Abstract
Chronic myelomonocytic leukemia (CMML) includes components of both myelodysplastic syndrome and myeloproliferative neoplasms and is associated with a characteristic peripheral monocytosis. CMML is caused by the proliferation of an abnormal hematopoietic stem cell clone and may be influenced by microenvironmental changes. The disease is rare and has undergone revisions in its classification. We review the recent classification strategies as well as diagnostic criteria, focusing on CMML’s genetic alterations and unique pathophysiology. We also discuss the latest molecular characterization of the disease, including how molecular factors affect current prognostic models. Finally, we focus on available treatment strategies, with a special emphasis on experimental and forthcoming therapies.
Keywords: chronic myelomonocytic leukemia, myelodysplastic syndrome, myeloproliferative neoplasms
1. Introduction
Chronic myelomonocytic leukemia (CMML) is generally recognized as a chronic leukemia with persistent monocytosis and components of both myeloproliferative neoplasms and myelodysplastic syndrome [1]. It has been recognized as a distinct disease for more than 40 years, although until 2002 it was grouped with myelodysplastic syndrome (MDS). In 1971, Saarni and Linman recognized qualitative abnormalities in more than one lineage in patients with monocytoid leukemic transformation; 36% of their patient series had peripheral monocytosis, and 31% demonstrated a preleukemic phase, sometimes for as long as 9 years [2, 3]. The term myelomonocytic leukemia denotes both the myeloid and the monocytoid features of the disease. In 1972, Zittoun et al. described 27 cases of subacute myelomonocytic leukemia [4, 5]. The first significant-size cohort of patients to be recognized as having CMML was described in 1975 and included 18 elderly patients with unexplained monocytosis, cytopenias, and splenomegaly [6, 7]. Five of the patients survived longer than 5 years. This finding indicated that intensive chemotherapy may not be needed in this patient population and led to recognition of CMML as a distinct entity by the French-American-British (FAB) Group in 1976 [8]. Since then, several groups have categorized the clinical manifestations and outcome of CMML as a subset of MDS. However, some patients with CMML may express features of myeloproliferative neoplasms (MPN; also known as myeloproliferative disorders or MPD) at the time of diagnosis or at another stage in the course of the disease. Consequently, CMML has remained under-researched and is often excluded from MDS and MPN clinical trials.
Here we discuss the classification and diagnosis of CMML, the clinical features and epidemiology of the disease, and current insights into its pathophysiology. We review established treatments for patients with CMML as well as state-of-the-art approaches.
2. Classification and diagnosis
2.1. FAB and WHO classifications
From the time CMML was first identified 50 years ago, debate has continued on its proper place in the classification of hematologic malignancies. In 1982, the FAB Group classified CMML as part of MDS, given the morphologic evidence of dysplastic hematopoiesis [9], but whether CMML should be classified as myeloproliferative or myelodysplastic remained unclear. Recognizing the heterogeneity of the clinical features of the disease, the FAB Group later proposed a reclassification of patients into two subtypes based on white blood cell (WBC) count at diagnosis [10]. The FAB classification is shown in Table 1. Patients with WBC counts of ≤13 × 109/L were considered to have myelodysplastic CMML (MD-CMML), and those with WBC counts of >13 × 109/L were considered to have myeloproliferative CMML (MP-CMML). The separation of patients by this classification system remains problematic because the two groups have overlapping features. However, since many studies have classified CMML according to FAB criteria, the differences between MD-CMML and MP-CMML are important to interpreting the studies’ findings.
Table 1.
FAB classification | WHO classification |
---|---|
MDS | MDS/MPD overlap syndrome |
PB Monocytes > 1 × 109/L (defining feature) | PB Monocytes > 1 × 109/L |
Myeloblasts < 20% in bone marrow | Myeloblasts < 20% in bone marrow |
PB blasts < 5% | No Philadelphia chromosome or BCR-ABL1 fusion gene |
Subclassification MD-CMML: WBC ≤ 13 × 109/L MP-CMML: WBC > 13 × 109/L |
No rearrangement of PDGFRA or PDGFRB (excluded especially in cases with eosinophilia) Dysplasia in at least one myeloid lineage. If myelodysplasia is absent or minimal, diagnosis can still be made if the other requirements are present and An acquired clonal cytogenetic or molecular genetic abnormality is present or The monocytosis has persisted for at least 3 months and all other causes of monocytosis have been excluded (e.g. infection, inflammation, malignancy) Subclassification CMML-1: PB blasts < 5%, BM blasts < 10% CMML-2: PB blasts 5-19%, BM blasts 10-19%, or Auer rods present CMML-1 or CMML-2 with eosinophilia: above criteria with PB eosinophils > 1.5 × 109/L |
FAB, French-American-British; WHO, World Health Organization, CMML, chronic myelomonocytic leukemia; MDS, myelodysplastic syndrome; MPD, myeloproliferative disorder; PB, peripheral blood; MD-CMML, myelodysplastic CMML; MP-CMML, myeloproliferative CMML.
Nosslinger et al. conducted a retrospective analysis of 91 patients with CMML who had been treated primarily with supportive care [11]. At the time of diagnosis, patients with MPCMML (n, 31; 34%) had higher lactate dehydrogenase (LDH) levels, absolute neutrophil counts, and bone marrow cellularity values than did patients with MD-CMML (n, 60; 66%). The median overall survival (OS) duration for the MP-CMML group (16 months) was significantly shorter than that for the MD-CMML group (31 months) (p-value, 0.03), with a higher risk of leukemia transformation in the MP-CMML group, indicating differences in outcomes between the two groups. Onida et al. retrospectively analyzed 213 patients with CMML treated with various approaches, including chemotherapy, and classified each patient’s CMML as MD-CMML (n, 74; 35%) or MP-CMML (n, 139; 65%) on the basis of the patients’ WBC counts [12]. Although the OS rates were similar in the first few months of treatment, a significant difference appeared after 16 months, with a higher OS rate for MD-CMML patients. The difference in rate of leukemic transformation, however, was not statistically significant [12]. Voglova et al. analyzed 69 patients with CMML, 31 (45%) classified as MD-CMML and 38 (55%) as MP-CMML [13]. Cytogenetic abnormalities were more frequent among patients with MP-CMML. The median OS was significantly longer in the MD-CMML group than in the MP-CMML group (30 vs. 16 months, respectively; p-value < 0.01), and there was no significant difference in leukemic transformation. However, in 24 patients with MD-CMML, the WBC count increased to more than 13 × 109/L over the course of the disease. The authors concluded that using the WBC count obtained at diagnosis as the single criterion for subclassification of CMML did not seem fully justified and that MD-CMML and MP-CMML should be considered different stages of the same disease, rather than two different diseases [13].
In an effort to resolve this discrepancy, and to eliminate the arbitrary use of WBC counts to classify patients with CMML, the World Health Organization (WHO) in 2002 recognized CMML as a distinct entity for the first time and moved it to a new category called MDS/MPD [3]. This category also included atypical chronic myeloid leukemia and juvenile myelomonocytic leukemia. The FAB and WHO diagnostic criteria for CMML are compared in Table 1. The FAB system includes both MD-CMML and MP-CMML and is based on the WBC count, while the WHO classification differentiates CMML-1 and CMML-2 and is based on blast percentages. The quantification of blast percentages for peripheral blood and bone marrow include myeloblasts, monoblasts, and promonocytes [3]. Most recently, Schuler et al. proposed classifying patients with less than 5% medullary blasts as CMML-0, and sub-classifying each group of patients based on dysplasia vs. proliferation, as these groups have distinct clinical outcomes [14].
2.2. Diagnostic considerations
2.2.1. Peripheral blood counts
Monocytosis in the bone marrow or peripheral blood represents a major diagnostic criterion for CMML. However, in some cases, eosinophilia may present as a part of the CMML disease process. Because monocytosis and/or eosinophilia are cardinal features of several other diseases, a thorough search for other monocytic diseases must be conducted and those diseases must be systematically ruled out when a new patient is evaluated for possible CMML. In addition, a proper evaluation for infections and chronic inflammatory states as well as other malignancies known to cause monocytosis or eosinophilia should be conducted before CMML is diagnosed [15].
2.2.2. Cytogenetic analysis
There is no pathognomonic cytogenetic abnormality in CMML [15, 16]. Previously, some cases of CMML were associated with translocation of chromosomes 5 and 12 [t(5;12)(q33;p13)] [17-20], which is associated with expression of a fusion transcript that links the ETS-variant gene TEL with PDGFRB [21]. These patients are best classified as “myeloid neoplasia with PDGFRB rearrangements” even if they have features of CMML.
Other chromosomal abnormalities have been described in patients with CMML, including t(3;6)(q12;24), t(5;7)(q33;q11.2), der(9)t(1;9)(q11;q34), der(14)t(1;14)(q12;p11), and t(11;19)(q23;p13.1) [22-27]. One study also identified translocations involving the RET oncogene, specifically t(10;22)(q11;q11) and t(6;10)(q27;q11), as responsible for two separate cases of CMML [28]. First, a large study using data from the Spanish Registry of Myelodysplastic Syndromes found abnormal cytogenetic profiles in 110 (27%) of 414 CMML patients [29]. Two subsequent and recent studies found abnormal karyotype in 30% and 28% of 417 and 409 CMML patients respectively, confirming that overall ~30% of CMML patients have an abnormal karyotype, while ~70% have normal karyotype [30, 31].
Single nucleotide polymorphism arrays (SNP-A) have been employed to study chromosomal lesions in patients with CMML [32, 33]. Uniparental disomy (UPD), which results from segmental DNA recombination during mitosis, was a common event in MDS/MPN (35%), especially in CMML patients tested (n = 24) [32]. In a comprehensive follow up study that included 54 CMML patients (36 CMML-1 and 18 CMML-2 patients), SNP-A again detected widespread acquired copy-neutral loss of heterozygosity (a result of UPD) [33]. Given the predominance of UPD in CMML, detection of these chromosomal abnormalities has been critical in identifying genes such as CBL and TET2 that are important in CMML pathogenesis [33-36].
2.2.3. Molecular phenotyping
More recently, key molecular tests have been identified that aid in the workup of patients with suspected CMML. Importantly, PDGFRB rearrangement at chromosome band 5q33 is present in some CMML patients and is associated with eosinophilia [16]. These patients may respond to tyrosine kinase inhibition (with imatinib mesylate therapy); therefore, knowledge about this genetic aberration can be critical to treatment (see section 5 below) [37, 38]. Though JAK2 V617F mutations are less frequent in CMML than in polycythemia vera, essential thrombocytosis, or primary myelofibrosis [39-42], in one series, Pich et al. found that 8 (10.2%) of 78 patients with CMML were positive for the JAK2 V617 mutation at diagnosis [43]. JAK2 inhibitors have been characterized as potentially useful for treatment of CMML, and limited numbers of patients have been treated with them (see section 5 below) [44, 45].
Both KRAS and NRAS mutations, important in multiple hematologic malignancies, have been implicated in approximately 30%-40% of patients with CMML [46-48]. Other common molecular abnormalities (with percentages of CMML patients in parentheses) include mutated TET2 (~30-60%), ASXL1 (~40%), SRSF2 (~45%), RUNX1 (~15%), CBL (~10%), IDH1/2 (~5-10%), U2AF35 (9%), ZRSF2 (8%), UTX (8%), DNMT3A (~2-10%), SF3B1 (6%), U2AF1 (5%), FLT3 (~3-4%), EZH2 (~5%), and SETBP1 (~4-5%) [49-58]. These genes are associated with epigenetic regulation, mRNA splicing, embryonic development, protein ubiquitination, and cellular proliferation. Several of these genes identified are involved in spliceosome function, and may be mutually exclusive [56]. The biological significance of these mutations is related to the clonal architecture of the disease (see subsection 3.2 below), and some of the mutations may be used as prognostic indicators (see subsection 4.1 below). Interestingly, ASXL1 and SF3B1 mutations tended to occur with an abnormal karyotype (p-values 0.04 and 0.03 respectively), while SRSF2 mutations more frequently occurred with a normal karyotype (p-value = 0.02) [31].
2.3. Epidemiology and presentation
CMML is rarely diagnosed in younger adults, with the median age at diagnosis being about 70 years, similar to the median age of patients at diagnosis for MDS [5, 59]. The clinical presentations are protean and can represent the full spectrum of both MDS and MPN [60]. Common presentations include signs and symptoms of bone marrow suppression (anemia, thrombocytopenia, neutropenia), leukocytosis, lymphadenopathy, and hepatosplenomegaly [15, 60]. Furthermore, some reports have described leukemia cutis as the initial manifestation of CMML or heralding transformation to acute myeloid leukemia (AML) [61]. In most reports, patients with MP-CMML were older than patients with MD-CMML, but the differences were not statistically significant [7, 11]. Furthermore, male predominance was observed more frequently in patients with MD-CMML than in those with MP-CMML, but again the differences were not significant [7, 11].
3. Biology and pathophysiology
The pathophysiology of CMML is still not fully understood, in large part because CMML has been frequently studied as a subtype of MDS rather than as a separate entity. The high variability of the clinical presentation and the course of the disease reflect the heterogeneity of CMML’s pathogenetic features. Multiple theories about the biology of CMML, based on cellular, cytogenetic, and molecular abnormalities and supported by animal models, were developed over the past two decades, and recently a number of models to investigate the pathogenesis of CMML have emerged.
3.1 Cellular biology
Decreased apoptosis as an alternative pathway to tumorigenesis has been proposed in the pathogenesis of MDS as well as CMML. A competitive reconstitution assay demonstrated that long-term repopulating cells deficient in Bid, a BH3-only proapoptotic protein, gave rise to expanded myelomonocytic cells in vivo [62]. A mouse model recapitulating this phenomenon is discussed below in subsection 3.2. In support of the idea that decreased cellular apoptosis is a manifest contributor to CMML, Boudad et al. reported increased expression of antiapoptotic proteins Bcl-2 and Bcl-xL in patients with CMML [63]. In another study, increased expression of the cell cycle gene CCND1 (encoding cyclin D1) was also demonstrated in CMML patients compared with other FAB subgroups of MDS [64]. These findings suggest that the combination of specific decreased-apoptosis pathways and critical cell cycle pathway dysregulation directly contributes to the pathogenesis of CMML.
Recent investigation into immune checkpoint tolerance genes in myeloid malignancies revealed that CMML has increased expression of PD-L1 compared to AML [65]. Treatment with hypomethylating agents increased expression of PD-L1 and other related immune checkpoint genes in patients [65]. The work suggests that therapies blocking immune checkpoints, especially in conjunction with hypomethylating agents, may be an option for patients with CMML.
Angiogenesis has been shown to play a pivotal role in human leukemia. Pruneri et al. reported significant increases in microvascular density (MVD) in patients with CMML and MDS [66]. Aguayo et al. demonstrated that plasma levels of vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) were elevated in patients with CMML compared with the levels in non-leukemic control patients and patients with other leukemias including MDS [67]. Likewise, Wimazal, et al. found increased MVD and elevated levels of VEGF in the bone marrows of patients with CMML; immature myeloid cells were the source of VEGF [68]. Alexandrakis et al. further confirmed the importance of angiogenesis in CMML by demonstrating increased MVD and increased levels of angiogenin and interleukin 6 in patients with CMML [69, 70]. Finally, Bellamy, et al. proposed an autocrine function of VEGF, demonstrating that some patients’ CMML cells expressed cytoplasmic VEGF as well as the membranous VEGF receptors Flt-1 and KDR. Inhibiting VEGF prevented CMML colony growth in vitro [71]. In total, these studies strongly suggest that microenvironmental influences, particularly angiogenesis, play a special role in the development and maintenance of CMML.
3.2 Molecular underpinnings
At the molecular level, CMML shares some characteristics with other myeloproliferative neoplasms. Activation of the JAK/STAT pathway has been the prominent molecular feature of polycythemia vera and is also a feature of primary myelofibrosis and essential thrombocythemia [72, 73]. Mutations in the JAK2 gene are less common in CMML [39, 74, 75], however GM-CSF dependent STAT5 hypersensitivity has been noted in some CMML patients [76]. These studies suggest reliance on GM-CSF and the JAK/STAT pathway, and the potential for therapeutic intervention with anti-GM-CSF or JAK2 inhibition, even in the absence of mutated JAK2 [76].
Other molecular and genetic markers have been sought in CMML. Among hematologic malignancies, CMML is associated with the highest incidence of RAS pathway mutations [74]. MacKenzie et al. have shown that irradiated mice that undergo transplantation with Nras-mutated bone marrow cells develop myeloproliferative-like disease. Along with RAS pathway mutated mouse models described below in section 3.3, this work supports the theory that the RAS activation is a significant contributor to myeloproliferation in CMML [77].
Driver mutations responsible for the development of CMML have been studied by analyzing mutational data generated to date. Using single-cell-derived colonies from 28 patients with CMML, Itzykson et al. were able to determine that early mutations in TET2 were likely responsible for development of a dominant granulomonocytic clone [78]. After the acquisition of additional mutations, in spliceosome and epigenetic factors, for instance, the proliferative advantage and abnormal differentiation of a hematopoietic stem/progenitor cell emerges, as does the clinical entity CMML [79]. The findings suggest that a clonal hierarchy might be identified so that treatments would specifically target abnormal stem cell clones, to allow normal stem cells to proliferate and differentiate appropriately [79]. TET2 is recognized as an especially important player in the development of CMML [80]. Disruption of TET2 was demonstrated to act on the hematopoietic system at the level of the stem cell, promoting self-renewal, increasing 5-hydroxymethylcytosine, preventing normal differentiation, and resulting in skewing towards myeloid/monocyte development [81-84].
Other genes found mutated in CMML have also been shown to promote myeloid differentiation through methylation effects on the hematopoietic stem cell. ASXL1 was determined to act through the loss of H3K27 methylation by polycomb repressive complex 2, and loss of ASXL1 promoted myeloid differentiation in a proliferative setting [85]. In support of the idea of driver mutations in the stem cell compartment, Cilloni et al. also reported abnormal activation of the tyrosine kinase ROS1 in the stem-like cell population of 70% of CMML patients, and this abnormal activation may drive oncogenicity [86].
3.3 Mouse models
Several mouse models have capitalized on findings from cellular and molecular studies, and recapitulated elements of CMML. A number of mouse models that overexpress Nras or Kras in the hematopoietic compartment have been shown to develop CMML-like disease [87-89]. One model has recently been used to show effectiveness of combined ERK/MEK inhibition in for treatment of the Nras-driven murine CMML mimic [90].
Mice that were deficient in Bid proapoptotic protein developed a myeloid leukemia that clinically and morphologically resembled adult CMML [62]. Additionally, enforcing production of Bcl-2 protein under the control of the human myeloid related protein 8 (hMRP8) promoter, induces proliferation of myelomonocytic cells, and has been proposed as a leukemic stem cell model for CMML [91, 92]. These model supports the idea that decreased apoptosis is a fundamental mechanism in the development of CMML.
Tet2 deficient mice were predisposed to develop a myeloproliferation and a CMML-like malignancy, and mice transplanted with Tet2 deleted hematopoietic cells developed monocytosis and splenomegaly [82, 84]. Asxl1 deficient mice have also recently been shown to develop myelodysplasia/CMML [93, 94]. In these models, Asxl1 haploinsufficiency led to a milder myeloproliferation, and combined Asxl1/Tet2 knockout led to a more aggressive MDS [93, 94]. Interestingly, methylation effects of Asxl1 knockout altered expression of apoptosis related genes such as Bcl2, suggesting this as a mechanism acting on hematopoietic stem cells leading to the observed phenotype [93]. Finally, hematopoietic-restricted deletion of the tumor suppressor Bap1, whose protein product is believed to cooperate with ASXL1 in epigenetic regulation, led to MDS and splenomegaly in mice [95].
Another model where mice developed CMML-like features was created with conditionally deleted TGF-β activated kinase 1 (Tak1) in the myeloid compartment [96]. Mice with myeloid-deficient Tak1 developed a myelomonocytic expansion, splenomegaly, and murine AML as they aged [96]. The deletion appeared to affect cytokine mediated signaling, and was associated with genomic instability [96]. In another recently developed mouse model, deletion of transcription intermediary factor 1γ (Tif1g) initiated age-dependent development of myeloproliferation reminiscent of CMML [97]. The gene was demonstrated to be an epigenetically regulated tumor suppressor gene [97].
Additional mouse models of MDS and MPN are especially promising in recapitulating clinical manifestations of CMML that are seen in patients, and may be helpful in elucidating the pathogenesis of CMML. PDGFRB mutant mice develop a disorder that is not unlike myeloproliferative CMML [38, 98, 99]. The over-production of the oncogenic portion of the Hmga2 gene induces hematopoiesis in stem cell progenitors and produces a mouse model that mimics MPN [100]. Similarly, the deletion of the Dido1 gene, when knocked out in a mouse model, produces a human-like MPN/MPD disease [101]. These mouse models may lead to additional understanding of myelomonocytosis at the level of cellular and molecular biology.
4. Prognosis
4.1. Prognostic scoring systems
One of the most important prognostic indicators in CMML is the blast count, which is the basis for differentiating CMML-1 and CMML-2 in the WHO classification. CMML-1 presents with <5% blasts in the blood and <10% blasts in the bone marrow and has an 18% chance of transformation to AML within 5 years. CMML-2 has 5-19% peripheral blood blasts and/or 10-19% bone marrow blasts and is associated with a 63% 5-year risk of progression to AML [102].
Several prognostic scoring systems used for patients with MDS have been employed for patients with CMML [12, 103-112]. All of those systems score patients on the basis of various critical hematologic, clinical, or biochemical indices such as complete blood counts, bone marrow biopsy characteristics, age, sex, splenomegaly, prior malignancies, antecedent hematologic disease, presence of RAS mutations, and levels of lactate dehydrogenase and β2-microglobulin. Some prognostic systems have specifically been proposed for patients with CMML [12, 51, 104, 110, 113, 114]. Most recently, Such et al. described the CMML-specific prognostic scoring system (CPSS), which relies on the WHO subclassification, the FAB subclassification, CMML-specific cytogenetic classification (low risk = normal and isolated −Y; intermediate risk = all other abnormalities not included in low or high risk; and high risk = trisomy 8, complex, and chromosome 7 abnormalities), and erythrocyte transfusion dependency [110]. Another recent and useful risk model, the Global MD Anderson Prognostic Scoring System (GMDAPS), was proposed for all MDS and CMML patients, regardless of specific disease type, prior therapy, or duration of disease. G-MDAPS is based on performance status, age, platelet count, hemoglobin value, percentage bone marrow blasts, white blood cell count, karyotype, and prior transfusion [111]. CPSS and G-MDAPS are summarized in Table 2. A refined MDAPS score, more specific for patients with CMML, was recently proposed [114]. It is also reported that therapy-related CMML carries a worse prognosis than de novo CMML [115].
Table 2.
CPSS (0-5 points) | |||
---|---|---|---|
Characteristic for score |
|||
Variable | 0 | 1 | 2 |
WHO subtype | CMML-1 | CMML-2 | - |
FAB subtype | MD-CMML | MP-CMML | - |
CMML-specific cytogenetic risk |
Low | Int | High |
RBC transfusion dependence |
No | Yes | - |
| |||
Risk groups for overall scores | |||
Low | 0 | ||
Intermediate-1 | 1 | ||
Intermediate-2 | 2 | ||
High | 3 |
G-MDAPS (0-17 points) | |
---|---|
Prognostic factor | Points |
Performance status ≥ 2 | 2 |
Age, yr | |
60-64 | 1 |
≥ 65 | 2 |
Platelets, × 109/L | |
<30 | 3 |
30-49 | 2 |
50-199 | 1 |
Hemoglobin < 12g/dL | 2 |
Bone marrow blasts, % | |
5-10 | 1 |
11-29 | 2 |
WBC > 20 × 109/L | 2 |
Karyotype: Chr7 abnormality or ≥ 3 abnormalities | 3 |
Prior transfusion | 1 |
Risk groups for overall scores | |
Low | 0-4 |
Intermediate-1 | 5-6 |
Intermediate-2 | 7-8 |
High | ≥9 |
CMML, chronic myelomonocytic leukemia; CPSS, CMML-specific prognostic scoring system; G-MDAPS, Global MD Anderson Prognostic Scoring system; WHO, World Health Organization; FAB, French-American-British; MD-CMML, myelodysplastic CMML; MP-CMML, myeloproliferative CMML; RBC, red blood cell; WBC, white blood cells; Chr7, chromosome 7.
4.2. Impact of cytogenetic and molecular studies on prognosis
4.2.1. Cytogenetics and prognosis
In a study using data from the Spanish Registry of Myelodysplastic Syndromes, CMML patients with an abnormal karyotype (27%) had a higher leukemia transformation rate (26% vs. 12% at 2 years and 36% vs. 27% at 5 years; p-value, 0.01) and shorter OS (median 16 vs. 36 months, p-value < 0.01) [29]. Three independent risk groups were created on the basis of cytogenetic abnormalities: low risk (-Y, diploid/normal), high risk (trisomy 8, chromosome 7 abnormalities, complex cytogenetic abnormalities), and intermediate risk (all other abnormalities). In a multivariate analysis based on these risk groups, cytogenetic stratification was identified as an independent prognostic factor for OS in patients with CMML [29]. This finding confirmed those of other studies showing the usefulness of cytogenetic risk stratification for prognosticating CMML outcomes [12, 109]. A subsequent confirmatory study determined that CMML patients who had abnormal karyotype (30%) had a higher rate of 2-year transformation to AML (29% vs. 14%, p <0.001), and shorter OS (median 19 vs. 33 months, p-value < 0.0001) [30]. Monosomal karyotype has been independently shown to confer worse prognosis in patients with CMML [116].
4.2.2. Molecular characterization and prognosis
Future prognostic scoring systems for CMML will likely use molecular analysis in combination with clinical and cytogenetic data. Mutational analysis is especially important considering that approximately 70% of CMML patients have normal diploid cytogenetics [29-31]. For example, TET2 mutations may not offer prognostic information except for certain patients with CMML-1, and SETBP1 mutations are associated with poor prognosis [53, 54, 117, 118]. CBL mutations in both CMML-1 and CMML-2 are associated with poorer outcomes, and CBL mutant patients tended to have splenomegaly [52]. Although FLT3 mutations occur in approximately one-third of AML patients, they are far less common in CMML (~4%) and do not predict outcomes [57]. Among the spliceosome mutations in SRSF2, SF3B1, and U2AF35 that have been identified in CMML (most commonly in SRSF2), none are prognostic for outcome [56, 58]. RUNX1 mutations, on the other hand, are thought to predict transformation to AML [119]. Some recent prognostic models have begun to take into account mutational information, such as the mutational status of ASXL1, which may confer an adverse prognosis when mutated [51, 113, 120]. These models are promising and will need continued streamlining and validation. A challenge to creating a uniform model is the bias created by inclusion or exclusion of patients in studies at different institutions since CMML shares features with both MDS and MPN [113].
5. Treatment
Treatment for CMML has been investigated mainly as part of MDS clinical trials that included patients with the CMML subtype. In the past several years, interest into the use of newer agents for CMML has been growing. Traditionally, patients are started on treatment when they begin to experience disease-related complications such as cytopenias or immunosuppression leading to fever or infection, or significant hematologic abnormalities including worsening transfusion dependence and increasing blast percentage. There are no currently accepted standards for determining the optimal time to initiate therapy for a particular patient. Even as new therapies emerge, supportive care remains integral to the management of patients with CMML. Primary measures include symptom management such as blood product transfusions for anemia. Growth factor support with erythropoietin may be also used, since there is anecdotal evidence that it may be of benefit [121].
5.1. Stem cell transplantation
Currently, the only known curative treatment for CMML is allogeneic hematopoietic stem cell transplantation (HSCT) [122]. For younger patients with good performance status, HSCT may be preferred. However, because most patients with CMML are older than 70 years and are thus poor candidates for stem cell transplantation, alternative treatments have been sought. Recent evidence suggests that the number of additional comorbidities may relate directly to the success or failure of transplantation in CMML [123].
All studies investigating HSCT in CMML have been retrospective. The largest study of its kind used the European Blood and Bone Marrow Transplant (EBMT) database and included 283 patients. At the time of analysis, 38% of the patients were alive and free of disease. In total, 25% of patients experienced a relapse; however, the risk of relapse was lower in patients who experienced grade II-IV acute graft-versus-host disease (GVHD). Of interest, several factors were ruled out as prognostic indicators for relapse-free survival or OS in transplant patients, including CMML subtype, cytogenetic abnormalities, conditioning regimen, use of total body irradiation, stem cell source, T-cell depletion, type or grade of GVHD, age or disease status at transplantation, and HLA (human leukocyte antigen) type or sex of donor. The non-relapse mortality rate was 37% and was found to be lower in patients who had received peripheral blood stem cells or who had undergone transplantation after 2002 [124]. Relapse rates were comparable to those in other studies [123, 125-132]. One earlier study with 21 CMML patients found a disease-free survival rate of 43% with a median follow-up of 6-9 years [132]. In total, those studies showed high treatment-related mortality but suggested the strong possibility of long-term disease-free survival.
Several studies included patients with CMML who received reduced-intensity conditioning with a transplant [124-126, 129, 131]. It has been suggested that this approach is appropriate for patients with CMML who are advanced in age and/or have other significant comorbidities. Relapse-free survival rates for CMML patients receiving HSCT have improved in recent years [15, 122]. This is hypothesized to be the result of advancements in supportive care, reduced-intensity conditioning, and better HLA matching protocols [122]. The utility of treating CMML patients undergoing transplantation in the same manner as MDS transplant patients has yet to be prospectively studied [15].
Some patients received donor lymphocyte infusions (DLI) after transplantation. Of six patients with CMML who received DLI in two studies, two benefited from DLI, achieving long-term complete remission (CR). However, there is no consensus about which CMML patients might benefit from DLI, and the patients who did benefit also had significant chronic GVHD [127, 133].
5.2. Cytotoxic chemotherapy
Cytoreductive therapy has traditionally been used to control early-stage disease. Early attempts to control disease with hydroxyurea were more successful than etoposide, and consequently hydroxyurea is still commonly used [134]. For progressive disease, low-dose cytarabine, a nucleoside analog, has also been used. However, very few studies have looked at cytarabine specifically in CMML, and most included other types of MDS. In cases of transformation into AML, traditional high-dose cytarabine-based regimens that are used for de novo AML are utilized. Other investigational cytotoxic chemotherapies are discussed below.
5.3. Hypomethylating agents
Some recent studies focused on hypomethylating agents for CMML, and several of the studies investigated 5-azacytidine or decitabine [135-141]. Both compounds were approved for CMML by the U.S. Food and Drug Administration. In the European Union, subcutaneous 5-azacytidine was approved for CMML patients not eligible for transplantation who have 10%-29% bone marrow blasts and do not have an MPD. The use of hypomethylating agents is further supported by a recent mutational analysis of 52 patients with CMML demonstrating mutations in genes known to regulate methylation, including TET2, UTX, EZH2, and DNMT3A [52].
Three studies with 18, 19, and 31 CMML patients treated with decitabine found CR rates of 10%, 50%, and 58% and objective response or hematologic improvement rates of 67%, 11%, and 19%, respectively [135-137]. More recently, a study of 39 patients with advanced CMML treated with decitabine found CR in 10% of patients and an overall response rate of 38% (median duration of response 13 months, range 4 to 21 months) [139]. Similarly, a study of 38 CMML patients treated with 5-azacytidine found CR in 11% of patients and HI in 25% of patients (median duration 6.5 months, range 3 to >50 months) [140]. The most common adverse effects were those associated with cytopenias.
5.4. Investigational agents
Investigational agents that have been used for treatment of CMML include topoisomerase I inhibitors, sapacitabine, clofarabine, tyrosine kinase inhibitors, thalidomide/lenalidomide, all-trans retinoic acid, histone deacetylase inhibitors, JAK2 inhibitors, and farnesyltransferase inhibitors. Treatments of CMML with these agents largely reflect the approaches used for MDS, and they are outlined in Table 3.
Table 3.
Reference [#] | Drug class | Specific drug | Other drugs used |
Regimen | No. of CMML patients |
CMML-1, CMML-2 |
Age | Cytogenetics/molec info | Nonhematologic toxicities | Response |
---|---|---|---|---|---|---|---|---|---|---|
Beran, 1996 [142] | Topoisomerase I inhibitor | Topotecan | None | 2 mg/m2 cont IV per day × 5 days repeated every 4-8 weeks |
25 | Not reported | 21 patients (84%) ≥ 60 years |
Diploid 10 (40%), abnormal 5/7 7 (28%), 11q 2 (8%), other 2 (8%) |
Mucositis, GI, neurotoxicity most common |
28% CR |
Beran, 1998 [143] | Topoisomerase I inhibitor | Topotecan | None | 2 mg/m2 cont IV per day × 5 days repeated every 4-8 weeks |
30 | Not reported | Including patients with MDS, 83% were > 60 years |
Not reported for CMML patients |
Mucositis, GI, neurotoxicity most common |
27% CR |
Beran, 1999 [144] | Topoisomerase I inhibitor | Topotecan | Cytarabine | Topotecan 1.25 mg/m2 cont IV per day on days 1-5 + cytarabine 1g/m2 IV over 2 hours on days 1-5 |
27 | Not reported | Median 64 years (range 21-80) for all patients including MDS |
Not reported | Mucositis, diarrhea, rash, nausea/vomiting, fever, infection |
44% CR |
Weihrauch, 2004 [145] |
Topoisomerase I inhibitor | Topotecan | Cytarabine | Topotecan 1.25 mg/m2 cont IV per day on days 1-5 + cytarabine 1g/m2 IV over 2 hours on days 1-5 |
1 | Not reported | Median 62 years (range 32-75) for all patients including MDS |
Not reported | GI, rash, fever, alopecia, pain | Non-CR |
Quintas- Cardama, 2006 [172] |
Topoisomerase I inhibitor | 9-Nitro- camptothecin |
None | 9-NC 2 mg/m2 PO daily × 5 d/wk as tolerated |
32 | 26 (81%) CMML-1, 5 (16%) CMML-2 |
Median 66 years | Diploid, trisomy 8, other reported for responders |
GI (reversible) | 40% ORR, 16% CR |
Ribrag, 2003 [173] |
Topoisomerase I inhibitor | CPT-11 | None | CPT-11 200 mg/m2 IV every 2 weeks |
5 | Not reported | Median 71 years, including patients with MDS (range, 51-77) |
Not reported for individual CMML patients |
TLS with ARF, invasive fungal infection, GI |
1 patient with CMML had disappearance of pleural effusion, splenomegaly and monocytosis but BM blasts > 5% |
Kantarjian, 2010 [148] |
Deoxycytidine analog | Sapacitabine | None | 75 mg PO BID × 7 days every 3-4 weeks |
Not reported |
Not reported | Median 65 years (range, 35-90), including all patients with CMML, AML, MDS, ALL |
Responders: del 13q, diploid, other (noncomplex) |
GI, alopecia, fatigue most common |
3 patients with h/o CMML responded; CMML→AML: CRp (2.2 mos); MDS- CMML: CR (3.3 mos); MDS-CMML: BM CR (2 mos) |
Faderl, 2010 [149] |
Nucleoside analog | Clofarabine | None | After toxicity evaluations, 20 mg/m2 PO daily × 5 days, every 4-8 weeks |
6 | 5 (15%) CMML- 1, 1 (3%) CMML-2 |
Median 70 years (range, 53-86), including all patients with MDS, CMML, AML |
Not reported | GI, rash, elevated LFTs, fatigue, headache most common |
33% CR |
Apperley, 2002 [37] |
Tyrosine kinase inhibitor | Imatinib | None | 400 mg PO daily | 3 adults, 1 child |
Not reported (PDGFRB subtype) |
6, 32, 50, and 68 years | All contained translocations involving Chr 5q33 |
No significant events reported | 100% CR |
Magnusson, 2002 [38] |
Tyrosine kinase inhibitor | Imatinib | None | 400 mg PO daily | 1 | Not reported (PDGFRB subtype) |
29 years | t(5;17)(q33;p13.3) | None | 100% CR |
Giles, 2003 [153] | Tyrosine kinase inhibitor | Semaxanib | None | 145 mg/m2 IV twice weekly | 6 | Not reported | Median 66 years (range, 29-85), including all patients with MPD, CMML, CML-BP, AMM |
Not reported | Abdominal pain, bone pain, dyspnea, headache, fatigue most common |
1 patient (17%) with stable disease >6 months |
Strupp, 2002 [154] |
Immunomodulator | Thalidomide | None | 100-400 mg PO daily | 3 | Not reported | 59, 69, 73 years | Poor, good, and good IPSS risk cytogenetics |
Fatigue, obstipation, edema most common |
33% hematologic improvement, platelets |
Raza, 2001 [155] | Immunomodulator | Thalidomide | None | 100-400 mg PO daily | 4 | Not reported | Median 67 years, including all patients with MDS and CMML |
Not reported for individual CMML patients |
Fatigue, constipation, dyspnea, edema, dizziness, rash, neuropathy, fever, headache, nausea |
0% response |
Raza, 2006 [146] | Immunomodulator followed by topoisomerase I inhibitor |
Thalidomide | Topotecan | Topotecan 1.25 mg/m2 on days 1-5 of a 21-day cycle for 3 cycles, and then evaluated. If blasts were less than 5% or had decreased by more than 50%, the patients were then treated with thalidomide 100 mg/day (dose escalated up to 300 mg/day) for up to 1 year |
6 | Not reported | Median 68 years (range, 52-79), including patients with MDS |
46, X, Y, del(7) (q11.2) in responding patient |
Pain, fatigue, fever, constipation most common |
17% (1 patient) PR with hematologic improvement in erythroid cells |
Bejanyan, 2011 [158] |
Immunomodulator + others | Thalidomide | Arsenic trioxide, dexamethasone, ascorbic acid |
TADA regimen: Thalidomide 50 mg/day × 2 weeks, then 100 mg/day × 6 months; ATO 0.25 mg/kg × 5 days, then 0.25 mg/kg twice per week × 11 weeks; Dex 4 mg/day × 5 days every 4 weeks; AA 1000 mg PO 2-3 hours prior to each ATO dose |
8 | All 8 had CMML-1 |
Median 66 years, including patients with MDS/MPN-u, PMF, and splenomegaly |
Not reported for individual CMML patients |
Fatigue, edema, cough, dyspnea, GI, neuropathy, infection, rash |
38% overall response (1 PR and 2 stable disease) |
Raza, 2008 [157] | Immunomodulator + hypomethylator |
Thalidomide | 5-Azacytadine | 5-Azacytadine 75 mg/m2 SC daily × 5 days every 28 days + thalidomide 50-100 mg PO daily |
4 | Not reported | Median 72 years (range, 49-84), including patients with MDS, AML |
Diploid: 2 patients; 46,xy,r(18)(p11.3q23)[1 0]/46,xy[10] and 47,xx,18[13]/46,xx[7] in the other 2 |
Fever, DVT, GI, infection | 50% with major hematologic improvement (both in patients with diploid CGs) |
Platzbecker, 2007 [159] |
Immunomodulator | Lenalidomide | None | 25 mg/day × 14 days | 1 | CMML-2 | 66 years | Single del(5q)(q31); FISH revealed loss of CSF1R gene at 5q33, and not EGR1 |
None reported | Hematologic improvement |
Buckstein, 2014 [174] |
Immunomodulator | Lenalidomide | Melphalan | Lenalidomide 10 mg PO daily; Melphalan 2 mg PO daily for 21 days of a 28- day cycle |
12 | 10 (83%) CMML-1; 2 (17%) CMML- 2 |
Median 73 (range, 52- 87), including patients with MDS |
Not reported for individual CMML patients |
Infection, neurological | 25% overall response (1 CR, 1 HI-platelets, 1 HI-erythroid); all 3 responders had proliferative CMML-1 |
Cambier, 1996 [175] |
Retinoid | All-trans retinoic acid |
+/− Hydroxyurea (for differentiation syndrome in 6 patients) |
45 mg/m2 daily | 10 | Not reported | Median 66 years (range 57-74) |
Not reported | Differentiation syndrome | 40% had reduction in transfusion requirement or increases in platelet count |
Kuendgen, 2004 [160] |
Histone deacetylase inhibitor +/− retinoid |
Valproic acid | +/− ATRA | VPA administered PO with dose escalation to reach serum concentrations of 346-693 μM during months 1-4; ATRA 80 mg/m2 PO daily in 2 divided doses on days 1-7 every other week during months 3-4 |
1 | Not reported | 68 years | Normal | Vertigo, tremor | Progressive disease after 4 months |
Siitonen, 2007 [161] |
Histone deacetylase inhibitor + retinoid |
Valproic acid | 13-cis retinoic acid + 1,25- dihydroxyvitamin D3 |
VPA administered PO with dose escalation to reach serum concentrations of 500-700 μmol/L; 13-cis retinoic acid 10 mg PO BID; VD3 1 μg PO daily |
4 | 3 (75%) CMML-1, 1 (25%) CMML- 2 |
73, 74, 76 years (CMML-1); 79 years (CMML-2) |
CMML-1 patients: normal; CMML-2 patient: t(1;3), +8 |
Cheilitis, dry skin, fatigue, elevated LFTs, pneumonitis |
CMML-2 patient had stable disease; CMML- 1 patients: 2 with stable disease and 1 with hematologic improvement |
Voso, 2009 [162] | Histone deacetylase inhibitor + hypomethylator |
Valproic acid | 5-Azacytadine +/− ATRA |
VPA 600 to 1,500 mg PO on day 0 to reach a final plasma concentration of >50 μg/mL; then 5-AZA was added at a standard dose of 75 mg/m2 SC daily × 7 days every 4 weeks × 8 cycles. In cases of minor response, stable disease, or failure after four cycles, ATRA was added at 30 mg/m2 PO daily on days 8 to 27 × 4 cycles |
4 | Not reported | Median 70 years (range, 53-83) including patients with MDS |
Not reported for individual CMML patients |
Allergy, cardiovascular, GI, dyspnea, elevated LFTs |
Not reported for individual CMML patients. CR/PR rate for patients who completed 8 cycles (n=26) was 31%. |
Ravoet, 2008 [176] |
Farnesyltransferase inhibitor |
Lonafarnib | None | 200 mg PO BID | 2 | Not reported | 63 and 77 years | Both normal | GI, fever, elevated LFTs | Both with stable disease |
Feldman, 2008 [166] |
Farnesyltransferase inhibitor |
Lonafarnib | None | 200-300 mg PO BID | 35 | 11 were reported as MDS-CMML and 24 as MPD-CMML |
Median 70 years (range, 44-86) including patients with MDS |
Not reported for individual CMML patients |
GI, dyspnea, weakness, dehydration, fever, hypokalemia |
29% overall response (4 HI in MDS-CMML; 3 HI, 1 PR, 2 CR in MPD-CMML) |
Kurzrock, 2003 [167] |
Farnesyltransferase inhibitor |
Tipifarnib | None | Increased from 300 mg PO BID on days 1-21 of a 28-day cycle |
10 | Not reported | Median 66 years (range 50-83), including patients with MDS |
Not reported for individual CMML patients |
Fatigue, rash, GI, elevated LFTs, bone pain |
30% in CMML patients (1 HI and 2 PR) |
Fenaux, 2007 [170] |
Farnesyltransferase inhibitor |
Tipifarnib | None | Increased from 300 mg PO BID on days 1-21 of a 28 day cycle |
17 | 8 (47%) CMML-1, 9 (53%) CMML-2 |
Median 67 years (range 39-86), including patients with MDS |
Not reported for individual CMML patients |
Fever, infection, rash, fatigue | 18% CR (1 CMML- 1 and 2 CMML-2 patients); individual HI, PR for CMML patients not reported, but median survival for all CMML patients was 14.5 months |
Borthakur, 2007 [177] |
Farnesyltransferase inhibitor |
FTS (s-trans, transfarnesylthiosalicylic acid) |
None | 200-600 mg PO BID on days 1-21 of a 28-day cycle |
1 | Not reported | Median 74 years (range 57-85), including patients with AML, MDS, and CML |
Not reported | Diarrhea | HI in CMML patient |
Eghtedar, 2010 [178] |
JAK2 inhibitor | Ruxolitinib | None | 25 mg PO BID | 4 | Not reported | Median 69 years (range, 45–88), including patients with sAML, AML, ALL, MDS, and CML |
Not reported | Elevated LFTs | 75% (3 CMML patients) had clinical benefit; 1 of these (with JAK2 mutation) had reduction of BM blasts <5% |
molec, molecular; cont, continuous; IV, intravenously; 5/7, chromosome 5 or 7; GI, gastrointestinal; 9-NC, 9-Nitro-camptothecin; PO, orally; ORR, overall response rate; TLS, tumor lysis syndrome; ARF, acute renal failure; BID, twice daily; ALL, acute lymphocytic leukemia; del, deletion; h/o, history of; CRp, complete remission without recovery of platelets; LFTs, liver function tests; Chr, chromosome; AMM, agnogenic myeloid metaplaisia; IPSS, international prognostic scoring system for MDS risk; PR, partial remission; ATO, arsenic trioxide; Dex, dexamethasone; AA, ascorbic acid; MDS/MPN-u, MDS/MPN unclassifiable; PMF, primary myelofibrosis; SC, subcutaneously; DVT, deep venous thrombosis; CGs, cytogenetics; FISH, fluorescent in situ hybridization; ATRA, all-trans retinoic acid; VPA, valproic acid; VD3, 1,25-dihydroxyvitamin D3; 5-AZA, 5-azacytidine; HI, hematologic improvement; sAML, post-myeloproliferative disorder acute myeloid leukemia; CML, chronic myeloid leukemia.
The most studied topoisomerase 1 inhibitor is topotecan, which has been used alone, in combination with cytarabine, and in combination with thalidomide [142-146]. Treatment with single-agent topotecan produced a CR rate of 28% in 25 patients with CMML [142], and addition of cytarabine in a combination regimen increased the CR rate to 44% in another study, which included 27 patients with CMML [144].
Sapacitabine is an orally administered deoxycytidine analog whose mechanism of action is distinct from that of cytarabine [147]. In a single study at The University of Texas MD Anderson Cancer Center, two patients with CMML treated with sapacitabine responded with CR or bone marrow CR for 2 and 3.3 months, respectively. A patient whose CMML transformed into AML also responded to sapacitabine with CR without recovery of platelets for 2.2 months. All three patients had been treated previously with decitabine, and the AML patient had also been treated previously with clofarabine [148]. Clofarabine, a second-generation nucleoside analog has also been tested in patients with MDS including those with CMML. In another MD Anderson study, out of 6 patients with CMML, 2 achieved CR [149]. Both clofarabine and sapacitabine are currently used when disease progresses despite the use of hypomethylators [150].
Tyrosine kinase inhibitors have been used specifically to treat CMML associated with a rearrangement of the platelet-derived growth factor receptor beta (PDGFRB) gene, which is a tyrosine kinase receptor in the MAP kinase pathway [37, 38]. Some CMML patients without a PDGFRB rearrangement who were treated with imatinib had no significant response [151, 152]. One study investigated the use of semaxanib, a multikinase inhibitor that selectively targets VEGF receptor 2 tyrosine kinase. Of the 6 patients with CMML in the study, 1 (17%) had stable disease for longer than 6 months. Cytogenetic abnormalities in treated patients were not reported [153]. It is worthwhile to note that aberrant activation of ROS1, a tyrosine kinase, was detected in the CD34+ cell compartment for 70% of CMML patients in one recent study [86].
The immunomodulator thalidomide and its derivative lenalidomide have both been used to treat CMML. In one series, thalidomide was studied in MDS patients, and 3 patients with CMML were included. One patient experienced hematologic improvement, with a platelet response [154]. In another study, none of four patients with CMML had a response [155]. A larger study included 13 CMML patients who received thalidomide alone or in combination with arsenic trioxide, topotecan, or ciprofloxacin plus dexamethasone. The overall response rate was approximately 21%; however, individual responses for patients with CMML were not reported [156].
One study combined two investigational therapies for patients with MDS and included 6 patients with CMML. Patients were treated with topotecan at 1.25 mg/m2 on days 1-5 of a 21-day cycle for three cycles and were then evaluated. If blasts were <5% or had decreased by >50%, the patients were then treated with thalidomide at 100 mg/day (with dose escalation up to 300 mg/day) for up to 1 year. Otherwise, the patients were retreated with topotecan until blast indices had reached the levels required to change the therapy to thalidomide. Of the 6 patients with CMML, 1 (with karyotype 46, XY, del(7)(q11.2)[20]) had a partial response with hematologic improvement in erythroid cells and became transfusion independent. The findings suggested that the combined regimen, which was generally well tolerated, or maintenance therapy with thalidomide once a decrease in blasts is achieved, may be appropriate for selected CMML patients [146].
Two studies that looked at thalidomide in combination with 5-azacytidine or as a part of the TADA regimen (thalidomide, arsenic trioxide, dexamethasone, and ascorbic acid) reported response rates of 50% and 38%, respectively [157, 158]. In the study by Bejanyan et al. the TADA regimen was evaluated also in 15 patients with MDS/MPN-unclassifiable, a large number of difficult-to-classify patients whose clinical characteristics overlap with CMML. In another study, lenalidomide was used to treat CMML-2 in a patient who had a del(5q) cytogenetic abnormality. A single 14-day course of lenalidomide at 25 mg/day, administered in preparation for allogeneic stem cell transplantation, resulted in cytoreduction and blast clearance in the patient [159].
Three studies investigating the histone deacetylase inhibitor valproic acid (VPA) for treatment of MDS and CMML obtained inconclusive results for CMML patients [160-162]. Vorinostat (suberoylanilide hydroxamic acid) is another histone deacetylase inhibitor that was approved by the Food and Drug Administration for cutaneous T-cell lymphoma. It was investigated for treatment of MDS alone, and no CMML patients were included [163]. Notably, the MDS/CMML–derived cell line P39 was inhibited by vorinostat alone (as well as by bortezomib alone) in a preclinical study [164].
As noted, some patients with CMML possess mutations in JAK2, suggesting that constitutive activation of the JAK/STAT pathway may be targetable for therapy. The JAK2 inhibitor ruxolitinib has been tested in a limited number of CMML patients [44]. Additionally, Padron et al. have recently reported a GM-CSF-dependent STAT5 hypersensitivity in up to 90% of CMML patients, with potential therapy targeting GM-CSF and the JAK/STAT pathway [76].
Because CMML cells often carry RAS mutations leading to increased RAS protein activity, the RAS-activation pathway has been the target of potential therapeutics [165]. The RAS-activating enzyme farnesyltransferase has been targeted with specific farnesyltransferase inhibitors (FTIs). In the largest multicenter study to date, 35 CMML patients were treated with the FTI lonafarnib at 200-300 mg twice daily. The overall response rate was 29%, with two patients with CMML-2 achieving CR [166]. Another FTI, tipifarnib, has been investigated in phase 1 and 2 trials, as well as in combination with idarubicin and cytarabine for treatment of MDS [167-170]. In a phase 2 study, 17 patients with CMML-1 or CMML-2 received tipifarnib at a dose of 300-600 mg twice daily, and 3 had CR (18%), with a median survival of 14.5 months [170]. Although myelosuppression was the most commonly observed adverse effect with tipifarnib, it is notable that some CMML patients have had rapid onset of leukocytosis with lonafarnib or tipifarnib treatment, which may be akin to leukemia differentiation syndrome [167, 171].
5.5. Conclusions
Although many therapies have been tested against CMML (additional experimental therapies are detailed in Table 3), much work remains to identify optimal treatment for individual patients. In addition, appropriate response criteria have not previously been defined specifically for patients with MDS/MPN, and as criteria are designed, treatments may be better assessed. As the classification of patients with CMML continues to evolve, so will treatment protocols. Stratification based on patient and disease characteristics has become increasingly important to treatment decisions. With the dawn of more sophisticated assays, such as next-generation sequencing, targeted therapy will be directed at patients who would maximally benefit from them. Simultaneously, an increasing understanding of the underlying mechanisms that drive the pathogenesis of this unique disorder will improve the classification, prognostication, and treatment of patients with CMML.
Acknowledgements
The authors wish to thank Arthur Gelmis for review of the manuscript, and The University of Texas MD Anderson Cancer Center for support.
Funding sources
None.
Biographies
Christopher B. Benton, M.D.
Dr. Christopher B. Benton is currently an Instructor and Advanced Scholar in the Department of Leukemia at The University of Texas MD Anderson Cancer Center. He completed his undergraduate training at Massachusetts Institute of Technology and medical training at Baylor College of Medicine. He completed a combined clinical/research residency at Baylor College of Medicine as a MeRIT scholar (Medical Resident Investigator Track) and hematology/oncology fellowship at MD Anderson Cancer Center. He has published and presented several first author papers in peer-reviewed journals and at international meetings. He was a recipient of the American Society of Hematology Research Training Award for Fellows and a Celgene Future Leaders in Hematology Award. He works as a physician-scientist in myeloid malignancies focused on stem cells that drive leukemia initiation and relapse.
Aziz Nazha, M.D.
Dr. Aziz Nazha, completed his medical school at Tishreen University, Lattakia, Syria in 2007. He completed his internal medicine residency at Thomas Jefferson University affiliated AtlantiCare Regional Medical Center in New Jersey in 2011 followed by Leukemia fellowship at The University of Texas MD Anderson Cancer Center in 2012. He is currently a second-year Fellow at Taussig Cancer Institute, Cleveland Clinic foundation. Dr. Nazha has authored/coauthored over 20 papers published in peer-reviewed journals. His research interest is in leukemia and the development of novel targeted therapies. He is also interested in the quality of life research and the treatment of cancer associated symptoms.
Naveen Pemmaraju, M.D.
Dr. Naveen Pemmaraju is currently an Assistant Professor of Medicine at The University of Texas MD Anderson Cancer Center. He graduated Cum Laude from Tulane University, and received his Medical Degree at The University of Arkansas for Medical Sciences. He completed Internal Medicine residency at Johns Hopkins Hospital and hematology/oncology fellowship at MD Anderson Cancer Center. He has received several honors and awards in the medical arena, including the Apple Award for Excellence in Patient Education and The Kimberly Patterson Fellowship in Leukemia Research at MD Anderson Cancer Center. His clinical and research interests include analyzing and improving patient outcomes as well as developing novel clinical trial designs and targeted therapies for myeloid leukemia patients. He is actively involved in the implementation of several clinical and lab protocols, and has coauthored approximately over 25 published papers and abstracts in peer-reviewed international journals.
Guillermo Garcia-Manero, M.D.
Dr. Guillermo Garcia-Manero is deputy chair for translational research, chief of the Myelodysplastic Syndromes section and professor in the Department of Leukemia at The University of Texas MD Anderson Cancer Center. He is a world leader in translational research, clinical investigation and treatment of myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML), and has built this section into the largest clinical program of its class. Together with Hagop Kantarjian, M.D., Dr. Garcia-Manero is co-leader and co-principal investigator for the AML/MDS initiative in MD Anderson’s comprehensive Moon Shots Program. He serves as co-director of MD Anderson’s DNA Methylation Core and his research focuses on improving outcomes for patients with AML/MDS by advancing our understanding of the molecular basis of these diseases to develop new therapies. He has conducted extensive research developing prognostic models for these cancers, including one to identify apparently low-risk MDS patients who actually have poor prognoses. Dr. Garcia-Manero also serves on the faculty of The University of Texas Graduate School of Biomedical Sciences at Houston, has twice received the MD Anderson Faculty Educator of the Month Award, and serves on multiple advisory committees for MD Anderson trainees. He has authored more than 300 peer-reviewed articles, dozens of invited articles, over 450 abstracts and several book chapters. He has been awarded numerous high-profile honors for his work at MD Anderson, including the Emil J. Frei Award for Excellence in Translational Research, the Faculty Scholar Award and the Physician-Scientist Program Award. The American Society of Hematology bestowed him with the Celgene Young Investigator Award for Clinical Research in Hematology and the American Society of Clinical Oncology honored him with the Career Development Award. After earning his medical degree from the Faculty of Medicine of the University of Zaragoza, Spain, in 1991, he completed a clinical internship and residency (1996) and a clinical fellowship in hematology and medical oncology (1999) at Thomas Jefferson University Hospital in Philadelphia, Pennsylvania. Since joining the faculty of MD Anderson in 1999, he has been the principal investigator, coinvestigator or collaborator for a large number of research projects and protocols, as well as serving as a National Institutes of Health (NIH) ad hoc grant reviewer.
Footnotes
Conflict of interest statement
The authors declare no competing conflicts of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- [1].Foucar K. Myelodysplastic/myeloproliferative neoplasms. American journal of clinical pathology. 2009;132:281–9. doi: 10.1309/AJCPJ71PTVIKGEVT. [DOI] [PubMed] [Google Scholar]
- [2].Saarni MI, Linman JW. Myelomonocytic leukemia: disorderly proliferation of all marrow cells. Cancer. 1971;27:1221–30. doi: 10.1002/1097-0142(197105)27:5<1221::aid-cncr2820270530>3.0.co;2-p. [DOI] [PubMed] [Google Scholar]
- [3].Vardiman JW, Harris NL, Brunning RD. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood. 2002;100:2292–302. doi: 10.1182/blood-2002-04-1199. [DOI] [PubMed] [Google Scholar]
- [4].Zittoun R, Bernadou A, Bilski-Pasquier G, Bousser J. Subacute myelo-monocytic leukemia. Study of 27 cases and review of the literature. La semaine des hôpitaux : organe fondé par l'Association d'enseignement médical des hôpitaux de Paris. 1972;48:1943–56. [PubMed] [Google Scholar]
- [5].Germing U, Gattermann N, Strupp C, Aivado M, Aul C. Validation of the WHO proposals for a new classification of primary myelodysplastic syndromes: a retrospective analysis of 1600 patients. Leukemia research. 2000;24:983–92. doi: 10.1016/s0145-2126(00)00088-6. [DOI] [PubMed] [Google Scholar]
- [6].Geary CG, Catovsky D, Wiltshaw E, Milner GR, Scholes MC, Van Noorden S, et al. Chronic myelomonocytic leukaemia. British journal of haematology. 1975;30:289–302. doi: 10.1111/j.1365-2141.1975.tb00544.x. [DOI] [PubMed] [Google Scholar]
- [7].Germing U, Gattermann N, Minning H, Heyll A, Aul C. Problems in the classification of CMML--dysplastic versus proliferative type. Leukemia research. 1998;22:871–8. doi: 10.1016/s0145-2126(97)00192-6. [DOI] [PubMed] [Google Scholar]
- [8].Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick HR, et al. Proposals for the classification of the acute leukaemias. French-American-British (FAB) co-operative group. British journal of haematology. 1976;33:451–8. doi: 10.1111/j.1365-2141.1976.tb03563.x. [DOI] [PubMed] [Google Scholar]
- [9].Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick HR, et al. Proposals for the classification of the myelodysplastic syndromes. British journal of haematology. 1982;51:189–99. [PubMed] [Google Scholar]
- [10].Bennett JM, Catovsky D, Daniel MT, Flandrin G, Galton DA, Gralnick H, et al. The chronic myeloid leukaemias: guidelines for distinguishing chronic granulocytic, atypical chronic myeloid, and chronic myelomonocytic leukaemia. Proposals by the French-American-British Cooperative Leukaemia Group. British journal of haematology. 1994;87:746–54. doi: 10.1111/j.1365-2141.1994.tb06734.x. [DOI] [PubMed] [Google Scholar]
- [11].Nösslinger T, Reisner R, Grüner H, Tüchler H, Nowotny H, Pittermann E, et al. Dysplastic versus proliferative CMML--a retrospective analysis of 91 patients from a single institution. Leukemia research. 2001;25:741–7. doi: 10.1016/s0145-2126(01)00014-5. [DOI] [PubMed] [Google Scholar]
- [12].Onida F, Kantarjian HM, Smith TL, Ball G, Keating MJ, Estey EH, et al. Prognostic factors and scoring systems in chronic myelomonocytic leukemia: a retrospective analysis of 213 patients. Blood. 2002;99:840–9. doi: 10.1182/blood.v99.3.840. [DOI] [PubMed] [Google Scholar]
- [13].Voglová J, Chrobák L, Neuwirtová R, Malasková V, Straka L. Myelodysplastic and myeloproliferative type of chronic myelomonocytic leukemia--distinct subgroups or two stages of the same disease? Leukemia research. 2001;25:493–9. doi: 10.1016/s0145-2126(00)00159-4. [DOI] [PubMed] [Google Scholar]
- [14].Schuler E, Schroeder M, Neukirchen J, Strupp C, Xicoy B, Kundgen A, et al. Refined medullary blast and white blood cell count based classification of chronic myelomonocytic leukemias. Leukemia research. 2014;38:1413–9. doi: 10.1016/j.leukres.2014.09.003. [DOI] [PubMed] [Google Scholar]
- [15].Bacher U, Haferlach T, Schnittger S, Kreipe H, Kröger N. Recent advances in diagnosis, molecular pathology and therapy of chronic myelomonocytic leukaemia. British journal of haematology. 2011 doi: 10.1111/j.1365-2141.2011.08631.x. [DOI] [PubMed] [Google Scholar]
- [16].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. 2009:937–51. doi: 10.1182/blood-2009-03-209262. [DOI] [PubMed] [Google Scholar]
- [17].Keene P, Mendelow B, Pinto MR, Bezwoda W, MacDougall L, Falkson G, et al. Abnormalities of chromosome 12p13 and malignant proliferation of eosinophils: a nonrandom association. British journal of haematology. 1987;67:25–31. doi: 10.1111/j.1365-2141.1987.tb02291.x. [DOI] [PubMed] [Google Scholar]
- [18].Berkowicz M, Rosner E, Rechavi G, Mamon Z, Neuman Y, Ben-Bassat I, et al. Atypical chronic myelomonocytic leukemia with eosinophilia and translocation (5;12). A new association. Cancer genetics and cytogenetics. 1991;51:277–8. doi: 10.1016/0165-4608(91)90142-h. [DOI] [PubMed] [Google Scholar]
- [19].Lerza R, Castello G, Sessarego M, Cavallini D, Pannacciulli I. Myelodysplastic syndrome associated with increased bone marrow fibrosis and translocation (5;12)(q33;p12.3) British journal of haematology. 1992;82:476–7. doi: 10.1111/j.1365-2141.1992.tb06450.x. [DOI] [PubMed] [Google Scholar]
- [20].Wessels JW, Fibbe WE, van der Keur D, Landegent JE, van der Plas DC, den Ottolander GJ, et al. t(5;12)(q31;p12). A clinical entity with features of both myeloid leukemia and chronic myelomonocytic leukemia. Cancer genetics and cytogenetics. 1993;65:7–11. doi: 10.1016/0165-4608(93)90051-m. [DOI] [PubMed] [Google Scholar]
- [21].Golub TR, Barker GF, Lovett M, Gilliland DG. Fusion of PDGF receptor beta to a novel ets- like gene, tel, in chronic myelomonocytic leukemia with t(5;12) chromosomal translocation. Cell. 1994;77:307–16. doi: 10.1016/0092-8674(94)90322-0. [DOI] [PubMed] [Google Scholar]
- [22].Ross TS, Bernard OA, Berger R, Gilliland DG. Fusion of Huntingtin interacting protein 1 to platelet-derived growth factor beta receptor (PDGFbetaR) in chronic myelomonocytic leukemia with t(5;7)(q33;q11.2) Blood. 1998;91:4419–26. [PubMed] [Google Scholar]
- [23].Suh B, Park TS, Kim JS, Song J, Kim J, Yoo J-H, et al. Chronic myelomonocytic leukemia with der(9)t(1;9)(q11;q34) as a sole abnormality. Annals of clinical and laboratory science. 2009;39:307–12. [PubMed] [Google Scholar]
- [24].Krishnan K, Sheldon S. A new translocation, t(3;6)(q12;24) associated with chronic myelomonocytic leukaemia and marrow fibrosis. Clinical and laboratory haematology. 1996;18:47–9. doi: 10.1111/j.1365-2257.1996.tb00738.x. [DOI] [PubMed] [Google Scholar]
- [25].Satake N, Ishida Y, Otoh Y, Hinohara S, Kobayashi H, Sakashita A, et al. Novel MLL-CBP fusion transcript in therapy-related chronic myelomonocytic leukemia with a t(11;16)(q23;p13) chromosome translocation. Genes, chromosomes & cancer. 1997;20:60–3. [PubMed] [Google Scholar]
- [26].Djordjevic V, Jankovic G, Suvajdzic N, Marisavljevic D, Pantic M, Bogdanovic A, et al. A der(14)t(1;14)(q12;p11) in chronic myelomonocytic leukemia. Cancer genetics and cytogenetics. 2005;160:89–93. doi: 10.1016/j.cancergencyto.2004.12.009. [DOI] [PubMed] [Google Scholar]
- [27].Kakihana K, Kubo F, Wakabayashi S, Kurosu T, Miki T, Murakami N, et al. A novel variant form of MLL-ELL fusion transcript with t(11;19)(q23;p13.1) in chronic myelomonocytic leukemia transforming to acute myeloid leukemia. Cancer genetics and cytogenetics. 2008;184:109–12. doi: 10.1016/j.cancergencyto.2008.04.001. [DOI] [PubMed] [Google Scholar]
- [28].Ballerini P, Struski S, Cresson C, Prade N, Toujani S, Deswarte C, et al. RET fusion genes are associated with chronic myelomonocytic leukemia and enhance monocytic differentiation. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2012;26:2384–9. doi: 10.1038/leu.2012.109. [DOI] [PubMed] [Google Scholar]
- [29].Such E, Cervera J, Costa D, Solé F, Vallespí T, Luño E, et al. Cytogenetic risk stratification in chronic myelomonocytic leukemia. Haematologica. 2011;96:375–83. doi: 10.3324/haematol.2010.030957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].Tang G, Zhang L, Fu B, Hu J, Lu X, Hu S, et al. Cytogenetic risk stratification of 417 patients with chronic myelomonocytic leukemia from a single institution. American journal of hematology. 2014;89:813–8. doi: 10.1002/ajh.23751. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Wassie EA, Itzykson R, Lasho TL, Kosmider O, Finke CM, Hanson CA, et al. Molecular and prognostic correlates of cytogenetic abnormalities in chronic myelomonocytic leukemia: a Mayo Clinic-French Consortium Study. American journal of hematology. 2014;89:1111–5. doi: 10.1002/ajh.23846. [DOI] [PubMed] [Google Scholar]
- [32].Gondek LP, Tiu R, O'Keefe CL, Sekeres MA, Theil KS, Maciejewski JP. Chromosomal lesions and uniparental disomy detected by SNP arrays in MDS, MDS/MPD, and MDS-derived AML. Blood. 2008;111:1534–42. doi: 10.1182/blood-2007-05-092304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Tiu RV, Gondek LP, O'Keefe CL, Elson P, Huh J, Mohamedali A, et al. Prognostic impact of SNP array karyotyping in myelodysplastic syndromes and related myeloid malignancies. Blood. 2011;117:4552–60. doi: 10.1182/blood-2010-07-295857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Dunbar AJ, Gondek LP, O'Keefe CL, Makishima H, Rataul MS, Szpurka H, et al. 250K single nucleotide polymorphism array karyotyping identifies acquired uniparental disomy and homozygous mutations, including novel missense substitutions of c-Cbl, in myeloid malignancies. Cancer Research. 2008;68:10349–57. doi: 10.1158/0008-5472.CAN-08-2754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Jankowska AM, Szpurka H, Tiu RV, Makishima H, Afable M, Huh J, et al. Loss of heterozygosity 4q24 and TET2 mutations associated with myelodysplastic/myeloproliferative neoplasms. Blood. 2009;113:6403–10. doi: 10.1182/blood-2009-02-205690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Tefferi A, Lim K-H, Abdel-Wahab O, Lasho TL, Patel J, Patnaik MM, et al. Detection of mutant TET2 in myeloid malignancies other than myeloproliferative neoplasms: CMML, MDS,MDS/MPN and AML. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2009;23:1343–5. doi: 10.1038/leu.2009.59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Apperley JF, Gardembas M, Melo JV, Russell-Jones R, Bain BJ, Baxter EJ, et al. Response to imatinib mesylate in patients with chronic myeloproliferative diseases with rearrangements of the platelet-derived growth factor receptor beta. New England Journal of Medicine. 2002;347:481–7. doi: 10.1056/NEJMoa020150. [DOI] [PubMed] [Google Scholar]
- [38].Magnusson MK, Meade KE, Nakamura R, Barrett J, Dunbar CE. Activity of STI571 in chronic myelomonocytic leukemia with a platelet-derived growth factor beta receptor fusion oncogene. Blood. 2002;100:1088–91. doi: 10.1182/blood-2002-01-0165. [DOI] [PubMed] [Google Scholar]
- [39].Jelinek J, Oki Y, Gharibyan V. JAK2 mutation 1849G>T is rare in acute leukemias but can be found in CMML, Philadelphia chromosome–negative CML, and megakaryocytic leukemia. 2005. [DOI] [PMC free article] [PubMed]
- [40].Jones AV, Kreil S, Zoi K, Waghorn K, Curtis C, Zhang L, et al. Widespread occurrence of the JAK2 V617F mutation in chronic myeloproliferative disorders. Blood. 2005;106:2162–8. doi: 10.1182/blood-2005-03-1320. [DOI] [PubMed] [Google Scholar]
- [41].Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJP, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7:387–97. doi: 10.1016/j.ccr.2005.03.023. [DOI] [PubMed] [Google Scholar]
- [42].Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365:1054–61. doi: 10.1016/S0140-6736(05)71142-9. [DOI] [PubMed] [Google Scholar]
- [43].Pich A, Riera L, Sismondi F, Godio L, Davico Bonino L, Marmont F, et al. JAK2V617Factivating mutation is associated with the myeloproliferative type of chronic myelomonocytic leukaemia. Journal of clinical pathology. 2009;62:798–801. doi: 10.1136/jcp.2009.065904. [DOI] [PubMed] [Google Scholar]
- [44].Naqvi K, Verstovsek S, Kantarjian H, Ravandi F. A potential role of ruxolitinib in leukemia. Expert opinion on investigational drugs. 2011;20:1159–66. doi: 10.1517/13543784.2011.589383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Ruiz-Argüelles GJ, Ruiz-Delgado GJ, Galo-Hooker E, Sanchez-Sosa S. Ruxolitinib Chronic Myelomonocytic Leukemia-Associated Myelofibrosis: A Case Report. J Bone Marrow Res. 2013;1:2. [Google Scholar]
- [46].Hirsch-Ginsberg C, LeMaistre AC, Kantarjian H, Talpaz M, Cork A, Freireich EJ, et al. RAS mutations are rare events in Philadelphia chromosome-negative/bcr gene rearrangement-negative chronic myelogenous leukemia, but are prevalent in chronic myelomonocytic leukemia. Blood. 1990;76:1214–9. [PubMed] [Google Scholar]
- [47].Padua RA, Carter G, Hughes D, Gow J, Farr C, Oscier D, et al. RAS mutations in myelodysplasia detected by amplification, oligonucleotide hybridization, and transformation. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 1988;2:503–10. [PubMed] [Google Scholar]
- [48].Wang J, Liu Y, Li Z, Wang Z, Tan LX, Ryu M-J, et al. Endogenous oncogenic Nras mutation initiates hematopoietic malignancies in a dose- and cell type-dependent manner. Blood. 2011;118:368–79. doi: 10.1182/blood-2010-12-326058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Bejar R, Stevenson K, Abdel-Wahab O, Galili N, Nilsson B, Garcia-Manero G, et al. Clinical effect of point mutations in myelodysplastic syndromes. New England Journal of Medicine. 2011;364:2496–506. doi: 10.1056/NEJMoa1013343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [50].Gelsi-Boyer V, Trouplin V, Adélaïde J, Bonansea J, Cervera N, Carbuccia N, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. British journal of haematology. 2009;145:788–800. doi: 10.1111/j.1365-2141.2009.07697.x. [DOI] [PubMed] [Google Scholar]
- [51].Itzykson R, Kosmider O, Renneville A, Gelsi-Boyer V, Meggendorfer M, Morabito M, et al. Prognostic Score Including Gene Mutations in Chronic Myelomonocytic Leukemia. Journal of Clinical Oncology. 2013 doi: 10.1200/JCO.2012.47.3314. [DOI] [PubMed] [Google Scholar]
- [52].Jankowska AM, Makishima H, Tiu RV, Szpurka H, Huang Y, Traina F, et al. Mutational spectrum analysis of chronic myelomonocytic leukemia includes genes associated with epigenetic regulation: UTX, EZH2 and DNMT3A. Blood. 2011 doi: 10.1182/blood-2010-10-311019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [53].Laborde RR, Patnaik MM, Lasho TL, Finke CM, Hanson CA, Knudson RA, et al. SETBP1mutations in 415 patients with primary myelofibrosis or chronic myelomonocytic leukemia: independent prognostic impact in CMML. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2013 doi: 10.1038/leu.2013.97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [54].Fernandez-Mercado M, Pellagatti A, Di Genua C, Larrayoz MJ, Winkelmann N, Aranaz P, et al. Mutations in SETBP1 are recurrent in myelodysplastic syndromes and often coexist with cytogenetic markers associated with disease progression. British journal of haematology. 2013 doi: 10.1111/bjh.12491. [DOI] [PubMed] [Google Scholar]
- [55].Visconte V, Rogers HJ, Singh J, Barnard J, Bupathi M, Traina F, et al. SF3B1haploinsufficiency leads to formation of ring sideroblasts in myelodysplastic syndromes. Blood. 2012;120:3173–86. doi: 10.1182/blood-2012-05-430876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Patnaik MM, Lasho TL, Finke CM, Hanson CA, Hodnefield JM, Knudson RA, et al. Spliceosome mutations involving SRSF2, SF3B1, and U2AF35 in chronic myelomonocytic leukemia: prevalence, clinical correlates, and prognostic relevance. American journal of hematology. 2013;88:201–6. doi: 10.1002/ajh.23373. [DOI] [PubMed] [Google Scholar]
- [57].Daver N, Strati P, Jabbour E, Kadia T, Luthra R, Wang S, et al. FLT3 mutations in myelodysplastic syndrome and chronic myelomonocytic leukemia. American journal of hematology. 2013;88:56–9. doi: 10.1002/ajh.23345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [58].Meggendorfer M, Roller A, Haferlach T, Eder C, Dicker F, Grossmann V, et al. SRSF2mutations in 275 cases with chronic myelomonocytic leukemia (CMML) Blood. 2012;120:3080–8. doi: 10.1182/blood-2012-01-404863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [59].Orazi A, Germing U. The myelodysplastic/myeloproliferative neoplasms: myeloproliferative diseases with dysplastic features. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2008;22:1308–19. doi: 10.1038/leu.2008.119. [DOI] [PubMed] [Google Scholar]
- [60].Cazzola M, Malcovati L, Invernizzi R. Myelodysplastic/myeloproliferative neoplasms. Hematology / the Education Program of the American Society of Hematology American Society of Hematology Education Program. 2011;2011:264–72. doi: 10.1182/asheducation-2011.1.264. [DOI] [PubMed] [Google Scholar]
- [61].Mahmood S, Cooper A, Ireland R, Pocock C. Leukaemia cutis with chronic myelomonocytic leukaemia. British journal of haematology. 2009;147:413. doi: 10.1111/j.1365-2141.2009.07683.x. [DOI] [PubMed] [Google Scholar]
- [62].Zinkel SS, Ong CC, Ferguson DO, Iwasaki H, Akashi K, Bronson RT, et al. Proapoptotic BID is required for myeloid homeostasis and tumor suppression. Genes & development. 2003;17:229–39. doi: 10.1101/gad.1045603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [63].Boudard D, Vasselon C, Berthéas M-F, Jaubert J, Mounier C, Reynaud J, et al. Expression and prognostic significance of Bcl-2 family proteins in myelodysplastic syndromes. American journal of hematology. 2002;70:115–25. doi: 10.1002/ajh.10108. [DOI] [PubMed] [Google Scholar]
- [64].Economopoulou C, Pappa V, Papageorgiou S, Kontsioti F, Economopoulou P, Charitidou E, et al. Cell cycle and apoptosis regulatory gene expression in the bone marrow of patients with de novo myelodysplastic syndromes (MDS) Annals of hematology. 2010;89:349–58. doi: 10.1007/s00277-009-0835-2. [DOI] [PubMed] [Google Scholar]
- [65].Yang H, Bueso-Ramos C, DiNardo C, Estecio MR, Davanlou M, Geng Q-R, et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2014;28:1280–8. doi: 10.1038/leu.2013.355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [66].Pruneri G, Bertolini F, Soligo D, Carboni N, Cortelezzi A, Ferrucci PF, et al. Angiogenesis in myelodysplastic syndromes. British journal of cancer. 1999;81:1398–401. doi: 10.1038/sj.bjc.6693515. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [67].Aguayo A, Kantarjian H, Manshouri T, Gidel C, Estey E, Thomas D, et al. Angiogenesis in acute and chronic leukemias and myelodysplastic syndromes. Blood. 2000;96:2240–5. [PubMed] [Google Scholar]
- [68].Wimazal F, Krauth M-T, Vales A, Böhm A, Agis H, Sonneck K, et al. Immunohistochemical detection of vascular endothelial growth factor (VEGF) in the bone marrow in patients with myelodysplastic syndromes: correlation between VEGF expression and the FAB category. Leukemia & Lymphoma. 2006;47:451–60. doi: 10.1080/10428190500353083. [DOI] [PubMed] [Google Scholar]
- [69].Alexandrakis MG, Passam FH, Kyriakou DS, Dambaki C, Katrinakis G, Tsirakis G, et al. Expression of the proliferation-associated nuclear protein MIB-1 and its relationship with microvascular density in bone marrow biopsies of patients with myelodysplastic syndromes. Journal of molecular histology. 2004;35:857–63. doi: 10.1007/s10735-004-2341-0. [DOI] [PubMed] [Google Scholar]
- [70].Alexandrakis MG, Passam FH, Pappa CA, Sfiridaki K, Tsirakis G, Damilakis J, et al. Relation between bone marrow angiogenesis and serum levels of angiogenin in patients with myelodysplastic syndromes. Leukemia research. 2005;29:41–6. doi: 10.1016/j.leukres.2004.05.002. [DOI] [PubMed] [Google Scholar]
- [71].Bellamy WT, Richter L, Sirjani D, Roxas C, Glinsmann-Gibson B, Frutiger Y, et al. Vascular endothelial cell growth factor is an autocrine promoter of abnormal localized immature myeloid precursors and leukemia progenitor formation in myelodysplastic syndromes. Blood. 2001;97:1427–34. doi: 10.1182/blood.v97.5.1427. [DOI] [PubMed] [Google Scholar]
- [72].James C, Ugo V, Le Couedic J-P, Staerk J, Delhommeau F, Lacout C, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434:1144–8. doi: 10.1038/nature03546. [DOI] [PubMed] [Google Scholar]
- [73].Pargade V, Darnige L, Gaussem P. Acquired mutation of JAK2 tyrosine kinase and polycythaemia vera. Annales de biologie clinique. 2006;64:3–9. [PubMed] [Google Scholar]
- [74].Gelsi-Boyer V, Trouplin V, Adélaïde J, Aceto N, Rémy V, Pinson S, et al. Genome profiling of chronic myelomonocytic leukemia: frequent alterations of RAS and RUNX1 genes. BMC cancer. 2008;8:299. doi: 10.1186/1471-2407-8-299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [75].Levine RL, Loriaux M, Huntly BJP, Loh ML, Beran M, Stoffregen E, et al. The JAK2V617Factivating mutation occurs in chronic myelomonocytic leukemia and acute myeloid leukemia, but not in acute lymphoblastic leukemia or chronic lymphocytic leukemia. Blood. 2005;106:3377–9. doi: 10.1182/blood-2005-05-1898. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [76].Padron E, Painter JS, Kunigal S, Mailloux AW, McGraw K, McDaniel JM, et al. GM-CSF-dependent pSTAT5 sensitivity is a feature with therapeutic potential in chronic myelomonocytic leukemia. Blood. 2013;121:5068–77. doi: 10.1182/blood-2012-10-460170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [77].MacKenzie KL, Dolnikov A, Millington M, Shounan Y, Symonds G. Mutant N-ras induces myeloproliferative disorders and apoptosis in bone marrow repopulated mice. Blood. 1999;93:2043–56. [PubMed] [Google Scholar]
- [78].Itzykson R, Kosmider O, Renneville A, Morabito M, Preudhomme C, Berthon C, et al. Clonal architecture of chronic myelomonocytic leukemias. Blood. 2013;121:2186–98. doi: 10.1182/blood-2012-06-440347. [DOI] [PubMed] [Google Scholar]
- [79].Itzykson R, Solary E. An evolutionary perspective on chronic myelomonocytic leukemia. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2013 doi: 10.1038/leu.2013.100. [DOI] [PubMed] [Google Scholar]
- [80].Kosmider O, Gelsi-Boyer V, Ciudad M, Racoeur C, Jooste V, Vey N, et al. TET2 gene mutation is a frequent and adverse event in chronic myelomonocytic leukemia. Haematologica. 2009:1676–81. doi: 10.3324/haematol.2009.011205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [81].Ko M, Bandukwala HS, An J, Lamperti ED, Thompson EC, Hastie R, et al. Ten-Eleven-Translocation 2 (TET2) negatively regulates homeostasis and differentiation of hematopoietic stem cells in mice. Proceedings of the National Academy of Sciences. 2011;108:14566–71. doi: 10.1073/pnas.1112317108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [82].Li Z, Cai X, Cai C, Wang J, Zhang W, Petersen BE, et al. Deletion of Tet2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies. Blood. 2011 doi: 10.1182/blood-2010-12-325241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [83].Quivoron C, Couronné L, Della Valle V, Lopez CK, Plo I, Wagner-Ballon O, et al. TET2inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis. Cancer Cell. 2011;20:25–38. doi: 10.1016/j.ccr.2011.06.003. [DOI] [PubMed] [Google Scholar]
- [84].Moran-Crusio K, Reavie L, Shih A, Abdel-Wahab O, Ndiaye-Lobry D, Lobry C, et al. Tet2loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. Cancer Cell. 2011;20:11–24. doi: 10.1016/j.ccr.2011.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [85].Abdel-Wahab O, Adli M, Lafave LM, Gao J, Hricik T, Shih AH, et al. ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell. 2012:180–93. doi: 10.1016/j.ccr.2012.06.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [86].Cilloni D, Carturan S, Bracco E, Campia V, Rosso V, Torti D, et al. Aberrant activation of ROS1 represents a new molecular defect in chronic myelomonocytic leukemia. Leukemia research. 2013;37:520–30. doi: 10.1016/j.leukres.2013.01.014. [DOI] [PubMed] [Google Scholar]
- [87].Braun BS, Tuveson DA, Kong N, Le DT, Kogan SC, Rozmus J, et al. Somatic activation of oncogenic Kras in hematopoietic cells initiates a rapidly fatal myeloproliferative disorder. Proceedings of the National Academy of Sciences of the United States of America. 2004;101:597–602. doi: 10.1073/pnas.0307203101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [88].Chan IT, Kutok JL, Williams IR, Cohen S, Kelly L, Shigematsu H, et al. Conditional expression of oncogenic K-ras from its endogenous promoter induces a myeloproliferative disease. Journal of Clinical Investigation. 2004;113:528–38. doi: 10.1172/JCI20476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [89].Wang J, Liu Y, Li Z, Du J, Ryu M-J, Taylor PR, et al. Endogenous oncogenic Nras mutation promotes aberrant GM-CSF signaling in granulocytic/monocytic precursors in a murine model of chronic myelomonocytic leukemia. Blood. 2010;116:5991–6002. doi: 10.1182/blood-2010-04-281527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [90].Kong G, Wunderlich M, Yang D, Ranheim EA, Young KH, Wang J, et al. Combined MEK and JAK inhibition abrogates murine myeloproliferative neoplasm. Journal of Clinical Investigation. 2014;124:2762–73. doi: 10.1172/JCI74182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [91].Lagasse E, Weissman IL. bcl-2 inhibits apoptosis of neutrophils but not their engulfment by macrophages. The Journal of experimental medicine. 1994;179:1047–52. doi: 10.1084/jem.179.3.1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [92].Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature. 2001;414:105–11. doi: 10.1038/35102167. [DOI] [PubMed] [Google Scholar]
- [93].Wang J, Li Z, He Y, Pan F, Chen S, Rhodes S, et al. Loss of Asxl1 leads to myelodysplastic syndrome-like disease in mice. Blood. 2014:541–53. doi: 10.1182/blood-2013-05-500272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [94].Abdel-Wahab O, Gao J, Adli M, Dey A, Trimarchi T, Chung YR, et al. Deletion of Asxl1 results in myelodysplasia and severe developmental defects in vivo. J Exp Med. 2013:2641–59. doi: 10.1084/jem.20131141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [95].Dey A, Seshasayee D, Noubade R, French DM, Liu J, Chaurushiya MS, et al. Loss of the tumor suppressor BAP1 causes myeloid transformation. Science. 2012:1541–6. doi: 10.1126/science.1221711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [96].Lamothe B, Lai Y, Hur L, Orozco NM, Wang J, Campos AD, et al. Deletion of TAK1 in the myeloid lineage results in the spontaneous development of myelomonocytic leukemia in mice. PLoS ONE. 2012;7:e51228. doi: 10.1371/journal.pone.0051228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [97].Aucagne R, Droin N, Paggetti J, Lagrange B, Largeot A, Hammann A, et al. Transcription intermediary factor 1γ is a tumor suppressor in mouse and human chronic myelomonocytic leukemia. Journal of Clinical Investigation. 2011;121:2361–70. doi: 10.1172/JCI45213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [98].Ritchie KA, Aprikyan AA, Bowen-Pope DF, Norby-Slycord CJ, Conyers S, Bartelmez S, et al. The Tel-PDGFRbeta fusion gene produces a chronic myeloproliferative syndrome in transgenic mice. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 1999;13:1790–803. doi: 10.1038/sj.leu.2401494. [DOI] [PubMed] [Google Scholar]
- [99].Cain JA, Grisolano JL, Laird AD, Tomasson MH. Complete remission of TEL-PDGFRB-induced myeloproliferative disease in mice by receptor tyrosine kinase inhibitor SU11657. Blood. 2004;104:561–4. doi: 10.1182/blood-2003-11-3801. [DOI] [PubMed] [Google Scholar]
- [100].Ikeda K, Mason PJ, Bessler M. 3'UTR-truncated Hmga2 cDNA causes MPN-like hematopoiesis by conferring a clonal growth advantage at the level of HSC in mice. Blood. 2011;117:5860–9. doi: 10.1182/blood-2011-02-334425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [101].Fütterer A, Campanero MR, Leonardo E, Criado LM, Flores JM, Hernández JM, et al. Dido gene expression alterations are implicated in the induction of hematological myeloid neoplasms. Journal of Clinical Investigation. 2005;115:2351–62. doi: 10.1172/JCI24177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [102].Germing U, Strupp C, Knipp S, Kuendgen A, Giagounidis A, Hildebrandt B, et al. Chronic myelomonocytic leukemia in the light of the WHO proposals. Haematologica. 2007;92:974–7. doi: 10.3324/haematol.11051. [DOI] [PubMed] [Google Scholar]
- [103].Mufti GJ, Stevens JR, Oscier DG, Hamblin TJ, Machin D. Myelodysplastic syndromes: a scoring system with prognostic significance. British journal of haematology. 1985;59:425–33. doi: 10.1111/j.1365-2141.1985.tb07329.x. [DOI] [PubMed] [Google Scholar]
- [104].Worsley A, Oscier DG, Stevens J, Darlow S, Figes A, Mufti GJ, et al. Prognostic features of chronic myelomonocytic leukaemia: a modified Bournemouth score gives the best prediction of survival. British journal of haematology. 1988;68:17–21. doi: 10.1111/j.1365-2141.1988.tb04173.x. [DOI] [PubMed] [Google Scholar]
- [105].Sanz GF, Sanz MA, Vallespí T, Cañizo MC, Torrabadella M, García S, et al. Two regression models and a scoring system for predicting survival and planning treatment in myelodysplastic syndromes: a multivariate analysis of prognostic factors in 370 patients. Blood. 1989;74:395–408. [PubMed] [Google Scholar]
- [106].Aul C, Gattermann N, Heyll A, Germing U, Derigs G, Schneider W. Primary myelodysplastic syndromes: analysis of prognostic factors in 235 patients and proposals for an improved scoring system. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 1992;6:52–9. [PubMed] [Google Scholar]
- [107].Morel P, Hebbar M, Laï JL, Duhamel A, Preudhomme C, Wattel E, et al. Cytogenetic analysis has strong independent prognostic value in de novo myelodysplastic syndromes and can be incorporated in a new scoring system: a report on 408 cases. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 1993;7:1315–23. [PubMed] [Google Scholar]
- [108].Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89:2079–88. [PubMed] [Google Scholar]
- [109].Germing U, Kundgen A, Gattermann N. Risk assessment in chronic myelomonocytic leukemia (CMML) Leukemia & Lymphoma. 2004;45:1311–8. doi: 10.1080/1042819042000207271. [DOI] [PubMed] [Google Scholar]
- [110].Such E, Germing U, Malcovati L, Cervera J, Kuendgen A, Della Porta MG, et al. Development and validation of a prognostic scoring system for patients with chronic myelomonocytic leukemia. Blood. 2013;121:3005–15. doi: 10.1182/blood-2012-08-452938. [DOI] [PubMed] [Google Scholar]
- [111].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. doi: 10.1002/cncr.23697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [112].González-Medina I, Bueno J, Torrequebrada A, López A, Vallespí T, Massagué I. Two groups of chronic myelomonocytic leukaemia: myelodysplastic and myeloproliferative. Prognostic implications in a series of a single center. Leukemia research. 2002;26:821–4. doi: 10.1016/s0145-2126(02)00021-8. [DOI] [PubMed] [Google Scholar]
- [113].Patnaik MM, Padron E, Laborde RR, Lasho TL, Finke CM, Hanson CA, et al. Mayo prognostic model for WHO-defined chronic myelomonocytic leukemia: ASXL1 and spliceosome component mutations and outcomes. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2013;27:1504–10. doi: 10.1038/leu.2013.88. [DOI] [PubMed] [Google Scholar]
- [114].Takahashi K, Pemmaraju N, Kantarjian H, Beran M, Quintas-Cardama A, Cortes J, et al. Refined MD Anderson Prognostic Scoring System (MDAPS-R) for Chronic Myelomonocytic Leukemia (CMML) Clinical lymphoma, myeloma & leukemia. 2013;13:S373–S. [Google Scholar]
- [115].Takahashi K, Pemmaraju N, Strati P, Nogueras-Gonzalez G, Ning J, Bueso-Ramos C, et al. Clinical characteristics and outcomes of therapy-related chronic myelomonocytic leukemia. Blood. 2013 doi: 10.1182/blood-2013-03-491399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [116].Alsahlawi A, Alkhateeb H, Patnaik M, Begna K, Elliott M, Hogan WJ, et al. Monosomal Karyotype Predicts Adverse Prognosis in Patients Diagnosed With Chronic Myelomonocytic Leukemia: A Single-Institution Experience. Clinical lymphoma, myeloma & leukemia. 2014 doi: 10.1016/j.clml.2014.06.007. [DOI] [PubMed] [Google Scholar]
- [117].Smith AE, Mohamedali AM, Kulasekararaj A, Lim Z, Gäken J, Lea NC, et al. Next-generation sequencing of the TET2 gene in 355 MDS and CMML patients reveals low-abundance mutant clones with early origins, but indicates no definite prognostic value. Blood. 2010;116:3923–32. doi: 10.1182/blood-2010-03-274704. [DOI] [PubMed] [Google Scholar]
- [118].Kosmider O, Gelsi-Boyer V, Ciudad M, Racoeur C, Jooste V, Vey N, et al. TET2 gene mutation is a frequent and adverse event in chronic myelomonocytic leukemia. Haematologica. 2009;94:1676–81. doi: 10.3324/haematol.2009.011205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [119].Kuo M-C, Liang D-C, Huang C-F, Shih Y-S, Wu J-H, Lin T-L, et al. RUNX1 mutations are frequent in chronic myelomonocytic leukemia and mutations at the C-terminal region might predict acute myeloid leukemia transformation. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2009;23:1426–31. doi: 10.1038/leu.2009.48. [DOI] [PubMed] [Google Scholar]
- [120].Patnaik MM, Parikh SA, Hanson CA, Tefferi A. Chronic myelomonocytic leukaemia: a concise clinical and pathophysiological review. British journal of haematology. 2014 doi: 10.1111/bjh.12756. [DOI] [PubMed] [Google Scholar]
- [121].Kerridge I, Spencer A, Azzi A, Seldon M. Response to erythropoietin in chronic myelomonocytic leukaemia. Internal medicine journal. 2001;31:371–2. doi: 10.1046/j.1445-5994.2001.00074.x. [DOI] [PubMed] [Google Scholar]
- [122].Cheng H, Kirtani VG, Gergis U. Current status of allogeneic HST for chronic myelomonocytic leukemia. Bone marrow transplantation. 2011 doi: 10.1038/bmt.2011.141. [DOI] [PubMed] [Google Scholar]
- [123].Eissa H, Gooley TA, Sorror ML, Nguyen F, Scott BL, Doney K, et al. Allogeneic hematopoietic cell transplantation for chronic myelomonocytic leukemia: relapse-free survival is determined by karyotype and comorbidities. Biology of blood and marrow transplantation: journal of the American Society for Blood and Marrow Transplantation. 2011;17:908–15. doi: 10.1016/j.bbmt.2010.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [124].Symeonidis A, van Biezen A, Mufti G, Finke J, Beelen D, Bornhauser M, et al. Allogeneic stem cell transplantation in patients with chronic myelomonocytic leukaemia: the impact of WHO classification and of the conditioning regimen on the transplantation outcome. Bone marrow transplantation. 2010;45:S241. [Google Scholar]
- [125].Krishnamurthy P, Lim ZY, Nagi W, Kenyon M, Mijovic A, Ireland R, et al. Allogeneic haematopoietic SCT for chronic myelomonocytic leukaemia: a single-centre experience. Bone marrow transplantation. 2010;45:1502–7. doi: 10.1038/bmt.2009.375. [DOI] [PubMed] [Google Scholar]
- [126].Ocheni S, Kröger N, Zabelina T, Zander AR, Bacher U. Outcome of allo-SCT for chronic myelomonocytic leukemia. Bone marrow transplantation. 2009;43:659–61. doi: 10.1038/bmt.2008.366. [DOI] [PubMed] [Google Scholar]
- [127].Elliott MA, Tefferi A, Hogan WJ, Letendre L, Gastineau DA, Ansell SM, et al. Allogeneic stem cell transplantation and donor lymphocyte infusions for chronic myelomonocytic leukemia. Bone marrow transplantation. 2006;37:1003–8. doi: 10.1038/sj.bmt.1705369. [DOI] [PubMed] [Google Scholar]
- [128].Kerbauy DMB, Chyou F, Gooley T, Sorror ML, Scott B, Pagel JM, et al. Allogeneic hematopoietic cell transplantation for chronic myelomonocytic leukemia. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation. 2005;11:713–20. doi: 10.1016/j.bbmt.2005.05.008. [DOI] [PubMed] [Google Scholar]
- [129].Mittal P, Saliba RM, Giralt SA, Shahjahan M, Cohen AI, Karandish S, et al. Allogeneic transplantation: a therapeutic option for myelofibrosis, chronic myelomonocytic leukemia and Philadelphia-negative/BCR-ABL-negative chronic myelogenous leukemia. Bone marrow transplantation. 2004;33:1005–9. doi: 10.1038/sj.bmt.1704472. [DOI] [PubMed] [Google Scholar]
- [130].Kröger N, Zabelina T, Guardiola P, Runde V, Sierra J, van Biezen A, et al. Allogeneic stem cell transplantation of adult chronic myelomonocytic leukaemia. A report on behalf of the Chronic Leukaemia Working Party of the European Group for Blood and Marrow Transplantation (EBMT) British journal of haematology. 2002;118:67–73. doi: 10.1046/j.1365-2141.2002.03552.x. [DOI] [PubMed] [Google Scholar]
- [131].Jurado M, Deeg H, Storer B, Anasetti C. … advanced myelodysplastic syndrome after conditioning with busulfan and fractionated total body irradiation is associated with low relapse rate but considerable …. … marrow transplantation. 2002. [DOI] [PubMed]
- [132].Zang DY, Deeg HJ, Gooley T, Anderson JE, Anasetti C, Sanders J, et al. Treatment of chronic myelomonocytic leukaemia by allogeneic marrow transplantation. British journal of haematology. 2000;110:217–22. doi: 10.1046/j.1365-2141.2000.02133.x. [DOI] [PubMed] [Google Scholar]
- [133].Depil S, Deconinck E, Milpied N, Sutton L, Witz F, Jouet JP, et al. Donor lymphocyte infusion to treat relapse after allogeneic bone marrow transplantation for myelodysplastic syndrome. Bone marrow transplantation. 2004;33:531–4. doi: 10.1038/sj.bmt.1704381. [DOI] [PubMed] [Google Scholar]
- [134].Wattel E, Guerci A, Hecquet B, Economopoulos T, Copplestone A, Mahé B, et al. A randomized trial of hydroxyurea versus VP16 in adult chronic myelomonocytic leukemia. Groupe Français des Myélodysplasies and European CMML Group. Blood. 1996;88:2480–7. [PubMed] [Google Scholar]
- [135].Kantarjian H, Oki Y, Garcia-Manero G, Huang X, O'Brien S, Cortes J, et al. Results of a randomized study of 3 schedules of low-dose decitabine in higher-risk myelodysplastic syndrome and chronic myelomonocytic leukemia. Blood. 2007;109:52–7. doi: 10.1182/blood-2006-05-021162. [DOI] [PubMed] [Google Scholar]
- [136].Aribi A, Borthakur G, Ravandi F, Shan J, Davisson J, Cortes J, et al. Activity of decitabine, a hypomethylating agent, in chronic myelomonocytic leukemia. Cancer. 2007;109:713–7. doi: 10.1002/cncr.22457. [DOI] [PubMed] [Google Scholar]
- [137].Wijermans PW, Rüter B, Baer MR, Slack JL, Saba HI, Lübbert M. Efficacy of decitabine in the treatment of patients with chronic myelomonocytic leukemia (CMML) Leukemia research. 2008;32:587–91. doi: 10.1016/j.leukres.2007.08.004. [DOI] [PubMed] [Google Scholar]
- [138].Oki Y, Jelinek J, Shen L, Kantarjian HM, Issa J-PJ. Induction of hypomethylation and molecular response after decitabine therapy in patients with chronic myelomonocytic leukemia. Blood. 2008;111:2382–4. doi: 10.1182/blood-2007-07-103960. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [139].Braun T, Itzykson R, Renneville A, de Renzis B, Dreyfus F, Laribi K, et al. Molecular predictors of response to decitabine in advanced chronic myelomonocytic leukemia: a phase II trial. Blood. 2011 doi: 10.1182/blood-2011-05-352039. [DOI] [PubMed] [Google Scholar]
- [140].Costa R, Abdulhaq H, Haq B, Shadduck RK, Latsko J, Zenati M, et al. Activity of azacitidine in chronic myelomonocytic leukemia. Cancer. 2011;117:2690–6. doi: 10.1002/cncr.25759. [DOI] [PubMed] [Google Scholar]
- [141].Garcia-Manero G, Gore SD, Cogle C, Ward R, Shi T, Macbeth KJ, et al. Phase I study of oral azacitidine in myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia. Journal of Clinical Oncology. 2011;29:2521–7. doi: 10.1200/JCO.2010.34.4226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [142].Beran M, Kantarjian H, O'Brien S, Koller C, al-Bitar M, Arbuck S, et al. Topotecan, a topoisomerase I inhibitor, is active in the treatment of myelodysplastic syndrome and chronic myelomonocytic leukemia. Blood. 1996;88:2473–9. [PubMed] [Google Scholar]
- [143].Beran M, Estey E, O'Brien SM, Giles FJ, Koller CA, Kornblau S, et al. Results of topotecan single-agent therapy in patients with myelodysplastic syndromes and chronic myelomonocytic leukemia. Leukemia & Lymphoma. 1998;31:521–31. doi: 10.3109/10428199809057611. [DOI] [PubMed] [Google Scholar]
- [144].Beran M, Estey E, O'Brien S, Cortes J. Topotecan and Cytarabine Is an Active Combination Regimen in Myelodysplastic Syndromes and Chronic Myelomonocytic Leukemia. Journal of clinical …. 1999 doi: 10.1200/JCO.1999.17.9.2819. [DOI] [PubMed] [Google Scholar]
- [145].Weihrauch MR, Staib P, Seiberlich B, Hoffmann M, Diehl V, Tesch H. Phase I/II clinical study of topotecan and cytarabine in patients with myelodysplastic syndrome, chronic myelomonocytic leukemia and acute myeloid leukemia. Leukemia & Lymphoma. 2004;45:699–704. doi: 10.1080/10428190310001593175. [DOI] [PubMed] [Google Scholar]
- [146].Raza A, Lisak L, Billmeier J, Pervaiz H, Mumtaz M, Gohar S, et al. Phase II study of topotecan and thalidomide in patients with high-risk myelodysplastic syndromes. Leukemia & Lymphoma. 2006;47:433–40. doi: 10.1080/10428190500353943. [DOI] [PubMed] [Google Scholar]
- [147].Matsuda A, Nakajima Y, Azuma A, Tanaka M, Sasaki T. Nucleosides and nucleotides. 100. 2'-C-cyano-2'-deoxy-1-beta-D-arabinofuranosyl-cytosine (CNDAC): design of a potential mechanism-based DNA-strand-breaking antineoplastic nucleoside. Journal of medicinal chemistry. 1991;34:2917–9. doi: 10.1021/jm00113a034. [DOI] [PubMed] [Google Scholar]
- [148].Kantarjian H, Garcia-Manero G, O'Brien S, Faderl S, Ravandi F, Westwood R, et al. Phase I clinical and pharmacokinetic study of oral sapacitabine in patients with acute leukemia and myelodysplastic syndrome. Journal of Clinical Oncology. 2010;28:285–91. doi: 10.1200/JCO.2009.25.0209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [149].Faderl S, Garcia-Manero G, Estrov Z, Ravandi F, Borthakur G, Cortes JE, et al. Oral clofarabine in the treatment of patients with higher-risk myelodysplastic syndrome. Journal of Clinical Oncology. 2010;28:2755–60. doi: 10.1200/JCO.2009.26.3509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [150].Garcia-Manero G. Treatment of higher-risk myelodysplastic syndrome. Seminars in oncology. 2011;38:673–81. doi: 10.1053/j.seminoncol.2011.07.001. [DOI] [PubMed] [Google Scholar]
- [151].Cortes J. CMML: a biologically distinct myeloproliferative disease. Current hematology reports. 2003;2:202–8. [PubMed] [Google Scholar]
- [152].Cortes J, Giles F, O'Brien S, Thomas D, Albitar M, Rios MB, et al. Results of imatinib mesylate therapy in patients with refractory or recurrent acute myeloid leukemia, high-risk myelodysplastic syndrome, and myeloproliferative disorders. Cancer. 2003;97:2760–6. doi: 10.1002/cncr.11416. [DOI] [PubMed] [Google Scholar]
- [153].Giles FJ, Cooper MA, Silverman L, Karp JE, Lancet JE, Zangari M, et al. Phase II study of SU5416--a small-molecule, vascular endothelial growth factor tyrosine-kinase receptor inhibitor--in patients with refractory myeloproliferative diseases. Cancer. 2003;97:1920–8. doi: 10.1002/cncr.11315. [DOI] [PubMed] [Google Scholar]
- [154].Strupp C, Germing U, Aivado M, Misgeld E, Haas R, Gattermann N. Thalidomide for the treatment of patients with myelodysplastic syndromes. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2002;16:1–6. doi: 10.1038/sj.leu.2402330. [DOI] [PubMed] [Google Scholar]
- [155].Raza A, Meyer P, Dutt D, Zorat F, Lisak L, Nascimben F, et al. Thalidomide produces transfusion independence in long-standing refractory anemias of patients with myelodysplastic syndromes. Blood. 2001;98:958–65. doi: 10.1182/blood.v98.4.958. [DOI] [PubMed] [Google Scholar]
- [156].Candoni A, Raza A, Silvestri F, Lisak L, Galili N, Mumtaz M, et al. Response rate and survival after thalidomide-based therapy in 248 patients with myelodysplastic syndromes. Annals of hematology. 2005;84:479–81. doi: 10.1007/s00277-005-1031-7. [DOI] [PubMed] [Google Scholar]
- [157].Raza A, Mehdi M, Mumtaz M, Ali F, Lascher S, Galili N. Combination of 5-azacytidine and thalidomide for the treatment of myelodysplastic syndromes and acute myeloid leukemia. Cancer. 2008;113:1596–604. doi: 10.1002/cncr.23789. [DOI] [PubMed] [Google Scholar]
- [158].Bejanyan N, Tiu RV, Raza A, Jankowska A, Kalaycio M, Advani A, et al. A phase 2 trial of combination therapy with thalidomide, arsenic trioxide, dexamethasone, and ascorbic acid (TADA) in patients with overlap myelodysplastic/myeloproliferative neoplasms (MDS/MPN) or primary myelofibrosis (PMF) Cancer. 2011 doi: 10.1002/cncr.26741. [DOI] [PubMed] [Google Scholar]
- [159].Platzbecker U, Mohr B, von Bonin M, Binder M, Schetelig J, Ehninger G, et al. Lenalidomide as induction therapy before allogeneic stem cell transplantation in a patient with proliferative CMML-2 and del(5q) not involving the EGR1 locus. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2007;21:2384–5. doi: 10.1038/sj.leu.2404811. [DOI] [PubMed] [Google Scholar]
- [160].Kuendgen A, Strupp C, Aivado M, Bernhardt A, Hildebrandt B, Haas R, et al. Treatment of myelodysplastic syndromes with valproic acid alone or in combination with all-trans retinoic acid. Blood. 2004;104:1266–9. doi: 10.1182/blood-2003-12-4333. [DOI] [PubMed] [Google Scholar]
- [161].Siitonen T, Timonen T, Juvonen E, Terävä V, Kutila A, Honkanen T, et al. Valproic acid combined with 13-cis retinoic acid and 1,25-dihydroxyvitamin D3 in the treatment of patients with myelodysplastic syndromes. Haematologica. 2007;92:1119–22. doi: 10.3324/haematol.11262. [DOI] [PubMed] [Google Scholar]
- [162].Voso MT, Santini V, Finelli C, Musto P, Pogliani E, Angelucci E, et al. Valproic acid at therapeutic plasma levels may increase 5-azacytidine efficacy in higher risk myelodysplastic syndromes. Clinical cancer research : an official journal of the American Association for Cancer Research. 2009;15:5002–7. doi: 10.1158/1078-0432.CCR-09-0494. [DOI] [PubMed] [Google Scholar]
- [163].Garcia-Manero G, Yang H, Bueso-Ramos C, Ferrajoli A, Cortes J, Wierda WG, et al. Phase 1 study of the histone deacetylase inhibitor vorinostat (suberoylanilide hydroxamic acid [SAHA]) in patients with advanced leukemias and myelodysplastic syndromes. Blood. 2008;111:1060–6. doi: 10.1182/blood-2007-06-098061. [DOI] [PubMed] [Google Scholar]
- [164].Galimberti S, Canestraro M, Khan R, Buda G, Orciuolo E, Guerrini F, et al. Vorinostat and bortezomib significantly inhibit WT1 gene expression in MO7-e and P39 cell lines. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2008;22:628–31. doi: 10.1038/sj.leu.2404918. [DOI] [PubMed] [Google Scholar]
- [165].Rowinsky EK, Windle JJ, Von Hoff DD. Ras protein farnesyltransferase: A strategic target for anticancer therapeutic development. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 1999;17:3631–52. doi: 10.1200/JCO.1999.17.11.3631. [DOI] [PubMed] [Google Scholar]
- [166].Feldman EJ, Cortes J, DeAngelo DJ, Holyoake T, Simonsson B, O'Brien SG, et al. On the use of lonafarnib in myelodysplastic syndrome and chronic myelomonocytic leukemia. Leukemia: official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2008;22:1707–11. doi: 10.1038/leu.2008.156. [DOI] [PubMed] [Google Scholar]
- [167].Kurzrock R, Kantarjian HM, Cortes JE, Singhania N, Thomas DA, Wilson EF, et al. Farnesyltransferase inhibitor R115777 in myelodysplastic syndrome: clinical and biologic activities in the phase 1 setting. Blood. 2003;102:4527–34. doi: 10.1182/blood-2002-11-3359. [DOI] [PubMed] [Google Scholar]
- [168].Kurzrock R, Albitar M, Cortes JE, Estey EH, Faderl SH, Garcia-Manero G, et al. Phase II study of R115777, a farnesyl transferase inhibitor, in myelodysplastic syndrome. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2004;22:1287–92. doi: 10.1200/JCO.2004.08.082. [DOI] [PubMed] [Google Scholar]
- [169].Jabbour E, Kantarjian H, Ravandi F, Garcia-Manero G, Estrov Z, Verstovsek S, et al. A phase 1-2 study of a farnesyltransferase inhibitor, tipifarnib, combined with idarubicin and cytarabine for patients with newly diagnosed acute myeloid leukemia and high-risk myelodysplastic syndrome. Cancer. 2011;117:1236–44. doi: 10.1002/cncr.25575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [170].Fenaux P, Raza A, Mufti GJ, Aul C, Germing U, Kantarjian H, et al. A multicenter phase 2 study of the farnesyltransferase inhibitor tipifarnib in intermediate- to high-risk myelodysplastic syndrome. Blood. 2007;109:4158–63. doi: 10.1182/blood-2006-07-035725. [DOI] [PubMed] [Google Scholar]
- [171].Buresh A, Perentesis J, Rimsza L, Kurtin S, Heaton R, Sugrue M, et al. Hyperleukocytosis complicating lonafarnib treatment in patients with chronic myelomonocytic leukemia. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2005;19:308–10. doi: 10.1038/sj.leu.2403569. [DOI] [PubMed] [Google Scholar]
- [172].Quintas-Cardama A, Kantarjian H, O'Brien S, Jabbour E, Giles F, Ravandi F, et al. Activity of 9-nitro-camptothecin, an oral topoisomerase I inhibitor, in myelodysplastic syndrome and chronic myelomonocytic leukemia. Cancer. 2006;107:1525–9. doi: 10.1002/cncr.22186. [DOI] [PubMed] [Google Scholar]
- [173].Ribrag V, Suzan F, Ravoet C, Feremans W, Guerci A, Dreyfus F, et al. Phase II trial of CPT-11 in myelodysplastic syndromes with excess of marrow blasts. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2003;17:319–22. doi: 10.1038/sj.leu.2402726. [DOI] [PubMed] [Google Scholar]
- [174].Buckstein R, Kerbel R, Cheung M, Shaked Y, Chodirker L, Lee CR, et al. Lenalidomide and metronomic melphalan for CMML and higher risk MDS: a phase 2 clinical study with biomarkers of angiogenesis. Leukemia research. 2014;38:756–63. doi: 10.1016/j.leukres.2014.03.022. [DOI] [PubMed] [Google Scholar]
- [175].Cambier N, Wattel E, Menot ML, Guerci A, Chomienne C, Fenaux P. All-trans retinoic acid in adult chronic myelomonocytic leukemia: results of a pilot study. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 1996;10:1164–7. [PubMed] [Google Scholar]
- [176].Ravoet C, Mineur P, Robin V, Debusscher L, Bosly A, André M, et al. Farnesyl transferase inhibitor (lonafarnib) in patients with myelodysplastic syndrome or secondary acute myeloid leukaemia: a phase II study. Annals of hematology. 2008;87:881–5. doi: 10.1007/s00277-008-0536-2. [DOI] [PubMed] [Google Scholar]
- [177].Borthakur G, Ravandi F, O'Brien S. Phase I study of s-trans, trans-farnesylthiosalicylic acid (FTS), a novel oral Ras inhibitor, in patients with refractory hematologic malignancies. Journal of Clinical Oncology. 2007;25:7071. doi: 10.1016/j.clml.2015.02.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [178].Eghtedar A, Verstovsek S, Cortes JE, Estrov Z, Burger JA, Bivins C, et al. Phase II Study of the JAK2 Inhibitor, INCB018424, In Patients with Refractory Leukemias Including Post-Myeloproliferative Disorder (MPD) Acute Myeloid Leukemia (sAML) ASH Annual Meeting Abstracts. 2010;116:509. [Google Scholar]