia. The prognostic value of SF3B1 mutations is unclear,
with some series suggesting no independent value and others suggesting a more favorable outcome even when accounting for other
clinicopathologic parameters. IPSS ¼ International Prognostic Scoring System. Adapted from the American Society of Hematology.101

transfusions have inferior survival compared noninvasively measuring hepatic, cardiac, and
with those who do not require transfusions, other organ iron concentrations. Quantitative
either because transfusions indicate more severe R2*/T2* magnetic resonance imaging of the liver
hematopoietic failure or because transfusions or heart may be useful in determining which pa-
themselves cause harm via iron overload, tients are appropriate candidates for iron chela-
immunomodulation, or another mechanism.54 tion therapy. T2* signals in the heart are
There is controversy about the magnitude of usually normal until patients have received at
risk from transfusion-related hemosiderosis in least 80 to 100 U of blood.58 Measurement of
patients with MDS and the importance of hemo- reactive oxygen species (nonetransferrin-bound
siderosis compared with other disease-associated iron) suggests that these volatile species are
risks.55,56 Lower-risk patients with MDS who elevated in transfused patients with MDS and
have a ferritin level greater than 1000 ng/mL decrease rapidly during chelation therapy, but
(to convert to pmol/L, multiply by 2.247) have the clinical significance is unclear.59
worse survival than patients with lower ferritin Iron chelation therapy with oral deferasirox
levels, and patients with ferritin levels greater or parenteral deferoxamine can be considered
than 2500 ng/mL have inferior survival with in patients with a relatively good MDS prognosis
HSCT.54,57 However, serum ferritin is an acute who have evidence of tissue iron overload. There
phase reactant that has only moderate correlation are currently no controlled prospective data indi-
with iron burden, so newer magnetic resonance cating benefit from iron chelation in MDS, and
imaging techniques have been developed for chelation therapy is expensive (deferasirox), is
n n
976 Mayo Clin Proc. July 2015;90(7):969-983 http://dx.doi.org/10.1016/j.mayocp.2015.04.001
www.mayoclinicproceedings.org
MYELODYSPLASTIC SYNDROMES: DIAGNOSIS AND TREATMENT

MDS initial diagnosis (using WHO 2008 diagnostic criteria,


supplemented by novel genomics approaches)

Individualized risk assessment, using Higher-risk


Clinical monitoring IPSS-R or other tools
Lower-risk
No
Is there a need for
Development of a treatment now?
need for therapy
Is the patient a
Yes transplant candidate?
No, other important
Is anemia cytopenias are also present
isolated, or the No Yes
major problem?
Azacitidine or decitabine AlloSCT, perhaps
Yes (ie, HMA) until disease with HMA or
progression, relapse, or chemotherapy as
drug intolerance bridging therapy
Is del5q No sEPO <500 No
present? U/L?
Relapse
Yes
Yes
Lenalidomide; if sEPO Optimal approach is
<500 U/L, ESA trial ESA ± G-CSF Donor lymphocyte
unclear; consider G-
before or after CSF or TPO agonist, infusion or second
Failure alloSCT, possibly after
Failure HMA, IST, clinical trial
cytoreductive therapy
Failure

Failure Failure
Optimal therapy unclear consider HMA, IST,
androgens, lenalidomide (if not already used),
or clinical trial
AlloSCT, enrollment in a clinical Enrollment in a clinical trial,
trial, or palliative/supportive care or supportive/palliative care

FIGURE 2. A general approach to myelodysplastic syndromes (MDS) therapy. All the patients should receive supportive care with
transfusions and antimicrobial agents as needed. Iron chelation therapy can be considered for selected red blood cell transfusione
requiring lower-risk patients. For lower-risk patients in whom the clinical picture is dominated by anemia, the initial therapeutic
choice depends on the karyotype and the serum erythropoietin (sEPO) level. For patients with del(5q), lenalidomide is an appropriate
first choice and is Food and Drug Administration approved for that indication. For patients without del(5q) but with sEPO levels less
than 500 U/L, the erythropoiesis-stimulating agents (ESAs) epoetin and darbepoetin are recommended. The most appropriate
therapy for lower-risk patients with anemia with sEPO levels greater than 500 U/L and without del(5q), pancytopenia, or a clinical
picture dominated by individual cytopenias other than anemia (ie, neutropenia or thrombocytopenia) is unclear. Hypomethylating
agents can be beneficial in some patients with lower-risk disease, although their effect on survival in this group is unclear, and some
reports indicate the potential for inferior survival. Patients with isolated thrombocytopenia may overlap with immune thrombocy-
topenia and may benefit from corticosteroids, romiplostim or eltrombopag, intravenous gamma globulin or other immune throm-
bocytopeniaedirected therapies. Immunosuppressive therapy, lenalidomide, supportive care alone, and hematopoietic stem cell
transplant (HSCT) are all reasonable choices in the other patient groups, depending on patient-specific factors. For higher-risk patients,
the treatment approach differs depending on whether the patient is an HSCT candidate. Higher-risk patients who are HSCT can-
didates should proceed with definitive HSCT therapy as soon as feasible. The HSCT may be preceded by a few treatment cycles of a
hypomethylating agent as a “bridging” therapy to try to cytoreduce or at least keep the disease stable until a donor is identified and
pretransplant screening tests are completed. Patients who are not HSCT candidates can be treated with a hypomethylating agent until
disease progression or intolerance. Some investigators prefer azacitidine over decitabine because of the demonstrated survival
advantage in this setting. Once initial therapy fails, no optimal second-line therapy is defined, and the choice depends on clinical
circumstances. Clinical trial enrollment is always appropriate if a well-designed study is available for which the patient is eligible.
alloSCT ¼ allogeneic stem cell transplant; G-CSF ¼ granulocyte colony-stimulating factor; HMA ¼ hypomethylating agent (azacitidine
or decitabine); IPSS-R ¼ 2012 Revised International Prognostic Scoring System; IST ¼ immunosuppressive therapy; TPO ¼
thrombopoietin; WHO ¼ World Health Organization. Partly based on European LeukemiaNet and National Comprehensive Cancer
Network guidelines; see Malcovati et al48 and http://www.nccn.org. Adapted from Blood.52

Mayo Clin Proc. n July 2015;90(7):969-983 n http://dx.doi.org/10.1016/j.mayocp.2015.04.001 977


www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

cumbersome (deferoxamine), and can have an endogenous TPO level less than 500 pg/mL
adverse effects. In elderly patients with MDS, a and are not heavily dependent on platelet trans-
dose of deferasirox high enough to cause a nega- fusions.69 One important safety concern is that
tive iron balance (ie, 20-30 mg/kg per day) blasts can have functional TPO receptors, so
often results in elevated creatinine levels or intol- patients treated with romiplostim or eltrombo-
erable gastrointestinal symptoms.59,60 pag may experience an increase in blood or
Bleeding is the second most common non- marrow blasts. In a placebo-controlled study
AML cause of death in MDS, after infection.61 of romiplostim, AML progression was observed
Platelet transfusions can decrease bleeding in 6% of romiplostim-treated patients and in
risk, but the development of alloimmunization 2.4% who received placebo, and most patients
is common with repeated platelet transfusions. who progressed to AML had excess blasts before
therapy.70 Romiplostim combined with azaciti-
Hematopoietic Growth Factors dine, decitabine, or lenalidomide has also been
Recombinant erythropoiesis-stimulating agents used to ameliorate thrombocytopenia. The antifi-
(ESAs; epoetin and darbepoetin) induce erythroid brinolytic drug epsiloneaminocaproic acid can
response rates in 20% to 50% of patients.62-64 decrease bleeding in thrombocytopenic patients
Combinations of an ESA and granulocyte who have mucosal hemorrhage, but it increases
colony-stimulating factor (G-CSF) may be more thrombosis risk when used systemically.71
effective than an ESA alone in ameliorating ane-
mia, especially in patients with refractory anemia Immunosuppressive and Immunomodulatory
with ring sideroblasts, for unclear reasons.62 Drugs
Although several retrospective studies suggest Autoreactive T cells, either clonal or polyclonal,
that ESAs may improve life expectancy in MDS, can contribute to suppressed hematopoiesis in
no prospective studies have found increased sur- some patients with MDS.72 This has prompted
vival. For patients with serum erythropoietin studies with antieT-cell immunosuppressive
levels less than 500 U/L, an 8- to 12-week trial treatment approaches similar to those used for
of an ESA is appropriate; patients with higher aplastic anemia, such as antithymocyte globulin
serum erythropoietin levels rarely respond.65 with a calcineurin inhibitor such as cyclosporine
The myeloid growth factors G-CSF (filgrastim or tacrolimus. The most difficult task is selection
and tbo-filgrastim) and granulocyte-macrophage of patients most likely to respond.73 Benefits are
CSF (sargramostim and molgramostim) increase primarily seen in younger patients (<60 years)
the neutrophil count in many patients but do with lower-risk disease who are not transfusion
not increase survival. In one study of 102 pa- dependent and have either normal cytogenetics
tients, progression to AML was similar between or trisomy 8.74 In some studies, marrow hypocel-
G-CSFetreated patients and the control group, lularity and HLA-DR15 have predicted a higher
but survival was shorter in patients with excess likelihood of response to immunosuppression.
blasts who received G-CSF.66 Patients with In the 1990s, thalidomide was used as an
recurrent infections are the best candidates for immunomodulatory agent and as an inhibitor
myeloid growth factors but may not be helped of neoangiogenesis. Favorable responses were
by these agents if the increased circulating neu- seen in 15% to 25% of patients, but adverse
trophils are dysfunctional. There are reports of events were problematic, especially sedation,
spontaneous splenic rupture and leukemoid re- constipation, and neuropathy.75,76 Lenalidomide
actions in MDS with pegfilgrastim, so this agent is a drug that is a minor chemical modification of
should be used only with caution.67 thalidomide and has an improved safety profile. It
One of the major shifts in clinical practice in is now known that the pleiotropic biological ef-
the past few years is the increased use of throm- fects of thalidomide and lenalidomide are medi-
bopoietin (TPO) receptor agonists in MDS.68 ated by modulation of the activity of cereblon, a
Although romiplostim and eltrombopag are component of an E3 ubiquitin ligase complex
currently not approved for use in MDS, their la- that targets cellular proteins for degradation.77
bel includes immune thrombocytopenia, which Lenalidomide specifically alters the degradation
some patients with MDS also have. These rate of casein kinase 1, a serine-threonine kinase
agents can improve platelet counts and reduce encoded on chromosome arm 5q that modulates
bleeding events, especially in patients who have Wnt/b-catenin signaling.78
n n
978 Mayo Clin Proc. July 2015;90(7):969-983 http://dx.doi.org/10.1016/j.mayocp.2015.04.001
www.mayoclinicproceedings.org
MYELODYSPLASTIC SYNDROMES: DIAGNOSIS AND TREATMENT

Lenalidomide is most effective in patients Median survival was 24 months in patients


with IPSS low-risk or intermediate-1erisk dis- receiving azacitidine and 15 months in patients
ease who have deletion of chromosome 5q31. receiving conventional care; thus, azacitidine
In this group, 67% achieve transfusion indepen- was the first choice. The complete response rate
dence, with a median time to response of 4.6 in the azacitidine-treated group was less than
weeks, a median increase in hemoglobin level 20%, but subsequent analysis found that a com-
of 5.4 g/dL, and a median duration of response plete response was not necessary for patients to
of more than 2 years.79 Cytogenetic remissions achieve a survival benefit.86 Azacitidine is FDA
are seen in up to one-half of patients, but approved for either intravenous administration
del(5q) hematopoietic stem cells are still detect- or subcutaneous dosing. Intravenous administra-
able even in patients in remission.80 For patients tion avoids injection site reactions but necessi-
who lack del(5q), responses are less frequentd tates venous access.
approximately 25% become transfusion free Decitabine is also clinically active in MDS,
during lenalidomide therapydand less durable, but neither an American nor a European multi-
with a median response of 40 weeks.81 The most center study of decitabine reported a survival
common adverse effect with lenalidomide is benefit.87,88 It is unclear whether this is because
myelosuppression. Patients with a low platelet decitabine is an inferior molecule to azacitidine
count (especially <50109/L), excess blasts, or or because the studies enrolled different patient
a complex karyotype are less likely to respond populations than AZA-001. The latter is sug-
to lenalidomide therapy. gested by the short (<10 months) survival of
the control arms in decitabine studies. Decita-
Hypomethylating Agents (DNA bine is given most commonly intravenously
Methyltransferase Inhibitors) for 5 consecutive days every 4 to 6 weeks.89
Epigenetic changes are those such as DNA Clinical response to hypomethylating
methylation or histone acetylation that alter agents may be slow, and an adequate therapeu-
gene expression without changing the DNA tic trial of either agent requires at least 4 treat-
sequence. Methylated cytidine residues cluster ment cycles with decitabine or 6 treatment
in CpG islands, which are located near the pro- cycles with azacitidine. The most common
moter regions of many genes. DNA methylation adverse events associated with these drugs are
is a dynamic process that affects transcription neutropenia and thrombocytopenia, which
rates of adjacent genes; when CpG islands are may improve with time as the MDS clones are
hypermethylated, expression of nearby genes is suppressed and normal hematopoiesis recovers.
silenced.82 The aza-substituted cytosine nucleo- The optimal maintenance dosing once patients
side analogues azacitidine and decitabine are in- achieve a response is unknown, but some main-
hibitors of DNA methyltransferase 1, the enzyme tenance therapy seems to be required to main-
that maintains cytidine methylation patterns. tain responses.90 Thus far, no therapy has
Azacitidine and decitabine treatment decreases been found to improve survival for patients
methylation of DNA and reverses gene silencing. with lower-risk MDS, although azacitidine or
These agents also induce DNA damage similar to decitabine can reduce transfusions and improve
cytarabine and other nucleoside analogues, and counts in some patients.
it remains unclear whether epigenetic changes Deacetylase inhibitors maintain chromatin in
or other activities are responsible for clinical ac- a transcriptionally active state by inhibiting deace-
tivity.83 Patients with TET2 or DNMT3A muta- tylation of histone tails on chromatin. In vitro,
tions have a higher likelihood of response to these agents reverse transcription repression and
DNA methyltransferase 1 inhibitor therapy.84,85 gene silencing and are synergistic with hypome-
AZA-001 was a multicenter trial in which 358 thylating agents. However, although several
patients with IPSS intermediate-2e or high-risk deacetylase inhibitors have been FDA approved
MDS were randomized to receive either azaciti- for lymphoma (panobinostat and vorinostat), in
dine, 75 mg/m2 subcutaneously for 7 consecutive 3 randomized cooperative group trials, the com-
days every 28 days, or conventional care (best bination of azacitidine plus a deacetylase inhibitor
supportive care alone, low-dose cytarabine, or (entinostat [MS-275] in E1905, vorinostat in
AML-like induction chemotherapy using in- S1117, and valproic acid in Aza-Plus) was not su-
fusional cytarabine and an anthracycline).50 perior to azacitidine monotherapy.91,92

Mayo Clin Proc. n July 2015;90(7):969-983 n http://dx.doi.org/10.1016/j.mayocp.2015.04.001 979


www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

Once hypomethylating agents fail the patient, suggests that the best strategy is to perform trans-
the prognosis is grim, with median survival of less plant early in the disease course in patients with
than 6 months.93,94 Switching from one failed higher-risk disease and to defer transplant in pa-
hypomethylating agent to the other agent or add- tients with lower-risk disease (ie, IPSS low and
ing lenalidomide or a deacetylase inhibitor is intermediate-1 risk or IPSS-R very low and low
rarely helpful. Responses are seen in some pa- risk) until the time of progression.97,98 It is unclear
tients with clofarabine use, AML-type induction what the best strategy is with IPSS-R intermediate-
chemotherapy, or low-dose cytarabine therapy, risk patients, and this is a group where molecular
but patients in whom hypomethylating agents genetic typing may assist in treatment selection.
are failing should instead be referred for HSCT No clear benefit has been found for the
or enrolled in clinical trials if possible. administration of 1 or more courses of cyto-
toxic chemotherapy or hypomethylating agent
Allogeneic HSCT therapy before HSCT, although pretransplant
Treatment of choice for children and young adults therapy may be useful to reduce the burden
with MDS is HSCT. Younger patients (ie, 40 of marrow blasts and clonal cells before
years) without excess blasts at the time of trans- HSCT.99 Most patients with higher-risk disease
plant may have long-term disease-free survival relapse after HSCT, and a variety of strategies to
exceeding 50% after an HLA-matched HSCT. Un- reduce relapse rates are being studied.100
less the patient has t-MDS, it is imperative to
perform a chromosome breakage assay to exclude CONCLUSION
Fanconi anemia before transplant in a child or The MDS are among the most common and
young adult because patients with Fanconi ane- serious hematologic disorders diagnosed in
mia cannot tolerate conventional conditioning older adults. Treatment options are limited,
regimens. Although most patients with Fanconi but HSCT offers the possibility of cure, and
anemia have short stature, radial ray anomalies, hematopoietic growth factors, azacitidine,
or other dysmorphic features, many do not. lenalidomide, decitabine, and supportive care
Advanced age, the presence of comorbidities, can provide palliative benefit. New insights
clinical inertia, and lack of a suitable donor limit into pathogenesis will, hopefully, lead to novel
the availability of allogeneic HSCT, but the therapies and improved patient outcomes.
growing use of reduced-intensity conditioning
approaches and alternative stem cell sources
Abbreviations and Acronyms: alloSCT = allogeneic stem
such as cord blood and mismatched donors cell transplant; AML = acute myeloid leukemia; ESA =
(including haploidentical donors) are expanding erythropoiesis-stimulating agent; FDA = Food and Drug
the roster of potentially transplant-eligible pa- Administration; FISH = fluorescence in situ hybridization;
tients.46 Therefore, patients with MDS who are G-CSF = granulocyte colony stimulating factor; HSCT =
potentially candidates for transplant should be hematopoietic stem cell transplant; ICUS = idiopathic cyto-
penia(s) of undetermined significance; IPSS = International
evaluated early in the disease course by a physi- Prognostic Scoring System; IPSS-R = revised International
cian with expertise in stem cell transplant. In Prognostic Scoring System; MDS = myelodysplastic syn-
many centers, reduced-intensity stem cell trans- drome; MPN = myeloproliferative neoplasm; RCUD =
plant is routinely performed for patients aged refractory cytopenias with unilineage dysplasia; t-MDS =
therapy-related myelodysplastic syndrome; TPO = throm-
up to the early 70s. Patients with high IPSS scores
bopoietin; WHO = World Health Organization
or treatment-resistant disease (including persis-
tence of excess blasts despite a hypomethylating Potential Competing Interests: Dr Steensma is a consultant
agent) have lower survival rates after HSCT. Pa- for Celgene, MEI Pharma, Astex, and H3/Eisai and serves on the
tients with a complex monosomal karyotype, Data Safety Monitoring Committee for Amgen and Novartis.
defined as 2 or more autosomal monosomies or
Statement About Off-label Drug Use: Only azacitidine, deci-
1 monosomy plus additional structural chromo- tabine, and lenalidomide are FDA approved for MDS. All other
somal abnormalities, have especially poor out- agents and uses are off label or experimental.
comes, as do those with t-MDS; when patients
Correspondence: Address to David P. Steensma, MD,
with complex karyotype also have a TP53 muta-
Adult Leukemia Program, Division of Hematological Malig-
tion, survival with HSCT is less than 10%.32,95,96 nancies, Department of Medical Oncology, Dana-Farber Can-
The optimal time to refer patients for trans- cer Institute, Brigham and Women’s Hospital, 450 Brookline
plant is unclear, but mathematical modeling Ave, D2037, Boston, MA 02215 (David_Steensma@DFCI.

n n
980 Mayo Clin Proc. July 2015;90(7):969-983 http://dx.doi.org/10.1016/j.mayocp.2015.04.001
www.mayoclinicproceedings.org
MYELODYSPLASTIC SYNDROMES: DIAGNOSIS AND TREATMENT

harvard.edu). Individual reprints of this article and a bound 22. Calado RT. Immunologic aspects of hypoplastic myelodysplas-
reprint of the entire Symposium on Neoplastic Hematology tic syndrome. Semin Oncol. 2011;38(5):667-672.
and Medical Oncology will be available for purchase from 23. Della Porta MG, Malcovati L, Boveri E, et al. Clinical relevance of
our website www.mayoclinicproceedings.org. bone marrow fibrosis and CD34-positive cell clusters in primary
myelodysplastic syndromes. J Clin Oncol. 2009;27(5):754-762.
24. Wang H, Chuhjo T, Yasue S, Omine M, Nakao S. Clinical sig-
The Symposium on Neoplastic Hematology and Medical nificance of a minor population of paroxysmal nocturnal
Oncology will continue in an upcoming issue. hemoglobinuria-type cells in bone marrow failure syndrome.
Blood. 2002;100(12):3897-3902.
25. Steensma DP. Dysplasia has a differential diagnosis: distinguish-
REFERENCES ing genuine myelodysplastic syndromes (MDS) from mimics,
1. Sekeres MA. The epidemiology of myelodysplastic syn- imitators, copycats and impostors. Curr Hematol Malig Rep.
dromes. Hematol Oncol Clin North Am. 2010;24(2):287-294. 2012;7(4):310-320.
2. Vardiman JW. Hematopathological concepts and controversies 26. van de Loosdrecht AA, Westers TM, Westra AH, Drager AM, van
in the diagnosis and classification of myelodysplastic syndromes. der Velden VH, Ossenkoppele GJ. Identification of distinct prog-
Hematology Am Soc Hematol Educ Program. 2006:199-204. nostic subgroups in low- and intermediate-1-risk myelodysplastic
3. Steensma DP, Bennett JM. The myelodysplastic syndromes: syndromes by flow cytometry. Blood. 2008;111(3):1067-1077.
diagnosis and treatment. Mayo Clin Proc. 2006;81(1):104-130. 27. Schanz J, Steidl C, Fonatsch C, et al. Coalesced multicentric
4. Swerdlow SH, Campo E, Harris NL, et al., eds. WHO Classifi- analysis of 2,351 patients with myelodysplastic syndromes in-
cation of Tumours of Haematopoietic and Lymphoid Tissues. 4th dicates an underestimation of poor-risk cytogenetics of mye-
ed. Lyon, France: IARC Press; 2008. lodysplastic syndromes in the international prognostic scoring
5. Ma X. Epidemiology of myelodysplastic syndromes. Am J Med. system. J Clin Oncol. 2011;29(15):1963-1970.
2012;125(7 suppl):S2-S5. 28. Greenberg PL, Tuechler H, Schanz J, et al. Revised interna-
6. Ma X, Does M, Raza A, Mayne ST. Myelodysplastic syn- tional prognostic scoring system for myelodysplastic syn-
dromes: incidence and survival in the United States. Cancer. dromes. Blood. 2012;120(12):2454-2465.
2007;109(8):1536-1542. 29. Ketterling RP, Wyatt WA, VanWier SA, et al. Primary myelo-
7. Cogle CR, Iannacone MR, Yu D, et al. High rate of uncaptured dysplastic syndrome with normal cytogenetics: utility of “FISH
myelodysplastic syndrome cases and an improved method of panel testing” and M-FISH. Leuk Res. 2002;26(3):235-240.
case ascertainment. Leuk Res. 2014;38(1):71-75. 30. Pedersen-Bjergaard J, Andersen MK, Christiansen DH,
8. Xie M, Lu C, Wang J, et al. Age-related mutations associated Nerlov C. Genetic pathways in therapy-related myelodysplasia
with clonal hematopoietic expansion and malignancies. Nat and acute myeloid leukemia. Blood. 2002;99(6):1909-1912.
Med. 2014;20(12):1472-1478. 31. Bejar R. Clinical and genetic predictors of prognosis in myelo-
9. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal he- dysplastic syndromes. Haematologica. 2014;99(6):956-964.
matopoiesis associated with adverse outcomes. N Engl J Med. 32. Bejar R, Stevenson KE, Caughey B, et al. Somatic mutations
2014;371(26):2488-2498. predict poor outcome in patients with myelodysplastic syn-
10. Genovese G, Kähler AK, Rose SA, et al. Clonal hematopoiesis and drome after hematopoietic stem-cell transplantation. J Clin
cancer risk in blood derived DNA sequence. N Engl J Med. In press. Oncol. 2014;32(25):2691-2698.
11. Van den Berghe H, Michaux L. 5q-, twenty-five years later: a 33. Valent P, Bain BJ, Bennett JM, et al. Idiopathic cytopenia of un-
synopsis. Cancer Genet Cytogenet. 1997;94(1):1-7. determined significance (ICUS) and idiopathic dysplasia of un-
12. Qian Z, Joslin JM, Tennant TR, et al. Cytogenetic and genetic certain significance (IDUS), and their distinction from low risk
pathways in therapy-related acute myeloid leukemia. Chem MDS. Leuk Res. 2012;36(1):1-5.
Biol Interact. 2010;184(1-2):50-57. 34. New test developed at Dana-Farber/Brigham and Women’s
13. Niemeyer CM, Baumann I. Myelodysplastic syndrome in chil- Cancer Center speeds treatment decisions for blood cancer pa-
dren and adolescents. Semin Hematol. 2008;45(1):60-70. tients. Dana-Farber Cancer Institute website. http://www.dana-
14. Owen CJ, Toze CL, Koochin A, et al. Five new pedigrees with farber.org/Newsroom/News-Releases/new-test-developed-at-
inherited RUNX1 mutations causing familial platelet disorder dana-farber-brigham-and-womens-cancer-center-speeds-
with propensity to myeloid malignancy. Blood. 2008;112(12): treatment-decisions-for-blood-cancer-patients.aspx. Publsihed
4639-4645. November 11, 2014. Accessed March 1, 2015.
15. Hahn CN, Chong CE, Carmichael CL, et al. Heritable GATA2 mu- 35. Papaemmanuil E, Gerstung M, Malcovati L, et al. Clinical and
tations associated with familial myelodysplastic syndrome and biological implications of driver mutations in myelodysplastic
acute myeloid leukemia. Nat Genet. 2011;43(10):1012-1017. syndromes. Blood. 2013;122(22):3616-3627; quiz 3699.
16. Savona MR, Malcovati L, Komrokji R, et al. An international 36. Della Porta MG, Malcovati L, Strupp C, et al. Risk stratification
consortium proposal of uniform response criteria for myelo- based on both disease status and extra-hematologic comor-
dysplastic/myeloproliferative neoplasms (MDS/MPN) in adults. bidities in patients with myelodysplastic syndrome. Haemato-
Blood. 2015;125(12):1857-1865. logica. 2011;96(3):441-449.
17. Greenberg P, Cox C, LeBeau MM, et al. International scoring 37. Santos FP, Kantarjian H, Garcia-Manero G, Ravandi F. The
system for evaluating prognosis in myelodysplastic syndromes. search for better prognostic models in myelodysplastic syn-
Blood. 1997;89(6):2079-2088. dromes. Curr Hematol Malig Rep. 2011;6(1):13-21.
18. Gibbons RJ, Pellagatti A, Garrick D, et al. Identification of 38. Malcovati L, Della Porta MG, Strupp C, et al. Impact of the de-
acquired somatic mutations in the gene encoding chromatin- gree of anemia on the outcome of patients with myelodys-
remodeling factor ATRX in the alpha-thalassemia myelodyspla- plastic syndrome and its integration into the WHO
sia syndrome (ATMDS). Nat Genet. 2003;34(4):446-449. classification-based Prognostic Scoring System (WPSS). Hae-
19. Tefferi A, Vardiman JW. Myelodysplastic syndromes. N Engl J matologica. 2011;96(10):1433-1440.
Med. 2009;361(19):1872-1885. 39. Kantarjian H, O’Brien S, Ravandi F, et al. Proposal for a new
20. Manoharan A, Brighton T, Gemmell R, Lopez K, Moran S, risk model in myelodysplastic syndrome that accounts for
Kyle P. Platelet dysfunction in myelodysplastic syndromes: a events not considered in the original International Prognostic
clinicopathological study. Int J Hematol. 2002;76(3):272-278. Scoring System. Cancer. 2008;113(6):1351-1361.
21. Boogaerts MA, Nelissen V, Roelant C, Goossens W. Blood 40. Garcia-Manero G, Shan J, Faderl S, et al. A prognostic score
neutrophil function in primary myelodysplastic syndromes. for patients with lower risk myelodysplastic syndrome. Leuke-
Br J Haematol. 1983;55(2):217-227. mia. 2008;22(3):538-543.

Mayo Clin Proc. n July 2015;90(7):969-983 n http://dx.doi.org/10.1016/j.mayocp.2015.04.001 981


www.mayoclinicproceedings.org
MAYO CLINIC PROCEEDINGS

41. Bejar R, Stevenson K, Abdel-Wahab O, et al. Clinical effect of 62. Park S, Grabar S, Kelaidi C, et al. Predictive factors of response
point mutations in myelodysplastic syndromes. N Engl J Med. and survival in myelodysplastic syndrome treated with eryth-
2011;364(26):2496-2506. ropoietin and G-CSF: the GFM experience. Blood. 2008;
42. Boultwood J, Wainscoat JS. Clonality in the myelodysplastic 111(2):574-582.
syndromes. Int J Hematol. 2001;73(4):411-415. 63. Moyo V, Lefebvre P, Duh MS, Yektashenas B, Mundle S.
43. Backx B, Broeders L, Touw I, Lowenberg B. Blast colony- Erythropoiesis-stimulating agents in the treatment of anemia
forming cells in myelodysplastic syndrome: decreased poten- in myelodysplastic syndromes: a meta-analysis. Ann Hematol.
tial to generate erythroid precursors. Leukemia. 1993;7(1): 2008;87(7):527-536.
75-79. 64. Steensma DP. Hematopoietic growth factors in myelodysplas-
44. Raza A, Gezer S, Mundle S, et al. Apoptosis in bone marrow tic syndromes. Semin Oncol. 2011;38(5):635-647.
biopsy samples involving stromal and hematopoietic cells in 65. Hellstrom-Lindberg E, Gulbrandsen N, Lindberg G, et al.
50 patients with myelodysplastic syndromes. Blood. 1995; A validated decision model for treating the anaemia of mye-
86(1):268-276. lodysplastic syndromes with erythropoietin þ granulocyte
45. Raaijmakers MH, Mukherjee S, Guo S, et al. Bone progenitor colony-stimulating factor: significant effects on quality of life.
dysfunction induces myelodysplasia and secondary leukaemia. Br J Haematol. 2003;120(6):1037-1046.
Nature. 2010;464(7290):852-857. 66. Greenberg P, Taylor K, Larson RA, et al. Phase III randomized
46. Deeg HJ, de Lima M. Hematopoietic stem cell transplantation multicenter trial of recombinant human G-CSF in MDS [ASH
for older patients with myelodysplastic syndromes. J Natl abstract 196]. Blood. 1993;82(11 suppl 1):196a.
Compr Canc Netw. 2013;11(10):1227-1233. 67. Arshad M, Seiter K, Bilaniuk J, et al. Side effects related to can-
47. Sekeres MA, Gerds AT. Established and novel agents for mye- cer treatment: CASE 2. Splenic rupture following pegfilgrastim.
lodysplastic syndromes. Hematology Am Soc Hematol Educ J Clin Oncol. 2005;23(33):8533-8534.
Program. 2014;2014(1):82-89. 68. Brierley CK, Steensma DP. Thrombopoiesis-stimulating agents
48. Malcovati L, Hellstrom-Lindberg E, Bowen D, et al. Diagnosis and myelodysplastic syndromes. Br J Haematol. 2015;169(3):
and treatment of primary myelodysplastic syndromes in adults: 309-323.
recommendations from the European LeukemiaNet. Blood. 69. Sekeres MA, Giagounidis A, Kantarjian H, et al. Development
2013;122(17):2943-2964. and validation of a model to predict platelet response to romi-
49. Fenaux P, Ades L. How we treat lower-risk myelodysplastic plostim in patients with lower-risk myelodysplastic syndromes.
syndromes. Blood. 2013;121(21):4280-4286. Br J Haematol. 2014;167(3):337-345.
50. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al. Efficacy of 70. Giagounidis A, Mufti GJ, Fenaux P, et al. Results of a ran-
azacitidine compared with that of conventional care regi- domized, double-blind study of romiplostim versus placebo
mens in the treatment of higher-risk myelodysplastic syn- in patients with low/intermediate-1-risk myelodysplastic
dromes: a randomised, open-label, phase III study. Lancet syndrome and thrombocytopenia. Cancer. 2014;120(12):
Oncol. 2009;10(3):223-232. 1838-1846.
51. Sekeres MA, Cutler C. How we treat higher-risk myelodys- 71. Salacz ME, Lankiewicz MW, Weissman DE. Management of
plastic syndromes. Blood. 2014;123(6):829-836. thrombocytopenia in bone marrow failure: a review. J Palliat
52. Bejar R, Steensma DP. Recent developments in myelodysplas- Med. 2007;10(1):236-244.
tic syndromes. Blood. 2014;124(18):2793-2803. 72. Epling-Burnette PK, Painter JS, Rollison DE, et al. Prevalence
53. Cazzola M, Della Porta MG, Malcovati L. Clinical relevance of and clinical association of clonal T-cell expansions in Myelo-
anemia and transfusion iron overload in myelodysplastic syn- dysplastic Syndrome. Leukemia. 2007;21(4):659-667.
dromes. Hematology Am Soc Hematol Educ Program. 2008: 73. Sloand EM, Wu CO, Greenberg P, Young N, Barrett J. Factors
166-175. affecting response and survival in patients with myelodysplasia
54. Malcovati L. Impact of transfusion dependency and secondary treated with immunosuppressive therapy. J Clin Oncol. 2008;
iron overload on the survival of patients with myelodysplastic 26(15):2505-2511.
syndromes. Leuk Res. 2007;31(suppl 3):S2-S6. 74. Saunthararajah Y, Nakamura R, Wesley R, Wang QJ,
55. Steensma DP, Gattermann N. When is iron overload delete- Barrett AJ. A simple method to predict response to immuno-
rious, and when and how should iron chelation therapy be suppressive therapy in patients with myelodysplastic syn-
administered in myelodysplastic syndromes? Best Pract Res drome. Blood. 2003;102(8):3025-3027.
Clin Haematol. 2013;26(4):431-444. 75. Raza A, Meyer P, Dutt D, et al. Thalidomide produces trans-
56. Steensma DP. The role of iron chelation therapy for patients fusion independence in long-standing refractory anemias of
with myelodysplastic syndromes. J Natl Compr Canc Netw. patients with myelodysplastic syndromes. Blood. 2001;98(4):
2011;9(1):65-75. 958-965.
57. Armand P, Kim HT, Cutler CS, et al. Prognostic impact of 76. Moreno-Aspitia A, Geyer S, Li C, et al. N998B: multicenter
elevated pretransplantation serum ferritin in patients undergo- phase II trial of thalidomide (Thal) in adult patients with
ing myeloablative stem cell transplantation. Blood. 2007; myelodysplastic syndromes (MDS) [abstract]. Blood. 2002;
109(10):4586-4588. 100:96a.
58. Pascal L, Beyne-Rauzy O, Brechignac S, et al. Cardiac iron 77. Ito T, Ando H, Suzuki T, et al. Identification of a primary target of
overload assessed by T2* magnetic resonance imaging and thalidomide teratogenicity. Science. 2010;327(5971):1345-1350.
cardiac function in regularly transfused myelodysplastic syn- 78. Fink EC, Kronke J, Hurst SN, et al. Lenalidomide induces ubiq-
drome patients. Br J Haematol. 2013;162(3):413-415. uitination and degradation of CSNK1A1 in MDS with Del(5q)
59. List AF, Baer MR, Steensma DP, et al. Deferasirox reduces [ASH abstract 4]. Blood. 2014;124(21):4.
serum ferritin and labile plasma iron in RBC transfusion- 79. List A, Dewald G, Bennett J, et al. Lenalidomide in the myelo-
dependent patients with myelodysplastic syndrome. J Clin dysplastic syndrome with chromosome 5q deletion. N Engl J
Oncol. 2012;30(17):2134-2139. Med. 2006;355(14):1456-1465.
60. Cappellini MD, Porter J, El-Beshlawy A, et al. Tailoring iron 80. Tehranchi R, Woll PS, Anderson K, et al. Persistent malignant
chelation by iron intake and serum ferritin: the prospective stem cells in del(5q) myelodysplasia in remission. N Engl J Med.
EPIC study of deferasirox in 1744 patients with transfusion- 2010;363(11):1025-1037.
dependent anemias. Haematologica. 2010;95(4):557-566. 81. Raza A, Reeves JA, Feldman EJ, et al. Phase 2 study of lenali-
61. Kantarjian H, Giles F, List A, et al. The incidence and impact of domide in transfusion-dependent, low-risk, and intermediate-
thrombocytopenia in myelodysplastic syndromes. Cancer. 1 risk myelodysplastic syndromes with karyotypes other than
2007;109(9):1705-1714. deletion 5q. Blood. 2008;111(1):86-93.

n n
982 Mayo Clin Proc. July 2015;90(7):969-983 http://dx.doi.org/10.1016/j.mayocp.2015.04.001
www.mayoclinicproceedings.org
MYELODYSPLASTIC SYNDROMES: DIAGNOSIS AND TREATMENT

82. McDevitt MA. Clinical applications of epigenetic markers and 92. Sekeres MA, Othus M, List AF, et al. A randomized phase II
epigenetic profiling in myeloid malignancies. Semin Oncol. study of azacitidine combined with lenalidomide or with
2012;39(1):109-122. vorinostat vs. azacitidine monotherapy in higher-risk myelo-
83. Gore SD. In vitro basis for treatment with hypomethylating agents dysplastic syndromes (MDS) and chronic myelomonocytic
and histone deacetylase inhibitors: can epigenetic changes be used leukemia (CMML): North American Intergroup Study
to monitor treatment? Leuk Res. 2009;33(suppl 2):S2-S6. SWOG S1117 [abstract 5]. Blood. 2014;124(21):5.
84. Bejar R, Lord A, Stevenson K, et al. TET2 mutations predict 93. Prebet T, Gore SD, Esterni B, et al. Outcome of high-risk mye-
response to hypomethylating agents in myelodysplastic syn- lodysplastic syndrome after azacitidine treatment failure. J Clin
drome patients. Blood. 2014;124(17):2705-2712. Oncol. 2011;29(24):3322-3327.
85. Itzykson R, Kosmider O, Cluzeau T, et al. Impact of TET2 mu- 94. Jabbour E, Garcia-Manero G, Batty N, et al. Outcome of pa-
tations on response rate to azacitidine in myelodysplastic syn- tients with myelodysplastic syndrome after failure of decita-
dromes and low blast count acute myeloid leukemias. bine therapy. Cancer. 2010;116(6):3830-3834.
Leukemia. 2011;25(7):1147-1152. 95. Litzow MR, Tarima S, Perez WS, et al. Allogeneic transplanta-
86. Gore SD, Fenaux P, Santini V, et al. A multivariate analysis of the tion for therapy-related myelodysplastic syndrome and acute
relationship between response and survival among patients myeloid leukemia. Blood. 2010;115(9):1850-1857.
with higher-risk myelodysplastic syndromes treated within aza- 96. Della Porta MG, Alessandrino EP, Bacigalupo A, et al. Predic-
citidine or conventional care regimens in the randomized tive factors for the outcome of allogeneic transplantation in
AZA-001 trial. Haematologica. 2013;98(7):1067-1072. patients with MDS stratified according to the revised IPSS-R.
87. Kantarjian H, Issa JP, Rosenfeld CS, et al. Decitabine improves Blood. 2014;123(15):2333-2342.
patient outcomes in myelodysplastic syndromes: results of a 97. Cutler CS, Lee SJ, Greenberg P, et al. A decision analysis of
phase III randomized study. Cancer. 2006;106(8):1794-1803. allogeneic bone marrow transplantation for the myelodysplas-
88. Lubbert M, Suciu S, Baila L, et al. Low-dose decitabine versus tic syndromes: delayed transplantation for low-risk myelodys-
best supportive care in elderly patients with intermediate- or plasia is associated with improved outcome. Blood. 2004;
high-risk myelodysplastic syndrome (MDS) ineligible for inten- 104(2):579-585.
sive chemotherapy: final results of the randomized phase III 98. Koreth J, Pidala J, Perez WS, et al. Role of reduced-intensity con-
study of the European Organisation for Research and Treat- ditioning allogeneic hematopoietic stem-cell transplantation in
ment of Cancer Leukemia Group and the German MDS older patients with de novo myelodysplastic syndromes: an inter-
Study Group. J Clin Oncol. 2011;29(15):1987-1996. national collaborative decision analysis. J Clin Oncol. 2013;31(21):
89. Steensma DP, Baer MR, Slack JL, et al. Multicenter study of 2662-2670.
decitabine administered daily for 5 days every 4 weeks to 99. Gerds AT, Gooley TA, Estey EH, Appelbaum FR, Deeg HJ,
adults with myelodysplastic syndromes: the alternative dosing Scott BL. Pretransplantation therapy with azacitidine vs induction
for outpatient treatment (ADOPT) trial. J Clin Oncol. 2009; chemotherapy and posttransplantation outcome in patients
27(23):3842-3848. with MDS. Biol Blood Marrow Transplant. 2012;18(8):1211-1218.
90. Voso MT, Breccia M, Lunghi M, et al. Rapid loss of response 100. Platzbecker U, Wermke M, Radke J, et al. Azacitidine for treat-
after withdrawal of treatment with azacitidine: a case series ment of imminent relapse in MDS or AML patients after allo-
in patients with higher-risk myelodysplastic syndromes or geneic HSCT: results of the RELAZA trial. Leukemia. 2012;
chronic myelomonocytic leukemia. Eur J Haematol. 2013; 26(3):381-389.
90(4):345-348. 101. Scheinberg P, Steensma DP. Acquired marrow failure syn-
91. Prebet T, Sun Z, Figueroa ME, et al. Prolonged administration dromes: aplastic anemia, paroxysmal nocturnal hemoglobin-
of azacitidine with or without entinostat for myelodysplastic uria, and myelodysplastic syndromes. In: Steensma DP,
syndrome and acute myeloid leukemia with myelodysplasia- McCrae KM, eds. American Society of Hematology Self-
related changes: results of the US Leukemia Intergroup Trial Assessment Program (ASH-SAP). 5th ed. Washington, DC:
E1905. J Clin Oncol. 2014;32(12):1242-1248. American Society of Hematology; 2013:369-400.

Mayo Clin Proc. n July 2015;90(7):969-983 n http://dx.doi.org/10.1016/j.mayocp.2015.04.001 983


www.mayoclinicproceedings.org

You might also like