Vous êtes sur la page 1sur 11

A JH

CME Information: Annual Clinical Updates 2015:


Myelodysplastic Syndromes
CME Editor: Ayalew Tefferi, M.D. and Author: Guillermo Garcia-Manero, M.D.

If you wish to receive credit for this activity, please refer to the website: www.wileyhealthlearning.com

Accreditation and Designation Statement


Blackwell Futura Media Services is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical
education for physicians.
Blackwell Futura Media Services designates this journal-based CME for a maximum of 1 AMA PRA Category 1 CreditTM. Physicians should
only claim credit commensurate with the extent of their participation in the activity.

Educational Objectives
Upon completion of this educational activity, participants will be better able to:

1. Understand the latest updates on molecular and cytogenetic alterations in Myelodysplastic Syndromes.
2. Understand the latest updates on the classification and treatment of patients with Myelodysplastic Syndromes.

Activity Disclosures
No commercial support has been accepted related to the development or publication of this activity.
CME Editor: Ayalew Tefferi, M.D. has no relevant financial relationships to disclose.
Author: Guillermo Garcia-Manero, M.D. has no relevant financial relationships to disclose.
This activity underwent peer review in line with the standards of editorial integrity and publication ethics maintained by American Journal of
Hematology. The peer reviewers have no conflicts of interest to disclose. The peer review process for American Journal of Hematology is single
blinded. As such, the identities of the reviewers are not disclosed in line with the standard accepted practices of medical journal peer review.
Conflicts of interest have been identified and resolved in accordance with Blackwell Futura Media Servicess Policy on Activity Disclosure and
Conflict of Interest. The primary resolution method used was peer review and review by a non-conflicted expert.

Instructions on Receiving Credit


This activity is intended for physicians. For information on applicability and acceptance of continuing medical education credit for this activity,
please consult your professional licensing board.
This activity is designed to be completed within one hour; physicians should claim only those credits that reflect the time actually spent in the
activity. To successfully earn credit, participants must complete the activity during the valid credit period, which is up to two years from initial
publication. Additionally, up to 3 attempts and a score of 70% or better is needed to pass the post test.
Follow these steps to earn credit:

 Log on to www.wileyhealthlearning.com
 Read the target audience, educational objectives, and activity disclosures.
 Read the activity contents in print or online format.
 Reflect on the activity contents.
 Access the CME Exam, and choose the best answer to each question.
 Complete the required evaluation component of the activity.
 Claim your Certificate.

This activity will be available for CME credit for twelve months following its launch date. At that time, it will be reviewed and potentially
updated and extended for an additional twelve months.

C 2015 Wiley Periodicals, Inc.


V

American Journal of Hematology, Vol. 90, No. 9, September 2015 831


ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES
A JH
Myelodysplastic syndromes: 2015 Update on diagnosis,
risk-stratification and management
Guillermo Garcia-Manero*

Disease overview: The myelodysplastic syndromes (MDS) are a very heterogeneous group of myeloid
disorders characterized by peripheral blood cytopenias and increased risk of transformation to acute
myelogenous leukemia (AML). MDS occurs more frequently in older males and in individuals with prior
exposure to cytotoxic therapy.
Diagnosis: Diagnosis of MDS is based on morphological evidence of dysplasia upon visual examination of a
bone marrow aspirate and biopsy. Information obtained from additional studies such as karyotype, flow
cytometry, or molecular genetics is complementary but not diagnostic.
Risk-stratification: Prognosis of patients with MDS can be calculated using a number of scoring systems. In
general, all these scoring systems include analysis of peripheral cytopenias, percentage of blasts in the
bone marrow, and cytogenetic characteristics. The most commonly used system still is probably the
International Prognostic Scoring System (IPSS). IPSS is being replaced by the new revised score IPSS-R.
Risk-adapted therapy: Therapy is selected based on risk, transfusion needs, percent of bone marrow blasts,
and more recently cytogenetic and mutational profiles. Goals of therapy are different in lower risk patients
than in higher risk. In lower risk, the goal is to decrease transfusion needs and transformation to higher risk
disease or AML, as well as to improve survival. In higher risk, the goal is to prolong survival. Current
available therapies include growth factor support, lenalidomide, hypomethylating agents, intensive
chemotherapy, and allogeneic stem cell transplantation. The use of lenalidomide has significant clinical
activity in patients with lower risk disease, anemia, and a chromosome 5 alteration. 5-Azacitidine and
decitabine have activity in higher risk MDS. 5-Azacitidine has been shown to improve survival in higher risk
MDS. A number of new molecular lesions have been described in MDS that may serve as new therapeutic
targets or aid in the selection of currently available agents. Additional supportive care measures may
include the use of prophylactic antibiotics and iron chelation.
Management of progressive or refractory disease: At the present time there are no approved interventions
for patients with progressive or refractory disease particularly after hypomethylating based therapy. Options
include participation in a clinical trial or cytarabine based therapy and stem cell transplantation.
Am. J. Hematol. 90:832841, 2015. V C 2015 Wiley Periodicals, Inc.

Disease Overview
MDS comprises a very heterogeneous group of myeloid malignancies with very distinct natural histories [13]. MDS occurs in three to four
individuals per 105 in the US population [4]. Prevalence increases with age. For instance, in individuals age 60 and above, prevalence is 735 per
105 [4]. Other series have reported higher rates [5]. MDS affects more frequently males than females [4]. Exposure to prior chemo or radiation
therapy is a risk for the development of MDS.
MDS is usually suspected by the presence of cytopenia on a routine analysis of peripheral blood. This prompts evaluation of bone marrow cell mor-
phology (aspirate) and cellularity (biopsy). A manual count of bone marrow blasts is fundamental for risk assessment. Cytogenetic analysis helps in pre-
dicting risk and in selecting therapy. Once this information is collected, the risk of the patient can be calculated. At the present time, the International
Prognostic Scoring System (IPSS) [6] is still commonly used. Natural history and therapeutic decisions are different for patients with lower risk disease
(low and INT-1) versus those with higher (INT-2 and high). In lower risk, disease interventions have been traditionally developed to improve transfu-
sion needs; whereas higher risk options were initially modeled following therapy of AML with remission induction being the goal. This concept was
modified by the better understanding of the natural history of MDS and the development of new therapies, in particular the hypomethylating agents.
Another important concept is that a large majority of patients with MDS die from causes intrinsic to the disease and not because of progression to
AML [7]. This has important implications for the development of therapies in MDS. The revised IPSS score (IPSS-R) was published in 2012 [8]. This

Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas


Conflict of interest: Nothing to report
*Correspondence to: Guillermo Garcia-Manero MD, Department of Leukemia MD Anderson Cancer Center Box 428 1515 Holcombe Blvd Houston, TX
77030. E-mail: ggarciam@mdanderson.org
Contract grant sponsor: Cancer Prevention & Research Institute of Texas; Contract grant number: RP100202.
Contract grant sponsors: Dr. Kenneth B. McCredie Chair in Clinical Leukemia Research endowment, Edward P. Evans Foundation, and the Fundacion Ramon
Areces.
Received for publication: 22 June 2015; Accepted: 24 June 2015
Am. J. Hematol. 90:832841, 2015.
Published online: in Wiley Online Library (wileyonlinelibrary.com).
DOI: 10.1002/ajh.24102

C 2015 Wiley Periodicals, Inc.


V

832 American Journal of Hematology, Vol. 90, No. 9, September 2015 doi:10.1002/ajh.24102
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES Myelodysplastic Syndromes: 2015 Update

Figure 1. Cytogenetic classification of MDS. Adapted from Schanz et al. [9]. [Color figure can be viewed in the online issue, which is available at wileyonline-
library.com.]

score includes a new cytogenetic risk classification of that divides nomenon is associated with increased risk of transformation to AML
patients into five cytogenetic categories (Fig. 1) [810]. The IPSS-R and worse survival [13]. This data has implications for the follow up of
divides patients in five risk subgroups including an intermediate group. patients at risk for clonal cytogenetic acquisition.
Because current therapies were approved using either FAB or IPSS crite- A number of other assays can be used to help in the diagnosis of
ria, I still use IPSS to decide therapy but IPSS-R to calculate prognosis. MDS. These include the use of flow cytometry and fluorescent in situ
hybridization (FISH). Flow cytometry can help in the identification of
abnormal phenotypic patterns and can be of help in cases of minimal
Diagnosis dysplasia. Because the significant heterogeneity of cytogenetic altera-
The diagnosis of MDS is generally suspected based on the presence tions in MDS, there is no evidence that a panel of FISH probes could
of an abnormal CBC. Diagnosis is confirmed by performing a bone replace routine 20 metaphase cytogenetic analysis. Thus, in my opinion
marrow aspiration and biopsy. Both procedures provide different FISH and flow cytometry should not be considered part of the standard
information. The bone marrow aspirate allows for detailed evaluation work up evaluation procedure of the patient with MDS and should be
of cellular morphology and evaluation of percent of blasts. The bone used in specific situations. More recently, and thanks to the discovery of
marrow biopsy allows for determination of bone marrow cellularity multiple genetic mutations in MDS, we now have access to clinical tools
and architecture. Diagnosis is established by the presence of dysplasia. for molecular annotation of patients with MDS.
A number of morphological classifications are in place to classify
patients with MDS. The most recent one being the 2008 WHO ver- Diagnostic problems in MDS
sion [11]. There is some controversy regarding the utility of bone Because diagnosis of MDS is based on morphological assessment it
marrow biopsy. We believe that biopsy helps in diagnosis and poten- can be subjective particularly in patients with early low risk disease. It is
tially in selecting therapy. For instance, in my practice we only use calculated that diagnostic discrepancy can occur at the time of initial
immunotherapy (ATG, cyclosporine, and steroids) in hypocellular presentation in close to 20% of patients [14]. This has obvious implica-
MDS (marrow cellularity less than 20%30% depending on age). Cel- tions for therapeutic decision making and patient counseling. In gen-
lularity is better assessed by bone marrow biopsy. eral, diagnosis is obvious in patients with excess blasts. The problem is
A number of additional tests are needed to complete the laboratory in patients without excess blasts where diagnosis is based on dysplasia.
evaluation of a patient with MDS, most important of which is the analy- Clinical assessment is needed in patients with minimal or not diagnostic
sis of bone marrow cytogenetics. It is well established that cytogenetic evidence of dysplasia. In these cases, it is recommended that other
patterns are very heterogeneous in MDS [12]. Cytogenetics are of causes of cytopenia be excluded. Routine tests include the analysis of
importance to calculate prognosis of patients and in some subsets of anemia and thrombocytopenia, and exclusion of cause of blood loss or
patients to select the most effective form therapy. The most recent cyto- inflammatory processes. When suspected, evaluation of GI tract needs
genetic risk classification in MDS includes five different subgroups to be considered. Once other potential causes of cytopenia are excluded,
including 20 different alterations (Fig. 1) [9]. Recent data has indicated patients with cytopenia but no dysplasia are considered in the subset of
that cytogenetic patterns are not stable in MDS and that a significant idiopathic cytopenia of unknown significance (ICUS). A fraction of
fraction of patients acquired additional cytogenetic changes. This phe- these patients may have cytogenetic abnormalities. The natural history

doi:10.1002/ajh.24102 American Journal of Hematology, Vol. 90, No. 9, September 2015 833
Garcia-Manero ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

TABLE I. The Global MDACC MDS Prognostic Model [19]


TABLE II. MDACC MDS Lower Risk Prognostic Model [20].
Prognostic factor Points
Characteristics Points
PS  2 2
Age Unfavorable cytogenetics 1
6064 1 Age  60 years 2
>64 2 Hemoglobin < 10 (g/dL) 1
Platelets x109/L Platelets
<30 3 < 50 x 109/L 2
30-49 2 50200 x 109/L 1
50199 1 Bone Marrow Blasts  4% 1
Hemoglobin <12 g/dL 2 Score Median survival 4-year OS (%)
BM blast % 0 NR 78
5 to 10 1 1 83 82
11 to 19 2 2 51 51
WBC > 20 x109/L 2 3 36 40
Alteration of chromosome 7 or  3 alterations 3 4 22 27
Prior transfusion 1 5 14 9
6 16 7
Abbreviations: PS, performance status; BM, bone marrow; WBC, white 7 9 N/A
blood cell count.
Patients with 0 to 4 points had a median survival of 54 months and a 3- Characteristics were selected from multivariate analysis model in patients
year 63% survival. Patients with 5 and 6 points had a median survival of with lower risk MDS. Each characteristic is associated with a number of
23 to 30 months and 3-year survival of 30% to 40%. Patients with 7 to 8 points. Score is calculated by adding all points. Each score allows calcula-
points had a median survival of 13 months and a 3-year survival rate of tion of median survival (in months) and probability of survival at 4 years.
13% to 19%. Patients with 9 or more points had a median survival of 5 to Adapted from Ref. 20.
10 months and a 2% 3-year survival. Adapted from Ref. 19.

of this group of patients with ICUS is not well known and observation evaluation of all patients that are considered as MDS at any time dur-
is recommended at the present time. Recently, the presence of clonal ing their course of their disease without needed WHO evaluation.
somatic mutations have been reported in hematopoietic cells from It has also become apparent that the natural history of patients
older individuals without evidence of hematological disorder [15,16]. with lower risk disease is very heterogeneous [20]. We evaluated
Patients with evidence of clonal hematopoiesis are at an increased risk outcomes in a large series of patients with low or int-1 disease by
to develop MDS [16]. The clinical implications of these findings are IPSS. We found that prognosis varied significantly in patients with
not understood at this point [17]. lower risk MDS and were able to develop a lower risk MDS specific
Finally, a subset of patients of importance are those with MDS/ prognostic score (Table II). This has significant implications for the
MPN features. These are patients with evidence of a myeloprolifera- development of specific interventions for patients with lower risk
tive component (with or without fibrosis). At the present time, we do disease. This model has been validated on several occasions and it
not fully understand the natural history of patients with MDS/MPN is being used to identify patients with poor prognosis lower risk
[18]. In our center they are currently treated as MDS, but studies are disease that could be candidates for early intervention. Bejar et al.
ongoing to clarify this issue. [21] published data indicating that patients with poorer prognosis
and lower risk disease accumulate a higher number of mutational
Risk Stratification events than the better risk counterpart. This data provides a poten-
tial molecular basis for the identification of this group of patients
The prognosis of patients with MDS is very heterogeneous and and poorer prognosis.
thus the need to develop prognostic systems that allow risk stratifica- MDS occurs in older patients that suffer from comorbidities more
tion and help in the timing and choice of therapy. Apart from the frequently. None of the systems discussed above include impact of
intrinsic prognostic value of morphological classifications [11], a comorbidity to the calculation of the natural history of MDS patients.
number of prognostic scores are currently in use in MDS. The IPSS To study this issue, we used a comprehensive comorbidity score
[6] has been in place since 1997. The prognostic score includes per- known as ACE-27in a cohort of 500 patients with MDS [22]. Pres-
cent of blasts, number of cytopenias and cytogenetics. IPSS is of fun- ence of comorbidity had a significant independent impact on survival
damental importance as it has allowed the prognostication of patients and a prognostic score could be developed that included age, IPSS,
for over two decades. This system is highly reproducible and very and ACE-27 score. This data indicates the need to add comorbidity
simple to use. The system has several limitations that have become scores in MDS. Recently, other groups have confirmed the impor-
evident over the years. The most important one is that it is not a tance of comorbidity scores in MDS [23]. We recently confirmed that
very precise predictor of prognosis in patients with lower risk disease ACE-27 adds to the prognostic value of IPSS-R [24].
and that it attributes relatively little weight to cytogenetics. The IPSS- Additional prognostic scoring systems include systems for hypocel-
R includes different cut off points of cytopenias and incorporates the lular MDS [25] and for therapy related MDS [26]. It should be noted
new cytogenetic MDS score [8]. IPSS-R should be the standard tool that prognosis in t-MDS is strongly associated with presence of cyto-
to assess risk although limitations still exist for its use as no drug genetic alterations: diploid patients have prognosis not dissimilar to
therapy has been approved yet using IPSS-R. A new molecular IPSS that of de novo patients [26].
system is expected soon.
Both the IPSS and IPSS-R were developed in a very specific subset
of patients: newly diagnosed patients at the time of initial presentation. Cytogenetic and Molecular Alterations
Patients with proliferative features and CMML or that have received Over the last 36 months, a number of very important studies have
prior therapy were excluded [6]. To overcome these limitations, the been published describing comprehensive analysis of the incidence
global MDACC model was developed [19]. This model is summarized and clinical impact of multiple genetic lesions in MDS [27]. Bejar
in Table I. The importance of the global MDACC model is that allows et al. [28] first published an analysis of 18 genes using different

834 American Journal of Hematology, Vol. 90, No. 9, September 2015 doi:10.1002/ajh.24102
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES Myelodysplastic Syndromes: 2015 Update

TABLE III. Reported Frequency of Genetic Lesions in MDS [28,3032]

Gene % Location Function

SF3B1 28 2q33 Splicing factor


TET2 21 4q24 Control of cytosine hydroxymethylation
ASXL1 14 20q11 Epigenetic regulator
SRSF2 12 17q25 Splicing factor
RUNX1 9 21q22 Transcription factor
TP53 8 17p13 Transcription factor
U2AF1 7 21q22 Splicing factor
EZH2 6 7q36 Polycomb group protein
NRAS 4 1p13 Signal transduction
JAK2 3 9p24 Tyrosine kinase
ETV6 3 12p13 Transcription factor
CBL 2 11q23 Signal transduction
IDH2 2 15q26 Cell metabolism, epigenetic regulation
NPM1 2 5q35 Phosphoprotein
IDH1 1 2q33 As IDH1
Figure 2. Distribution of mutational events in MDS. Adapted from Papaem- KRAS <1 12p12 Signal transduction
manuil et al. [29]. [Color figure can be viewed in the online issue, which is GNAS <1 20q13 G protein
available at wileyonlinelibrary.com.] PTPN11 <1 12q24 Protein phosphatase
BRAF <1 7q34 Raf kinase
PTEN <1 10q23 Phosphatase
techniques on 439 patients. More recently, two major studies have CDKN2A <1 9q121 Cell cycle control
described the mutational landscape of MDS in larger series analyzing
more genes [29,33] (Fig. 2). For instance, a European consortium has
reported an analysis of mutations in 111 genes using next generation
sequencing technology in a cohort of 738 patients [29]. Frequency of
common mutations is shown in Table III. This emerging data will
have a significant impact not only on our ability to prognosticate
patients with MDS but also in potentially selecting therapy. For
instance, it has been reported that patients with TET2 mutations may
have higher response rates to azacitidine than those without mutation
[34,35]. In addition, the results of molecular mutations is aiding in
the development of clinical trials for instance for patients with IDH1,
IDH2, Flt-3, and RAS. Recently, very poor outcomes with allogeneic
stem cell transplantation have been associated with presence of p53
and DNMT3A mutations [36].
Figure 3. Proposed treatment algorithm of patients with MDS. Once diag-
Risk Adapted Therapy nosis is confirmed, patients are divided into a lower and a higher risk cat-
egory. Options for patients with lower risk disease include growth factors,
At the present time, we still use IPSS to decide the choice of ther- lenalidomide, and azanucleosides. Treatment in general is sequential:
apy for an individual patient. Below is a summary of options and rec- patients that do not respond to growth factors can be treated with lenali-
ommendations for specific subsets of patients [2]. A treatment domide or azanuclesoides, if appropriate. Patients that fail lenalidomide
can subsequently be treated with azanucleosides. There is little experi-
algorithm is shown in Fig. 3. ence in terms of outcomes with this approach. Patients that fail all three
therapies should be considered for alloSCT and/or clinical trial. For
Options for newly diagnosed patients with lower risk patients with higher risk MDS options are alloSCT, AML-like therapy, or aza-
MDS nucleoside. Prognosis of patients that fail any of these approaches is
poor, particularly for those exposed to azanucleosides. In this setting
Therapy in this subset of patients is based on the transfusion needs alloSCT and clinical trial should be strongly considered. [Color figure can
of the patients. Patients that are transfusion independent are usually be viewed in the online issue, which is available at wileyonlinelibrary.com.]
observed until they become transfusion dependent. Below is a
description of agents currently available for patients with lower risk
MDS. A new important concept in lower risk MDS is the idea of group also evaluated the impact of ESA on survival in a retrospective
early intervention in patients with poor prognosis lower risk MDS. study of 284 patients and compared it with a group of patients that
We are currently testing this concept in prospective clinical trials formed the IPSS cohort [41]. In this study patients treated with ESA had
using very low doses of hypomethylating agents. a better survival (HR for death was 0.43, 95% CI 0.25-0.72] [41]. Ques-
Erythroid growth factor support. The use of erythroid stimulating tions on potential tumorigenic effect of these drugs haves resulted in
agents (ESA) is common practice in community practice [37]. It should increased scrutiny of their use. G-CSF is not approved by the FDA for
be noted that no randomized study has ever proven than this interven- patients with anemia of MDS in the US.
tion positively affects the natural history of patients with MDS. A num- Recent data on the results with romiplostin [42] questions its use
ber of ESAs are available. Reported response rates range from 30% to in patients with lower risk MDS, particularly because of complications
60% depending on study [38]. Data from the Swedish group indicated related to disease transformation and marrow fibrosis. A number of
that addition of G-CSF to erythropoietin increases responses rates. In a studies are now evaluating eltrombopag in MDS, another TPO ago-
retrospective observational study, early introduction of this combina- nist, but results are not known at this point.
tion in patients with low risk disease and minimally transfusion depend-
ent patients had an impact on survival [39]. The Swedish group also Recommendation. I believe that a course of ESA with or without G-
developed an algorithm to predict response to ESA [40]. The French CSF is not contraindicated in most patients with low risk MDS with

doi:10.1002/ajh.24102 American Journal of Hematology, Vol. 90, No. 9, September 2015 835
Garcia-Manero ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

significant anemia without other cytopenia. Data indicates that early III trial in patients with lower risk MDS and significant cytopenias
incorporation of these agents is more effective. I maintain therapy for (NCT01566695).
at least 3 months to judge efficacy. In responding patients, I continue There is relatively little data with decitabine in patients with lower
therapy until transfusion effect is lost. risk disease. In the initial randomized trial of decitabine [49], 31
Lenalidomide. Lenalidomide is approved for patients with lower patients with INT-1 disease were treated. Four of the 28 patients
risk MDS, anemia, and alteration of chromosome 5 [43]. Phase I results achieved some type of response. A randomized phase II trial was con-
[44] were confirmed in a subsequent phase II study of lenalidomide in ducted exploring a 3-day subcutaneous schedule versus a weekly 3 3
patients with anemia and alteration of chromosome 5 [44]. In that study monthly schedule. This study did not find significant differences
148 patients received 10 mg of lenalidomide for 21 days every 4 weeks between both arms, although it declared the daily 3 3 schedule as an
or daily. Of those, 112 had decrease need for transfusions (76%; 95% CI initial winner. Low dose decitabine used in this fashion was associ-
6882) and 99 patients (67%; 95% CI, 5974) became transfusion inde- ated with close to 60% trilineage transfusion independence with mini-
pendent. Response was fast: median time 4.6 weeks. The median rise of mal hematopoietic toxicity [52].
hemoglobin was 5.4 g/dL. Of interest, cytogenetic responses were
observed in close to 50% informative patients. Predictors of response Recommendation. Both 5-azacitidine and decitabine are used in the
included presence of a platelet count of 100 3 109/L and less than four US in patients with lower risk disease that are transfusion dependent.
prior units of red cells transfused. It should be noted that in this study Most patients treated with these agents have failed or were not candi-
patients with a platelet count of less than 50 3 109/L were excluded. dates for growth factor support or lenalidomide. Further studies of
Subsequently, these results were confirmed in a phase III known as these agents in lower risk MDS are needed. An oral formulation of 5-
AZA-004 comparing two different doses of lenalidomide (5 and 10 mg azacitidine is being studied for patients with lower risk MDS [[51]].
orally daily) versus placebo. This study was also designed to clarify the Lower dose of azacitidine or decitabine can be considered for these
issue of transformation to AML. Ten micrograms daily was the superior patients.
arm and that there was no increased incidence of transformation to Immune therapy. This is an area of controversy. It is accepted that
AML in patients treated with lenalidomide [45]. Of importance, a recent a subset of patients with MDS are characterized by deregulation of both
report from the initial MDS-003 trial of lenalidomide has indicated a cellular and innate immunity [5355]. Based on this, it will be logical
longer survival for patients responding to therapy [46]. This is addi- that the use of immune-modulatory agents could have therapeutic bene-
tional evidence that lenalidomide can change the natural history of fit in MDS. The NIH group pioneered these approaches. Agents studied
patients with 5q- MDS. include antithymocyte globulin (ATG), cyclosporine, and steroids.
In parallel, lenalidomide has been investigated in patients without These therapies have been modeled after therapy of aplastic anemia
chromosome 5 alterations [47]. In an initial study, 214 patients [53]. The NIH group also developed an algorithm to predict response to
received lenalidomide 10 mg orally daily or 10 mg on days 1 to 21 of these classes of agents [[56]]. This model included younger age, HLA-
a 28-day cycle. Fifty six (26%) patients achieved transfusion inde- DR15 positivity, and shorter duration of transfusion dependency. Using
pendence after a median of 4.8 weeks of therapy. Median response this algorithm, the NIH group reported that alemtuzumab, an antibody
duration was 41 weeks. Preliminary results of a phase III trial in this against CD52, has significant activity in patients with MDS predicted to
patient population were presented at ASH 2014 (Santini et al., unpub- respond to immune suppressive therapy [57]. The group at Moffitt Can-
lished) and supported the findings of the initial phase II trial [47]. cer Center has suggested that a CD4/CD8 ratio could also be used to
predict response [58]. Our group has not been capable to reproduce
Recommendation. The degree of response with lenalidomide in some of the data discussed above. Response rates with ATG observed at
patients with lower risk MDS, anemia, good platelets, and del5q makes MDACC are significantly lower than those of the NIH [59]. The most
it the standard of care for this subset of patients. This is further rein- important predictor for response for us has been the presence of mar-
forced by the recent data on survival in responding patients [46]. I do row hypocellularity [60]. This is consistent with the results of Mufti and
not consider this agent in patients with thrombocytopenia. No recom- coworkers in London [61].
mendation can be made for patients without an alteration of chromo- Also recently the impact of ATG based therapy on survival has
some 5 at the present time. It is likely that a subset of patients with been questioned. Data from a Swiss study comparing ATG versus
anemia, low risk disease, and diploid cytogenetics could also benefit supportive care indicated a higher response rate, but no survival ben-
from this compound. This will be clarified with the full report the study efit [62]. In this study, patients were randomized to a combination of
by Santini et al (ASH 2014, unpublished). horse ATG with cyclosporine versus best supportive care (BSC).
Azanucleosides. Two azanuclesoides are approved for MDS: 5- Forty-five patients received ATG1CSA and 43 patients received BSC.
azacitidine [48] and 5-aza-20 -deoxycitidine (decitabine) [49]. 5-Azacitidine By month 6, 13 of the 45 patients on ATG1CSA had a hematologic
is approved for all subsets of MDS, whereas decitabine for those with INT- response compared with four of the 43 patients on BSC (P 5 0.0156).
1 disease and above. There is little data in the use of these compounds in Despite higher response rates no significant effect on survival or
lower risk MDS. None of them have been shown to modify the natural transformation was observed.
history of patients with lower risk disease. Recently, the NIH group has also reported on the clinical activity
Different schedules of 5-azacitidine have been explored in MDS. In of eltrombopag in patients with aplastic anemia as salvage therapy
a community study a 5-day schedule was compared with a 7 day 5-2- [63]. This data was of importance because of the fact that responses
2 schedule (weekend off) or a 5-2-5 schedule of 10 days [50]. Fifty were multilineage and not just restricted to platelets. A number of
patients were assigned to each arm (except 5-2-2 was 51 patients). studies are investigating this agent in MDS.
Most patients had lower risk disease. Hematologic improvement was
achieved by 44% to 56% of patients in each arm. Transfusion inde- Recommendation. This is a particular difficult group of patients. A
pendency was documented in 50% to 64% of patients. There was a majority of older patients cannot tolerate ATG and most patients are
trend for better response rates and less toxicity with the 5-day sched- treated with some form of supportive care that could include cyclospo-
ule of 5-azacitidine. Therefore it is reasonable to use a shorter (5-day rine, growth factors, and steroids. The impact of it is not known either.
schedule) of 5-azacitidine in lower risk MDS. The results of a phase 1 In younger patients with severe hypoplastic MDS, allogeneic stem cell
trial of an oral derivative of azacitidine have been published [51]. transplantation (alloSCT) should be considered as soon as possible. For
Oral azacitidine is currently being studied in an international phase those that are not candidates, a combination with horse ATG is

836 American Journal of Hematology, Vol. 90, No. 9, September 2015 doi:10.1002/ajh.24102
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES Myelodysplastic Syndromes: 2015 Update

Figure 4. Impact on survival of stem cell transplantation in MDS. Adapted from Koreth et al. [65]. A: Monte Carlo analysis for low/intermediate-1 international
prognostic scoring system (IPSS) myelodysplastic syndromes (MDS). Simulated Kaplan-Meier survival plots (n 5 10,000; with log-rank P value) are indicated
for the modeled 10-year time period, comparing a strategy of early reduced-intensity conditioning (RIC) transplantation (blue line) versus no early RIC trans-
plantation (gold line). The results graphically indicate survival benefit of the nontransplantation strategy in low/intermediate-1 IPSS MDS quality-adjusted life
expectancy (QALE): two-way sensitivity analysis. B: Two-way sensitivity analysis plot for the utilities of the Markov states alive after RIC transplantation and
alive with MDS without RIC transplantation is shown. The gold area indicates the range in which nontransplantation therapy produces superior QALE. The
blue area indicates the range in which RIC transplantation produces superior QALE. The red square indicates the plausible range of quality of life (QoL) for
alive with low/intermediate-1 IPSS MDS and for alive after RIC transplantation and does not cross the threshold line. This result is interpreted as insensi-
tive, that is, the conclusion regarding benefit does not change within the plausible QoL range. C: Monte Carlo analysis for intermediate-2/high IPSS MDS.
Simulated Kaplan-Meier survival plots (n 5 10,000; with log-rank P value) are indicated for the modeled 10-year time period, comparing a strategy of early
RIC transplantation (blue line) versus no early RIC transplantation (gold line). The results graphically indicate survival benefit of the early RIC transplantation
strategy in intermediate-2/high IPSS MDS. D: Intermediate-2/high IPSS MDS QALE: two-way sensitivity analysis. Two-way sensitivity plot for the utilities of the
Markov states alive after RIC transplantation and alive with MDS without early RIC transplantation is shown. The gold area indicates the range in which
nontransplantation therapy produces superior QALE. The blue area indicates the range in which RIC transplantation produces superior QALE. The red square
indicates the plausible range of QoL for alive with intermediate-2/high IPSS MDS and for alive after RIC transplantation and does not cross the threshold
line. This result is interpreted as insensitive, that is, the conclusion regarding benefit does not change within the plausible QoL range. HCT, hematopoietic
cell transplantation. [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

recommended. I cannot recommend the use of alemtuzumab at the formal recommendation for any of these interventions. In my experi-
present time until more data from other clinical trials is reported. The ence patients with isolated neutropenia and MDS not receiving ther-
data on eltrombopag is of interest. apy are not at significantly increased risk of infection to support
Allogeneic stem cell transplantation. AlloSCT is usually not recom- recommendation of prophylactic antibiotics. Prophylactic antibiotics
mended in patients with lower risk disease even if they are young. This are commonly used in the context of active therapy for these
is based on data from Cutler et al. using a Markov model [64]. The patients.
anticipated early mortality with alloSCT cannot be overcome by the The role of iron chelation in MDS is more complicated. Data from
potential beneficial survival effect of alloSCT. This concept was con- thallasemias indicates that iron chelation has an important role in
firmed by Koreth et al. at recently (Fig. 4) [65]. Using another Markov this setting. Iron accumulation is frequent in MDS. The consequences
model, the investigators analyzed the impact of reduced intensity trans- of this are not fully understood in MDS. I do not see in my practice
plant in older patients with MDS. Patients with lower risk disease did patients with MDS and iron deposition that develop liver cirrhosis or
not benefit from this less toxic transplant approach [65]. cardiomyopathy as frequently as reported by other groups [66]. Iron
accumulation could have a role in transformation to AML [67] and
Recommendation. I generally do not recommend alloSCT in patients in the increased risk of infectious complications known to occur on
with lower risk disease at initial presentation. That said because of these patients. The NCCN guidelines recommend the use of chelation
time required for donor identification, I refer all potential candidate therapy in patients with ferritin levels above 2500 ng/mL [68]. A large
patients for a transplant consult in anticipation of future needs. phase III (NCT00940602) study is evaluating the role of iron chela-
Patients that are candidates for alloSCT and that had been exposed to tion in MDS.
multiple therapies (growth factors, lenalidomide, azanuclesoides, etc.)
should be considered for transplantation. These patients are also candi- Recommendations. I do not routinely recommend antibiotics in
dates for clinical trials. Patients with hypoplastic MDS that are young patients with isolated neutropenia and MDS that are not receiving
should be considered for alloSCT up front. some form of cytotoxic or immunosuppressive therapy. I use prophylac-
Supportive care measures in MDS. A number of interventions can tic antibiotics in patients receiving active therapy. I use iron chelation
be used in patients with MDS. These include the use of prophylactic in patients with ferritin levels in excess of 2500 ng/mL but I consider
antibiotics and iron chelation. No randomized data exists to make all these patients for a clinical trial of iron chelation.

doi:10.1002/ajh.24102 American Journal of Hematology, Vol. 90, No. 9, September 2015 837
Garcia-Manero ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

Options for patients with relapsed or refractory lower azacitidine resulted in improved survival compared with a menu of
risk MDS and investigational new agents in lower risk standard of care options [48]. These included BSC, low dose cytara-
bine (ara-C) or AML-like therapy. In AZA-001, 358 patients with
MDS higher-risk MDS were randomized to either 5-azacitidine (as per
Treatment of patients with lower risk MDS is sequential. A com- CALGB9221 schedule) or to standard of care. Median age of patients
mon practice is to start growth factor support and then consider was 69 years. Median survival was significantly better in patients
lenalidomide or an azanucleoside. Patients that fail either lenalido- treated with 5-azacitidine versus standard of care options: 24.5
mide or azanucleoside are candidates for clinical trials or alloSCT. months versus 15 months (P 5 0.0001). Progression to AML was sig-
There is no drug approved in the US for patients with MDS and nificantly delayed, and RBC transfusion requirements and rate of
HMA failure. Recently the survival of these patients was calculated to infections were also significantly improved with 5-azacitidine. The
be between 14 and 17 months [69]. survival advantage with 5-azacitidine was irrespective of age (includ-
A number of agents are being studied for lower risk MDS. These ing patients older than 75 years), percent of marrow blasts (including
include oral azacitidine [51] and oral decitabine, agents that modulate patients with 20%30% blasts, now classified as AML using WHO
TGF-B pathway such as ACE-536 and ACE-011 [70,71], proteasome criteria) or karyotype. This effect was significant when compared with
inhibitors and antagonists of toll-like receptor signaling [55,72]. BSC and low dose ara-C. The number of patients treated with AML-
like therapy was too small to allow comparison with 5-azacitidine.
Options for newly diagnosed patients with higher risk Biomarkers of response to azanucleosides. An area of active
MDS research is the identification of biomarkers predictive of response to
Options for patients with higher risk MDS have evolved signifi- azanucleosides. Due to their capacity to induce DNA hypomethyla-
cantly over the last decade. Before that period most patients were tion, a number of groups have focused in the identification of methyl-
treated with some of cytarabine based therapy modeled after AML. ation patterns that would predict for response. Recently, a large-scale
The used of azanucleosides has modified this practice [2]. methylation analysis has resulted in the identification of 167 differen-
Azanucleosides. Decitabine was studied in an initial randomized tially methylated regions associated with response to hypomethylating
comparing it to BSC [49]. In this study the dose of decitabine was agent based therapy [79]. Of interest, most of these regions were
15 mg/m2 IV infused over 3 hours every 8 hours for 3 days (at a localized in nonpromoter regions. Two candidate biomarkers and a
dose of 135 mg/m2 per course) and repeated every 6 weeks. Although clinical model have been proposed. Levels of miR29b [76] and muta-
there was no clear benefit in terms of survival in this study, the use tions on TET2 [80] have been reported to be associated with response
of decitabine was associated with a complete response rate of 9% and to decitabine and azacitidine respectively. miR29b regulates expres-
overall response rate of 17%. These results led to the approval of dec- sion levels of DNMT1 [76]. TET2 is a protein involved in the conver-
itabine in the US. Based on the results of a phase I trial of decitabine sion of 5mC to 5OHmC and could therefore have resulted in passive
performed at MDACC, a Bayesian randomized phase II trial of three induction of DNA methylation [81]. None of these two biomarkers
different doses and schedules of decitabine was conducted [73]. In have been confirmed in other larger studies. Our group was not able
this study, a 5-day schedule of decitabine administered daily at a dose to correlate miRNA29b levels with response [82]. The French group
of 20 mg/m2 was shown to be superior to a 10-day or subcutaneous reported that previous low dose ara-C, bone marrow blasts more
schedule. A multicenter phase II trial of decitabine (ADOPT) using than 15% and abnormal karyotype predicted for lower response rate
the 5-day schedule confirmed the safety of this schedule, although to 5-azacitidine [83]. Poor performance status, intermediate and poor
response rates were lower than those reported by the MDACC [74]. risk cytogenetics, circulating blasts, and more than four units of red
In the ADOPT study the median number of courses administered blood cells transfuse every 8 weeks were associated with worse sur-
was five, the CR rate was 17% and the median survival was 19.4 vival [83]. In the near future a combination of genetic and clinical
months. No randomized survival study of a 5-day schedule of decita- markers could be used to define who may benefit from this type of
bine has been conducted in MDS. In parallel with this work, Euro- therapy. The incorporation of new molecular markers is going to
pean investigators developed a randomized study of decitabine using have a profound impact on our ability to prognosticate and select
the initial 3-day schedule. The major objective of the study was sur- therapy for patients with MDS.
vival. Unfortunately, use of decitabine at this dose and schedule was
not associated with improved survival in patients with higher risk Recommendation. The azanucleosides are the standard of care for
MDS [75]. Despite all this data, the final dose and schedule of decita- most patients with higher risk disease. No study has compared 5-
bine is not fully understood. Blum et al. have indicated that a 10-day azacitidine vs decitabine. Although response rates appear to be similar,
schedule of decitabine has significant activity in AML [76]. only 5-azacitidine has been associated with improvement of survival in
5-Azacitidine has been studied in higher-risk MDS in two major a randomized trial. Based on this, I consider 5-azacitidine standard
randomized multicenter trials: CALGB 9221 [77] and AZA-001 [48]. therapy for front line treatment in higher risk MDS.
In the CALGB 9221 [77] study, 191 patients with MDS were random- AML-like chemotherapy. AML-like protocols in higher risk MDS
ized between 5-azacitidine (75 mg/m2/day for 7 consecutive days have generally used classical anthracycline-araC combinations similar
every 28 days) and best supportive care (BSC). Median age was 68 to those used in de novo AML [84,85]. When used in MDS or AML
years. Sixty percent of the patients in the 5-azacitidine group, com- post-MDS, AML-like therapy results in lower CR rates (40 to 60%),
pared with 5% of control arm patients, responded to treatment shorter CR duration (median duration of 10 to 12 months) and tend
(P < 0.0001). The median time to leukemic transformation or death to be associated with more prolonged periods of aplasia than
was 21 months in patients treated with 5-azacitidine versus 12 observed in AML. In addition, the feasibility of AML-like therapy is
months in the BSC arm (P 5 0.007). No significant difference in sur- also reduced by the advanced median age of patients with MDS. The
vival was observed. A landmark analysis suggested a survival advant- most important prognostic factor of response to AML-like therapy is
age for patients initially on 5-azacitidine or who had crossed-over to karyotype: patients with unfavorable karyotype (27/del 7q or com-
5-azacitidine within 6 months of inclusion on study (P 5 0.03). A sig- plex karyotype) have a low CR rate and short duration of response.
nificant improvement in quality of life was documented in patients This is of importance as, at least in the AZA-001 study [48], patients
treated with 5-azacitidine compared with BSC [78]. AZA-001 was a with alterations of chromosome 7 had a significant benefit with 5-
randomized study designed to test the concept that treatment with 5- azacitidine versus other therapies. Currently, AML-like therapy is

838 American Journal of Hematology, Vol. 90, No. 9, September 2015 doi:10.1002/ajh.24102
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES Myelodysplastic Syndromes: 2015 Update

Figure 5. Impact on survival of mutation of p53 or DNMT3A in patients with MDS treated with allogeneic stem cell transplantation (adapted from Bejar et al.
[36]). [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.]

only recommended for relatively younger patients with favorable consider that when marrow blasts > 10% at the time of transplant,
karyotype that are candidates for alloSCT. because of the very high relapse risk post transplant, pretransplant
therapy is required. A recent report from the EBMTR has indicated
Recommendation. The randomized AZA-001 study was not powered that long-term survival of patients with monosomy 7 is very poor
to demonstrate the superiority of 5-azacitidine versus AML-like ther- with alloSCT [87]. Although this data needs to be validated in more
apy. The reason for this being that most investigators did not consider recent series, these results have significant implications for the use of
their patients candidates for such therapy. Therefore the question is alloSCT in MDS, as this suggests that the current practice of reserv-
who may be a candidate for AML therapy. In our practice this is ing transplant for poor prognostic features may not be indicated. In
restricted to younger patients with a high likelihood of response to the patients at higher risk for relapsed post alloSCT, data from a single
therapy, such as diploid patients or candidates for alloSCT. I rarely use arm trial indicated that post-transplant hypomethylating use can
AML therapy in older patients or in those with poor risk cytogenetics. improve outcome [88].
AlloSCT. AlloSCT is reported to be the only curative treatment of To complicate more the decision making regarding alloSCT in
higher-risk MDS. Results from selected studies report prolonged DFS MDS, a recent report has associated presence of p53 and DNMT3A
in about 30%50% of the patients [86]. However its use is mainly mutations with poor prognosis. At this time no consensus exists in
restricted to younger patients with an appropriate donor. Different terms of how to use this data [36] (Fig. 5).
transplant modalities of different intensities and donor sources are
now in use. Most of them remain investigational and therefore in my Recommendations. All patients potential candidates for alloSCT
opinion all patients should be transplanted in the setting of a clinical should be counseled regarding the possibilities of undergoing alloSCT.
trial. Current advances in transplant technology are allowing the con- Optimally patients will be enrolled in an MDS specific clinical trial of
sideration of older patients and alternative donors. This should result alloSCT. A national study to study this issue is being developed. I con-
in greater number of older patients benefitting from this potentially sider the use of lower doses of 5-azacitidine in patients at high risk for
curative treatment modality. relapse post transplantation, including more recently now those with
There are several relevant practical questions regarding alloSCT in higher risk mutations such as p53 or DNMT3A.
MDS. These include timing of transplant; and what to do with
patients that achieved a complete response to hypomethylating agent
Investigational new options for patients with higher
prior to alloSCT. A study from the IBMTR indicated that early trans-
plantation in higher-risk MDS was associated with longer life expect- risk MDS, including HMA failures
ancy [64,65] (Fig. 4). Although data suggests longer survival with Because the HMAs are the standard of care in front line higher
SCT in patients with higher risk disease, it should be noted that risk MDS, a number of studies are being conducted to test combina-
curves cross close to 3 years after initiation of therapy when com- tions. Agents used in combinations include the histone deacetylase
pared with hypomethylating agents. It will be important to identify inhibitors [89,90] and lenalidomide. So far none of these combina-
who are long-term survivors that benefit the most from transplant. A tions have been shown to be superior to single agent HMA. A second
prospective study is comparing azanucleosides use versus transplant generation HMA known as SGI-110 is currently under development.
in MDS. In terms of what to do in responding patients, no recom- SGI-110 is a dinucleotide form of decitabine.
mendation can be given at this time. Other questions include whether At the present time there is no therapy approved for patients with
or not alloSCT should be preceded by a cytoreductive regimen (with higher risk MDS that fail hypomethylating agents or relapsed after
chemotherapy or perhaps hypomethylating agents). Many authors AML-like therapy or alloSCT. The group of patients that failed

doi:10.1002/ajh.24102 American Journal of Hematology, Vol. 90, No. 9, September 2015 839
Garcia-Manero ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

hypomethylating agents has particularly poor prognosis with an esti- use of immune checkpoint inhibitors in MDS [93]. Multiple studies
mated survival of 46 months [91]. This is an area of active research are investigating the role of this new class of agents in MDS. Finally,
that is complicated by lack of understanding of the mechanisms of it is becoming apparent that a fraction of patients with higher risk
resistance to HMAs. It also should be emphasized that outcomes of MDS are characterized by targetable mutations such as Flt-3 [94],
patients in response to HMA that electively stopped HMA is poor RAS, and IDH1and IDH2. Multiple agents blocking these mutations
[92]. In a phase III study of rigorsetib (Garcia-Manero, ASH 2014, are currently under development [95].
unpublished) versus best supportive care, the study drug was not
shown to improve survival. Very low dose clofarabine may have a Recommendations. All patients with higher risk disease that have
role in patients with HMA failure and diploid cytogenetics (Jabbour, relapsed or refractory disease should be considered for an investiga-
ASH 2014, unpublished). A recent area of particular interest is the tional clinical trial and potentially alloSCT.

REFERENCES 17. Steensma DP, Bejar R, Jaiswal S, et al. Clonal


hematopoiesis of indeterminate potential and its
33. Haferlach T, Nagata Y, Grossmann V, et al.
Landscape of genetic lesions in 944 patients
1. Tefferi A, Vardiman JW. Myelodysplastic syn- distinction from myelodysplastic syndromes. with myelodysplastic syndromes. Leukemia
dromes. N Engl J Med 2009;361(19):18721885. Blood, in press. 2014;28(2):241247.
2. Garcia-Manero G, Fenaux P. Hypomethylating 18. DiNardo CD, Daver N, Jain N, et al. Myelodys- 34. Itzykson R, Kosmider O, Cluzeau T, et al.
agents and other novel strategies in myelodys- plastic/myeloproliferative neoplasms, unclassifi- Impact of TET2 mutations on response rate to
plastic syndromes. J Clin Oncol 2011;29(10):516 able (MDS/MPN, U): Natural history and azacitidine in myelodysplastic syndromes and
523. clinical outcome by treatment strategy. Leuke- low blast count acute myeloid leukemias. Leuke-
3. Garcia-Manero G. Myelodysplastic syndromes: mia 2014;28(4):958961. mia. 2011;25(7):11471152.
2014 Update on diagnosis, risk-stratification, 19. Kantarjian H, OBrien S, Ravandi F, et al. Pro- 35. Bejar R, Lord A, Stevenson K, et al. TET2 muta-
and management. Am J Hematol 2014;89(1):97 posal for a new risk model in myelodysplastic tions predict response to hypomethylating
108. syndrome that accounts for events not consid- agents in myelodysplastic syndrome patients.
4. Rollison DE, Howlader N, Smith MT, et al. Epi- ered in the original International Prognostic Blood 2014;124(17):27052712.
demiology of myelodysplastic syndromes and Scoring System. Cancer 2008;113(6):13511361. 36. Bejar R, Stevenson KE, Caughey B, et al.
chronic myeloproliferative disorders in the 20. Garcia-Manero G, Shan J, Faderl S, et al. A prog- Somatic mutations predict poor outcome in
United States, 20012004, using data from the nostic score for patients with lower risk myelo- patients with myelodysplastic syndrome after
NAACCR and SEER programs. Blood 2008; dysplastic syndrome. Leukemia 2008;22(3):538 hematopoietic stem-cell transplantation. J Clin
112(1):4552. 543. Oncol 2014;32(25):26912698.
5. Goldberg SL, Chen E, Corral M, et al. Incidence 21. Bejar R, Stevenson KE, Caughey BA, et al. Vali- 37. Sekeres MA, Schoonen WM, Kantarjian H, et al.
and clinical complications of myelodysplastic dation of a prognostic model and the impact of Characteristics of US patients with myelodys-
syndromes among United States Medicare bene- mutations in patients with lower-risk myelodys- plastic syndromes: Results of six cross-sectional
ficiaries. J Clin Oncol 2011;28(17):28472852. plastic syndromes. J Clin Oncol. 2012;30(27): physician surveys. J Natl Cancer Inst 2008;
6. Greenberg P, Cox C, LeBeau MM, et al. Interna- 33763382. 100(21):15421551.
tional scoring system for evaluating prognosis in 22. Naqvi K, Garcia-Manero G, Sardesai S, et al. 38. Moyo V, Lefebvre P, Duh MS, et al. Erythropoi-
myelodysplastic syndromes. Blood 1997;89(6): Association of comorbidities with overall sur- esis-stimulating agents in the treatment of ane-
20792088. vival in myelodysplastic syndrome: Development mia in myelodysplastic syndromes: A meta-
7. Dayyani F, Conley AP, Strom SS, et al. Cause of of a prognostic model. J Clin Oncol 2011; analysis. Ann Hematol 2008;87(7):527536.
death in patients with lower-risk myelodysplas- 29(16):22402246. 39. Jadersten M, Malcovati L, Dybedal I, et al.
tic syndrome. Cancer 2010;116(9):21742179. 23. Della Porta MG, Malcovati L, Strupp C, et al. Erythropoietin and granulocyte-colony stimulat-
8. Greenberg PL, Tuechler H, Schanz J, et al. Risk stratification based on both disease status ing factor treatment associated with improved
Revised international prognostic scoring system and extra-hematologic comorbidities in patients survival in myelodysplastic syndrome. J Clin
for myelodysplastic syndromes. Blood 2012; with myelodysplastic syndrome. Haematologica Oncol 2008;26(21):36073613.
120(12):24542465. 2011;96(3):441449. 40. Jadersten M, Montgomery SM, Dybedal I, et al.
9. Schanz J, Tuchler H, Sole F, et al. New compre- 24. Daver N, Naqvi K, Jabbour E, et al. Impact of Long-term outcome of treatment of anemia in
hensive cytogenetic scoring system for primary comorbidities by ACE-27 in the revised-IPSS for MDS with erythropoietin and G-CSF. Blood
myelodysplastic syndromes (MDS) and oligo- patients with myelodysplastic syndromes. Am J 2005;106(3):803811.
blastic acute myeloid leukemia after MDS Hematol 2014;89(5):509516. 41. Park S, Grabar S, Kelaidi C, et al. Predictive fac-
derived from an international database merge. 25. Tong WG, Quintas-Cardama A, Kadia T, et al. tors of response and survival in myelodysplastic
J Clin Oncol 2012;30(8):820829. Predicting survival of patients with hypocellular syndrome treated with erythropoietin and G-
10. Greenberg PL, Attar E, Bennett JM, et al. Mye- myelodysplastic syndrome: Development of a CSF: The GFM experience. Blood 2008;111(2):
lodysplastic syndromes: Clinical practice guide- disease-specific prognostic score system. Cancer 574582.
lines in oncology. J Natl Compr Canc Netw 2012;118(18):44624470. 42. Kantarjian HM, Giles FJ, Greenberg PL, et al.
2013;11(7):838874. 26. Quintas-Cardama A, Daver N, Kim H, et al. A Phase 2 study of romiplostim in patients with
11. Vardiman JW, Thiele J, Arber DA, et al. The prognostic model of therapy-related myelodys- low- or intermediate-risk myelodysplastic syn-
2008 revision of the WHO classification of mye- plastic syndrome for predicting survival and drome receiving azacitidine therapy. Blood 2010;
loid neoplasms and acute leukemia: Rationale transformation to acute myeloid leukemia. Clin 116(17):31633170.
and important changes. Blood 2009;114(5):937 Lymphoma Myeloma Leuk 2014;14(5):401410. 43. List A, Dewald G, Bennett J, et al. Lenalidomide
951. 27. Bejar R, Levine R, Ebert BL. Unraveling the in the myelodysplastic syndrome with chromo-
12. Haase D, Germing U, Schanz J, et al. New molecular pathophysiology of myelodysplastic some 5q deletion. N Engl J Med 2006;355(14):
insights into the prognostic impact of the karyo- syndromes. J Clin Oncol 2011;29(5):504515. 14561465.
type in MDS and correlation with subtypes: Evi- 28. Bejar R, Stevenson K, Abdel-Wahab O, et al. 44. List A, Kurtin S, Roe DJ, et al. Efficacy of lenali-
dence from a core dataset of 2124 patients. Clinical effect of point mutations in myelodys- domide in myelodysplastic syndromes. N Engl J
Blood 2007;110(13):43854395. plastic syndromes. N Engl J Med 2011;364(26): Med 2005;352(6):549557.
13. Jabbour E, Takahashi K, Wang X, et al. Acquisi- 24962506. 45. Fenaux P, Giagounidis A, Selleslag D, et al. A
tion of cytogenetic abnormalities in patients 29. Papaemmanuil E, Gerstung M, Malcovati L, randomized phase 3 study of lenalidomide ver-
with IPSS defined lower-risk myelodysplastic et al. Clinical and biological implications of sus placebo in RBC transfusion-dependent
syndrome is associated with poor prognosis and driver mutations in myelodysplastic syndromes. patients with low-/intermediate-1-risk myelodys-
transformation to acute myelogenous leukemia. Blood 2013;122(22)36163627. plastic syndromes with del5q. Blood 2011;
Am J Hematol 2013;88(10):831837. 30. Malcovati L, Papaemmanuil E, Bowen DT, et al. 118(14):37653776.
14. Naqvi K, Jabbour E, Bueso-Ramos C, et al. Clinical significance of SF3B1 mutations in mye- 46. List AF, Bennett JM, Sekeres MA, et al.
Implications of discrepancy in morphologic lodysplastic syndromes and myelodysplastic/ Extended survival and reduced risk of AML
diagnosis of myelodysplastic syndrome between myeloproliferative neoplasms. Blood 2011; progression in erythroid-responsive lenalido-
referral and tertiary care centers. Blood 2011; 118(24):62396246. mide-treated patients with lower-risk del(5q)
118(17):46904693. 31. Papaemmanuil E, Cazzola M, Boultwood J, et al. MDS. Leukemia. 2014;28(5):10331040.
15. Busque L, Patel JP, Figueroa ME, et al. Recur- Somatic SF3B1 mutation in myelodysplasia with 47. Raza A, Reeves JA, Feldman EJ, et al. Phase 2
rent somatic TET2 mutations in normal elderly ring sideroblasts. N Engl J Med 2011;365(15): study of lenalidomide in transfusion-dependent,
individuals with clonal hematopoiesis. Nat 13841395. low-risk, and intermediate-1 risk myelodysplas-
Genet 2012;44(11):117921181. 32. Thol F, Kade S, Schlarmann C, et al. Frequency tic syndromes with karyotypes other than dele-
16. Jaiswal S, Fontanillas P, Flannick J, et al. Age- and prognostic impact of mutations in SRSF2, tion 5q. Blood 2008;111(1):8693.
related clonal hematopoiesis associated with U2AF1, and ZRSR2 in patients with myelodys- 48. Fenaux P, Mufti GJ, Hellstrom-Lindberg E,
adverse outcomes. N Engl J Med 2014;371(26): plastic syndromes. Blood 2012;119(15):3578 et al. Efficacy of azacitidine compared with that
24882498. 3584. of conventional care regimens in the treatment

840 American Journal of Hematology, Vol. 90, No. 9, September 2015 doi:10.1002/ajh.24102
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES Myelodysplastic Syndromes: 2015 Update

of higher-risk myelodysplastic syndromes: A 65. Koreth J, Pidala J, Perez WS, et al. Role of 81. Ko M, Huang Y, Jankowska AM, et al. Impaired
randomised, open-label, phase III study. Lancet reduced-intensity conditioning allogeneic hema- hydroxylation of 5-methylcytosine in myeloid
Oncol 2009;10(3):223232. topoietic stem-cell transplantation in older cancers with mutant TET2. Nature
49. Kantarjian H, Issa JP, Rosenfeld CS, et al. Deci- patients with de novo myelodysplastic syn- 2010;468(7325):839843.
tabine improves patient outcomes in myelodys- dromes: An international collaborative decision 82. Yang H, Fang Z, Wei Y, et al. Levels of miR-
plastic syndromes: Results of a phase III analysis. J Clin Oncol 2013;31(21):26622670. 29b do not predict for response in patients with
randomized study. Cancer 2006;106(8):1794 66. Takatoku M, Uchiyama T, Okamoto S, et al. acute myelogenous leukemia treated with the
1803. Retrospective nationwide survey of Japanese combination of 5-azacytidine, valproic acid, and
50. Lyons RM, Cosgriff TM, Modi SS, et al. Hema- patients with transfusion-dependent MDS and ATRA. Am J Hematol 2011;86(2):237238.
tologic response to three alternative dosing aplastic anemia highlights the negative impact 83. Itzykson R, Thepot S, Quesnel B, et al. Prognos-
schedules of azacitidine in patients with myelo- of iron overload on morbidity/mortality. Eur J tic factors for response and overall survival in
dysplastic syndromes. J Clin Oncol 2009;27(11): Haematol 2007;78(6):487494. 282 patients with higher-risk myelodysplastic
18501856. 67. Sanz G, Nomdedeu B, Such E, et al. Independ- syndromes treated with azacitidine. Blood 2011;
51. Garcia-Manero G, Gore SD, Cogle C, et al. ent Impact of Iron Overload and Transfusion 117(2):403411.
Phase I study of oral azacitidine in myelodys- Dependency on Survival and Leukemic Evolu- 84. Wattel E, De Botton S, Luc Lai J, et al. Long-
plastic syndromes, chronic myelomonocytic leu- tion in Patients with Myelodysplastic Syndrome. term follow-up of de novo myelodysplastic syn-
kemia, and acute myeloid leukemia. J Clin Blood. 2008;112:640. dromes treated with intensive chemotherapy:
Oncol 2011;29(18):25212527. 68. Greenberg PL, Attar E, Bennett JM, et al. Mye- Incidence of long-term survivors and outcome
52. Garcia-Manero G, Jabbour E, Borthakur G, lodysplastic syndromes. J Natl Compr Canc of partial responders. Br J Haematol 1997;98(4):
et al. Randomized open-label phase II study of Netw 2011;9(1):3056. 983991.
decitabine in patients with low- or intermediate- 69. Jabbour EJ, Garcia-Manero G, Strati P, et al. 85. Beran M, Shen Y, Kantarjian H, et al. High-
risk myelodysplastic syndromes. J Clin Oncol Outcome of patients with low-risk and dose chemotherapy in high-risk myelodysplastic
2013;31(20):25482553. intermediate-1-risk myelodysplastic syndrome syndrome: Covariate-adjusted comparison of
53. Young NS, Calado RT, Scheinberg P. Current after hypomethylating agent failure: A report on five regimens. Cancer 2001;92(8):19992015.
concepts in the pathophysiology and treatment behalf of the MDS Clinical Research Consor- 86. Chang C, Storer BE, Scott BL, et al. Hematopoi-
of aplastic anemia. Blood 2006;108(8):2509 tium. Cancer 2015;121(6):876882. etic cell transplantation in patients with myelo-
2519. 70. Attie KM, Allison MJ, McClure T, et al. A phase dysplastic syndrome or acute myeloid leukemia
54. Olnes MJ, Sloand EM. Targeting immune dysre- 1 study of ACE-536, a regulator of erythroid arising from myelodysplastic syndrome: Similar
gulation in myelodysplastic syndromes. JAMA differentiation, in healthy volunteers. Am J outcomes in patients with de novo disease and
2011; 305(8):814819. Hematol 2014;89(7):766770. disease following prior therapy or antecedent
55. Ganan-Gomez I, Wei Y, Starczynowski DT, 71. Suragani RN, Cadena SM, Cawley SM, et al. hematologic disorders. Blood 2007;110(4):1379
et al. Deregulation of innate immune and Transforming growth factor-beta superfamily 1387.
inflammatory signaling in myelodysplastic syn- 87. Van Gelder M, Schetelig J, Volin L, et al. Mono-
ligand trap ACE-536 corrects anemia by pro-
dromes. Leukemia, in press. somal karyotype predicts poor outcome for
moting late-stage erythropoiesis. Nat Med 2014;
56. Saunthararajah Y, Nakamura R, Wesley R, et al. MDS/sAML patients with chromosome 7 abnor-
20(4):408414.
A simple method to predict response to immu- malities after allogeneic stem cell transplantation
72. Wei Y, Dimicoli S, Bueso-Ramos C, et al. Toll-
nosuppressive therapy in patients with myelo- for MDS/sAML. A study of the MDS subcom-
like receptor alterations in myelodysplastic syn- mittee of the Chronic Leukemia Working Party
dysplastic syndrome. Blood 2003;102(8):3025 drome. Leukemia 2013;27(9):18321840.
3027. of the European Group for Blood and Marrow
73. Kantarjian H, Oki Y, Garcia-Manero G, et al. Transplantation (EBMT). Blood 2009;114:293.
57. Sloand EM, Olnes MJ, Shenoy A, et al. Alemtu- Results of a randomized study of 3 schedules of
zumab treatment of intermediate-1 myelodys- 88. de Lima M, Giralt S, Thall PF, et al. Mainte-
low-dose decitabine in higher-risk myelodysplas- nance therapy with low-dose azacitidine after
plasia patients is associated with sustained
tic syndrome and chronic myelomonocytic leu- allogeneic hematopoietic stem cell transplanta-
improvement in blood counts and cytogenetic
kemia. Blood 2007;109(1):5257. tion for recurrent acute myelogenous leukemia
remissions. J Clin Oncol 2011;28(35):51665173.
74. Steensma DP, Baer MR, Slack JL, et al. Multi- or myelodysplastic syndrome: A dose and
58. Zou JX, Rollison DE, Boulware D, et al. Altered
center study of decitabine administered daily for schedule finding study. Cancer 2010;116(23):
naive and memory CD41 T-cell homeostasis
and immunosenescence characterize younger 5 days every 4 weeks to adults with myelodys- 54205431.
patients with myelodysplastic syndrome. Leuke- plastic syndromes: The alternative dosing for 89. Garcia-Manero G, Gore SD. Future directions
mia 2009;23(7):12881296. outpatient treatment (ADOPT) trial. J Clin for the use of hypomethylating agents. Semin
59. Kadia TM, Borthakur G, Garcia-Manero G, Oncol 2009;27(23)38423848. Hematol 2005;42(3 Suppl 2):S50S59.
et al. Final results of the phase II study of rabbit 75. Lubbert M, Suciu S, Baila L, et al. Low-dose 90. Sekeres MA, List AF, Cuthbertson D, et al.
anti-thymocyte globulin, ciclosporin, methyl- decitabine versus best supportive care in elderly Phase I combination trial of lenalidomide and
prednisone, and granulocyte colony-stimulating patients with intermediate- or high-risk myelo- azacitidine in patients with higher-risk myelo-
factor in patients with aplastic anaemia and dysplastic syndrome (MDS) ineligible for inten- dysplastic syndromes. J Clin Oncol 2010;28(13):
myelodysplastic syndrome. Br J Haematol 2012; sive chemotherapy: Final results of the 22532258.
157(3):312320. randomized phase III study of the European 91. Jabbour E, Garcia-Manero G, Batty N, et al.
60. Garg R, Faderl S, Garcia-Manero G, et al. Phase Organisation for Research and Treatment of Outcome of patients with myelodysplastic syn-
II study of rabbit anti-thymocyte globulin, Cancer Leukemia Group and the German MDS drome after failure of decitabine therapy. Cancer
cyclosporine and granulocyte colony-stimulating Study Group. J Clin Oncol 2011;29(15):1987 2010;116(16):38303834.
factor in patients with aplastic anemia and mye- 1996. 92. Cabrero M, Jabbour E, Ravandi F, et al. Discon-
lodysplastic syndrome. Leukemia 2009;23(7): 76. Blum W, Garzon R, Klisovic RB, et al. Clinical tinuation of hypomethylating agent therapy in
12971302. response and miR-29b predictive significance in patients with myelodysplastic syndromes or
61. Lim ZY, Killick S, Germing U, et al. Low IPSS older AML patients treated with a 10-day sched- acute myelogenous leukemia in complete remis-
score and bone marrow hypocellularity in MDS ule of decitabine. Proc Natl Acad Sci U S A. sion or partial response: Retrospective analysis
patients predict hematological responses to 2010;107(16):74737478. of survival after long-term follow-up. Leukemia
antithymocyte globulin. Leukemia 2007;21(7): 77. Silverman LR, Demakos EP, Peterson BL, et al. Res 2015;39(5):520524.
14361441. Randomized controlled trial of azacitidine in 93. Yang H, Bueso-Ramos C, DiNardo C, et al.
62. Passweg JR, Giagounidis AA, Simcock M, et al. patients with the myelodysplastic syndrome: A Expression of PD-L1, PD-L2, PD-1 and CTLA4
Immunosuppressive therapy for patients with study of the cancer and leukemia group B. in myelodysplastic syndromes is enhanced by
myelodysplastic syndrome: A prospective J Clin Oncol 2002;20(10):24292440. treatment with hypomethylating agents. Leuke-
randomized multicenter phase III trial compar- 78. Kornblith AB, Herndon JE II, Silverman LR, mia 2014;28(6):12801288.
ing antithymocyte globulin plus cyclosporine et al. Impact of azacytidine on the quality of life 94. Takahashi K, Jabbour E, Wang X, et al.
with best supportive careSAKK 33/99. J Clin of patients with myelodysplastic syndrome Dynamic acquisition of FLT3 or RAS alterations
Oncol 2011;29(3):303309. treated in a randomized phase III trial: A Can- drive a subset of patients with lower risk MDS
63. Olnes MJ, Scheinberg P, Calvo KR, et al. cer and Leukemia Group B study. J Clin Oncol to secondary AML. Leukemia 2013;27(10):2081
Eltrombopag and improved hematopoiesis in 2002;20(10):24412452. 2083.
refractory aplastic anemia. N Engl J Med 2012; 79. Meldi K, Qin T, Buchi F, et al. Specific molecu- 95. Montalban-Bravo G, Garcia-Manero G. Novel
367(1):1119. lar signatures predict decitabine response in drugs for older patients with acute myeloid leu-
64. Cutler CS, Lee SJ, Greenberg P, et al. A decision chronic myelomonocytic leukemia. J Clin Invest kemia. Leukemia 2015;29(4):760769.
analysis of allogeneic bone marrow transplanta- 2015;125(5):18571872.
tion for the myelodysplastic syndromes: Delayed 80. Itzykson R, Kosmider O, Cluzeau T, et al. Pres-
transplantation for low-risk myelodysplasia is ence of TET2 Mutation predicts a higher
associated with improved outcome. Blood 2004; response rate to azacitidine in MDS and AML
104(2):579585. post MDS. Blood 2010:abstract 439.

doi:10.1002/ajh.24102 American Journal of Hematology, Vol. 90, No. 9, September 2015 841

Vous aimerez peut-être aussi