Skip to content
Publicly Available Published by De Gruyter January 14, 2016

Laboratory and clinical risk assessment to treat myelodysplatic syndromes

  • Antonio Gidaro , Giorgio Lambertenghi Deliliers , Paolo Gallipoli , Massimo Arquati , Maddalena Alessandra Wu and Roberto Castelli EMAIL logo

Abstract

Myelodisplastic syndromes (MDS) are heterogeneous myeloid disorders characterized by peripheral cytopenias and increased risk of transformation into acute myelogenous leukemia (AML). MDS are generally suspected in the presence of cytopenia on routine analysis and the evaluation of bone marrow cells morphology and cellularity leads to correct diagnosis of MDS. The incidence of MDS is approximately five cases per 100,000 people per year in the general population, but it increases up to 50 cases per 100,000 people per year after 60 years of age. Typically MDS affect the elderly, with a median age at diagnosis of 65–70 years. Here the current therapeutic approaches for MDS are evaluated by searching the PubMed database. Establishing the prognosis in MDS patients is a key element of therapy. In fact an accurate estimate of prognosis drives decisions about the choice and timing of the therapeutic options. Therapy is selected based on prognostic risk assessment, cytogenetic pattern, transfusion needs and biological characteristics of the disease, comorbidities and clinical condition of the patients. In lower-risk patients the goals of therapy are different from those in higher-risk patients. In lower-risk patients, the aim of therapy is to reduce transfusion needs and transformation to higher risk disease or AML, improving the quality of life and survival. In higher-risk patients, the main goal of therapy is to prolong survival and to reduce the risk of AML transformation. Current therapies include growth factor support, lenalidomide, immunomodulatory and hypomethylating agents, intensive chemotherapy, and allogenic stem cell transplantation. The challenge when dealing with MDS patients is to select the optimal treatment by balancing efficacy and toxicity.

Introduction

Myelodisplastic syndromes (MDS) are a group of clonal myeloid disorders characterized by progressive cytopenia due to ineffective hematopoiesis, with a variable risk of transformation into acute myeloid leukemia (AML) [1, 2].

The incidence of MDS is estimated to be around five cases per 100,000 people per year in the general population, but after 60 years it increases up to 50 cases per 100,000 people per year. Typically MDS affect elderly people (median age at diagnosis of 65–70 years), while they occur in <10% of patients under 50 years of age [3]. The annual incidence of MDS increases logarithmically after 20 years of age, from <1.0 per million persons to 20 per 100,000 persons in septuagenarians. Males are affected approximately 1.5 times as often as females. The incidence is widely distributed with no ethnic differences, but in the Asian population MDS occur at an earlier age, as compared to the Western population [3].

MDS are generally suspected in the presence of cytopenia on routine analysis of peripheral blood, which triggers bone marrow evaluation. The evaluation of bone marrow cells morphology and cellularity generally shows hypercellular bone marrow, with variable grades of dysplasia, with or without immature blood cells. Parameters such as the percentage of blasts in the bone marrow, the number of cell lines involved in peripheral cytopenia, and karyotype abnormalities can significantly affect the natural course of the disease and its prognosis [4]. The International Prognostic Scoring System (IPSS) on the basis of the above mentioned parameters identifies four risk groups of patients with different prognosis: low, intermediate-1, intermediate-2, and high risk [5]. The median overall survival of MDS intermediate-2 or high-risk patients is 1.2 and 0.4 years, respectively; these patients are at high risk of developing AML, which means that the aim of the treatment is to modify the natural history of the disease and extend survival. Over the years more accurate prognostic scores, including an evaluation of transfusion requirement and assessment of patients’ comorbidities, have been developed, in order to obtain a deeper risk assessment to guide treatment in MDS patients [512].

The available therapies range from the treatment of symptomatic cytopenias in the low-risk group of MDS patients, to immunomodulatory agents, chemotherapy or allogenic stem cell transplantation in high-risk patients. The aim of this article is to review the role of oldest and newer laboratory assays in association with clinical and hematological parameters in stratifying the risk of MDS patients.

Materials and methods

We reviewed the medical literature for published studies evaluating “Myelodysplastic syndromes and risk assessment, and laboratory evaluation and Therapy and Prognosis”. The PubMed electronic database was searched without temporal limits using an English language restriction. The key words used were: myelodysplastic syndromes, risk assessment, laboratory evaluation, therapy and prognosis. References of most recent papers on myelodysplastic syndromes were also cross-referenced to identify potentially relevant papers not captured in our initial literature search. Data of pediatric patients are not considered in the present paper. Search terms were also applied to abstracts from the latest international hematological and oncological congresses.

The reference lists of the trials as well as articles were reviewed for additional publications.

When there was duplication of publications, we reviewed each article and included only the most recent or the complete version of the trial for analysis. In situations in which there was a discrepancy in the data, we considered the safety report from the most recent package insert to be the most accurate and used that report instead of the original publication for our review article.

Clinical and laboratory diagnosis

MDS patients complain about symptoms which are usually consistent with the type and severity of the peripheral blood cytopenias. They commonly report fatigue and decreased exercise tolerance due to anemia. Less often, patients show bleeding, easy bruisability, or recurrent bacterial infections as initial complaint [4]. Hepatomegaly or splenomegaly occur in approximately 5 or 10% of patients, respectively.

Blood and bone marrow examination

Anemia is present in >85% of patients and it is generally macrocytic. Red cell shape abnormalities include oval, elliptical, tear-drop, spherical, and fragmented cells. Reticulocyte counts are usually lower than expected on the basis of the degree of anemia. This latter finding is consistent with ineffective erythropoiesis.

Approximately 50% of patients at the time of diagnosis show neutropenia [13]. The percentage of monocytes is often slightly increased, and monocytosis per se can be the dominant manifestation of the hematopoietic abnormality for months or years [14]. Morphologic abnormalities of neutrophils can occur, sometimes resulting in the acquired Pelger-Huët anomaly. Approximately 25% of patients have mild to moderate thrombocytopenia at the time of diagnosis [4]. Mild thrombocytosis can also occur. Platelets may be abnormally large, may present poor granulation, or have large, fused central granules. Abnormal platelet function, with decreased platelet aggregation in response to collagen or epinephrine, can be responsible for a prolonged bleeding time, or easy bruising [15], Table 1 represents blood and bone marrow findings in MDS [6].

Table 1

Peripheral blood and bone marrow findings in myelodysplastic syndromes [6].

Peripheral bloodBone marrow examination
ErythropoiesisAnisocytosisErythroid hyperplasia
Elliptical red cellsPathologic sideroblasts
Fragmented cellsMegaloblastoid erythropoiesis
Macrocytic or dismorphic red cellsProerythroblasts may be present in excess
Ringed sideroblasts
GranulopoiesisAcquired Pelger-Huët anomalyGranulocytic hyperplasia
Neutrophils with condensed chromatinHypogranulation
Unilobed or bilobed nucleiImmature myeloid cells
Defective primary granules of abnormal sizeAcquired Pelger-Huët anomaly
ThrombopoiesisThrombocytopeniaMicromegakaryocytes
Abnormally large plateletsMegakaryocytes with unilobed or bilobed nuclei
Abnormal platelet function (decreased platelet aggregation in response to collagen or epinephrine)

Bone marrow analysis has a pivotal role for the diagnosis of MDS. The bone marrow is generally hypercellular, shows dysplastic features in one or several myeloid series, generally different from those observed in megaloblastic anemia due to vitamin B12 or folate deficiency. The bone marrow blast percentage should be assessed on at least 500 nucleated cells. Ring sideroblasts count, after Prussian blue staining, is mandatory, considering the differential diagnosis with refractory anemia with ringed sideroblasts (RARS). The trephine biopsy is essential when bone marrow fibrosis is suspected, and when a differential diagnosis with aplastic anemia or AML is needed [4]. Table 2 diagnostic approach to myelodysplastic syndromes and their diagnostic significance [6].

Table 2

Diagnostic approach to myelodysplastic syndromes and their diagnostic significance [6].

Clinical utility
Mandatory
 Bone marrow aspirateMorphological evidence of dysplasia
Blast count
Ringed sideroblasts count
 Conventional cytogenetic analysisCytogenetics are of importance to establish clonal haematopoiesis, calculate prognosis of patients and in some subsets of patients to drive specific therapy (e.g. 5q- syndrome and lenalidomide)
Bone marrow cellularity
Immature CD 34+ myeloid cells
 Bone marrow biopsyReticulin and collagen fibers evaluation
In dry tap or hypoplastic MDS essential to diagnosis
Optional
 Flow cytometryCan be of help in the identification of abnormal phenotypic patterns and can be of help in cases of minimal dysplasia
 Molecular analysisAssessment of specific genetic abnormalities with diagnostic and prognostic significance
 Fluorescent in situ hybridization (FISH)To establish clonal hematopoiesis, after conventional cytogenetic failure

Cytogenetic findings

Chromosomal abnormalities are described in MDS [16, 17]. Cytogenetic analysis has been shown to be of major prognostic value for MDS, being part of IPSS scoring system. Table 3 describes frequencies of cytogenetic abnormalities and prognostic IPSS-R risk category in MDS [7, 18].

Table 3

Cytogenetic findings in patients with myelodysplastic syndromes, by their prognostic value [7, 18].

IPSS-R risk categoryProportion of patients, %KaryotypeMedian survival, yearsTime to 25% acute myeloid leukaemia evolution, years
Very good4-Y, del(11q)5–4Not reached
Good72Normal, del(5q), del(12p), del(20q), double including del(5q)4–89–4
Intermediate13del(7q), +8, +19, i(17q), any other single or double independent clones2–72–5
Poor4–7, inv(3)/t(3q)/del(3q), double including –7/del(7q); complex: 3 abnormalities1–51–7
Very poor7Complex >3 abnormalities0–70–7

Furthermore, in addition to prognostic value, cytogenetic analysis has a pivotal role in confirming or in ruling out the diagnosis of MDS, when hematological findings are ambiguous. Common abnormalities include an extra chromosome 8; loss of the long arm of the chromosome 5, 7, 9, 20, or 21; and monosomy for chromosomes 7 and 9. Losses of part or all of chromosomes 5 and 7 and complex chromosome aberrations are particularly common in the oligoblastic myelogenous leukemias (and the overt leukemias) associated with prior treatment with cytotoxic drugs, radiation, or exposure to benzene [19]. Categories of cytogenetic abnormalities correlated with median survival have been determined. The more favorable risk category includes a normal karyotype and isolated deletions of 5q32-33.3, 20q, or Y. The poor-risk category includes -5q31.1, -7, del (7q), and complex chromosomal abnormalities. The intermediate-risk group includes other abnormalities. In treatment-induced MDS, complex cytogenetic abnormalities are very common, whereas in de novo MDS abnormalities occur in approximately 15% of cases [20].

Fluorescence in situ hybridization (FISH) is generally considered useful to identify the chromosome involved when low number of mitoses are available [13].

5q syndrome

Abnormalities in chromosome 5 occur in more than 15% of MDS patients, but the incidence of the originally described 5q- syndrome is much less frequent [21, 22].

The 5q- syndrome is characterized by refractory macrocytic anemia, normal or increased platelet counts, increased numbers of megakaryocytes in the bone marrow, and deletion of the long arm of chromosome 5 (5q-) as the unique cytogenetic abnormality. Survival is relatively long, and is characterized by low rate of leukemic transformation. The majority of patients are older women. Iron overload due to transfusion requirement can become a clinical problem in these patients, and iron chelating therapy may be required [22].

Patients with del(5q), either as an isolated abnormality or often as part of a complex karyotype, have a higher rate of concomitant TP53 mutations. These mutations are associated with diminished response or relapse after treatment with lenalidomide. In these cases, TP53 mutations may be secondary events and are often present in small subclones that can expand during treatment. More sensitive techniques may be required to identify the presence of sub-clonal, low-abundance TP53 mutations before treatment [23, 24].

Cytopenias with myelodysplastic changes can be seen in a variety of conditions, some of which are reversible. It is crucial to exclude these reversible causes before giving a patient the diagnosis of myelodysplasia or starting treatment for MDS.

The diagnostic approach recommended by the WHO in the case of a patient with suspected MDS includes the integration of the cytological evaluation of peripheral blood smears, evaluation of bone marrow aspirates and bone marrow biopsy. Data resulting from the analysis of conventional cytogenetics, FISH and immunophenotyping can complete the diagnosis. Diagnosis can be difficult when cytopenias are moderate, especially when a mild bone marrow dysplasia coexist. It is calculated that diagnostic discrepancy can occur at the time of initial presentation in 20%–30% of patients [4].

Classification

The integration of morphological, histopathological and cytogenetic tests allows to define the diagnosis of MDS in accordance with the current classification proposed by the WHO in 2008 [25].

Seven distinct categories have been identified according to the WHO classification: refractory anemia (RA), refractory anemia with ringed sideroblasts, refractory cytopenia with multilineage dysplasia (RCMD), refractory cytopenia and ringed sideroblasts (RCMD-RS), refractory anemia with excess blasts-1 (RAEB I) and refractory anemia with excess blasts-2 (RAEB-2), myelodispastic syndrome unclassified (MDS-U), MDS associated with isolated del(5q). This classification is a useful tool for the definition of different subtypes characterized by different prognosis. Therapy-related myelodysplastic syndromes are separately classified, together with therapy-related AML.

Variants of myelodysplastic syndromes

Therapy-related myelodysplasia

Therapy-related myelodysplasia (t-MDS) currently accounts for ~10% to 15% of MDS cases [26] and it develops in patients who were previously receiving chemotherapy or radiotherapy. Hypocellular MDS and MDS with myelofibrosis are more common in patients with t-MDS. Most patients have a relatively brief myelodysplastic phase and progress to overt leukemia within a few months. There is a high incidence of adverse cytogenetic abnormalities, particularly abnormalities involving chromosomes 5 and 7.

Hypocellular myelodysplasia

Approximately 10%–15% of patients with myelodysplasia have a hypocellular bone marrow [27]. The challenge is to distinguish this form from aplastic anemia: the presence of striking myelodysplastic changes together with cytogenetic analysis are the key elements for differential diagnosis. A trial of immunosuppressant agents may be warranted.

Risk stratification

The prognostic score based on the French American British (FAB) classification called IPSS includes percentage of blasts, number of cytopenias, and cytogenetics [5]. This system is highly reproducible and very simple to use. In order to overcome the main limitation of the system (imprecise predictor of prognosis in low-risk patients) a new scoring system called IPSS-R has been developed [7]. Bone marrow cytogenetics, bone marrow blast percentage, and cytopenias remain the basis of the new system but novel components are included (five rather than three cytogenetic prognostic subgroups, splitting the low bone marrow blast percentage value and depth of cytopenias). This model defined five rather than the four major prognostic categories that are present in the IPSS. The WHO classification-based prognostic scoring system (WPSS) represents another commonly used scoring system [8]. This system was developed when it was clear that red cell transfusion dependency is an independent predictor of prognosis in MDS. This system was also developed as time-dependent model, meaning that it can be used sequentially at any time during the course of the disease. The WPSS requires WHO classification of the disease and prior information on transfusion needs. Recently, the WPSS score was modified to include hemoglobin levels instead of transfusion needs. Both the IPSS and WPSS were developed in a very specific subset of patients: newly diagnosed patients at the time of initial presentation. Recently the global MD Anderson Cancer Centre (MDACC) model was developed [9]. Poor performance, older age, thrombocytopenia, anemia, increased bone marrow blasts, leukocytosis, chromosome 7 or complex (≥3) abnormalities and prior transfusions are considered the main predictors of prognosis in MDS patients in a multivariate analysis of prognostic factors. The new MDS prognostic model divided patients into four prognostic groups with significantly different outcomes. The model was found applicable to any patient with MDS at any time during the course of MDS. Low-risk MDS patients are challenging due to their heterogeneous prognosis which does not allow to distinguish between longer survival patients and those with intermediate outcome. In a study enrolling 600 MDS patients, the assessment of comorbidities performed with the adult comorbidity evaluation showed that patients with severe diseases had 50% lower survival than did those without co-morbidities, independently of age and IPSS risk group [10]. The impact of comorbidities on prognosis of MDS has been largely underlined also by other groups [11].

Amongst other prognostic factors, age is an important predictor of poor prognosis and bone-marrow fibrosis has been shown to be independently associated with poorer prognosis, in both lower-risk and higher-risk MDS patients [28]. Recurrent mutations in genes encoding components of the splicing machinery, including SF3B1, SRSF2, U2AF1, and ZRSR2, have also been reported in myelodysplastic syndromes [29, 30] SF3B1 mutations are strictly linked with ring sideroblastic subtypes of myelodysplastic syndromes; directly contributing to formation of ringed sideroblasts and abnormal iron retention. Furthermore, SF3B1 mutations seem to be predictors of favorable clinical outcome [31].

Somatic mutations also strongly affect survival. The presence of any of the mutations TP53, RUNX1, EZH2, or ETV6 worsens outcome, independently of IPSS [32, 33]. ASXL1 and SRSF2 mutations are also associated with a poorer outcome according to the literature [33, 34]. These genes can be divided into four main groups: (1) transcription factors (TP53, RUNX1, ETV6); (2) epigenetic regulators and chromatin-remodeling factors (TET2, DNMT3A, ASXL1, EZH2, IDH1/2); (3) pre-mRNA splicing factors (SF3B1, SRSF2, U2AF1, ZRSR2); and (4) signaling molecules (NRAS, CBL, JAK2, SETBP1). The most frequently mutated genes were TET2, SF3B1, ASXL1, DNMT3A, SRSF2, RUNX1, TP53, U2AF1, EZH2, ZRSR2, STAG2, CBL and NRAS, although no single mutated gene was found in more than a third of patients. Several of these gene mutations are associated with adverse clinical features, such as complex karyotypes (TP53), excess bone marrow blast proportion (RUNX1, NRAS, and TP53) and severe thrombocytopenia (RUNX1, NRAS, and TP53). Despite associations with clinical features considered by prognostic scoring systems, mutations in several genes hold independent prognostic value as shown in Table 4. Mutations of TP53, EZH2, ETV6, RUNX1, and ASXL1 have been shown to predict decreased OS in multivariable models adjusted for IPSS or IPSS-R risk groups in several studies. Within IPSS risk groups, a mutation in one or more of these genes identifies patients whose risk resembles that of patients in the next highest IPSS risk group (e.g. the survival curve for INT-1-risk patients with an adverse gene mutation was similar to that of patients assigned to the INT-2-risk group by the IPSS) [35].

Table 4

Reported frequency of genetic lesions in MDS [27, 30, 3436].

GeneFrequency, %LocationFunctionPrognosis
SF3BI282q33Splicing factorFavorable
TET2214q24Control of cytosine hydroxymethylationNeutral
ASXL11420q11Epigenic regulatorUnfavorable
SRSF21217q25Splicing factorNeutral
RUNX1921q22Transcription factorUnfavorable
TP53817q13Transcription factorUnfavorable
U2AF1721q22Splicing factorUnfavorable
EZH267q36Polycomb group proteinUnfavorable
NRAS4Ip13Signal transductionUnfavorable
JAK239p24Tyrosine KinaseFavorable
ETV6312p13Transcription factorUnfavorable
CBL211q23Signal transductionUnknown
IDH2215q26Cell metabolism, epigenetic regulationUnfavorable
NPM125q35PhosphoproteinUnknown
IDH112Q33As IDH2Unfavorable
KRAS<112q12Signal transductionUnfavorable
GNAS<120q13G proteinUnknown
PTPN<112q24Protein phospataseUnknown
BRAF<17q34Raf KinaseUnknown
PTEN11<110q23PhosphataseUnknown
CDKN2A<19q21Cell cycle controlUnknown

When applied to patients stratified by the IPSS-R, the presence of a mutation in one or more of these five genes was associated with shorter OS for patients in the low- and intermediate-risk groups. Other mutated genes have been associated with decreased OS, including DNMT3A, U2AF1, SRSF2, CBL, PRPF8, SETBP1, and KRAS.

Mutations of SF3B1 have been associated with a more favorable prognosis, but this may not be an independent risk factor.

For example, SF3B1 mutated patients are likely to present reduced hemoglobin levels leading to a higher transfusion dependence, while patients harboring SRSF2 mutations clustered in RAEB-1 and RAEB-2 subtypes and had pronounced thrombocytopenias [36]. Table 4 reports the frequency of genetic mutations in MDS [4, 27, 30, 3438].

Hyperferritininemia and high levels of lactate dehydrogenase are associated with poorer prognosis and higher cardiac or extra hematological mortality [39].

The introduction of new therapies that can modify the clinical course of MDS revealed new factors able to predict the response to these treatments, in addition to the traditional ones.

For example, patients with low levels of blood erythropoietin (EPO) respond better to erythroid stimulating agents (ESA) and an early resistance to ESA seems to be associated with a worse outcome [3941]. Response to hypomethylating agents can be predicted by performance status, karyotype, erythrocyte transfusion requirements, presence of circulating immature bone marrow elements or TP53 mutations [42, 43]. Although screening for such molecular defects on a routine basis cannot currently be recommended, the spread of massive genotyping technology will allow clinicians to detect a broad range of genetic aberrations in peripheral blood at a reasonable cost in the near future, making it easier to confirm the diagnosis in patients with suspected MDS.

Treatment strategies

In the recent years, despite the improvement of treatment strategies, MDS remain challenging diseases. New drugs such as lenalidomide, demethylating agents and iron chelators, helped slowing the natural history of the disease and improving the quality of life of patients who are not eligible for transplantation [44, 45]. Many factors make identification of an univocal treatment strategy in MDS patients very difficult. First of all, MDS are a heterogeneous group of hematological diseases, with different clinical and prognostic features but they are all linked by a clonal disorder of stem cells, ineffective haematopoiesis and a variable risk of transformation into AML. In addition, the complexity of cytogenetic and molecular abnormalities (except for 5q-) does not allow to identify targeted therapies. Most patients with MDS are old and affected by many comorbidities, therefore less able to tolerate aggressive therapies. Some patients with MDS may have prolonged survival; older patients with low-grade myelodysplasia may be more likely to die of illnesses other than MDS.

Establishing the prognosis for patients with MDS is a key step of their management. In fact, possible benefit from therapy has to be carefully balanced against the risks of complications. Treatment for myelodysplasia has to be highly individualized. An accurate estimate of prognosis drives decisions about timing and choice of the therapeutic options. In low-risk and unfit patients control of symptoms is the primary goal of therapy. In younger or healthier patients with high-risk MDS aggressive therapy in attempt to achieve cure might be warranted.

Treatment strategies in low-risk/intermediate-1 MDS (IPSS), very low, low, intermediate MDS (IPSS-R), very low, low, intermediate MDS (WPSS)

The approach for treatment of low-risk MDS is aimed at correcting cytopenias. Chronic anemia adversely affects the quality of life of MDS patients and the clinical course of disease while transfusion dependence leads to reduced survival [46, 47]. Erythropoietic stimulating agents (ESAs) increase the hemoglobin level in approximately 15%–25% of MDS patients. The multivariate analysis in several studies confirmed that predictive factors of major erythroid response after treatment with ESAs were baseline serum levels of EPO <100 IU/L, favorable cytogenetics, low number of blasts, no or low transfusion requirement [4648]. Anti-apoptotic effects on erythroid progenitors are probably the most important mechanism of action of ESAs. However, in responding patients the median duration of response is only approximately 2 years [47]. A recent study from our group has shown that biosimilar epoetin-α is effective for the treatment of anemia in MDS patients with comparable efficacy to that of other ESAs [49]. Several studies have shown that ESAs have no effects on the risk of progression to AML [5052].

In non-responding MDS patients there is some evidence that adding low-dose granulocyte colony stimulating factors (G-CSFs) has a synergistic effect with erythropoiesis stimulating agents (ESAs) [48, 53, 54]. No randomized study has shown improvement in survival for patients with MDS treated with epoetin-α plus G-CSF and further randomized trials are needed to optimize dosing. Greenberg et al. [48] showed that combination of G-CSFs (initial dose 1 μg/kg per day SQ) plus epoetin-α (150–300 units/kg per day SQ) results in a substantial erythroid response (i.e. decreased transfusion requirements and increased hemoglobin concentrations) in approximately 40%–47% of patients.

Lenalidomide directly acts on del5q- clones, inducing hematopoiesis and erythropoiesis stimulation, whereas other effects including immunomodulation, anti-inflammatory activity, and angiogenesis inhibition [55, 56] are similar to those of thalidomide. Based on this evidence lenalidomide has been licensed in the USA and in Europe for transfusion-dependent anemia in MDS patients with documented 5q-abnormality. Cytogenetic responses are reported in 50%–70% of the treated population [5759]. List et al. [60] showed that lenalidomide was effective in inducing an erythroid response as well as reversing cytological and cytogenetic abnormalities in 148 MDS patients with del5q31 and transfusion-dependency. The response to lenalidomide was rapid and long-lasting, and the median duration of transfusion independence had not been reached after a median of 104 weeks of follow-up. Moderate-to-severe neutropenia (in 55% of the patients) and thrombocytopenia (in 44%) were the most frequent reasons for interrupting treatment or adjusting the dose of lenalidomide. Fenaux et al. [56] carried out a randomized double-blind study of the efficacy and safety of lenalidomide in 205 patients with MDS who were RBC transfusion-dependent and at IPSS low-risk or intermediate-1 risk and carried del5q31. The researchers concluded that lenalidomide is beneficial and has an acceptable safety profile in transfusion-dependent 5q- patients who were with low to intermediate-1-risk. Lenalidomide reverses transfusion dependence in 25%–30% of lower-risk MDS patients resistant to ESAs [56] but it is not approved for this indication outside clinical trials. Preliminary results suggest that the combination of lenalidomide and ESAs can lead to high rates of independence from erythrocyte transfusion in patients resistant to ESAs alone [61, 62]. Treatment of anemia in patients resistant to ESAs (or lenalidomide in patients with deleted 5q) or after relapse remains challenging [21]. Hypomethylating agents in low risk MDS patients [63] are effective and are shown to lead to transfusion independence in about 40% of patients. However, despite these treatments, many low-risk MDS patients eventually need erythrocyte transfusion to control anemia.

In selected younger patients with low-risk MDS, with limited erythrocyte transfusion history, normal karyotype (or possibly trisomy 8), no excess marrow blasts or HLA DR15 genotype, no deleted 5q, limited exposure to previous treatments, and possibly hypocellular marrow, antithymocyte globulins, with or without ciclosporin, can be used to obtain an erythroid response, with response of other cytopenias (especially thrombocytopenia) in 25%–40% of MDS patients [6467].

The experience gained with thalassemic patients suggests the use of chelation therapy in patients with MDS undergoing transfusion therapy for which prolonged life expectancy is not already affected by leukemic transformation. In lower-risk myelodysplastic patients, receiving more than 20–40 red-blood-cell concentrates, or when serum ferritin rises over 1000 ng/mL [68], iron chelation therapy is considered, in order to prevent iron overload. However, clinically significant iron overload associated with heart failure is quite frequent in MDS patients, especially when elderly, receiving 100 or more red blood cell concentrates. Other papers suggest that potential benefits of iron chelation should be lowering of infection risk, improvement of the outcome of allogeneic hematopoietic stem cell transplantation and delay of leukemic transformation [69]. Deferasirox, an oral iron chelator, has shown efficacy and acceptable tolerability in MDS setting and has also been shown to improve peripheral cytopenia in 10%–20% of MDS patients [70, 71]. However, desferasirox has a potential renal toxicity and is contraindicated in patients with renal failure. According to literature data, we suggest a starting dose of 10–20 mg/kg/per day with dose escalation up to 40 mg/kg. Young MDS patients suitable for bone marrow transplantation need to be especially controlled for iron overload. Several studies have shown that iron overload can affect the outcome of bone marrow transplantation [7274].

Although MDS patients have a high incidence of infection due to neutropenia and granulocyte dysfunction and infection is the principal cause of death in patients with MDS, the use of granulocyte-colony stimulating factors (G-CSFs) [7577] or granulocyte macrophage colony stimulating factors (GM-CSFs) [7880] in efficacy trials, including randomized trials, has been disappointing. Antibiotics are indicated for bacterial infections, but no routine prophylaxis is recommended (with the exception of patients with recurrent infections).

Severe bleeding is a rare problem in low-risk MDS patients and platelet counts below 50×109 cell/L are generally observed in 30%–50% of patients. Rarely and only in advanced stages MDS patients require repeated platelet transfusions. The current availability of thrombopoietin-receptor agonists romiplostim and eltrombopag approved for therapy of autoimmune thrombocytopenia suggested their use in the setting of MDS patients [8183]. The development of myeloid malignancies is a concern when administering thrombopoietin receptor agonists. Among 168 MDS subjects treated with romiplostim, progression from MDS to AML was observed in 10 (6%) patients [84]. In another clinical trial a transient increase in bone marrow blasts has been reported in 15% [79]. In conclusion thrombopoietin mimetics should be considered in the setting of MDS patients with bleeding due to low platelet counts who do not respond to transfusions. Further studies to evaluate their safety in the setting of MDS patients with thrombocytopenia are ongoing. Figure 1 summarizes the therapeutic approach to this risk class of patients.

Figure 1: Therapeutic strategies for the treatment of myelodysplastic syndromes.Treatment strategies for patients with myelodisplastic syndromes according to the International prognostic scoring system (IPSS), WHO Prognostic Scoring System (WPSS) and Revised IPSS (R-IPSS). Anemic very low and low-intermediate myelodisplastic (MDS) patients carrying del 5q should be treated with lenalidomide in order to reduce transfusion request and to obtain cytogenetic response. Lenalidomide should be avoided in patients with a clinically significant decrease in neutophils or platelet count. Although the cost of lenalidomide is higher than that of other treatments, the reduction in the number of transfusions and in transfusion dependence partially offsets the expense. Symptomatic patients with no del(5q) or patients not responding to lenalidomide should be treated according to endogenous levels of eritropoietin (Epo) with erytropoietic stimulating agents (ESAs), eventually associated with granulocyte colony stimulating factors (G-CSFs). Non responding patients should be treated with immusuppressive therapy or with anti-timocytes globulin (ATG) or cyclosporine. Allo hematopoietic stem cell transplantation (allo-HSCT) should be considered in selected fit patients affected by intermediate-1 myelodysplastic syndromes. In selected patients carrying high levels of endogenous Epo who are unlikely to respond to ESAs immunosuppressive therapy should be considered. For patients with poor probability to respond to immunosuppressive therapy alternative therapies with Azacytidine/Decitidine/Lenalidomide should be considered. Red blood cell (leuko-reduced) transfusions are recommended for symptomatic anemia. Platelet transfusions are recommended for thrombocytopenic bleeding. However, they should not be used routinely in patients with thrombocytopenia in the absence of bleeding unless platelet count is <10,000/mm3. Irradiated products are suggested for transplant candidates. If >20 to 30 red blood cells transfusions have been received, consider daily chelation with deferoxamine subcutaneously or deferasirox orally to decrease iron overload, particularly for low/intermediate-1 and for potential transplant patients. Patients with low creatinine clearance (<40 mL/min) should not be treated with deferasirox. IPSS, International Prognostic Scoring System; WPSS, World Health Organization Prognostic Scoring System; IPSS-R, Revised IPSS; EPO, erythropoietin; ATG, anti timocytes globulins; allo HSCT, allo hematopoietic stem cell transplantation; MDS, myelodisplastic syndromes.
Figure 1:

Therapeutic strategies for the treatment of myelodysplastic syndromes.

Treatment strategies for patients with myelodisplastic syndromes according to the International prognostic scoring system (IPSS), WHO Prognostic Scoring System (WPSS) and Revised IPSS (R-IPSS). Anemic very low and low-intermediate myelodisplastic (MDS) patients carrying del 5q should be treated with lenalidomide in order to reduce transfusion request and to obtain cytogenetic response. Lenalidomide should be avoided in patients with a clinically significant decrease in neutophils or platelet count. Although the cost of lenalidomide is higher than that of other treatments, the reduction in the number of transfusions and in transfusion dependence partially offsets the expense. Symptomatic patients with no del(5q) or patients not responding to lenalidomide should be treated according to endogenous levels of eritropoietin (Epo) with erytropoietic stimulating agents (ESAs), eventually associated with granulocyte colony stimulating factors (G-CSFs). Non responding patients should be treated with immusuppressive therapy or with anti-timocytes globulin (ATG) or cyclosporine. Allo hematopoietic stem cell transplantation (allo-HSCT) should be considered in selected fit patients affected by intermediate-1 myelodysplastic syndromes. In selected patients carrying high levels of endogenous Epo who are unlikely to respond to ESAs immunosuppressive therapy should be considered. For patients with poor probability to respond to immunosuppressive therapy alternative therapies with Azacytidine/Decitidine/Lenalidomide should be considered. Red blood cell (leuko-reduced) transfusions are recommended for symptomatic anemia. Platelet transfusions are recommended for thrombocytopenic bleeding. However, they should not be used routinely in patients with thrombocytopenia in the absence of bleeding unless platelet count is <10,000/mm3. Irradiated products are suggested for transplant candidates. If >20 to 30 red blood cells transfusions have been received, consider daily chelation with deferoxamine subcutaneously or deferasirox orally to decrease iron overload, particularly for low/intermediate-1 and for potential transplant patients. Patients with low creatinine clearance (<40 mL/min) should not be treated with deferasirox. IPSS, International Prognostic Scoring System; WPSS, World Health Organization Prognostic Scoring System; IPSS-R, Revised IPSS; EPO, erythropoietin; ATG, anti timocytes globulins; allo HSCT, allo hematopoietic stem cell transplantation; MDS, myelodisplastic syndromes.

Treatment strategies in intermediate-2 high-risk MDS (IPSS), intermediate high, very high-risk MDS (IPSS-R)

The median overall survival of MDS intermediate-2 or high-risk patients is 1.2 and 0.4 years, respectively; these patients show high risk of developing AML, which means that the aim of the treatment is to modify the natural history of the disease and extend survival [3].

AML-like chemotherapies

AML-like therapy is only recommended for relatively younger patients with favorable karyotype that are candidates for AlloSCT. AML-like protocols in higher-risk MDS patients have generally used classical anthracycline-cytarabin combinations similar to those used in de-novo AML [80]. When used in MDS or AML post-MDS, AML-like therapy results in lower complete remission (CR) rates (40%–60%), shorter CR duration (median duration of 10–12 months) and is associated with more prolonged periods of aplasia. Patients with high risk karyotype have lower CR and shorter durations of remission (DOR) [81]. No chemotherapeutic regimen including fludarabine or topotecan, or gemtuzumab ozogamicin, with cytarabine, with or without G-CSF has shown any survival advantage over classic anthracycline-cytarabine regimens [8288].

Fifteen percent to 20% of complete or partial remissions are obtained with low-dose cytarabine (20 mg/m2 daily, 14–21 days every month) in higher-risk MDS patients, but no proven survival advantages [89] have been observed.

Hypomethylating agents

Oligoblastic and secondary myelogenous leukemias have a high prevalence of tumor suppressor gene hypermethylation. 5-Azacytidine is a pyrimidine analog inhibiting DNA methyltransferase, reducing cytosine methylation, and inducing maturation of some leukemic cell lines [90]. Antiproliferative properties have been showed by inhibiting the release of oncostatin-M, IL-6, and IL-11 from mononuclear cells in patients with clonal anemia [91]. 5-Azacytidine at a dose of 75 mg/m2 once per day given subcutaneously for 7 consecutive days each month provided significantly more frequent benefit to two thirds of patients than did supportive care [63]. Quality of life was improved and disease progression was delayed. 5-Azacitidine has been studied in higher-risk MDS patients in two major randomized multicenter trials: CALGB 9221 [63] and AZA-001 [81]. In the CALGB 9221 study, 191 patients (median age 68 years) with MDS were randomized to receive 5-azacitidine (75 mg/m2 per day for 7 consecutive days every 28 days) or best supportive care (BSC). Sixty percent of the patients in the 5-azacitidine group, compared with 5% of control arm patients, responded to treatment (p<0.0001). The median time to leukemic transformation or death was 21 months in patients treated with 5-azacitidine vs. 12 months in the BSC arm (p<0.007). A survival benefit due to delayed transition to AML was obtained in these studies treating MDS patients with 5-azacytidine treatment (75 mg/m2 daily subcutaneously, 7 days every 4 weeks). Age, bone marrow blast percentage, and karyotype seem to be independent factors of response. Moreover, these agents have shown a reduction in erythrocyte transfusion requirement. Achievement of any type of hematological improvement, even in the absence of complete or partial remission, was significantly associated with better outcome. The median duration of response to 5-azacitidine was 13.6 months. The median number of cycles was 15 in responders. These data suggest that long-term treatment is needed to obtain a survival benefit.

Moreover, the French group reported that previous therapy with low dose cytarabine, bone marrow blasts >15% and abnormal karyotype were predictors of lower response rate to 5-azacitidine [92]. Poor performance status, intermediate and poor risk cytogenetics, circulating blasts, and more than four units of red blood cells transfused every 8 weeks were associated with worse survival.

Decitabine is another hypomethylating agent that was tested in the EORTC/German MDS trial [92]. During this trial 233 patients with MDS (93% intermediate-2 or high IPSS) were randomly assigned to best supportive care with or without decitabine. Decitabine (15 mg/m2) was given intravenously over 4 hours three times a day for 3 days in 6-week cycles. At a median follow-up of 2.5 years, median overall survival was 8.5 months for BSC vs. 10.1 months for decitabine and acute myeloid leukemia-free survival was 6.1 months for BSC vs. 8.8 months for decitabine (these differences were both not statistically significant). The statistically significant achieved goals with treatment with decitabine were prolonged progression-free survival (median PFS, 6.6 vs. 3.0 months, respectively) and reduced AML transformation at 1 year (from 33% with BSC to 22% with decitabine). Decitabine treatment was also associated with improvements in patient-reported quality-of-life parameters.

The effectiveness of hypomethylating agents in inducing hematologic improvements and also true remissions with low toxicity justifies the hypothesis of their use in the pre-transplant phase instead of conventional chemotherapy.

Taken together these trials show that the pyrimidine nucleoside analogs of cytidine are the standard of care for patients with higher-risk disease, even in patients eligible for BMT as bridging therapy.

Now there is no therapy approved for patients with higher-risk MDS that do not respond to hypomethylating agents or relapse after AML-like therapy or AlloSCT. The group of patients for whom hypomethylating agents failed has a particularly poor prognosis. For these patients, investigational treatments within carefully designed clinical trials should be considered.

Allogeneic bone marrow transplant (BMT)

Allogeneic bone marrow transplant (BMT) represents the only potentially curative therapy for MDS. Unfortunately, the use of allogeneic BMT for MDS is limited by the older age of most patients and by the fact that only a minority of patients has histocompatible bone marrow donors. However, the use of allogeneic BMT is being extended to older patients, and the use of a national bone marrow donor registry has allowed matched unrelated transplants. Several trials treating MDS with allogeneic BMT have been performed; ~40% of patients in these trials have long-term disease-free survival and may be cured [93]. Patients with MDS show high transplant-related mortality rates (~30% to 35%) due to infections, graft-vs.-host disease and multi organ failure [94]. It is now generally accepted that AlloSCT with myeloablative conditioning is generally indicated only in few patients with MDS while for most other patients, particularly older patients, a reduced-intensity transplant can be still offered from an HLA-identical donor [95]. Different transplant modalities of different intensities and donor sources are now active. Most of them are still investigational. There are several relevant concerns regarding AlloSCT in MDS. These include timing of transplant and choice of the best approach for patients that achieved a complete response to hypomethylating agents prior to AlloSCT. A study from the International Blood and Bone Marrow Transplant Registry (IBMTR) indicated that early transplantation in higher-risk MDS patients was associated with better outcome [96].

Due to the observation that blast percentage before transplantation (especially if >10%) is clinically associated with higher risk of relapse [97], a cytoreductive regimen (chemotherapy or hypomethylating agents) are generally administered to patients with excess of blasts. Prospective studies on the topic are lacking to drive definitive conclusions.

Regarding the timing of BMT, both in younger higher-risk patients treated with myeloablative conditioning transplantation and in older patients receiving non-myeloablative regimens, early stem-cell transplantation was associated with a survival advantage compared with other therapeutic options [97, 98]. By contrast, early stem-cell transplantation had an adverse effect on survival in lower-risk patients [96, 97].

The evolution of transplantation techniques, today characterized by a better control of graft-vs.-host disease (GVHD) and infectious complications, the consequent increased availability of unrelated donors with the use of peripheral blood stem cells and the overall lower toxicity of the transplant have extended the availability of this procedure to patients older than 60 years if clinically fit [99].

There is no consensus regarding the optimal treatment of patients with intermediate-1 IPSS and intermediate IPSS-R risk and this remains a burning issue in the treatment of MDS patients. Patients who want to focus on quality rather than quantity of remaining years may favor supportive care and hypomethylating agents. In contrast, more intensive chemotherapy with or without transplantation may be chosen by younger, fit patients who prefer to deal with the higher risk of treatment-related mortality and morbidity in order to achieve an increase in survival. The possible role of hypomethylating agents as a bridge therapy to bone marrow transplantation is under consideration. Treatment with hypomethylating agents may delay progression to AML before HSCT. Such a strategy is especially relevant for MDS patients with intermediate-2 or high-risk IPSS disease, for whom the average time to AML progression may be short [100].

Figure 2 summarizes the approach to treatment in these subgroups of patients.

Figure 2: Treatment strategies for patients with myelodysplastic syndromes according to the International Prognostic Scoring System (IPSS), WHO Prognostic Scoring System (WPSS) and Revised IPSS (R-IPSS).Intermediate-2, high or very high risk myelodysplastic patients have different treatment strategies based on patients’ characteristics and their elegibility for high intensity therapy. If they are candidate to bone marrow transplant and a donor is available, patients should be treated with allo hematopoietic stem cell transplantation. A bridging therapy should be considered in order to decrease marrow blasts to an acceptable level prior to transplant. High intensity chemotherapy (only in selected fit patients) or azacytdine/decitabine treatment should be considered for fit patients eligible for high intensity chemotherapy. Not high intensity therapy candidates should be treated with azacytdine/decitabine. Supportive care or clinical trials should be considered for non-responding or relapsing patients. IPSS, International Prognostic Scoring System; WPSS, World Health Organization Prognostic Scoring System; IPSS-R, Revised IPSS; allo HSCT, allo hematopoietic stem cell transplantation.
Figure 2:

Treatment strategies for patients with myelodysplastic syndromes according to the International Prognostic Scoring System (IPSS), WHO Prognostic Scoring System (WPSS) and Revised IPSS (R-IPSS).

Intermediate-2, high or very high risk myelodysplastic patients have different treatment strategies based on patients’ characteristics and their elegibility for high intensity therapy. If they are candidate to bone marrow transplant and a donor is available, patients should be treated with allo hematopoietic stem cell transplantation. A bridging therapy should be considered in order to decrease marrow blasts to an acceptable level prior to transplant. High intensity chemotherapy (only in selected fit patients) or azacytdine/decitabine treatment should be considered for fit patients eligible for high intensity chemotherapy. Not high intensity therapy candidates should be treated with azacytdine/decitabine. Supportive care or clinical trials should be considered for non-responding or relapsing patients. IPSS, International Prognostic Scoring System; WPSS, World Health Organization Prognostic Scoring System; IPSS-R, Revised IPSS; allo HSCT, allo hematopoietic stem cell transplantation.

Conclusions and future directions

The central problem with MDS is their heterogeneity. MDS are a heterogeneous group of hematological diseases, with different clinical and prognostic features. The MDS are challenging for clinicians and pathologists due to the clinicopathologic heterogeneity of the disease and overlapping features with other benign and malignant disorders. Currently, the initial evaluation of a patient with suspected MDS focuses on a detailed medical history, review of the peripheral blood and bone marrow by an expert hematopathologist and risk stratification using laboratory results, morphology and cytogenetics. More sophisticated technologies, including multi-color flow cytometry, FISH, next-generation sequencing, and others are emerging and promise to offer significant refinements in diagnostic, prognostic and, hopefully, therapeutic information.

Since MDS range from indolent conditions with a long natural history to subtypes analogous to AML, clinical decision-making concerning treatment modalities and timing of interventions is challenging. Currently the prognosis of patients with MDS can be predicted 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 common used system was IPSS and tends to shift to IPSS-R. Although parameters such as hemoglobin level, blast count, and high-risk cytogenetic abnormalities will continue to retain strong independent prognostic value, the current era of genomics will provide us with additional parameters including new molecular markers, which may significantly contribute to a refined risk assessment of MDS and allow us to move towards a more patient-tailored therapeutic approach. Newer technologies with next-generation targeted deep sequencing and whole-genome and -exone sequencing have identified several recurrent mutations that play a pivotal role in the pathophysiology of MDS and the impact of these genetic changes on disease phenotype.

In recent years, several gene mutations have been identified among patients with MDS that may, at least partly, explain the clinical heterogeneity of the disease course and may influence prognosis. A large variety of gene mutations will be present in most patients with newly diagnosed MDS, including most patients with normal cytogenetics. Several studies examining large numbers of MDS bone marrow or peripheral blood samples have identified more than 40 recurrently mutated genes, with more than 80% of patients harboring at least one mutation.

Thus, the combined analysis of these gene mutations and the IPSS or IPSS-R may improve the risk stratification provided by these prognostic models alone.

Future molecular analysis could predict not only the risk of disease, but also the response to therapy allowing a molecular based tailored therapy. Other important goals include the determination of the clinical impact of all these mutations on response to therapy and MDS patients’ survival in large cohorts of patients. With the introduction of more sophisticated molecular techniques like gene expression profiling, it might become possible not only to predict the natural course of the disease, but also to identify patient populations that are prone to respond to specific drugs especially designed for specific genetic lesions. Sequencing-based studies suggest that multiple mutations may play a role in the progression of MDS to AML. Further work is necessary to understand the molecular basis of leukemic transformation in MDS syndromes.


Corresponding author: Roberto Castelli, MD, Department of Biomedical and Clinical Sciences Luigi Sacco, University of Milan, Luigi Sacco Hospital Milan, Milan, Italy, Phone: +39/02/50319830, Fax: +39/02/50319828, E-mail:

  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: None declared.

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Tefferi A, Vardiman JW. Myelodysplastic syndromes. N Engl J Med 2009;361:1872–85.10.1056/NEJMra0902908Search in Google Scholar

2. Cazzola M, Malcovati L. Myelodysplastic syndromes–coping with ineffective hematopoiesis. N Engl J Med 2005;352:536–8.10.1056/NEJMp048266Search in Google Scholar

3. Neukirchen J, Schoonen WM, Strupp C, Gattermann N, Aul C, Haas R, et al. Incidence and prevalence of myelodysplastic syndromes: data from the Düsseldorf MDS-registry. Leuk Res 2011;35:1591–6.10.1016/j.leukres.2011.06.001Search in Google Scholar

4. Adès L, Itzykson R, Fenaux P. Myelodysplastic syndromes. Lancet 2014;383:2239–52.10.1016/S0140-6736(13)61901-7Search in Google Scholar

5. Greenberg P, Cox C, Le Beau MM, Fenaux P, Morel P, Sanz G, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood 1997;89:2079–88.10.1182/blood.V89.6.2079Search in Google Scholar

6. Killick SB, Carter C, Culligan D, Dalley C, Das-Gupta E, Drummond M, et al. Guidelines for the diagnosis and management of adult myelodysplastic syndromes. Br J Haematol 2014;164:503–25.10.1111/bjh.12694Search in Google Scholar PubMed

7. Greenberg PL, Tuechler H, Schanz J, Sanz G, Garcia-Manero G, Solé F, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood 2012;120:2454–65.10.1182/blood-2012-03-420489Search in Google Scholar PubMed PubMed Central

8. Malcovati L, Germing U, Kuendgen A, Della Porta MG, Pascutto C, Invernizzi R, et al. Time-dependent prognostic scoring system for predicting survival and leukemic evolution in myelodysplastic syndromes. J Clin Oncol 2007;25:3503–10.10.1200/JCO.2006.08.5696Search in Google Scholar PubMed

9. Kantarjian H, O’Brien S, Ravandi F, Cortes J, Shan J, Bennett JM, et al. Proposal for a new risk model in myelodysplastic syndrome that accounts for events not considered in the original International Prognostic Scoring System. Cancer 2008;113:1351–61.10.1002/cncr.23697Search in Google Scholar PubMed PubMed Central

10. Naqvi K, Garcia-Manero G, Sardesai S, Oh J, Vigil CE, Pierce S, et al. Association of comorbidities with overall survival in myelodysplastic syndrome: development of a prognostic model. J Clin Oncol 2011;29:2240–6.10.1200/JCO.2010.31.3353Search in Google Scholar PubMed PubMed Central

11. Della Porta MG, Malcovati L, Strupp C, Ambaglio I, Kuendgen A, Zipperer E, et al. Risk stratification based on both disease status and extra-hematologic comorbidities in patients with myelodysplastic syndrome. Haematologica 2011;96:441–9.10.3324/haematol.2010.033506Search in Google Scholar

12. Morel P, Declercq C, Hebbar M, Bauters F, Fenaux P. Prognostic factors in myelodysplastic syndromes: critical analysis of the impact of age and gender and failure to identify a very-low-risk group using standard mortality ratio techniques. Br J Haematol 1996;94:116–9.10.1046/j.1365-2141.1996.6122056.xSearch in Google Scholar

13. Noël P, Solberg LA Jr. Myelodysplastic syndromes. Pathogenesis, diagnosis and treatment. Crit Rev Oncol Hematol 1992;12:193–215.10.1016/1040-8428(92)90054-TSearch in Google Scholar

14. Group Française de Morphologie Hématologique: French registry of acute leukemia and myelodysplastic syndromes. Cancer 1987;60:1385–94.10.1002/1097-0142(19870915)60:6<1385::AID-CNCR2820600637>3.0.CO;2-ISearch in Google Scholar

15. Zeidman A, Sokolover N, Fradin Z, Cohen A, Redlich O, Mittelman M. Platelet function and its clinical significance in the myelodysplastic syndromes. Hematol J 2004;5:234–8.10.1038/sj.thj.6200364Search in Google Scholar

16. Bacher U, Haferlach T, Kern W, Haferlach C, Schnittger S. A comparative study of molecular mutations in 381 patients with myelodysplastic syndrome and in 4130 patients with acute myeloid leukemia. Haematologica 2007;92:744–52.10.3324/haematol.10869Search in Google Scholar

17. Olney HJ, Le Beau MM. The cytogenetics and molecular Biology of myelodysplastic syndromes. In: Bennett JM, editors. The Myelodysplastic Syndromes. New York: Marcel Dekker, 2002:89.Search in Google Scholar

18. Haase D, Germing U, Schanz J, Pfeilstöcker M, Nösslinger T, Hildebrandt B, et al. New insights into the prognostic impact of the karyotype in MDS and correlation with subtypes: evidence from a core dataset of 2124 patients. Blood 2007;110:4385–95.10.1182/blood-2007-03-082404Search in Google Scholar

19. Zhang L, Rothman N, Wang Y. Increased aneusomy and long arm deletion of chromosomes 5 and 7 in the lymphocytes of Chinese workers exposed to benzene. Carcinogenesis 1998;19:1955–61.10.1093/carcin/19.11.1955Search in Google Scholar

20. Smith SH, Le Beau MM, Huo D, Karrison T, Sobecks RM, Anastasi J, et al. Clinical-cytogenetic associations in 306 patients with therapy-related myelodysplasia and myeloid leukemia: the University of Chicago series. Blood 2003;102:43–52.10.1182/blood-2002-11-3343Search in Google Scholar

21. Padron E, Komrokji R, List AF. Biology and treatment of the 5q- syndrome. Expert Rev Hematol 2011;4:61–9.10.1586/ehm.11.2Search in Google Scholar

22. Nimer SD, Golde DW. The 5q- abnormality. Blood 1987;70:1705–12.10.1182/blood.V70.6.1705.1705Search in Google Scholar

23. Mallo M, Del Rey M, Ibanez M, Calasanz MJ, Argellinas L, Larrayoz MY, et al. Response to lenalidomide in myelodysplastic syndromes with del (5q): influence of cytogenetics and mutations. Br J Haematol 2013;162:74–86.10.1111/bjh.12354Search in Google Scholar PubMed

24. Jadersten M, Saft L, Pellagatti A, Gohring G, Wainscoat JS, Boultwood J, et al. Clonal heterogeneity in the 5q-syndrome: p53 expressing progenitors prevail during lenalidomide treatment and expand at disease progression. Haematologica 2009;94:1762–6.10.3324/haematol.2009.011528Search in Google Scholar PubMed PubMed Central

25. Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood 2009;114:937–51.10.1182/blood-2009-03-209262Search in Google Scholar PubMed

26. Graubert T. Therapy-related myelodysplastic syndrome: models and genetics. Biol Blood Marrow Transplant 2010;16 (1 Suppl):S45–7.10.1016/j.bbmt.2009.10.019Search in Google Scholar PubMed PubMed Central

27. Tuzuner N, Cox C, Rowe JM, Watrous D, Bennett JM. Hypocellular myelodysplastic syndromes (MDS): new proposals. Br J Haematol 1995;91:612–7.10.1111/j.1365-2141.1995.tb05356.xSearch in Google Scholar PubMed

28. Sperr WR, Wimazal F, Kundi M, Baumgartner C, Nösslinger T, Makrai A, et al. Comorbidity as prognostic variable in MDS: comparative evaluation of the HCT-CI and CCI in a core dataset of 419 patients of the Austrian MDS Study Group. Ann Oncol 2010;21:114–9.10.1093/annonc/mdp258Search in Google Scholar PubMed

29. Papaemmanuil E, Cazzola M, Boultwood J, Malcovati L, Wyas P, Bowen D, et al. Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts. N Engl J Med 2011;365:1384–95.10.1056/NEJMoa1103283Search in Google Scholar PubMed PubMed Central

30. Yoshida K, Sanada M, Shiraishi Y, Nowak D, Nagata Y, Yamamoto R, et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 2011;478:64–9.10.1038/nature10496Search in Google Scholar PubMed

31. Malcovati L, Papaemmanuil E, Bowen DT, Boultwood J, Della Porta MG, Pascutto C, et al. Clinical significance of SF3B1 mutations in myelodysplastic syndromes andmyelodysplastic/myeloproliferative neoplasms. Blood 2011;118:6239–46.10.1182/blood-2011-09-377275Search in Google Scholar PubMed PubMed Central

32. Della Porta MG, Malcovati L, Boveri E, Travaglino E, Pietra D, Pascutto C, et al. Clinical relevance of bone marrow fibrosis and CD34-positive cell clusters in primary myelodysplastic syndromes. J Clin Oncol 2009 10;27:754–62.10.1200/JCO.2008.18.2246Search in Google Scholar

33. Bejar R, Stevenson KE, Caughey BA, Abdel Wahab O, Stensma DP, Gallilli N, et al. Validation of a prognostic model and the impact of mutations in patients with lower-risk myelodysplastic syndromes. J Clin Oncol 2012;30:3376–82.10.1200/JCO.2011.40.7379Search in Google Scholar

34. Thol F, Friesen I, Damm F, Yun H, Weissinger EM, Krauter J, et al. Prognostic significance of ASXL1 mutations in patients with myelodysplastic syndromes. J Clin Oncol 2011;29:2499–506.10.1200/JCO.2010.33.4938Search in Google Scholar

35. Kim DY, Lee JH, Park YH, Lee JH, Kim SD, Choi Y, et al. Feasibility of hypomethylating agents followed by allogeneic hematopoietic cell transplantation in patients with myelodysplastic syndrome. Bone Marrow Transplant 2012;47:374–9.10.1038/bmt.2011.86Search in Google Scholar

36. Patnaik MM, Itzykson R, Lasho TL, Kosmider O, Finke CM, Hanson CA, et al. ASXL1 and SETBP1 mutations and their prognostic contribution in chronic myelomonocytic leukemia: a two-center study of 466 patients. Leukemia 2014;28:2206–12.10.1038/leu.2014.125Search in Google Scholar

37. Bejar R, Stevenson K, Abdel-Wahab O, Galili N, Nilsson B, Garcia-Manero G, et al. Clinical effect of point mutations in myelodysplastic syndromes. N Engl J Med 2011;364:2496–6.10.1056/NEJMoa1013343Search in Google Scholar

38. Thol F, Kade S, Schlarmann C, Löffeld P, Morgan M, Krauter J, et al. Frequency and prognostic impact of mutations in SRSF2, U2AF1, and ZRSR2 in patients with myelodysplastic syndromes. Blood 2012;119:3578–84.10.1182/blood-2011-12-399337Search in Google Scholar

39. Malcovati L. Impact of transfusion dependency and secondary iron overload on the survival of patients with myelodysplastic syndromes. Leuk Res 2007;31(Suppl 3):S2–6.10.1016/S0145-2126(07)70459-9Search in Google Scholar

40. Mittelman M, Oster HS, Hoffman M, Neumann D. The lower risk MDS patient at risk of rapid progression. Leuk Res 2010;34:1551–5.10.1016/j.leukres.2010.05.023Search in Google Scholar PubMed

41. Kelaidi C, Park S, Sapena R, Beyne-Rauzy O, Coiteux V, Vey N, et al. Long-term outcome of anemic lower-risk myelodysplastic syndromes without 5q deletion refractory to or relapsing after erythropoiesis-stimulating agents. Leukemia 2013;27:1283–90.10.1038/leu.2013.16Search in Google Scholar PubMed

42. Ferrini PR, Grossi A, Vannucchi AM, Barosi G, Guarnone R, Piva N, et al. A randomized double-blind placebo-controlled study with subcutaneous recombinant human erythropoietin in patients with low-risk myelodysplastic syndromes. Br J Haematol 1998;103:1070–4.10.1046/j.1365-2141.1998.01085.xSearch in Google Scholar PubMed

43. Itzykson R, Thepot S, Quesnel B, Dreyfus F, Beyne-Rauzy O, Turlure P, et al. Prognostic factors for response and overall survival in 282 patients with higher-risk myelodysplastic syndromes treated with azacitidine. Blood 2011;117:403–11.10.1182/blood-2010-06-289280Search in Google Scholar PubMed

44. Castelli R, Cassin R, Cannavò A, Cugno M. Immunomodulatory drugs: new options for the treatment of myelodysplastic syndromes. Clin Lymphoma Myeloma Leuk 2013;13:1–7.10.1016/j.clml.2012.09.016Search in Google Scholar PubMed

45. Melchert M, List A. Targeted therapies in myelodysplastic syndrome. Semin Hematol 2008;45:31–8.10.1053/j.seminhematol.2007.10.001Search in Google Scholar PubMed

46. Park S, Grabar S, Kelaidi C, Beyne–Rauzy O, Picard F, Bardet V, et al. Predictive factors of response and survival in myelodysplastic syndrome treated with erythropoietin and G-CSF: the GFM experience. Blood 2008;111:574–82.10.1182/blood-2007-06-096370Search in Google Scholar PubMed

47. Jädersten M, Malcovati L, Dybedal I, Della Porta MG, Invernizzi R, Montgomery SM, et al. Erythropoietin and granulocyte-colony stimulating factor treatment associated with improved survival in myelodysplastic syndrome. J Clin Oncol 2008;26:3607–13.10.1200/JCO.2007.15.4906Search in Google Scholar PubMed

48. Greenberg PL, Sun Z, Miller KB, Bennet JM, Tallman MS, Dewald G, et al. Treatment of myelodysplastic syndrome patients with erythropoietin with or without granulocyte colony-stimulating factor: results of a prospective randomized phase 3 trial by the Eastern Cooperative Oncology Group (E1996). Blood 2009;114:2393–400.10.1182/blood-2009-03-211797Search in Google Scholar PubMed PubMed Central

49. Castelli R, Deliliers GL, Colombo R, Moreo G, Gallipoli P, Pantaleo G. Biosimilar epoetin in elderly patients with low-risk myelodysplastic syndromes improves anemia, quality of life, and brain function. Ann Hematol 2014;93:1523–9.10.1007/s00277-014-2070-8Search in Google Scholar PubMed

50. Villegas A, Arrizabalaga B, Fernández-Lago C, Castro M, Mayans JR, González-Porras JR, et al. Darbepoetin alfa for anemia in patients with low or intermediate-1 risk myelodysplastic syndromes and positive predictive factors of response. Curr Med Res Opin 2011;27:951–60.10.1185/03007995.2011.561834Search in Google Scholar PubMed

51. Santini V, Alessandrino PE, Angelucci E, Barosi G, Billio A, Di Maio M, et al. Italian Society of Hematology Clinical management of myelodysplastic syndromes: update of SIE, SIES, GITMO practice guidelines. Leuk Res 2010;34:1576–88.10.1016/j.leukres.2010.01.018Search in Google Scholar PubMed

52. Musto P, Falcone A, Sanpaolo G, Bodenizza C, La Sala A, Perla G, et al. Efficacy of a single, weekly dose of recombinant erythropoietin in myelodysplastic syndromes. Br J Haematol 200;122:269–71.10.1046/j.1365-2141.2003.04435.xSearch in Google Scholar PubMed

53. Gotlib J, Lavori P, Quesada S, Stein RS, Shahnia S, Greenberg A. Phase II intra-patient dose-escalation trial of weight-based darbepoetin alfa with or without granulocyte-colony stimulating factor in myelodysplastic syndromes. Am J Hematol 2009;84:15–20.10.1002/ajh.21316Search in Google Scholar PubMed PubMed Central

54. Mantovani L, Lentini G, Hentschel B, Wickramanayake PD, Loeffler M, Diehl V, et al. Treatment of anaemia in myelodysplastic syndromes with prolonged administration of recombinant human granulocyte colony-stimulating factor and erythropoietin. Br J Haematol 2000;109:367–75.10.1046/j.1365-2141.2000.02016.xSearch in Google Scholar PubMed

55. Raza A, Reeves JA, Feldman EJ, Dewald GW, Bennett JM, Deeg HJ, et al. Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. N Engl J Med 2006;355:1456–65.10.1056/NEJMoa061292Search in Google Scholar PubMed

56. Fenaux P, Giagounidis A, Selleslag D, Beyne-Rauzy O, Mufti G, Mittelman M, et al. A randomized phase 3 study of lenalidomide versus placebo in RBC transfusion-dependent patients with low-/intermediate-1-risk myelodysplastic syndromes with del5q. Blood 2011;118:3765–76.10.1182/blood-2011-01-330126Search in Google Scholar PubMed

57. Chang DH, Liu N, Klimek V, Hassoun H, Mazumder A, Nimer SD, et al. Enhancement of ligand-dependent activation of human natural killer T cells by lenalidomide: therapeutic implications. Blood 2006;108:618–21.10.1182/blood-2005-10-4184Search in Google Scholar PubMed PubMed Central

58. Chan AC, Neeson P, Leeansyah E, Tainton K, Quach H, Prince HM, et al. Testing the NKT cell hypothesis in lenalidomide- treated myelodysplastic syndrome patients. Leukemia 2010;24:592–600.10.1038/leu.2009.279Search in Google Scholar PubMed

59. Ximeri M, Galanopoulos A, Klaus M, Parcharidou A, Giannikou K, Psyllaki M, et al. Effect of lenalidomide therapy on hematopoiesis of patients with myelodysplastic syndrome associated with chromosome 5q deletion. Haematologica 2010;95:406–14.10.3324/haematol.2009.010876Search in Google Scholar PubMed PubMed Central

60. List A, Dewald G, Bennett J, Giagounidis A, Raza A, Feldman E, et al. Myelodysplastic syndrome-003 study investigators. Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. N Engl J Med 2006;355:1456–65.10.1056/NEJMoa061292Search in Google Scholar

61. Raza A, Reeves JA, Feldman EJ, Dewald GW, Bennet JM, Deeg HJ, et al. Phase 2 study of lenalidomide in transfusion-dependent, low-risk, and intermediate-1 risk myelodysplastic syndromes with karyotypes other than deletion 5q. Blood 2008;111:86–93.10.1182/blood-2007-01-068833Search in Google Scholar PubMed

62. Komrokji RS, Lancet JE, Swern AS, Chen N, Paleveda J, Lush R, et al. Combined treatment with lenalidomide and epoetin alfa in lower-risk patients with myelodysplastic syndrome. Blood 2012;120:3419–24.10.1182/blood-2012-03-415661Search in Google Scholar PubMed

63. Silverman LR, Demakos EP, Peterson BL, Kornblith AB, Holland JC, Odchimar-Reissig R, et al. Randomized controlled trial of azacitidine in patients with the myelodisplastic syndrome: a study of the cancer and leukemia group B. J Clin Oncol 2002;20:2429–40.10.1200/JCO.2002.04.117Search in Google Scholar PubMed

64. Passweg JR, Giagounidis AA, Simcock M, Aul C, Dobbelstein C, Stadler M, et al. Immunosuppressive therapy for patients with myelodysplastic syndrome: a prospective randomized multicenter phase III trial comparing antithymocyte globulin plus cyclosporine with best supportive care–SAKK 33/99. J Clin Oncol 2011;29:303–9.10.1200/JCO.2010.31.2686Search in Google Scholar PubMed

65. Molldrem JJ, Caples M, Mavroudis D, Plante M, Young NS, Barrett AJ. Antithymocyte globulin for patients with myelodysplastic syndrome. Br J Haematol 1997;99:699–705.10.1046/j.1365-2141.1997.4423249.xSearch in Google Scholar PubMed

66. Sloand EM, Wu CO, Greenberg P, Young N, Barrett J. Factors affecting response and survival in patients with myelodysplasia treated with immunosuppressive therapy. J Clin Oncol 2008;26:2505–11.10.1200/JCO.2007.11.9214Search in Google Scholar PubMed PubMed Central

67. Saunthararajah Y, Nakamura R, Wesley R, Wang QJ, Barrett AJ. A simple method to predict response to immunosuppressive therapy in patients with myelodysplastic syndrome. Blood 2003;102:3025–7.10.1182/blood-2002-11-3325Search in Google Scholar PubMed

68. List AF, Baer MR, Steensma DP, Raza A, Esposito J, Martinez-Lopez N, et al. Deferasirox reduces serum ferritin and labile plasma iron in RBC transfusion-dependent patients with myelodysplastic syndrome. J Clin Oncol 2012;30:2134–9.10.1200/JCO.2010.34.1222Search in Google Scholar PubMed

69. Pullarkat V. Objectives of iron chelation therapy in myelodysplastic syndromes: more than meets the eye? Blood 2009;114:5251–5.10.1182/blood-2009-07-234062Search in Google Scholar PubMed

70. Gattermann N, Porter J, Lopes L, Seymour J. Consensus statement on iron overload in myelodysplastic syndromes. Hematol Oncol Clin North Am 2005;19(Suppl 1):18–25.Search in Google Scholar

71. Greenberg PL. Myelodysplastic syndromes: iron overload consequences and current chelating therapies. J Natl Compr Canc Netw 2006;4:91–6.10.6004/jnccn.2006.0010Search in Google Scholar

72. Alessandrino EP, Della Porta MG, Bacigalupo A, Malcovati L, Angelucci E, Van Lint MT, et al. Prognostic impact of pre-transplantation transfusion history and secondary iron overload in patients with myelodysplastic syndrome undergoing allogeneic stem cell transplantation: a GITMO study. Haematologica 2010;95:476–84.10.3324/haematol.2009.011429Search in Google Scholar PubMed PubMed Central

73. Armand P, Kim HT, Cutler CS, Ho VT, Koreth J, Alyea EP, et al. Prognostic impact of elevated pre-transplant serum ferritin in patients undergoing myeloablative stem cell transplantation. Blood 2007;109:4586–8.10.1182/blood-2006-10-054924Search in Google Scholar PubMed PubMed Central

74. Alessandrino EP, Della Porta MG, Bacigalupo A, Van Lint MT, Falda M, Onida F, et al. WHO classification and WPSS predict post trans-plantation outcome in patients with myelodysplastic syndrome: a study from the Gruppo Italiano Trapianto di Midollo Osseo (GITMO). Blood 2008;112:895–902.10.1182/blood-2008-03-143735Search in Google Scholar PubMed

75. Negrin RS, Haeuber DH, Nagler A, Olds LC, Donlon T, Souza LM, et al. Treatment of myelodysplastic syndromes with recombinant human granulocyte colony-stimulating factor. A phase I-II trial. Ann Intern Med 1989;110:976–84.10.7326/0003-4819-110-12-976Search in Google Scholar PubMed

76. Negrin RS, Haeuber DH, Nagler A, Kobayashi Y, Sklar J, Donlon T, et al. Maintenance treatment of patients with myelodysplastic syndromes using recombinant human granulocyte colony-stimulating factor. PL Blood 1990;76:36–43.10.1182/blood.V76.1.36.36Search in Google Scholar

77. Sultana TA, Harada H, Ito K, Tanaka H, Kyo T, Kimura A. Expression and functional analysis of granulocyte colony-stimulating factor receptors on CD34++ cells in patients with myelodysplastic syndrome (MDS) and MDS-acute myeloid leukaemia. Br J Haematol 2003;121:63–75.10.1046/j.1365-2141.2003.04261.xSearch in Google Scholar PubMed

78. Economopoulos T, Papageorgiou E, Stathakis N, Asprou N, Karmas P, Dervenoulas J, et al. Treatment of myelodysplastic syndromes with human granulocytic-macrophage colony stimulating factor (GM-CSF) or GM-CSF combined with low-dose cytosine arabinoside. Eur J Haematol 1992;49:138–42.10.1111/j.1600-0609.1992.tb00918.xSearch in Google Scholar PubMed

79. Gerhartz HH, Marcus R, Delmer A, Zwierzina H, Suciu S, Dardenne M, et al. A randomized phase II study of low-dose cytosine arabinoside (LD-AraC) plus granulocyte-macrophage colony-stimulating factor (rhGM-CSF) in myelodysplastic syndromes (MDS) with a high risk of developing leukemia. EORTC Leukemia Cooperative Group. Leukemia 1994;8:16–23.Search in Google Scholar

80. Miller KB, Kim K, Morrison FS, Winter JN, Bennett JM, Neiman RS, et al. The evaluation of low-dose cytarabine in the treatment of myelodysplastic syndromes: a phase-III intergroup study. Ann Hematol 1992;65:162–8.10.1007/BF01703109Search in Google Scholar PubMed

81. Sekeres MA, Giagounidis A, Kantarjian H, Mufti GJ, Fenaux P, Jia C, et al. Development and validation of a model to predict platelet response to romiplostim in patients with lower-risk myelodysplastic syndromes. Br J Haematol 2014;167:337–45.10.1111/bjh.13037Search in Google Scholar PubMed

82. Platzbecker U, Sekeres MA, Kantarjian H, Giagounidis A, Mufti GJ, Jia C, et al. Relationship of different platelet response criteria and patient outcomes in a romiplostim myelodysplastic syndromes trial. Leukemia 2014;28:2418–21.10.1038/leu.2014.253Search in Google Scholar PubMed PubMed Central

83. Giagounidis A, Mufti GJ, Fenaux P, Sekeres MA, Szer J, Platzbecker U, et al. Results of a randomized, double-blind study of romiplostim versus placebo in patients with low/intermediate-1-risk myelodysplastic syndrome and thrombocytopenia. Cancer 2014;120:1838–46.10.1002/cncr.28663Search in Google Scholar PubMed PubMed Central

84. Kantarjian H, Fenaux P, Sekeres MA. Phase 1/2 study of AMG 531 in thrombocytopenic patients (pts) with low-risk myelodysplastic syndrome (MDS): update including extended treatment. ASH Annual Meeting Abstracts 2007;110:250.10.1182/blood.V110.11.250.250Search in Google Scholar

85. Knipp S, Hildebrand B, Kündgen A, Giagunidis A, Krobbe G, Haas R, et al. Intensive chemotherapy is not recommended for patients aged >60 years who have myelodysplastic syndromes or acute myeloid leukemia with high-risk karyotypes. Cancer 2007;110:345–52.10.1002/cncr.22779Search in Google Scholar

86. Kantarjian H, Beran M, Cortes J, O’Brien S, Giles F, Pierce S, et al. Long-term follow-up results of the combination of topotecan and cytarabine and other intensive chemotherapy regimens in myelodysplastic syndrome. Cancer 2006;106:1099–109.10.1002/cncr.21699Search in Google Scholar

87. Estey EH, Thall PF, Cortes JE, Giles FJ, O’Brien S, Pierce SA, et al. Comparison of idarubicin + ara-C-, fludarabine + ara-C-, and topotecan + ara-C-based regimens in treatment of newly diagnosed acute myeloid leukemia, refractory anemia with excess blasts in transformation, or refractory anemia with excess blasts. Blood 2001;98:3575–8.10.1182/blood.V98.13.3575Search in Google Scholar

88. Estey EH, Thall PF, Giles FJ, Wang XM, Cortes JE, Beran M, et al. Gemtuzumab ozogamicin with or without interleukin 11 in patients 65 years of age or older with untreated acute myeloid leukemia and high-risk myelodisplastic syndrome: comparison with idarubicin plus continuous-infusion, high-dose cytosine arabinoside. Blood 2002;99:4343–9.10.1182/blood.V99.12.4343Search in Google Scholar

89. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, Santini V, Finelli C, Giagounidis A, et al. Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. Lancet Oncol 2009;10:223–32.10.1016/S1470-2045(09)70003-8Search in Google Scholar

90. Leone G, Teofili L, Voso M, Lubbert M. DNA methylation and demethylating drugs in myelodysplastic syndromes and secondary leukemias. Haematologica 2002;87:1324–41.Search in Google Scholar

91. López-Karpovitch X, Barrales-Benítez O, Flores M, Piedras J. Effect of azacytidine in the release of leukemia inhibitory factor, oncostatin m, interleukin (IL)-6, and IL-11 by mononuclear cells of patients with refractory anemia. Cytokine 2002;20:154–62.10.1006/cyto.2002.1998Search in Google Scholar PubMed

92. Lübbert M, Suciu S, Baila L, Ruter BH, Platzbecker U, Giagounidis A, et al. Low-dose decitabine versus best supportive care in elderly patients with intermediate- or high-risk myelodysplastic syndrome (MDS) ineligible for intensive chemotherapy: final results of the randomized phase III study of the European Organisation for Research and Treatment of Cancer Leukemia Group and the German MDS Study Group. J Clin Oncol 2011;29:1987–96.10.1200/JCO.2010.30.9245Search in Google Scholar PubMed

93. Anderson JE, Litzow MR, Appelbaum FR, Schoch G, Fisher LD, Buckner CD. Allogeneic bone marrow transplantation for 93 patients with myelodysplastic syndrome. Blood 1993;82:677–81.10.1182/blood.V82.2.677.677Search in Google Scholar

94. O’Donnell MR, Long GD, Parker PM, Niland J, Nademanee A, Amylon M, et al. Busulfan/cyclophosphamide as conditioning regimen for allogeneic bone marrow transplantation for myelodysplasia. J Clin Oncol 1995;13:2973–9.10.1200/JCO.1995.13.12.2973Search in Google Scholar PubMed

95. Sierra J, Pérez WS, Rozman C, Carreras E, Klein JP, Davies SM, et al. Bone marrow transplantation from HLA-identical siblings as treatment for myelodysplasia. Blood 2002;100:1997–2004.Search in Google Scholar

96. Koreth J, Pidala J, Perez WS, Deeg HJ, Garcia Manero G, Malcovati L, et al. Role of reduced-intensity conditioning allogeneic hematopoietic stem-cell transplantation in older patients with de novo myelodysplastic syndromes: an international collaborative decision analysis. J Clin Oncol 2013;31:2662–700.10.1200/JCO.2012.46.8652Search in Google Scholar PubMed PubMed Central

97. Cutler CS, Lee SJ, Greenberg P, Deeg HJ, Perez WS, Anasetti C, et al. A decision analysis of allogeneic bone marrow transplantation for the myelodysplastic syndromes: delayed transplantation for low-risk myelodysplasia is associated with improved outcome. Blood 2004;104:579–85.10.1182/blood-2004-01-0338Search in Google Scholar PubMed

98. Anderson JE, Appelbaum FR, Schoch G, Gooley T, Anasetti C, Bensinger WI, et al. Allogeneic marrow transplantation for myelodysplastic syndrome with advanced disease morphology: a phase II study of busulfan, cyclophosphamide, and total-body irradiation and analysis of prognostic factors. J Clin Oncol 1996;14:220–6.10.1200/JCO.1996.14.1.220Search in Google Scholar PubMed

99. Sorror ML, Sandmaier BM, Storer BE, Maris MB, Baron F, Maloney DG, et al. Comorbidity and disease status based risk stratification of outcomes among patients with acute myeloid leukemia or myelodysplasia receiving allogeneic hematopoietic cell transplantation. J Clin Oncol 2007;25: 4246–54.10.1200/JCO.2006.09.7865Search in Google Scholar PubMed

100. Itzykson R, Kosmider O, Renneville A, Gelsi-Boyer V, Meggendorfer M, Morabito M, et al. Prognostic score including gene mutations in chronic myelomonocytic leukemia. J Clin Oncol 2013;31:2428–36.10.1200/JCO.2012.47.3314Search in Google Scholar PubMed

Received: 2015-8-14
Accepted: 2015-11-11
Published Online: 2016-1-14
Published in Print: 2016-9-1

©2016 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 25.4.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2015-0789/html
Scroll to top button