Open access peer-reviewed chapter

Myelodysplastic Syndromes: An Update on Pathophysiology and Management

Written By

Wanxing Chai-Ho and Gary J. Schiller

Submitted: 29 August 2018 Reviewed: 22 October 2018 Published: 27 November 2018

DOI: 10.5772/intechopen.82166

From the Edited Volume

Recent Developments in Myelodysplastic Syndromes

Edited by Ota Fuchs

Chapter metrics overview

1,885 Chapter Downloads

View Full Metrics

Abstract

Myelodysplastic syndromes (MDS) comprise a set of clonal hematopoietic stem cell (HSC) disorders characterized by ineffective hematopoiesis that manifest as cytopenia of variable severity. The result often is an increased risk of infection, transfusion dependence, and a potential to transform to acute myeloid leukemia (AML). For the past decade, hypomethylating agents remain the only FDA-approved therapy. Given that MDS is more prevalent in the elderly who often have comorbid conditions, supportive care remains the mainstay of therapy. Curative treatments are restricted to younger, healthy individuals with histocompatible-matched donors for allogeneic transplant able to tolerate more intensive chemotherapeutic treatment. Understanding of the pathophysiology of MDS advanced over the past decade, which leads to an increasing array of new agents under clinical investigation. This review focuses on our recent enhanced understanding of MDS molecular biology, and promising novel agents that go beyond the hypomethylating agent.

Keywords

  • myelodysplastic syndrome (MDS)
  • acute myeloid leukemia (AML)
  • bone marrow transplant
  • hypomethylating agent
  • somatic mutation

1. Introduction

The myelodysplastic syndromes (MDS) comprise an heterogeneous group of malignant hematopoietic stem cell disorders characterized by dysplastic and ineffective blood cell production and a variable risk of transformation to acute leukemia. Based on the United States Surveillance, Epidemiology, and End Results (SEER) Program, the incidence of MDS is about 4.1–4.6 cases per 100,000 population per year, with approximately 86% of patients aged ≥60 years at time of diagnosis (median age 76 years). The incidence rate is higher in men than women. [1] The prevalence is slightly lower in Europe with reported 1.24–3.7 cases per 100,000 population per year, also with observed male predominance.[2, 3] With an aging population and improved awareness of disease, it is likely that the number of new patients diagnosed with MDS each year will increase in the future.

Pathogenesis of MDS is incompletely understood. Studies have revealed age, male gender, alcohol, cigarette smoking, ionizing radiation, chemotherapy such as alkylating agents and topoisomerase II inhibitor, immunosuppressive therapy, viral infection, benzene and other environmental/occupational exposures as possible implicating factors. [4, 5, 6, 7, 8] However, disease caused by these risk factors are estimated to account for only 20–30% of cases, which are described as secondary MDS, with remainders as primary MDS [4]. The major subsets of secondary MDS are therapy-related MDS (t-MDS) and MDS with predisposition to familial myeloid neoplasm.

The risk for MDS and AML is increased in certain familial predisposition syndromes, such as inherited bone marrow failure disorders like Diamond-Blackfan syndrome, Fanconi anemia, dyskeratosis congenital, Shwachman-Diamond syndrome, and Down syndrome, Noonan syndrome/Noonan syndrome-like disorders and neurofibromatosis [9]. Accurate diagnosis and recognition of these syndromic disorders allows opportunities to improve clinical care. Genetic counseling should be offered to family members of affected individual. One should avoid using heterozygous sibling as bone marrow transplant donor. Recently, a growing number of germline mutations including CEBPA, DDX41, ANKRD26, ETV6, GATA2, RUNX1 were identified to associate with familial thrombocytopenia and development of MDS and acute leukemia in up to 40% of patients [10, 11]. Special attention should be noted that many patients with familial MDS and acute leukemia predisposition syndromes develop disease in adulthood rather than childhood. To increase awareness of this entity of disease, myeloid neoplasm with above mentioned germline predisposition was incorporated into the updated WHO 2016 classification [12].

Advertisement

2. Diagnosis

2.1. Clinical presentation

MDS usually presents as cytopenia in one or more lineage. Fatigue, dyspnea on exertion, infection, easy bruising or bleeding are the most common symptoms. Lymphadenopathy and hepatosplenomegaly are infrequent and should raise suspicion for chronic myelomonocytic leukemia (CMML) [13, 14]. It has been estimated that various autoimmune features such as subacute vasculitis, fever, arthritis, peripheral edema, and pulmonary infiltrates, may be present in up to 10% of patients [15, 16, 17, 18]. Certain autoimmune syndromes have correlated with distinct cytogenetic abnormalities; including Behcet’s disease with trisomy 8, Sweet’s syndrome and pyoderma gangrenosum with del(5q) [19]. Acquired hemoglobin H disease has been documented in approximately 8% of cases of MDS [20, 21, 22]. An acquired somatic mutation of ATRX, an X-linked gene encoding a chromatin-associated protein, has been linked to this entity, [21] as have acquired deletions of the alpha globin loci.

2.2. Pathology evaluation and WHO criteria

Bone marrow aspiration and biopsy are critical to the diagnosis of MDS. In general, the marrow is normo- or hypercellular due to ineffective hematopoiesis. However, up to 20% of MDS patients have hypocellular marrow, making it difficult to distinguish from aplastic anemia or paroxysmal nocturnal hematuria [23, 24]. Dysplastic neutrophils are commonly found in the peripheral blood smear. These cells may demonstrate reduced segmentation, increased size, the so-called pseudo-Pelger-Huet cell [25], often accompanied by reduced or absent granulation [26], and are associated with del(17)p [27]. Hypersegmentation with greater than 5 nuclear lobes is another feature of neutrophil dysplasia [28]. Red cells are usually normocytic or macrocytic, although ring sideroblasts, ovalomacrocytosis, teardrops, stomatocytes or acanthocytes may be seen [28]. Platelet morphology is usually normal, but micromegakaryocytes, mononuclear megakaryocytes, dumbbell-shaped nuclei, multinuclearity with multiple isolated nuclei (“Pawn ball” changes) may be seen [29].

Classification of MDS has been a challenge. In 1982, the French-American-British (FAB) Cooperative Group published the first seminal classification system that distinguished five subcategories of MDS based on marrow morphological criteria and myeloblasts proportions: refractory anemia, refractory anemia with ring sideroblasts (RARS), refractory anemia with excess of blasts (RAEB), RAEB “in transformation” (RAEB-T), and chronic myelomonocytic leukemia (CMML) [30]. Presence of more than 30% blasts in the bone marrow was defined as AML.

In 2001, World Health Organization (WHO) published new classification system on myeloid malignancy with modifications to the FAB system: The diagnosis of AML requires 20% myeloblasts. RAEB-T is classified as AML, and CMML is categorized as a new entity of myeloid neoplasms with both MDS and myeloproliferative features. In addition, MDS with isolated del(5q) is acknowledged as distinctive features in forms of disease with a low blast count, severe anemia and thrombocytosis (5q- syndrome) [31]. The revised 2008 WHO criteria maintained these modifications [32]. In the absence of definitive morphologic features of MDS, MDS-defining cytogenetic abnormalities were included in the diagnostic criteria (Table 1). The presence of chromosome 7, Y, or del(20q) does not meet criteria as an MDS-defining abnormality.

Unbalanced abnormalities Balanced abnormalities
−7 or del(7q) t(11;16)(q23;p13.3)
−5 or del(5q) t(3;21)(q26.2;q22.1)
i(17q) or t(17p) t(1;3)(p36.3;q21.1)
−13 or del(13q) t(2;11)(p21;q23)
del(11q) inv(3)(q21q26.2)
del(12p) or t(12p) t(6;9)(p23;q34)
del(9q)
idic(X)(q13)

Table 1.

Recurring chromosomal abnormalities considered as presumptive evidence of MDS in the setting of persistent cytopenia or undetermined origin in the absence of definitive morphologic features of MDS, according to World Health Organization 2008 and 2016 criteria.

Complex karyotype (3 or more chromosomal abnormalities) involving one or more of the above abnormalities.

The 2016 revision of WHO (Table 2) incorporated rapidly accumulating molecular genetic information into the classification [12]. The same cytogenetic abnormalities listed in the 2008 WHO classification remain MDS-defining in a cytopenic patient. Given recent data showing 1 chromosomal abnormality in addition to the del(5q) poses no adverse effect [33, 34, 35], the entity “5q- syndrome” may be diagnosed if there is 1 additional cytogenetic abnormality besides the del(5q), unless that abnormality is monosomy 7 or del(7q). Mutations like SF3B1, TET2, SRSF2, ASXL1, DNMT3A, RUNX1, U2AF1, TP53, and EZH2 can be found in 80–90% MDS patients [36, 37]. Importantly, acquired clonal mutations identical to those seen in MDS can occur in the hematopoietic cells of healthy older individuals without MDS, so-called “clonal hematopoiesis of indeterminate potential” (CHIP), or patients with mild cytopenia but without dysplastic changes or specific cytogenetic and/or genetic abnormalities considered as presumptive evidence of MDS (idiopathic cytopenia of undetermined significance, ICUS) [38, 39]. Although some CHIP and ICUS subsequently develop MDS, there have not been sufficient data to support using the presence of aforementioned mutations as surrogate diagnostic marker of MDS. Based on the link between ring sideroblasts and an SF3B1 mutation, MDS with ring sideroblasts and multilineage dysplasia, marked thrombocytosis, lacking excess blasts or an isolated del(5q) abnormality is included into the category of MDS with ring sideroblasts, and correlates with a favorable prognosis [40, 41, 42, 43].

Myeloproliferative neoplasms
Myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB, or FGFR1, or with PCM1-JAK2
Myelodysplastic/myeloproliferative neoplasms (MDS/MPN)
Chronic myelomonocytic leukemia(CMML)
Atypical chronic myeloid leukemia (aCML), BCR-ABL1
Juvenile myelomonocytic leukemia (JMML)
MDS/MPN with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T)
MDS/MPN, unclassifiable
Myelodysplastic syndrome
MDS with single lineage dysplasia
MDS with ring sideroblasts (MDS-RS)
MDS-RS and single lineage dysplasia
MDS-RS and multilineage dysplasia
MDS with multilineage dysplasia
MDS with excess blasts
MDS with isolated del(5q)
MDS, unclassifiable
Provisional entity: Refractory cytopenia of childhood
Myeloid neoplasms with germline predisposition
Acute myeloid leukemia (AML) and related neoplasms
Includes AML with myelodysplasia-related changes and therapy-related myeloid neoplasms
Blastic plasmacytoid dendritic cell neoplasm
Acute leukemias of ambiguous lineage
B-lymphoblastic leukemia/lymphoma
T-lymphoblastic leukemia/lymphoma

Table 2.

Classification of myeloid neoplasms and acute leukemia, according to World Health Organization 2016 criteria.

2.3. Differential diagnosis

MDS must be distinguished from other marrow dysplasia secondary to reversible causes, such as folate and vitamin B12 deficiency, viral infections (e.g. HIV), antibiotics, benzene, ethanol, or lead poisoning. Other primary bone marrow disorders presenting as pancytopenia, such as aplastic anemia, paroxysmal nocturnal hematuria, hairy cell leukemia, large granular lymphocytic leukemia can be distinguished by marrow morphology, flow cytometry features and gene mutation profile [8].

2.4. Risk stratification

The natural history of MDS in patients varies. The heterogeneity reflects both known and unknown differences in the pathophysiology of specific disease subtypes and patient related characteristics. Several prognostic scoring systems were developed and validated for MDS patients. In 1996, the International Prognostic Scoring System (IPSS) was developed by the International MDS Risk Analysis Workshop based on FAB classification [44]. Based on percent bone marrow blasts, specific cytogenetic abnormalities, and the number of cell lines involved with dysplasia and cytopenia, individual patient are placed into 4 groups: low, intermediate-1, intermediate-2, and high. The median survival in these four risk categories is 5.7 years for low risk, 3.5 years for intermediate-1 risk, 1.2 years for intermediate-2 risk, and 0.4 year for high risk.

In 2012, a revised IPSS (IPSS-R) was developed based upon data from 7012 patients with primary MDS diagnosed using the FAB or WHO classifications [45]. It incorporated new cytogenetic categories [35], and differentially weighed the degree of cytopenias in newly diagnosed patients. Patient age is an optional variable that can be incorporated to predict overall survival, but not evolution to AML. Individual patient was categorized into five risk groups: very low, low, intermediate, high and very high risk, that translates into median survival of 8.8, 5.3, 3.0, 1.6 and 0.8 years respectively. IPSS-R is simple to use, and is perhaps the most commonly used prognostication system today. However, there are several potential limitations to the IPSS-R. Both IPSS and IPSS-R were developed using data from patients who were observed without treatment. While outcomes might be different now that a variety of interventions are available, an analysis of a separate population suggested that the predictive value of the IPSS-R also applies to those treated with lenalidomide and azacitidine. [46] The prognosticating system only considered patients with de novo MDS. It is well recognized that patients with secondary MDS are more likely to have shorter survival. Much of this reflects the association between secondary MDS and “unfavorable cytogenetics”. In addition, the IPSS-R seems to be most reliable at predicting outcomes at initial disease diagnosis, as the hazards in mortality and leukemia transformation diminishes over time in higher-risk but remains stable in lower-risk patients [47]. With increasing knowledge of MDS clonal genetics, the future risk stratification system might incorporate the prognostic value of mutation profile, which will be discussed in the next section.

WHO prognostic scoring system (WPSS) was designed to include information on red blood cell (RBC) transfusion need and cytogenetic information [48]. A subset of patients in the study cohort had data from multiple time points for a time-dependent analysis, therefore had the advantage over the IPSS of being able to be used at any time during the disease course.

The MD Anderson Cancer Center (MADCC) MDS model was developed based on a retrospective analysis of 856 patients with de novo or therapy-related MDS [49]. Age, cytogenetics, degree of anemia and thrombocytopenia, bone marrow blast percentage were identified as prognostic markers. Subsequently it was prospectively validated in 1915 patients, accounted for the duration of MDS and prior therapy [49]. One should take note that the MDACC model should only be applied to the population of patients with lower-risk (low or intermediate-1 IPSS) MDS, and patients who received various of MDS treatment, from which it was derived [50].

Advertisement

3. Pathogenesis

The pathogenesis of MDS is considered as a multistep process involving sequential acquisition of oncogenic mutations [51, 52]. The interplay between genetically altered HSCs and an abnormal bone marrow microenvironment may allow for selection of a predominant dysplastic clone [51, 52, 53, 54, 55, 56].

3.1. Clonal heterogeneity and evolution

MDS is driven by a multistep process characterized by recurrent mutations affecting basic cellular pathways, including RNA splicing, epigenome regulation, myeloid transcription coordination, DNA damage response and growth factor signaling. It has been long recognized that HSC with certain pathogenic alterations has a competitive advantage and drives clonal expansion at the stem cell level. Clonal cytogenetic abnormalities are detected in up to 50% of de novo MDS cases and 80% of therapy-related cases [57, 58]. Over the past decade, a number of large, MDS-focused sequencing studies further demonstrated that MDS is a genetically complex and heterogeneous disease [36, 37, 42, 59, 60, 61].

However, clonality alone is not sufficient to cause or diagnose disease, because increased clonal hematopoiesis can remain functionally intact [38, 39]. Recently published data on a large cohort of cytopenia (ICUS) patients delineated the natural history of patients with clonal vs. nonclonal cytopenia [62]. Patients with clonal ICUS had a much higher rate of progression than patient with nonclonal ICUS. Spliceosome gene mutations such as SF3B1 SRSF2 and U2AF1 and co-mutation patterns involving TET2, DNMT3A or ASXL1 had clinical characteristics resemble low-risk MDS patients, and higher progression to myeloid neoplasm when comparing with patient with somatic TET2, DNMT3A and ASXL1 mutation alone [39].

The diversity of clinical MDS phenotypes associated with specific mutations may be related to differential coregulation of the HSC self-renewal and lineage-specific differentiation capacity. Accurate prediction of the natural history of individual patient is certainly of high clinic interest. Our growing knowledge suggests that individual mutations occur in highly stereotyped order and strong patterns of co-mutation association and exclusivity (Figure 1) [36, 60, 63]. Mutations affecting epigenetic modifier genes (DNMT3A, TET2, ASXL1, EZH2, etc.) or RNA spliceosome components (SF3B1, SRSF2, and U2AF1) tend to arise in the initiation and early progression phase of MDS and rarely occur at the time of transformation. By contrast, mutations in growth factor signaling pathways (NRAS, KRAS, PTPN11, FLT3, etc.) are rarely found in early phase of disease, and instead, they are frequently acquired and expanded in subclones at time of progression to high-grade MDS or secondary AML [63, 64, 65, 66]. A recent study [67] suggested that at the time of secondary AML transformation, the founding clone persisted at high variant allele fraction, but there was selective emergence and dominance of at least one genetically distinct subclone. In t-MDS, mutations in PPM1D or TP53 were present in 46% of patients, and they were the only gene mutations that were significantly associated with t-MDS [63, 68, 69, 70, 71].

Figure 1.

Gene mutations have stereotyped positions in the MDS clonal hierarchy. Recent knowledge suggests that individual mutations occurs in highly stereotyped order and strong patterns of co-mutation association and exclusivity (Mutations affecting epigenetic modifier genes (DNMT3A, TET2, ASXL1, EZH2, etc.) or RNA spliceosome components (SF3B1, SRSF2, and U2AF1) tend to arise in the initiation and early progression phase of MDS and rarely occur at the time of transformation. Mutations in growth factor signaling pathways (NRAS, KRAS, PTPN11, FLT3, etc.) are frequently acquired and expanded in subclones at time of progression to high-grade MDS or secondary AML.

Various studies have also assessed the value of risk stratification based on MDS mutation profile [36, 42, 72]. TP53, ETV6, ASXL1, EZH2 and RUNX1 mutation confers adverse outcomes that are independent of IPSS risk assessment. SF3B1, which is frequently mutated in patients with ring sideroblasts, is associated with distinct and favorable clinical features [37, 38, 40].

3.2. Bone marrow microenvironment

HSC and genetically altered HSC all reside in a highly complex and dynamic cellular microenvironment in the bone marrow, that is composed of endothelial cells, multipotent mesenchymal stem cells, and sympathetic nerve fibers. There have been many in vivo studies demonstrated the concept of niche-induced disease initiation of MDS [73, 74] or AML [75, 76]. Evidence to support this in humans is mainly based on the not-so-rare occurrence of donor-derived leukemia in bone marrow transplant recipients, where changes in the preexisting niche in the host is thought to be leukemogenic [77]. In the review by Pleyer et al., [78] a variety of functional and molecular alterations were observed in ex vivo expanded mesenchymal stromal cells from MDS and AML patients, including their differentiation potential and HSC supportive activities, as well as chromosomal aberrations, transcriptional, and epigenetic changes. In vivo evidence also suggested that endothelial cells-specific gene alterations causes myeloproliferative disorders [79, 80].

3.3. Dysregulated immune pathways

Regulators of inflammation and innate immunity have always been thought to play an important role in pathogenesis of malignancies, but only until recent have the specific immune effectors and their cell-intrinsic functional roles in MDS stem cell biology been elucidated [81, 82]. Therapeutic targeting of over-activated innate immune components such as Toll-like receptors [83], IL-1 receptor–associated kinase/tumor necrosis factor receptor–associated factor-6 [84], IL8/CXCR2 [85], and IL1RAP [86] signaling pathways in MDS HSCs is being attempted pre-clinically.

Advertisement

4. Treatment

Treatment for MDS is guided by clinical symptoms, disease risk classification, patient age, comorbidities and performance status. Supportive care with transfusion and timely treatment of infection with antibiotics are important adjuncts for all MDS patients. Incorporating iron chelation therapy for patients requiring chronic transfusion and all candidates for allogeneic stem cell transplant is being increasingly emphasized to prevent cardiac and liver damage from iron overload [87, 88]. Pharmacologic treatment is usually reserved for symptomatic patients. Treatment goal for lower-risk MDS patients is to minimize symptoms, improve quality of life, and avoid toxicity from therapy. Erythropoiesis stimulating agent (ESA) can be used for symptomatic anemia and a low serum erythropoietin [89, 90, 91, 92]. Together with low-dose G-CSF, hemoglobin improvement can be seen in up to 40% of lower-risk patients [93, 94]. Immunosuppressive therapy with antithymocyte globulin and cyclosporine A can produce response in a selected subset of patients. Those most likely to benefit are younger than 60 years, with less than 5% bone marrow blasts, hypoplastic MDS, presence of a paroxysmal nocturnal hemoglobinuria clone, trisomy 8, human leukocyte antigen DR15 positive, and with short duration of transfusion dependence [95]. Low-dose lenalidomide at 10 mg daily is FDA-approved for lower-risk MDS characterized as the 5q- syndrome. Transfusion independence was achieved in 67% of patients in the phase 2 trial [96], and 56% in the phase 3 trial [97]. For ESA refractory lower-risk MDS patients without 5q- syndrome, lenalidomide in combination with ESA also demonstrated efficacy at reducing transfusion need [98, 99, 100]. So far, the only FDA approved therapies for higher-risk MDS are the HMAs azacitidine and decitabine. The use of these agents results in complete (CR) and partial response (PR) each in 10–20% patients, with median duration of response about 10 months [101, 102, 103, 104]. An additional 20–30% patients achieve hematologic improvement without an objective response. Despite survival benefit demonstrated with azacitidine in high-risk patients [101], HMAs are not curative. For young and fit patients, allogeneic stem cell transplant is the only curative treatment option. Therefore, there remains a significant unmet therapeutic need beyond HMAs. Novel agents under clinical investigation and the use of allogeneic stem cell transplant will be discussed here.

4.1. Next-generation hypomethylating agents

HMAs are S-phase specific. Conventional HMAs all have a very short half-lives (less than 30 min) due to rapid clearance of azanucleoside by cytidine deaminase. The focus of newer generation HMA development has been to meet the need of prolonged drug exposure, allowing greater drug incorporation into DNA.

Oral film-coated azacitidine (CC-486) was first studied in an open-label pilot trial. It demonstrated 17% mean bioavailability after a single dose at 80 mg [105]. In a subsequent phase 1 dose finding study in MDS, CMML and AML patients, overall response rate was 73% in previously untreated patients, and 35% in previously treated patients [106]. Extended dosing schedule of CC-486 for 14 or 21 days is being evaluated in a phase 3 trial (NCT01566695) in lower-risk MDS [107]. CC-486 is also being studied in combination with immune check point inhibitor in the second line setting (NCT02281084).

ASTX727 is a novel formulation of oral decitabine paired with an oral cytidine deaminase inhibitor E7727 to overcome the rapid clearance from cytidine deaminase in gut and liver. In the early phase studies with intermediate- or high-risk MDS, ASTX727 (35 mg decitabine, 100 mg E7227) successfully emulated the pharmacokinetic profile of intravenous decitabine [108, 109]. In the phase 2 trial, clinical benefit was observed in 62% patients, with 16% CR, 28% marrow complete response (mCR), and 18% hematologic improvement [109].

Another strategy to circumvent the rapid degradation of azanucleotide is to develop a novel formulation that is relatively resistant to cytidine deamination. Guadecitabine (SGI-110) is a novel dinucleotide of decitabine and deoxyguanosine, linked by a phosphodiester bond, that leads to a slower release of the active decitabine moiety, prolonging cellular exposure to the drug [110]. In the phase 2 study with guadecitabine in intermediate and high risk MDS and CMML patients, CR was observed in 7/49 treatment naïve patients (14%) while CR + mCR were observed in 11/53 previously treated patients (21%) [111].

4.2. Histone deacetylase inhibition

Both DNA-promoter hypomethylation as well as post-translational modification of histone tails (e.g., deacetylation) lead to transcriptional silencing of tumor-suppressor genes and genes involved in differentiation and apoptosis [112, 113]. Histone deacetylase inhibitors (HDACi) have limited single-agent efficacy in both high risk MDS and AML [114, 115, 116]. Preclinical evidence supported synergy between HMAs and HDACi [117]. However, a few phase 2 randomized clinical trials failed to demonstrate improvement in response rates or survival when azacitidine was combined with HDACi entinostat, vorinostat, valproic acid, or pracinostat [118, 119, 120, 121, 122]. Currently, a few clinical trials in MDS are ongoing using HDACi in combination with other novel agents such as immune checkpoint inhibitors (NCT 02936752) or pracinostat in combination with azacitidine using different dosing scheme (NCT 03151304). At this moment, how to best incorporate HDACi in MDS treatment remains uncertain.

4.3. Other epigenetic modification agents

Beyond targeting DNA methylation and HDAC recruitment, there has also been an increasing effort to develop epigenetic modification agents targeting posttranslation or posttranscription pathways, to mitigate malignant myeloid transformation in MDS.

Bromodomain and extraterminal (BET) proteins are epigenetic readers that recognize acetylated lysine tails of histones, and thus areas of open chromatin structure. It has been suggested that AML relies on BET protein BRD4 [123, 124], therefore led to great interest in utilizing BET inhibitors in myeloid malignancy. Various clinical trials are investigating the use of JQ1, the first selective BET inhibitor, in myeloid malignancy including MDS (NCT 02158858, NCT 02308761).

Overexpression of the mono and dimethyl lysine demethylase, LSD1 has been implicated in myeloid malignancies [125]. Clinical trials are ongoing evaluating LSD1 inhibitors in combination with ATRA or HMA in previously treated AML and MDS patients (NCT02273102, NCT02717884, NCT02929498).

4.4. Immune checkpoint inhibition

Upregulation of immune checkpoint molecules like PD-1/PDL-1 and CTLA4 is commonly observed in many malignancies, including AML and MDS [126, 127] to evade immune surveillance. However, preliminary experience suggested limited activity of immune checkpoint inhibitor use as single agent after HMA failure in MDS patients [128]. Several clinical trials are ongoing evaluating the efficacy of immune checkpoint inhibitors plus HMAs or HDACis (NCT02530463, NCT03092674, NCT02775903, NCT03094637, NCT02599649).

4.5. Other targeted therapies: extrapolating experience from AML

Based on the mutation profile, FLT3 inhibitor and IDH1/2 inhibitors are now FDA approved for AML. However, these mutations are less common in MDS [129]. The early phase ½ studies of IDH1 and IDH2 inhibitors included MDS patients, with reported response [130, 131]. Especially given their tolerability profile and single agent activity, these agents deserves further investigation in MDS.

Spliceosome mutations, such as SF3B1, SRSF2 and U2AF1 are the most common mutations in MDS [37]. Based on the encouraging activity in preclinical study [132], there is now a phase 1 study in myeloid malignancies including MDS, with splicing modulator H3B-8800, an oral modulator of the SF3B complex (NCT02841540).

Venetoclax, a selective BCL-2 inhibitor was granted breakthrough designation by FDA in combination with decitabine in 2017 for treatment–naive AML patients age greater than 65 years. This decision was based on result from two ongoing phase ½ clinical trials [133]. This combination is now being evaluated in higher-risk MDS in both frontline and HMA failure settings (NCT02966782, NCT02942290).

4.6. Management of anemia in lower-risk MDS

Luspatercept and sotatercept are modified activin receptor type II (ActRII) chimeric fusion proteins that consist of the modified extracellular domain of ActRIIB and ActRIIA respectively, trap TGF-β superfamily ligands to promote late-stage erythropoiesis [134, 135]. In the phase 2 trial of luspatercept for patients with lower-risk MDS who were ineligible for or refractory to ESAs, RBC transfusion independence was seen in 38% patients, and 63% hematologic improvement [136]. Similar efficacy was seen in the phase 2 trial for sotatercept, with 47% hematologic response in patients with high transfusion burden, and 58% with low transfusion burden [137]. Ongoing phase 3 clinical trial is evaluating the efficacy of ActRII antagonist in lower-risk MDS and MDS with ring sideroblasts who require regular RBC transfusions (NCT 02631070).

Rigosertib is a PI3K and polo-like kinase pathways small-molecule inhibitor. In the recent phase 2 study for transfusion-dependent lower-risk MDS patients, 20 of 62 (32%) patients achieved transfusion independence lasting for more than 8 weeks [138]. Validation of these results in future clinical trials is anticipated.

Roxadustat is a drug which acts as a HIF prolyl-hydroxylase inhibitor and thereby increases endogenous production of erythropoietin, which stimulates production of hemoglobin and RBCs. Roxadustat is shown to be safe and effective as anemia treatment for patient with underlying chronic kidney disease, not on dialysis [139]. A phase 3 trial is ongoing to evaluate the efficacy of roxadustat in low-risk MDS patients with low transfusion burden (NCT03263091).

4.7. Allogeneic stem cell transplant

Allogeneic stem cell transplant is the only curative therapy for MDS, but restricted to younger and fit patients. Disease free survival rates are approximately 30–50%. Treatment failure is attributed by transplant-related mortality in low-risk patients, and relapse in higher-risk patients [140]. In general, bone marrow transplant is offered to intermediate-2 and high-risk MDS patients. Over the past decade, reduced-intensity conditioning transplant made more older patients eligible for transplant [141]. An ongoing clinical trial is comparing the efficacy of reduced intensity allogeneic stem cell transplant to HMA in patients aged 50–75 with higher-risk disease [142]. In the study by Della Porta et al. [143], IPSS-R was prognostic for outcomes of patients in the high and very high-risk groups, but not in the low- and intermediate-risk groups.

There have been emerging data on the prognostic value of mutation profile and minimal residual disease pre- and post-transplantation. It was shown that only a minority of patients with MDS was in deep hematologic remission by flow cytometry minimal residual disease (MRD) and cytogenetic analysis before transplant [144]. For myeloablative conditioning, MRD positive and MRD negative patients had similar post-transplant outcome. However, relapse rate was higher for MRD positive patient who received non-myeloablative conditioning. Multiple studies have shown that TP53 mutation is an independent marker for short survival post-transplant [59, 61, 145]. EZH2, ETV6, RUNX1, ASXL1, JAK2, and mutations in the RAS signaling pathway have all been implicated to associate with short relapse-free interval post-transplant [59, 61, 145, 146].

Advertisement

5. Conclusion

Over the past decade, knowledge was gained in understanding the pathogenesis of MDS. However, many gaps remain to change the natural history of MDS. With increasing number of novel treatments under investigation, it is likely that we are getting closer to more therapeutics options for MDS in the near future.

Advertisement

Conflict of interest

Wanxing Chai-Ho reports no conflict-of-interest.

Gary Schiller receives research funding from Forma, Janssen, Onconova, Pfizer, Incyte and BioMed Valley Discoveries, and receives consulting fees from Agios, Amgen, Astellas, Bioline RX, Celgene, Gamida, Fanssen, Karyopharm, Leukemia Lymphoma Society, Medallion, Medeor, Merck, Pharmacyclics, Sanofi, Seattle Genetics, Spectrum, Stanford University Health, Sunesis, and Takeda.

References

  1. 1. Ma X. Epidemiology of myelodysplastic syndromes. The American Journal of Medicine. 2012;125(7 Suppl):S2-S5. DOI: 10.1016/j.amjmed.2012.04.014
  2. 2. Sant M, Allemani C, Tereanu C, et al. HAEMACARE Working GroupIncidence of hematologic malignancies in Europe by morphologic subtype: Results of the HAEMACARE project. Blood. 2010;116(19):3724-3734. DOI: 10.1182/blood-2010-05-282632
  3. 3. Smith A, Howell D, Patmore R, et al. Incidence of haematological malignancy by sub-type: A report from the Haematological Malignancy Research Network. British Journal of Cancer. 2011;105(11):1684-1692. DOI: 10.1038/bjc.2011.450
  4. 4. Finch SC. Myelodysplasia and radiation. Radiation Research. 2004;161:603-606
  5. 5. Nisse C, Haguenoer JM, Grandbastien B, et al. Occupational and environmental risk factors of the myelodysplastic syndromes in the North of France. British Journal of Haematology. 2001;112:927-935
  6. 6. Mundle S, Allampallam K, Rashid KA, et al. Presence of activation-related m-RNA for EBV and CMV in the bone marrow of patients with myelodysplastic syndromes. Cancer Letters. 2001;164:197-205
  7. 7. Dalamaga M, Petridou E, Cook FE, et al. Risk factors for myelodysplastic syndromes: A case-control study in Greece. Cancer Causes & Control. 2002;13:603-608
  8. 8. Catenacci DV, Schiller GJ. Myelodysplasic syndromes: A comprehensive review. Blood Reviews. 2005;19(6):301-319. DOI: 10.1016/j.blre.2005.01.004
  9. 9. Owen C, Barnett M, Fitzgibbon J. Familial myelodysplasia and acute myeloid leukaemia—A review. British Journal of Haematology. 2008;140:123-132. DOI: 10.1111/j.1365-2141.2007.06909.x
  10. 10. West AH, Godley LA, Churpek JE. Familial myelodysplastic syndrome/acute leukemia syndromes: A review and utility for translational investigations. Annals of the New York Academy of Sciences. 2014;1310:111-118. DOI: 10.1111/nyas.13543
  11. 11. Babushok DV, Bessler M, Olson TS. Genetic predisposition to myelodysplastic syndrome and acute myeloid leukemia in children and young adults. Leukemia & Lymphoma. 2016;57(3):520-536. DOI: 10.3109/10428194.2015.1115041
  12. 12. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20):2391-2405. DOI: 10.1182/blood-2016-03-643544
  13. 13. Cortes J. CMML: A biologically distinct myeloproliferative disease. Current Hematology Reports. 2003;2(3):202-208
  14. 14. Bennett J. The myelodysplastic/myeloproliferative disorders: The interface. Hematology/Oncology Clinics of North America. 2003;17(5):1095-1100
  15. 15. Enright H, Jacob H, Vercellotti G, et al. Paraneoplastic autoimmune phenomena in patients with myelodysplastic syndromes: Response to immunosuppressive therapy. British Journal of Haematology. 1995;91(2):403-408
  16. 16. Saif MW, Hopkins JL, Gore SD. Autoimmune phenomena in patients with myelodysplastic syndromes and chronic myelomonocytic leukemia. Leukemia and Lymphoma. 2002;43(11):2083-2092
  17. 17. Giannouli M, Voulgarelis M, Zintzaras E, et al. Autoimmune phenomena in myelodysplastic syndromes: A 4-yr prospective study. Rheumatology. 2004;43:626-632. DOI: 10.1093/rheumatology/keh136
  18. 18. Wolach O, Stone R. Autoimmunity and inflammation in myelodysplastic syndromes. Acta Haematologica. 2016;136(2):108. DOI: 10.1159/000446062
  19. 19. Lee S, Park J, Lee E, et al. Certain autoimmune manifestations are associated with distinctive karyotypes and outcomes in patients with myelodysplastic syndrome: A retrospective cohort study. Medicine (Baltimore). 2016;95(13):e3091. DOI: 10.1097/MD.0000000000003091
  20. 20. Steensma DP, Higgs DR, Fisher CA, et al. Acquired somatic ATRX mutations in myelodysplastic syndrome associated with alpha thalassemia (ATMDS) convey a more severe hematologic phenotype than germline ATRX mutations. Blood. 2004;103(6):2019. DOI: 10.1182/blood-2003-09-3360
  21. 21. Steensma DP, Porcher JC, Hanson CA, et al. Prevalence of erythrocyte haemoglobin H inclusions in unselected patients with clonal myeloid disorders. British Journal of Haematology. 2007;139(3):439. DOI: 10.1111/j.1365-2141.2007.06831.x
  22. 22. Higgs DR. Gene regulation in hematopoiesis: New lessons from thalassemia. Hematology. American Society of Hematology. Education Program. 2004;2004(1):1-13. DOI: 10.1182/asheducation-2004.1.1
  23. 23. Wong KF, So CC. Hypoplastic myelodysplastic syndrome—A clinical, morphologic, or genetic diagnosis. Cancer Genetics and Cytogenetics. 2002;138:85-88
  24. 24. Sloand EM. Hypocellular myelodysplasia. Hematology/Oncology Clinics of North America. 2009;23(2):347-360. DOI: 10.1016/j.hoc.2009.01.015
  25. 25. Kuriyama K, Tomonaga M, Matsuo T, et al. Diagnostic significance of detecting pseudo-Pelger-Huët anomalies and micro-megakaryocytes in myelodysplastic syndrome. British Journal of Haematology. 1986;63(4):665
  26. 26. Hast R, Nilsson I, Widell S, Ost A. Diagnostic significance of dysplastic features of peripheral blood polymorphs in myelodysplastic syndromes. Leukemia Research. 1989;13(2):173-178
  27. 27. Soenen V, Preudhomme C, Roumier C, et al. 17p deletion in acute myeloid leukemia and myelodysplastic syndrome: Analysis of breakpoints and deleted segments by fluorescence in situ. Blood. 1998;91:1008-1015
  28. 28. Giagounidis A, Haase D. Morphology, cytogenetics and classification of MDS. Best Practice & Research. Clinical Haematology. 2013;26(4):337-353. DOI: 10.1016/j.beha.2013.09.004
  29. 29. Wong KF, Chan JK. Are ‘dysplastic’ and hypogranular megakaryocytes specific markers for myelodysplastic syndrome? British Journal of Haematology. 1991;77(4):509
  30. 30. Bennett JM, Catovsky D, Daniel MT, et al. Proposals for the classification of the myelodysplastic syndromes. British Journal of Haematology. 1982;51:189-199
  31. 31. Vardiman JW, Brunning RD, Harris NL. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood. 2002;100(7):2292-2302. DOI: 10.1182/blood-2002-04-1199
  32. 32. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization classification of myeloid neoplasms and acute leukemia: Rationale and important changes. Blood. 2009;114:937-951. DOI: 10.1182/blood-2009-03-209262
  33. 33. Germing U, Lauseker M, Hildebrandt B, et al. Survival, prognostic factors and rates of leukemic transformation in 381 untreated patients with MDS and del(5q): A multicenter study. Leukemia. 2012;26(6):1286-1292. DOI: 10.1038/leu.2011.391
  34. 34. Mallo M, Cervera J, Schanz J, et al. Impact of adjunct cytogenetic abnormalities for prognostic stratification in patients with myelodysplastic syndrome and deletion 5q. Leukemia. 2011;25(1):110-120. DOI: 10.1038/leu.2010.231
  35. 35. Schanz J, Tüchler H, Solé F, et al. New comprehensive cytogenetic scoring system for primary myelodysplastic syndromes (MDS) and oligoblastic acute myeloid leukemia after MDS derived from an international database merge. Journal of Clinical Oncology. 2012;30(8):820-829. DOI: 10.1200/JCO.2011.35.6394
  36. 36. Papaemmanuil E, Gerstung M, Malcovati L, et al. Clinical and biological implications of driver mutations in myelodysplastic syndromes. Blood. 2013;122(22):3616-3627. DOI: 10.1182/blood-2013-08-518886
  37. 37. Haferlach T, Nagata Y, Grossmann V, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia. 2014;28(2):241-247. DOI: 10.1038/leu.2013.336
  38. 38. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. The New England Journal of Medicine. 2014;371(26):2488-2498. DOI: 10.1056/NEJMoa1408617
  39. 39. Genovese G, Kähler AK, Handsaker RE, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. The New England Journal of Medicine. 2014;371(26):2477-2487. DOI: 10.1056/NEJMoa1409405
  40. 40. Malcovati L, Papaemmanuil E, Bowen DT, et al. Clinical significance of SF3B1 mutations in myelodysplastic syndromes and myelodysplastic/myeloproliferative neoplasms. Blood. 2011;118(24):6239-6246. DOI: 10.1182/blood-2011-09-377275
  41. 41. Papaemmanuil E, Cazzola M, Boultwood J, et al. Somatic SF3B1 mutation in myelodysplasia with ring sideroblasts. The New England Journal of Medicine. 2011;365(15):1384-1395. DOI: 10.1056/NEJMoa1103283
  42. 42. Bejar R, Stevenson KE, Caughey BA, et al. Validation of a prognostic model and the impact of mutations in patients with lower-risk myelodysplastic syndromes. Journal of Clinical Oncology. 2012;30(27):3376-3382. DOI: 10.1200/JCO.2011.40.7379
  43. 43. Cazzola M, Rossi M, Malcovati L, et al. Biologic and clinical significance of somatic mutations of SF3B1 in myeloid and lymphoid neoplasms. Blood. 2013;121(2):260-269. DOI: 10.1182/blood-2012-09-399725
  44. 44. Greenberg P, Cox C, LeBeau MM, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89(6):2079-2088
  45. 45. Greenberg PL, Tuechler H, Schanz J, et al. Revised international prognostic scoring system for myelodysplastic syndromes. Blood. 2012;120(12):2454. DOI: 10.1182/blood-2012-03-420489
  46. 46. Voso MT, Fenu S, Latagliata R, et al. Revised international prognostic scoring system (IPSS) predicts survival and leukemic evolution of myelodysplastic syndromes significantly better than IPSS and WHO prognostic scoring system: Validation by the Gruppo Romano Mielodisplasie Italian Regional Database. Journal of Clinical Oncology. 2013;31(21):2671. DOI: 10.1200/JCO.2012.48.0764
  47. 47. Pfeilstöcker M, Tuechler H, Sanz G, et al. Time-dependent changes in mortality and transformation risk in MDS. Blood. 2016;128(7):902-910. DOI: 10.1182/blood-2016-02-700054
  48. 48. Malcovati L, Germing U, Kuendgen A, et al. Time-dependent prognostic scoring system for predicting survival and leukemic evolution in myelodysplastic syndromes. Journal of Clinical Oncology. 2007;25(23):3503-3510. DOI: 10.1200/JCO.2006.08.5696
  49. 49. Kantarjian H, O'Brien S, Ravandi F, 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(6):1351-1361. DOI: 10.1002/cncr.23697
  50. 50. Garcia-Manero G, Shan J, Faderl S, et al. A prognostic score for patients with lower risk myelodysplastic syndrome. Leukemia. 2008;22(3):538-543. DOI: 10.1038/sj.leu.2405070
  51. 51. Warlick ED, Smith BD. Myelodysplastic syndromes: Review of pathophysiology and current novel treatment approaches. Current Cancer Drug Targets. 2007;7:541-558
  52. 52. Fenaux P. Myelodysplastic syndromes: From pathogenesis and prognosis to treatment. Seminars in Hematology. 2004;41(2 Suppl. 4):13-20
  53. 53. Lindsley RC. Uncoding the genetic heterogeneity of myelodysplastic syndrome. Hematology. American Society of Hematology. Education Program. 2017;2017(1):447-452. DOI: 10.1182/asheducation-2017.1.447
  54. 54. Shastri A, Will B, Steidl U, Verma A. Stem and progenitor cell alterations in myelodysplastic syndromes. Blood. 2017;129(12):1586-1594. DOI: 10.1182/blood-2016-10-696062
  55. 55. Li AJ, Calvi LM. The microenvironment in myelodysplastic syndromes: Niche-mediated disease initiation and progression. Experimental Hematology. 2017;55:3-18. DOI: 10.1016/j.exphem.2017.08.003
  56. 56. Medyouf H. The microenvironment in human myeloid malignancies: Emerging concepts and therapeutic implications. Blood. 2017;129(12):1617-1626. DOI: 10.1182/blood-2016-11-696070
  57. 57. Maurtizson N, Albin N, Rylander L, et al. Pooled analysis of clinical and cytogenetic features in treatment-related and de novo adult acute myloid leukemia and myelodysplastic syndromes based on a consecutive series of 761 patients analyzed 1976-1993 and on 5098 unselected cases reported in the literature 1974-2001. Leukemia. 2002;16(12):2366-2378. DOI: 10.1038/sj.leu.2402713
  58. 58. Olney HJ, LeBeau MM. The cytogenetics of myelodysplastic syndromes. Best Practice & Research Clinical Haematology. 2001;14(3):479-495. DOI: 10.1053/beha.2001.0151
  59. 59. Lindsley RC, Saber W, Mar BG, et al. Prognostic mutations in myelodysplastic syndrome after stem-cell transplantation. The New England Journal of Medicine. 2017;376(6):536-547. DOI: 10.1056/NEJMoa1611604
  60. 60. Makishima H, Yoshizato T, Yoshida K, et al. Dynamics of clonal evolution in myelodysplastic syndromes. Nature Genetics. 2017;49(2):204-212. DOI: 10.1038/ng.3742
  61. 61. Yoshizato T, Nannya Y, Atsuta Y, et al. Genetic abnormalities in myelodysplasia and secondary acute myeloid leukemia: Impact on outcome of stem cell transplantation. Blood. 2017;129(17):2347-2358. DOI: 10.1182/blood-2016-12-754796
  62. 62. Malcovati L, Gallì A, Travaglino E, et al. Clinical significance of somatic mutation in unexplained blood cytopenia. Blood. 2017;129(25):3371-3378. DOI: 10.1182/blood-2017-01-763425
  63. 63. Lindsley RC, Mar BG, Mazzola E, et al. Acute myeloid leukemia ontogeny is defined by distinct somatic mutations. Blood. 2015;125(9):1367-1376. DOI: 10.1182/blood-2014-11-610543
  64. 64. Corces-Zimmerman MR, Hong W-J, Weissman IL, Medeiros BC, Majeti R. Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(7):2548-2553. DOI: 10.1073/pnas.1324297111
  65. 65. Badar T, Patel KP, Thompson PA, et al. Detectable FLT3-ITD or RAS mutation at the time of transformation from MDS to AML predicts for very poor outcomes. Leukemia Research. 2015;39(12):1367-1374. DOI: 10.1016/j.leukres.2015.10.005
  66. 66. Kim T, Tyndel MS, Kim HJ, et al. The clonal origins of leukemic progression of myelodysplasia. Leukemia. 2017;31(9):1928-1935. DOI: 10.1038/leu.2017.17
  67. 67. Walter MJ, Shen D, Ding L, et al. Clonal architecture of secondary acute myeloid leukemia. The New England Journal of Medicine. 2012;366(12):1090-1098. DOI: 10.1056/NEJMoa1106968
  68. 68. Takahashi K, Wang F, Kantarjian H, et al. Preleukaemic clonal haemopoiesis and risk of therapy-related myeloid neoplasms: A case-control study. The Lancet Oncology. 2017;18(1):100-111. DOI: 10.1016/S1470-2045(16)30626-X
  69. 69. Wong TN, Ramsingh G, Young AL, et al. Role of TP53 mutations in the origin and evolution of therapy-related acute myeloid leukaemia. Nature. 2015;518(7540):552-555. DOI: 10.1038/nature13968
  70. 70. Ok CY, Patel KP, Garcia-Manero G, et al. Mutational profiling of therapy-related myelodysplastic syndromes and acute myeloid leukemia by next generation sequencing, a comparison with de novo diseases. Leukemia Research. 2015;39(3):348-354. DOI: 10.1016/j.leukres.2014.12.006
  71. 71. Shih AH, Chung SS, Dolezal EK, et al. Mutational analysis of therapy-related myelodysplastic syndromes and acute myelogenous leukemia. Haematologica. 2013;98(6):908-912. DOI: 10.3324/haematol.2012.076729
  72. 72. Cargo CA, Rowbotham N, Evans PA, et al. Targeted sequencing identifies patients with preclinical MDS at high risk of disease progression. Blood. 2015;126(21):2362-2365. DOI: 10.1182/blood-2015-08-663237
  73. 73. Schepers K, Campbell TB, Passegue E. Normal and leukemic stem cell niches: Insights and therapeutic opportunities. Cell Stem Cell. 2015;16(3):254-267. DOI: 10.1016/j.stem.2015.02.014
  74. 74. Wiseman DH. Donor cell leukemia: A review. Biology of Blood and Marrow Transplantation. 2011;17(6):771-789. DOI: 10.1016/j.bbmt.2010.10.010
  75. 75. Blau O, Baldus CD, Hofmann WK, et al. Mesenchymal stromal cells of myelodysplastic syndrome and acute myeloid leukemia patients have distinct genetic abnormalities compared with leukemic blasts. Blood. 2011;118(20):5583-5592. DOI: 10.1182/blood-2011-03-343467
  76. 76. Blau O, Hofmann WK, Baldus CD, et al. Chromosomal aberrations in bone marrow mesenchymal stroma cells from patients with myelodysplastic syndrome and acute myeloblastic leukemia. Experimental Hematology. 2007;35(2):221-229. DOI: 10.1016/j.exphem.2006.10.012
  77. 77. Huang JC, Basu SK, Zhao X, et al. Mesenchymal stromal cells derived from acute myeloid leukemia bone marrow exhibit aberrant cytogenetics and cytokine elaboration. Blood Cancer Journal. 2015;5:e302. DOI: 10.1038/bcj.2015.17
  78. 78. Pleyer L, Valent P, Greil R. Mesenchymal stem and progenitor cells in normal and dysplastic hematopoies is masters of survival and clonality? International Journal of Molecular Sciences. 2016;17(7):1009. DOI: 10.3390/ijms17071009
  79. 79. Peled A, Petit I, Kollet O, et al. Dependence of human stem cell engraftment and repopulation of NOD/SCID mice on CXCR4. Science. 1999;283(5403):845-848
  80. 80. Foster K, Lassailly F, Anjos-Afonso F, et al. Different motile behaviors of human hematopoietic stem versus progenitor cells at the osteoblastic niche. Stem Cell Reports. 2015;5(5):690-701. DOI: 10.1016/j.stemcr.2015.09.003
  81. 81. Chen X, Eksioglu EA, Zhou J, et al. Induction of myelodysplasia by myeloid-derived suppressor cells. The Journal of Clinical Investigation. 2013;123(11):4595-4611. DOI: 10.1172/JCI67580
  82. 82. Verma A, List AF. Cytokine targets in the treatment of myelodysplastic syndromes. Current Hematology Reports. 2005;4(6):429-435
  83. 83. Varney ME, Melgar K, Niederkorn M, et al. Deconstructing innate immune signaling in myelodysplastic syndromes. Experimental Hematology. 2015;43(8):587-598. DOI: 10.1016/j.exphem.2015.05.016
  84. 84. Gañán-Gómez I, Wei Y, Starczynowski DT, et al. Deregulation of innate immune and inflammatory signaling in myelodysplastic syndromes. Leukemia. 2015;29(7):1458-1469. DOI: 10.1038/leu.2015.69
  85. 85. Schinke C, Giricz O, Li W, et al. IL8-CXCR2 pathway inhibition as a therapeutic strategy against MDS and AML stem cells. Blood. 2015;125(20):3144-3152. DOI: 10.1182/blood-2013-03-492884
  86. 86. Ågerstam H, Hansen N, von Palffy S, et al. IL1RAP antibodies block IL-1-induced expansion of candidate CML stem cells and mediate cell killing in xenograft models. Blood. 2016;128(23):2683-2693. DOI: 10.1182/blood-2015-01-621631
  87. 87. Malcovati L, Hellström-Lindberg E, Bowen D, et al. Diagnosis and treatment of primary myelodysplastic syndromes in adults: Recommendations from the European LeukemiaNet. Blood. 2013;22:2943-2964. DOI: 10.1182/blood-2013-03-492884
  88. 88. NCCN (National Comprehensive Cancer Network). NCCN Clinical Practice Guidelines in Oncology: Myelodysplastic Syndromes. Version 1. 2019. Available from: https://www.nccn.org/professionals/physician_gls/pdf/mds.pdf [Accessed: 2016-10-12]
  89. 89. Stenke L, Wallvik J, Celsing F, et al. Prediction of response to treatment with human recombinant erythropoietin in myelodysplastic syndromes. Leukemia. 1993;7:1324-1327
  90. 90. Stasi R, Brunetti M, Bussa S, et al. Response to recombinant human erythropoietin in patients with myelodysplastic syndromes. Clinical Cancer Research. 1997;3:733-739
  91. 91. Rossi Ferrini PR, Grossi A, Vannucchi AM, et al. A randomized double-blind placebo-controlled study with subcutaneous recombinant human erythropoietin in patients with low-risk myelodysplastic syndromes. British Journal of Haematology. 1998;103:1070-1074
  92. 92. Terpos E, Mougiou A, Kouraklis A, et al. Prolonged administration of erythropoietin increases erythroid response rate in myelodysplastic syndromes: A phase II trial in 281 patients. British Journal of Haematology. 2002;118:174-180
  93. 93. Greenberg PL, Sun Z, Miller KB, 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(12):2393. DOI: 10.1182/blood-2009-03-211797
  94. 94. Mantovani L, Lentini G, Hentschel B, et al. Treatment of anaemia in myelodysplastic syndromes with prolonged administration of recombinant human granulocyte colony-stimulating factor and erythropoietin. British Journal of Haematology. 2000;109(2):367
  95. 95. Sloand EM, Wu CO, Greenberg P, et al. Factors affecting response and survival in patients with myelodysplasia treated with immunosuppressive therapy. Journal of Clinical Oncology. 2008;26(15):2505. DOI: 10.1200/JCO.2007.11.9214
  96. 96. List A, Dewald G, Bennett J, et al. Lenalidomide in the myelodysplastic syndrome with chromosome 5q deletion. The New England Journal of Medicine. 2006;355(14):1456. DOI: 10.1056/NEJMoa061292
  97. 97. Fenaux P, Giagounidis A, Selleslag D, 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(14):3765. DOI: 10.1182/blood-2011-01-330126
  98. 98. Komrokji RS, Lancet JE, Swern AS, et al. Combined treatment with lenalidomide and epoetin alfa in lower-risk patients with myelodysplastic syndrome. Blood. 2012;120:3419-3424. DOI: 10.1182/blood-2012-03-415661
  99. 99. Sibon D, Cannas G, Baracco F, et al. Lenalidomide in lower-risk myelodysplastic syndromes with karyotypes other than deletion 5q and refractory to erythropoiesis-stimulating agents. British Journal of Haematology. 2012;156:619-625. DOI: 10.1111/j.1365-2141.2011.08979.x
  100. 100. Toma A, Kosmider O, Chevret S, et al. Lenalidomide with or without erythropoietin in transfusion dependent erythropoiesis-stimulating agent-refractory lower risk MDS without 5qdeletion. Leukemia. 2016;30:897-905. DOI: 10.1038/leu.2015.296
  101. 101. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, 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. The Lancet Oncology. 2009;10(3):223-232. DOI: 10.1016/S1470-2045(09)70003-8
  102. 102. Silverman LR, Fenaux P, Mufti GJ, et al. Continued azacitidine therapy beyond time of first response improves quality of response in patients with higher-risk myelodysplastic syndromes. Cancer. 2011;117(12):2697-2702. DOI: 10.1002/cncr.25774
  103. 103. Kantarjian H, Issa JP, Rosenfeld CS, et al. Decitabine improves patient outcomes in myelodysplastic syndromes: Results of a phase III randomized study. Cancer. 2006;106(8):1794-1803. DOI: 10.1002/cncr.21792
  104. 104. Steensma DP, Baer MR, Slack JL, et al. Multicenter study of decitabine administered daily for 5 days every 4 weeks to adults with myelodysplastic syndromes: The alternative dosing for outpatient treatment (ADOPT) trial. Journal of Clinical Oncology. 2009;27(23):3842-3848. DOI: 10.1200/JCO.2008.19.6550
  105. 105. Garcia-Manero G, Stoltz ML, Ward MR, et al. A pilot pharmacokinetic study of oral azacitidine. Leukemia. 2008;22(9):1680-1684. DOI: 10.1038/leu.2008.145
  106. 106. Garcia-Manero G, Gore SD, Cogle C, et al. Phase I study of oral azacitidine in myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia. Journal of Clinical Oncology. 2011;29(18):2521-2527. DOI: 10.1200/JCO.2010.34.4226
  107. 107. Garcia-Manero G, Gore SD, Kambhampati S, et al. Efficacy and safety of extended dosing schedules of CC-486 (oral azacitidine) in patients with lower-risk myelodysplastic syndromes. Leukemia. 2016;30(4):889-896. DOI: 10.1038/leu.2015.265
  108. 108. Garcia-Manero G, Odenike O, Amrin PC, et al. Successful emulation of IV decitabine pharmacokinetics with an oral fixed-dose combination of the oral cytidine deaminase inhibitor (CDAi) E7727 with oral decitabine, in subjects with myelodysplastic syndromes (MDS): Final data of phase 1 study. In the 58th American Society of Hematology annual meeting and exposition, 3-6 December 2016, San Diego, CA. Blood. 2016;128(22):114
  109. 109. Garcia-Manero G, Griffiths EA, Roboz GJ, et al. A phase 2 dose-confirmation study of oral ASTX727, a combination of oral decitabine with a cytidine deaminase inhibitor (CDAi) cedazuridine (E7727), in subjects with myelodysplastic syndromes (MDS). In the 59th American Society of Hematology annual meeting and exposition, 9-12 December 2017, Atlanta, GA. Blood. 2017;130:4274
  110. 110. Issa JJ, Roboz G, Rizzieri D, et al. Safety and tolerability of guadecitabine (SGI-110) in patients with myelodysplastic syndrome and acute myeloid leukaemia: A multicentre, randomised, dose-escalation phase 1 study. The Lancet Oncology. 2015;16(9):1099-1110. DOI: 10.1016/S1470-2045(15)00038-8
  111. 111. Garcia-Manero GRE, Walsh K, et al. First clinical results of a randomized phase 2 dose-response study of SGI-110, a novel subcutaneous (SC) hypomethylating agent (HMA), in 102 patients with intermediate (int) or high risk (HR) myelodysplastic syndromes (MDS) or chronic myelomonocytic leukemia (CMML). In the 56th American Society of Hematology annual meeting and exposition, 6-9 December 2014, San Francisco, CA. Blood. 2014;124(21):529
  112. 112. Herman JG, Baylin SB. Gene silencing in cancer in association with promoter hypermethylation. The New England Journal of Medicine. 2003;349(21):2042-2054. DOI: 10.1056/NEJMra023075
  113. 113. Stahl M, Kohrman N, Gore SD, et al. Epigenetics in cancer: A hematological perspective. PLoS Genetics. 2016;12:e1006193. DOI: 10.1371/journal.pgen.1006193
  114. 114. Stahl M, Gore SD, Vey N, Prebet T. Lost in translation? Ten years of development of histone deacetylase inhibitors in acute myeloid leukemia and myelodysplastic syndromes. Expert Opinion on Investigational Drugs. 2016;25:307-317. DOI: 10.1517/13543784.2016.1146251
  115. 115. Schaefer EW, Loaiza-Bonilla A, Juckett M, et al. A phase 2 study of vorinostat in acute myeloid leukemia. Haematologica. 2009;94:1375-1382. DOI: 10.3324/haematol.2009.009217
  116. 116. DeAngelo DJ, Spencer A, Bhalla KN, et al. Phase Ia/II, 2-arm, open-label, dose-escalation study of oral panobinostat administered via 2 dosing schedules in patients with advanced hematologic malignancies. Leukemia. 2013;27:1628-1636. DOI: 10.1038/leu.2013.38
  117. 117. Cameron EE, Bachman KE, Myöhänen S, et al. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nature Genetics. 1999;21(1):103-107
  118. 118. Tan P, Wei A, Mithraprabhu S, et al. Dual epigenetic targeting with panobinostat and azacitidine in acute myeloid leukemia and high risk myelodysplastic syndrome. Blood Cancer Journal. 2014;4:e170. DOI: 10.1038/bcj.2013.68
  119. 119. Prebet T, Sun Z, Figueroa ME, et al. Prolonged administration of azacitidine with or without entinostat for myelodysplastic syndrome and acute myeloid leukemia with myelodysplasia-related changes: Results of the US leukemia intergroup trial E1905. Journal of Clinical Oncology. 2014;32:1242-1248. DOI: 10.1200/JCO.2013.50.3102
  120. 120. Mikkael A, Sekeres MO, List AF, et al. Additional analyses of a randomized phase II study of azacitidine combined with lenalidomide or with vorinostat vs. azacitidine monotherapy in higher-risk myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML): North American intergroup study SWOG S1117. In the 57th American Society of Hematology annual meeting and exposition, 5-8 December 2015, Orlando, FL. Blood. 2015;126:908
  121. 121. Issa JP, Garcia-Manero G, Huang X, et al. Results of phase 2 randomized study of low-dose decitabine with or without valproic acid in patients with myelodysplastic syndrome and acute myelogenous leukemia. Cancer. 2015;121:556-561. DOI: 10.1002/cncr.29085
  122. 122. Garcia-Manero G, Montalban-Bravo G, Berdeja JG, et al. Phase 2, randomized, double-blind study of pracinostat in combination with azacitidine in patients with untreated, higher-risk myelodysplastic syndromes. Cancer. 2017;123(6):994-1002. DOI: 10.1002/cncr.30533
  123. 123. Dawson MA, Prinjha RK, Dittmann A, et al. Inhibition of BET recruitment to chromatin as an effective treatment for MLL-fusion leukaemia. Nature. 2011;478(7370):529-533. DOI: 10.1038/nature10509
  124. 124. Zuber J, Shi J, Wang E, et al. RNAi screen identifies Brd4 as a therapeutic target in acute myeloid leukaemia. Nature. 2011;478(7370):524-528. DOI: 10.1038/nature10334
  125. 125. Schenk T, Chen WC, Göllner S, et al. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nature Medicine. 2012;18(4):605-611. DOI: 10.1038/nm.2661
  126. 126. Zhang L, Gajewski TF, Kline J. PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model. Blood. 2009;114(8):1545-1552. DOI: 10.1182/blood-2009-03-206672
  127. 127. Yang H, Bueso-Ramos C, DiNardo C, et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia. 2014;28(6):1280-1288. DOI: 10.1038/leu.2013.355
  128. 128. Garcia-Manero G, Daver N, Montalban-Bravo G, et al. A phase II study evaluating the combination of nivolumab (Nivo) or ipilimumab (Ipi) with azacitidine in pts with previously treated or untreated myelodysplastic syndromes (MDS). In the 58th american Society of Hematology annual meeting and exposition, 3-6 December 2016, San Diego, CA. Blood. 2016;128(22):344
  129. 129. DiNardo CD, Jabbour E, Ravandi F, et al. IDH1 and IDH2 mutations in myelodysplastic syndromes and role in disease progression. Leukemia. 2016;30(4):980-984. DOI: 10.1038/leu.2015.211
  130. 130. Stein EM, DiNardo CD, Pollyea DA, et al. Enasidenib in mutant IDH2 relapsed or refractory acute myeloid leukemia. Blood. 2017;130(6):722-731. DOI: 10.1182/blood-2017-04-779405
  131. 131. CD DN, de Botton S, Stein EM, et al. Determination of IDH1 mutational burden and clearance via next-generation sequencing in patients with IDH1 mutation-positive hematologic malignancies receiving AG-120, a first-in-class inhibitor of mutant IDH1. In the 58th american Society of Hematology annual meeting and exposition, 3-6 December 2016, San Diego, CA. Blood. 2016;128(22):1070
  132. 132. Buonamici SY, Yoshimi A, Thomas M, et al. H3B-8800, an orally bioavailable modulator of the SF3b complex, shows efficacy in spliceosome-mutant myeloid malignancies. In the 58th American Society of Hematology annual meeting and exposition, 3-6 December 2016, San Diego, CA. Blood. 2016;128(22):966
  133. 133. DiNardo C, Pollyea DA, Pratz K, et al. A phase 1b study of venetoclax (ABT-199/GDC-0199) in combination with decitabine or azacitidine in treatment-naive patients with acute myelogenous leukemia who are ≥ to 65 years and not eligible for standard induction therapy. In the 58th American Society of Hematology annual meeting and exposition, 3-6 December 2016, San Diego, CA. Blood. 2015;126:327
  134. 134. Suragani RN, Cadena SM, Cawley SM, et al. Transforming growth factor-β superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. Nature Medicine. 2014;20(4):408-414. DOI: 10.1038/nm.3512
  135. 135. Dussiot M, Maciel TT, Fricot A, et al. An activin receptor IIA ligand trap corrects ineffective erythropoiesis in β-thalassemia. Nature Medicine. 2014;20(4):398-407. DOI: 10.1038/nm.3468
  136. 136. Platzbecker U, Germing U, Götze KS, et al. Luspatercept for the treatment of anaemia in patients with lower-risk myelodysplastic syndromes (PACE-MDS): A multicentre, open-label phase 2 dose-finding study with long-term extension study. The Lancet Oncology. 2017;18(10):1338-1347. DOI: 10.1016/S1470-2045(17)30615-0
  137. 137. Komrokji R, Garcia-Manero G, Ades L, et al. Sotatercept with long-term extension for the treatment of anaemia in patients with lower-risk myelodysplastic syndromes: A phase 2, dose-ranging trial. The Lancet Haematology. 2018;5(2):e63-e72. DOI: 10.1016/S2352-3026(18)30002-4
  138. 138. Azra R, Al-Kali A, Tibes R, et al. Rigosertib oral in transfusion dependent lower risk myelodysplastic syndromes (LR-MDS): Optimization of dose and rate of transfusion Independence (TI) or transfusion reduction (TR) in a single-arm phase 2 study. In the 59th American Society of Hematology annual meeting and exposition, 9-12 December 2017, Atlanta, GA. Blood. 2017;130:1689
  139. 139. Besarab A, Provenzano R, Hertel J, et al. Randomized placebo-controlled dose-ranging and pharmacodynamics study of roxadustat (FG-4592) to treat anemia in nondialysis-dependent chronic kidney disease (NDD-CKD) patients. Nephrology, Dialysis, Transplantation. 2015;30(10):1665-1673. DOI: 10.1093/ndt/gfv302
  140. 140. Sierra J, Perez WS, Rozman C, et al. Bone marrow transplantation from HLA-identical siblings as treatment for myelodysplasia. Blood. 2002;100(6):1997-2004
  141. 141. Alyea EP, Kim HT, Ho V, et al. Comparative outcome of nonmyeloablative and myeloablative allogeneic hematopoietic cell transplantation for patients older than 50 years of age. Blood. 2005;105:1810-1814. DOI: 10.1182/blood-2004-05-1947
  142. 142. Saber W, Le Rademacher J, Sekeres M, et al. Multicenter biologic assignment trial comparing reduced-intensity allogeneic hematopoietic cell transplant to hypomethylating therapy or best supportive care in patients aged 50 to 75 with intermediate-2 and high-risk myelodysplastic syndrome: Blood and marrow transplant clinical trials network #1102 study rationale, design, and methods. Biology of Blood and Marrow Transplantation. 2014;20(10):1566-1572. DOI: 10.1016/j.bbmt.2014.06.010
  143. 143. Della Porta MG, Alessandrino EP, Bacigalupo A, et al. Predictive factors for the outcome of allogeneic transplantation in patients with MDS stratified according to the revised IPSS-R. Blood. 2014;123(15):2333-2342. DOI: 10.1182/blood-2013-12-542720
  144. 144. Festuccia M, Deeg HJ, Gooley TA, et al. Minimal identifiable disease and the role of conditioning intensity in hematopoietic cell transplantation for MDS and AML evolving from MDS. Biology of Blood and Marrow Transplantation. 2016;22(7):1227-1233. DOI: 10.1016/j.bbmt.2016.03.029
  145. 145. Bejar R, Stevenson KE, Caughey B, et al. Somatic mutations predict poor outcome in patients with myelodysplastic syndrome after hematopoietic stem-cell transplantation. Journal of Clinical Oncology. 2014;32(25):2691-2698. DOI: 10.1200/JCO.2013.52.3381
  146. 146. Bejar R, Stevenson K, Abdel-Wahab O, et al. Clinical effect of point mutations in myelodysplastic syndromes. The New England Journal of Medicine. 2011;364(26):2496-2506. DOI: 10.1056/NEJMoa1013343

Written By

Wanxing Chai-Ho and Gary J. Schiller

Submitted: 29 August 2018 Reviewed: 22 October 2018 Published: 27 November 2018