Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic malignancy that may deserve specific management. Defined by a persistent peripheral blood monocytosis ≥1 × 109/L and monocytes accounting for ≥10% of the white blood cells, this aging-associated disease combines cell proliferation as a consequence of myeloid progenitor hypersensitivity to granulocyte-macrophage colony-stimulating factor with myeloid cell dysplasia and ineffective hematopoiesis. The only curative option for CMML remains allogeneic stem cell transplantation. When transplantation is excluded, CMML is stratified into myelodysplastic (white blood cell count <13 × 109/L) and proliferative (white blood cell count ≥13 × 109/L) CMML. In the absence of poor prognostic factors, the management of myelodysplastic CMML is largely inspired from myelodysplastic syndromes, relying on erythropoiesis-stimulating agents to cope with anemia, and careful monitoring and supportive care, whereas the management of proliferative CMML usually relies on cytoreductive agents such as hydroxyurea, although ongoing studies will help delineate the role of hypomethylating agents in this patient population. In the presence of excessive blasts and other poor prognostic factors, hypomethylating agents are the preferred option, even though their impact on leukemic transformation and survival has not been proved. The therapeutic choice is illustrated by 4 clinical situations among the most commonly seen. Although current therapeutic options can improve patient’s quality of life, they barely modify disease evolution. Improved understanding of CMML pathophysiology will hopefully lead to the exploration of novel targets that potentially would be curative.

In patients with a peripheral blood (PB) monocytosis, defined by a monocyte count >1 × 109/L, a clonal hematopoietic disorder must be considered as soon as reactive causes have been excluded. Chronic myelomonocytic leukemia (CMML), a clonal hematopoietic malignancy characterized by persistent monocytosis, combines myeloid cell proliferation with myeloid cell dysplasia and ineffective hematopoiesis, and has thus been classified by the World Health Organization (WHO) as a myelodysplastic syndrome (MDS)/myeloproliferative neoplasm (MPN).1  This disease, which demonstrates a propensity to transform into acute myeloid leukemia (AML), has long been considered an MDS. Most of the efforts to delineate the specificities of CMML and dedicated therapeutic strategies started recently.

The disease incidence rate ranges between 3.5 and 4.1/1 000 000 per year in the United States and in Europe.2-5  Median age at diagnosis is 72 years, CMML being rare in young adults. Whole genome sequencing of leukemic cell DNA identified 2 age-related molecular signatures,6  supporting the idea that aging is the main cause of the disease. CMML demonstrates a male predominance (2.3 males for 1 female), which is more apparent among older patients. The clinical, biological, and pathological expression of CMML is heterogeneous; latent forms may be neglected and the disease incidence underestimated.

How we diagnose CMML

According to the WHO,1  CMML diagnosis requires a persistent monocytosis with monocytes accounting for ≥10% of the white blood cell (WBC) differential count (Table 1, “WHO-defined criteria”). Other disorders must be excluded: the percentage of blast cells in the bone marrow (BM) and the PB, including myeloblasts, monoblasts, and promonocytes, must be <20%; BCR-ABL1 rearrangement and WHO criteria for essential thrombocytosis, polycythemia vera, and primary myelofibrosis must be absent; in rare persistent monocytosis with eosinophilia, a rearrangement involving PDGFRA (4q12), PDGFRB (5q31-33), FGFR1 (8p11-12), or JAK2 (9p24, PCM1-JAK2 fusion) gene must be excluded cytogenetically. Dysplasia in 1 or more myeloid lineages is commonly observed on BM aspirate, but it can be absent or subtle.1  In this latter situation, the diagnosis may be made if an acquired clonal cytogenetic or molecular genetic abnormality is detected in hematopoietic cells (Figure 1).1  Cytogenetic abnormalities are detected in ∼30% of patients, the most frequent being trisomy 8, loss of chromosome Y, monosomy 7, deletion 7q, trisomy 21, and del(20q).6 

Figure 1.

Our diagnostic workup for CMML diagnosis. ET, essential thrombocytosis; NGS, next-generation sequencing; PV, polycythemia vera; PMF, primary myelofibrosis.

Figure 1.

Our diagnostic workup for CMML diagnosis. ET, essential thrombocytosis; NGS, next-generation sequencing; PV, polycythemia vera; PMF, primary myelofibrosis.

Close modal

Molecular aberrations affect mostly epigenetic, splicing, and signaling genes.7  Typically, the disease genomic fingerprint combines mutations in TET2, SRSF2, and ASXL1 and genes of the RAS signaling pathway (NRAS, KRAS, CBL).8  The association of SRSF2 and TET2 mutations may be highly specific for CMML.9  In clinical practice, sequencing of a limited number of genes detects a clonal abnormality in >90% of cases (Table 2).8-11 

In the absence of myeloid cell dysplasia, CMML is diagnosed when monocytosis persists for at least 3 months and other causes of monocytosis are excluded.1  A flow cytometry assay that analyzes monocyte subsets allows exclusion of a benign reactive monocytosis without waiting for 3 months (Table 1, “Additional criteria”). The expression of CD14 and CD16 distinguishes classical (CD14+/CD16), intermediate (CD14+/CD16+), and nonclassical (CD14/CD16+) monocytes. In healthy conditions, up to 94% of monocytes are classical monocytes. CMML is characterized by an increase in this fraction ≥94%, whereas benign reactive monocytosis is due to the accumulation of the 2 minor subsets (Figure 2).12-14  Of interest, this abnormal monocyte subset repartition can be observed in a fraction of MDS patients, with half of them developing a monocytosis in the following year.15 

Figure 2.

Monocyte subset repartition in patients with a monocytosis. Multiparameter flow cytometry analysis of monocyte (MO) subsets13  in a healthy donor and in the 4 reported cases, showing an increased fraction of classical monocytes (>94%), except in case report 2 in which an associated inflammatory disease alters the characteristic repartition.

Figure 2.

Monocyte subset repartition in patients with a monocytosis. Multiparameter flow cytometry analysis of monocyte (MO) subsets13  in a healthy donor and in the 4 reported cases, showing an increased fraction of classical monocytes (>94%), except in case report 2 in which an associated inflammatory disease alters the characteristic repartition.

Close modal

With the improvement of diagnostic tools, CMML is increasingly recognized following the incidental finding of an elevated PB monocyte count. Clinical expression of CMML otherwise includes varied combinations of constitutional symptoms, clinical effects of cytopenias or spleen size increase, extramedullary myelomonocytic infiltrates, and associated autoimmune and inflammatory diseases.16,17  The PB leukocytosis may include monocyte or neutrophil predominance, anemia and thrombocytopenia are common and sometimes immune related, and leukocytosis requesting urgent therapy and severe neutropenia are rare. In a hypercellular BM, dysplasia is inconstant, blast cell percentage is heterogeneous, and reticulin fibrosis, nodules of plasmacytoid dendritic cells, ring sideroblasts, and mast cell infiltrates can be observed.18-21  This heterogeneous presentation was recently opposed to a lower genetic heterogeneity.22 

The median survival of CMML patients ranges from <1 year in population-based studies of older patients2  to almost 3 years in other subgroups.7,8,11  Leukemic transformation rate varies between 15% and 30%.7,8,11  It is therefore useful to detect at diagnosis the patients at high risk of rapid death or transformation into hard-to-treat AML.

The WHO recognizes 2 prognostic parameters.1  A WBC count ≥13 × 109 g/L separates MPN-CMML, in which the RAS/MAPK signaling pathway is frequently activated, from MDS-CMML, whose outcome is better.1,8  The other is blast cell percentage with 3 groups: CMML-0 (<2% blasts in PB and <5% in BM), CMML-1 (2% to 4% in PB and/or 5% to 9% in BM), and CMML-2 (5% to 19% blasts in PB and/or 10% to 19% in BM and/or Auer rods are present). The distinction between promonocytes, which must be counted as blast cells, dysplastic monocytes, and dysplastic granulocytes requires expertise.23,24 

A number of prognostic scoring systems have been established.8,11,25-32  The International Prognostic Scoring System developed to stratify MDS excludes proliferative CMML.33  A combination of clinical, cytogenetic, and molecular information may improve the accuracy of CMML prognostication.7,8,34,35  Three cytogenetic categories were identified as independent prognostic variables: low risk (normal karyotype or loss of Y), high risk (trisomy 8 or abnormal chromosome 7 or complex karyotype), and intermediate risk (the others).6  These groups were introduced in a CMML-specific prognostic scoring system.31  Interrogation of several models in a common database showed that all the scoring systems that do not include genetic information have comparable performance.36  We have shown that, in addition to older age, elevated WBC, and low platelet count, ASXL1 frameshift or nonsense mutations (but not missense mutations) were an independent predictor of poor survival.8  Recently, cytogenetic abnormalities and mutations in ASXL1, RUNX1, NRAS, and SETBP1 were independently associated with overall survival, providing a genetic score that, combined with red blood cell transfusion dependency, WBC, and marrow blasts, defines 4 risk groups.11  Detailed comparison with previously established prognostication tools using a common, large, and robust data set, is now mandatory to define an international, consensus scoring system. Meanwhile, any of the described stratification tools may be used for clinical decision-making in routine practice.

Except for allogeneic stem cell transplantation (ASCT), there is no disease-modifying treatment in CMML. Current therapies aim to improve symptom burden using a personalized strategy guided by cytopenia-induced or proliferation-associated symptoms. Commonly used drugs include erythropoiesis-stimulating agents (ESAs) in anemic patients, cytoreductive drugs in proliferative diseases, and hypomethylating agents (HMAs) in the most severe CMML, especially when cytopenias predominate (Figure 3). The therapeutic choice is illustrated by 4 common clinical situations.

Figure 3.

Current therapeutic strategies in CMML. Given the clinical and biological heterogeneity of the disease, current treatment goes from a watch-and-wait attitude with active monitoring of symptoms and complications resulting from HMAs or allogeneic stem cell transplantation (transplant), depending on age and comorbidities. Cytoreductive, usually hydroxyurea.

Figure 3.

Current therapeutic strategies in CMML. Given the clinical and biological heterogeneity of the disease, current treatment goes from a watch-and-wait attitude with active monitoring of symptoms and complications resulting from HMAs or allogeneic stem cell transplantation (transplant), depending on age and comorbidities. Cytoreductive, usually hydroxyurea.

Close modal

Case report 1 (part 1)

A 60-year-old man with a medical record of asthma and peptic ulcer was fortuitously diagnosed with isolated monocytosis. His physician observed a stable monocyte count ∼2 × 109/L. After 2 years, the patient was referred to a hematology department. His physical examination was normal. His blood cell count showed a normal WBC count (6.7 × 109/L) and confirmed the monocytosis (1.6 × 109/L) with normal hemoglobin level (135 g/L) and platelet count (184 × 109/L). His BM aspirate was normocellular, showing 1% blasts, 8% monocytes, and minimal signs of dysplasia, including hypolobulated megakaryocytes and some dysgranulopoiesis. His karyotype was normal.

Stable isolated monocytosis

According to the WHO criteria, this patient has a typical CMML-0.1  His age younger than 60 years; the absence of elevated leukocytosis, anemia, or thrombocytopenia; the lack of blast excess in the BM; and circulating immature myeloid cells are good prognostic factors.7,8,36  Flow cytometry analysis of PB monocyte subsets13,14  and molecular analysis of a panel of genes using new-generation sequencing7,8,10  could now complete the evaluation. Given his young age, ASCT could be considered if the disease evolves. In the absence of any progression and poor prognostic factor, we recommend a wait-and-watch attitude because a small fraction of CMML remains stable for many years.

Case report 1 (part 2)

The patient has been seen twice a year in the outpatient unit for 6 years without any therapeutic intervention. His blood cell count remains stable. Flow cytometry analysis of his PB monocytes showed a typical increase in the classical monocyte fraction (Figure 2), and molecular analyses detected mutations in TET2 and SRSF2. These results enforced CMML diagnosis without modifying the watch-and-wait attitude.

Case report 2 (part 1)

An 80-year-old woman was referred for anemia. She had a history of Hashimoto thyroiditis and complained of chronic inflammatory pain in the cervical spine and shoulders. Her blood cell results revealed a normal WBC count (5.1 × 109/L) with a normal neutrophil count (1.87 × 109/L) but an increased monocyte count (1.87 × 109/L), a mild normocytic anemia (hemoglobin 93 g/L, mean cell volume 89 fL), and thrombocytopenia (112 × 109/L). C reactive protein (12 mg/L) and fibrinogen level (4.6 g/L) were increased. Immune tests were negative for antinuclear and anti–double-stranded DNA antibodies and rheumatoid factor. Her serum electrophoresis showed 2 small spikes. Upon physical examination, there was no joint swelling, no spleen or liver enlargement, and no serous effusion or skin lesion. The PB smear showed pseudo-Pelger-Huët neutrophils with hypogranulation, red cell anisocytosis, and poikilocytosis. BM aspirate showed an average cellularity, normal megakaryocytes, lack of monocyte excess (3%), hypogranulated promyelocytes and myelocytes, and a few agranular blast cells (1%). Plasma cells (2%) were morphologically normal. The karyotype of BM cells was normal.

Cytopenias in CMML

At CMML diagnosis, the hemoglobin level is <100 g/L in ∼40% of patients, with 25% having red cell transfusion requirements. Anemia impairs overall survival and is incorporated in diverse scoring systems.8,11  Clinical experience and individual cases suggested that ESAs provided comparable results in MD-CMML and MDS.37,38  A recent retrospective analysis of their efficacy demonstrated that a response was observed in ∼60% of patients and red cell transfusion independence in ∼30%. A better response was detected in anemic patients with low endogenous serum erythropoietin level and transfusion independence.39  Thrombocytopenia is common in CMML patients because of megakaryocyte dysplasia40,41  or autoimmune mechanisms.42-44  The orally bioavailable, small nonpeptide thrombopoietin receptor agonist eltrombopag demonstrates some efficacy. Because safety concerns have been raised,45  this drug should be used only in clinical trials (Table 3).

CMML and lymphoid malignancies

A polyclonal hypergammaglobulinemia is commonly observed in CMML patients,46,47  likely reflecting elevated levels of inflammatory cytokines. The combination of a monoclonal gammopathy of undetermined significance, a multiple myeloma, or a lymphoid malignancy with CMML is rare.47-52  Of note, TET2 mutations can be detected in both myeloid and lymphoid malignancies.53  In TET2-mutated lymphoid malignancies, the detection of a mutated allele in immature myeloid progenitors suggests that a common genetic background could generate both myeloid and lymphoid diseases.54,55 

Case report 2 (part 2)

Flow cytometry analysis of PB monocytes showed that classical monocytes represented only 73.2% of total monocytes, resulting from an increased fraction of intermediate monocytes (Figure 2). Sequencing of monocyte DNA identified somatic mutations in TET2, SRSF2, and RUNX1. A diagnosis of CMML-0 was made. One year later, while polymyalgia rheumatica was controlled by prednisone and a sustained erythroid response to ESA had been obtained, her WBC count increased to 16 × 109/L. Without circulating immature myeloid cells, she developed a splenomegaly, and the thrombocytopenia worsened to 32 × 109/L with easy bruising. A control BM aspirate showed no blast excess or new cytogenetic lesion.

Inflammation, autoimmune diseases, and CMML

Retrospective studies suggest that the prevalence of inflammatory diseases (10% to 30%), which can be diagnosed concomitantly or shortly before or after CMML, is increased.56,57  It remains difficult to determine if they are the ground for CMML or if CMML predisposes to these diseases.58-60  The most frequently observed include systemic vasculitis, connective tissue diseases, polychondritis, seronegative arthritis, and immune thrombocytopenia.17,42-45,61  In such an inflammatory setting, the typical CMML phenotype can be transiently masked by an increased fraction of CD14+,CD16+ intermediate monocytes (Figure 1). When the inflammatory disease is controlled (eg, with steroids), the phenotype comes back to a typical accumulation of classical monocytes. If uncertainty exists, a search for somatic mutations in PB cells by sequencing a panel of frequently mutated genes (Table 3) allows practitioners to confidently determine whether a patient with normal karyotype and subtle dysplasia truly has a clonal disease. First-line treatment of inflammatory and immune diseases consists mostly of steroids, with ∼85% response. Steroid dependence and recurrence are observed in ∼50% of cases. Most additional immunosuppressive therapies may increase the risk of severe cytopenia and infections. However, HMAs, which have immunomodulatory functions, may be efficient and decrease steroid amounts and steroid dependence.43,62  It remains unclear if these events impact CMML overall survival.

Case 3 report (part 1)

A 41-year-old male was referred by a primary care facility for a newly diagnosed CMML. This house painter had been a heavy smoker for 20 years. His father had died of lung cancer, and 1 of his 3 sisters had been treated for breast cancer, but he had no family history of hematopoietic malignancy. The diagnosis of CMML was made after a 3-year follow-up for thrombocytopenia. His PB count showed an increased WBC count (29.6 × 109/L), including monocytosis (4.1 × 109/L) with decreased hemoglobin level (108 g/L) and platelet count (52 × 109/L). Neutrophils were increased (12.7 × 109/L), with 14% immature myeloid cells and 9% peripheral blasts. His physical examination was normal except for bruises. His BM aspirate showed 12% blasts, including promonocytes and features of dysgranulopoiesis; his karyotype was normal.

CMML in younger patients

Although CMML patients younger than 65 years have a better survival,8  median survival remains short (months) and treatment outcome is better with ASCT than with HMAs.63  CMML cases in young adults can develop on a background of germ line mutations in RUNX1,41 GATA2,64 ANKRD26,65  or DDX41.66  Preferred interactions between mutational events may explain the frequent acquisition of somatic ASXL1 on a GATA2-mutated background.67  About 10% of CMMLs develop after exposure to chemotherapies or radiation, with a median latency of 6 years,68  These therapy-related CMMLs could be the accelerated expansion of a clonal hemopoiesis of indeterminate potential.69 

Case 3 report (part 2)

Molecular testing on BM mononucleated cells collected at diagnosis revealed 2 mutations in RUNX1, 1 being a stop gain (variant allele frequency [VAF], 48%) whose somatic origin was uncertain, the other being subclonal. Additional somatic mutations were in TET2, SRSF2, ASXL1, NRAS, and ZRSR2 (VAF, 27% to 44%). He also had somatic mutations in BCOR and PHF6, with VAFs in the 75% to 85% range, compatible with these genes being located on the X chromosome. In the days following referral, the patient’s WBC count increased to 39 g/L. The 2 healthy sisters were haploidentical to the patient. Databases indicated a high probability of finding a matched unrelated donor. Based on BM and PB blast excess, normal cytogenetics and high matched unrelated donor probability, the patient was treated with an intensive chemotherapy regimen. Eight-week assessment showed a hypocellular marrow with the persistence of a blast excess (7%). The patient received fludarabine/melphalan–conditioned ASCT from a matched unrelated donor. Transplant was complicated with grade 2 acute graft-versus-host disease. CMML relapsed 8 months posttransplantation, with a 10% blast excess in the BM.

ASCT

ASCT remains the unique potentially curative therapy and the preferred therapeutic option in younger patients with high-risk CMML. It is therefore essential to assess the disease risk at diagnosis, using WHO criteria and 1 of the currently established scoring systems. A minority of high-risk patients is eligible, owing to advanced age and comorbidities. The advent of reduced intensity conditioning and alternate donor sources may increase the number of transplanted patients. All of the studies so far have been retrospective. Except a few recent reports,70,71  most have included a small number of patients.72-77  Current recommendations are therefore based on expert opinion rather than evidence.78,79  The median age of transplanted patients ranges from 50 to 56 years. About one-third of these patients are alive a few years later. Deaths are due almost equally to treatment-related mortality or posttransplant disease relapse. Indirect evidence for a graft-versus-CMML effect comes from correlations between graft-versus-host disease and reduction in relapse.71  The main risk factor for transplant-related mortality is the disease status. Although there is no consensus about the correct timing of ASCT in CMML patients, transplantation should preferably be performed early after diagnosis and after establishing the best possible remission status.70  There is no evidence whether the use of cytoreductive therapy before transplant could improve the outcome compared with frontline ASCT. Expert opinion suggests treatment before transplantation, especially when marrow blast cells are >10% and in patients with high-risk CMML according to available scoring systems.78,79  The best treatment of reducing tumor burden before transplantation, either intensive chemotherapy or HMAs, remains a controversial issue.80,81  Many experts currently consider pretreatment with an HMA in the case of CMML-2, even though evidence from prospective clinical trials is lacking. Some transplant candidates are eventually not transplanted owing to pretreatment toxicity or disease progression before transplant.

Case report 4 (part 1)

A 75-year-old male with a long history of high blood pressure, arrhythmia, and arteritis was referred for an elevated WBC count (27.6 × 109/L) and thrombocytopenia (50 × 109/L). WBC included a majority of neutrophils (10.5) and monocytes (11.8 × 109/L) including 5% circulating immature myeloid cells. The hemoglobin level was normal (140 g/L). Physical examination detected a splenomegaly (16 cm craniocaudal length by ultrasonography). BM aspiration detected 11% blast cells, including myeloblasts and promonocytes, indicating a CMML-2. Cytogenetic analysis of BM cells showed an isolated monosomy 7. Molecular analyses detected somatic mutations in ASXL1, ETV6, KRAS, NRAS, RIT1, SRSF2, and TET2. The patient was considered ineligible for transplant based on age and comorbidities.

Myeloproliferative CMML

A fraction of CMML patients demonstrate a rise in their peripheral leukocyte count, which can be dramatic without overt transformation into AML. This increase can be transiently majored by an associated infectious or inflammatory disease. In juvenile myelomonocytic leukemia, constitutive activation of the Ras pathway82,83  induces a hypersensitivity of myeloid progenitors to granulocyte-macrophage colony-stimulating factor (GM-CSF) with a specifically evoked STAT5 signature.84  In CMML, most of the disease-associated molecular events also converge within a GM-CSF/pSTAT5 pathway.85,86  Spontaneous formation of colony-forming unit GM at disease diagnosis could be a surrogate parameter of RAS pathway hyperactivation that correlates with a worse outcome.87,88  Therapeutic approaches targeting this signaling pathway are currently being assessed,89  including the use of ruxolitinib that could control inflammatory symptoms (Table 3).90 

Extramedullary disease

MPN-CMML patients can experience constitutional symptoms, including fatigue, diffuse bone pain, and night sweats, and develop a splenomegaly (∼30% of cases), which is occasionally troublesome, resulting from extramedullary hematopoiesis.91  Extramedullary disease, which occasionally infiltrates the skin,92,93  lymph nodes,94,95  gingiva,96  kidneys,97  pericardium,98  and other sites,99,100  could regress upon treatment with hydroxyurea,96  methylprednisolone,101  or HMA.102-104  Nevertheless, splenomegaly is a poor prognostic factor in CMML treated with HMAs.105 

Cytoreductive therapy

Oral cytoreductive agents have long been the main treatment of proliferative CMML. They remain required frontline in patients with initial leukocytosis >50 × 109/L, severe constitutional symptoms, splenomegaly, or extramedullary hematopoiesis.99,105  Even when HMAs are initiated, an initial cytoreduction may be required.106  A randomized trial demonstrated that the outcome was better in patients treated with hydroxyurea compared with oral etoposide.105  Low-risk CMML patients with MPN-like features can be effectively managed with hydroxyurea. There is no formal demonstration that control of myeloproliferative features improves outcome, and molecular studies have shown continuing clonal evolution under hydroxyurea,86  but this phenomenon has also been reported with HMA.6  There is no recommended target WBC or monocyte count, but we usually try to reduce WBC count below 15 × 109/L. Cytarabine is used occasionally as a debulking agent. Induction chemotherapy with anthracyclines and cytarabine or newer regimens including clofarabine have been used to bridge to ASCT.107 

HMAs

Clinical trials that established the efficacy of azacitidine (AZA) and decitabine (DAC) in MDS included a limited number of CMML patients.29,80,108  Small dedicated clinical trials showed overall response rates according to International Working Group (IWG) criteria109  ranging between 30% and 60%, with a complete response in fewer than 15% of patients. Median overall survival ranged between 12 and 37 months.106,110-112  AZA is licensed for nonproliferative CMML-2 in many countries; DAC is licensed in the United States only. These drugs are globally well tolerated, with myelosuppression being the major toxicity. Responses are generally not sustained but, in the absence of alternative, treatment is maintained until progression. Survival after loss of response is dismal.113  Somatic mutations are not predictive of treatment efficacy.106,114  Gene expression pattern, DNA methylation pattern, and platelet doubling after the first cycle could be more informative.106,114-116 

Case 4 report (part 2)

The patient was treated with DAC on a conventional schedule in the ongoing Decitabine with or without Hydroxyurea versus Hydroxyurea in CMML trial (EudraCT: 2014-000200-10). After 6 cycles, he was in complete response according to IWG criteria. After 12 cycles, he developed grade 2 thrombocytopenia at day 28 of each cycle, lowering to 20 g/L at day 28 of cycle 16. The BM aspirate was of average cellularity with few megakaryocytes and persistent dysgranulopoiesis, without excess of blasts or monocytes. Cycle 17 was delayed by 4 weeks, and further cycles spaced every 6 weeks. The patient remains stable after 21 cycles of DAC at 2 years from diagnosis.

The limits of HMA in CMML

Although HMAs demonstrate cytotoxic effects, their epigenetic activity plays a central role in restoring a balanced hematopoiesis. AZA and DAC induce dramatic changes in DNA methylation and gene expression in leukemic cells of responding patients.116  However, they do not reduce the mutated allele burden, nor do they permit the reexpansion of wild-type hematopoietic cells. In addition, the response to AZA or DAC does not prevent the accumulation of genetic damage in the leukemic clone.6 

Response criteria

IWG response criteria for MDS are not completely suited for adult patients with MDS/MPN106  (eg, they do not capture the treatment effects on the proliferative component). Also, scales that have not been specifically developed for CMML barely reflect the drug effects on constitutional symptoms.90  An international consortium recently proposed specific end points to better measure benefit to the patient.21  The proposed criteria may capture meaningful improvements not seen with IWG criteria, but the more stringent definition of complete response leads to divergent response rates compared with IWG criteria.117 

Because there is an unmet need for therapeutic options that modify CMML course and the disease specificities are increasingly identified, new therapeutic approaches are explored (Table 3). One objective may be to restore a wild-type hematopoiesis. Many drugs have been tested in cohorts of MDS to which CMML was added, which may preclude identification of CMML-specific effects.118  Short homogeneous series of CMML patients have been treated with recombinant α-2b interferon, which reduced monocytosis without improving cytopenias,119  or with all-trans retinoic acid, which improved cytopenias without reducing proliferation.120  Homogeneous cohorts of CMML deserving therapy remain difficult to recruit.

Guadecitabine is a dinucleotide of DAC and deoxyguanosine resists deamination by cytidine deaminase. The half-life of DAC after subcutaneous injection of guadecitabine is longer than that of DAC delivered intravenously.121  A phase 2 study is ongoing in patients with high-risk MDS (Table 3). Because HMAs could increase the expression of surface immune checkpoint proteins at the surface of MDS and CMML CD34+ cells,122  a clinical trial explores the potential of immune checkpoint inhibitors in combination with AZA in a cohort of MDS and CMML patients. IDH1 and IDH2 gene mutations are detected in only a small fraction of CMML patients,123  but their presence can be suspected by detecting an elevated level of 2-hydroxyglutarate in the patient serum124 ; the currently developed IDH inhibitors could be an option in these situations.125 

Molecular aberrations detected in CMML converge within the GM-CSF signaling pathway.85  A human GM-CSF fused to a truncated diphtheria toxin selectively kills leukemic progenitors of CMML patients but has never been developed clinically.126  Lenzilumab, a recombinant monoclonal antibody that neutralizes GM-CSF binding to its cognate receptor and could control cell proliferation, is currently being tested.85  GM-CSF hypersensitivity of CMML myeloid progenitors correlates with mutations in genes of the Ras signaling pathway. The farnesyltransferase inhibitor tifiparnib127  is currently being tested in a phase 2 trial. An alternative could be to inhibit Ras effectors that include the MEK/extracellular signal-regulated kinase and the phosphoinositide-3′-OH kinase/Akt cascades.128-130  Chemical inhibitors of JAK2, a sentinel kinase in this pathway,85  could inhibit the growth of CMML myeloid progenitors,131  and ruxolitinib was shown to be safe to decrease the size of the spleen and to induce responses that were not restricted to patients with somatic mutations in signaling genes while improving constitutional symptoms.90  Pacritinib, a JAK2/FLT3 inhibitor, also deserves to be tested in proliferative CMML.132,133 

Because activation of the Hedgehog signaling pathway could contribute to the development of leukemic stem cells, CMML patients can be included in a phase 1 study of a smoothened antagonist.134  Finally, high expression of interleukin-3 receptor (CD123) in leukemic stem cells led exploration of the efficacy of SL-401, a diphtheria toxin-interleukin-3 fusion protein,135  in patients with hematological malignancies including CMML. Another characteristic feature of CMML is the frequent mutations of a splicing gene.6,8  The gain-of-function mutation in SRSF2 results in transcriptome-wide missplicing,136  and cells with a mutated allele are more sensitive to splicing inhibitory molecules than wild-type cells,137  providing a therapeutic window that is explored in clinics.

Regarding cytopenias, transforming growth factor-β modulators such as sotatercept are explored to treat anemia,138  whereas trials testing eltrombopag in thrombocytopenic patients are under way. Proliferative CMML could develop leukocytosis with circulating blast cells upon therapy, an effect whose long-term consequences has to be evaluated.62 

Finally, molecules that target the mature cells of the clone have been identified preclinically. An adenosine-containing cyclic dinucleotide induces the selective apoptosis of monocytes, in vitro and in vivo,139  whereas trabectedin, a DNA minor groove binder that has been tested in solid tumor patients, could selectively deplete myelomonocytic cells by inhibiting their growth and triggering their apoptosis.140 

Stepwise linear regression models suggest that somatic mutations account for only 15% to 24% of variability of clinical phenotypes.11  These mutations occur in the hematopoietic stem and progenitor cell compartment and accumulate linearly in these cells with clonal dominance in this compartment.141  Therapies may aim at eradicating the founder mutant clones in this cell compartment and restore a wild-type hematopoiesis. Azanucleosides restore a balanced hematopoiesis but fail to decrease the mutation allele burden in leukemic cells, suggesting that epigenetic alterations account for CMML phenotypes.6  Mature cells of the leukemic clone, including monocytes, dysplastic granulocytes, and others,22  may contribute to disease installation or progression through the cytokines they produce.142-144  Finally, the role played by the microenvironment145  and the macroenvironment, including gut microbiota146  in this disease, remains unknown. Current therapeutic approaches improve the patient quality of life, but we have still to identify a disease-modifying therapeutic approach.

The authors thank the patients with chronic myelomonocytic leukemia who agree to participate in their investigations and the physicians who collect information and send biological samples for analyses. Most of these physicians are members of the Groupe Francophone des Myelodysplasies. The authors are also grateful to Dorothée Selimoglu-Buet for providing illustrations of flow cytometry analyses and to Jeffie Lafosse for collecting clinical information.

This work is supported by the Ligue Nationale Contre le Cancer (équipe labellisée).

Contribution: E.S. and R.I. conceived and wrote the manuscript.

Conflict-of-interest disclosure: E.S. received research grants from CIT Pharma and Servier. R.I. declares no competing financial interests.

Correspondence: Eric Solary, Gustave Roussy Cancer Center, 114 rue Edouard Vaillant, 94805 Villejuif, France; e-mail: eric.solary@gustaveroussy.fr; or Raphael Itzykson, Hôpital Saint-Louis, 1, Ave Claude Vellefaux, 75010 Paris, France; e-mail: raphael.itzykson@aphp.fr.

1.
Arber
DA
,
Orazi
A
,
Hasserjian
R
, et al
.
The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia
.
Blood
.
2016
;
127
(
20
):
2391
-
2405
.
2.
Guru Murthy
GS
,
Dhakal
I
,
Mehta
P
.
Incidence and survival outcomes of chronic myelomonocytic leukemia in the United States
.
Leuk Lymphoma
.
2017
;
58
(
7
):
1648
-
1654
.
3.
Srour
SA
,
Devesa
SS
,
Morton
LM
, et al
.
Incidence and patient survival of myeloproliferative neoplasms and myelodysplastic/myeloproliferative neoplasms in the United States, 2001-12
.
Br J Haematol
.
2016
;
174
(
3
):
382
-
396
.
4.
Maynadié
M
,
Girodon
F
,
Manivet-Janoray
I
, et al
.
Twenty-five years of epidemiological recording on myeloid malignancies: data from the specialized registry of hematologic malignancies of Cote d’Or (Burgundy, France)
.
Haematologica
.
2011
;
96
(
1
):
55
-
61
.
5.
Sant
M
,
Allemani
C
,
Tereanu
C
, et al
;
HAEMACARE Working Group
.
Incidence of hematologic malignancies in Europe by morphologic subtype: results of the HAEMACARE project
.
Blood
.
2010
;
116
(
19
):
3724
-
3734
.
6.
Such
E
,
Cervera
J
,
Costa
D
, et al
.
Cytogenetic risk stratification in chronic myelomonocytic leukemia
.
Haematologica
.
2011
;
96
(
3
):
375
-
383
.
7.
Merlevede
J
,
Droin
N
,
Qin
T
, et al
.
Mutation allele burden remains unchanged in chronic myelomonocytic leukaemia responding to hypomethylating agents
.
Nat Commun
.
2016
;
7
:
10767
.
8.
Itzykson
R
,
Kosmider
O
,
Renneville
A
, et al
.
Prognostic score including gene mutations in chronic myelomonocytic leukemia
.
J Clin Oncol
.
2013
;
31
(
19
):
2428
-
2436
.
9.
Malcovati
L
,
Papaemmanuil
E
,
Ambaglio
I
, et al
.
Driver somatic mutations identify distinct disease entities within myeloid neoplasms with myelodysplasia
.
Blood
.
2014
;
124
(
9
):
1513
-
1521
.
10.
Mughal
TI
,
Cross
NC
,
Padron
E
, et al
.
An International MDS/MPN Working Group’s perspective and recommendations on molecular pathogenesis, diagnosis and clinical characterization of myelodysplastic/myeloproliferative neoplasms
.
Haematologica
.
2015
;
100
(
9
):
1117
-
1130
.
11.
Elena
C
,
Gallì
A
,
Such
E
, et al
.
Integrating clinical features and genetic lesions in the risk assessment of patients with chronic myelomonocytic leukemia
.
Blood
.
2016
;
128
(
10
):
1408
-
1417
.
12.
Vuckovic
S
,
Fearnley
DB
,
Gunningham
S
,
Spearing
RL
,
Patton
WN
,
Hart
DN
.
Dendritic cells in chronic myelomonocytic leukaemia
.
Br J Haematol
.
1999
;
105
(
4
):
974
-
985
.
13.
Selimoglu-Buet
D
,
Wagner-Ballon
O
,
Saada
V
, et al
;
Francophone Myelodysplasia Group
.
Characteristic repartition of monocyte subsets as a diagnostic signature of chronic myelomonocytic leukemia
.
Blood
.
2015
;
125
(
23
):
3618
-
3626
.
14.
Talati
C
,
Zhang
L
,
Shaheen
G
, et al
.
Monocyte subset analysis accurately distinguishes CMML from MDS and is associated with a favorable MDS prognosis
.
Blood
.
2017
;
129
(
13
):
1881
-
1883
.
15.
Selimoglu-Buet
D
,
Badaoui
B
,
Benayoun
E
, et al
.
Accumulation of classical monocytes defines a subgroup of MDS that frequently evolve into CMML [published online ahead of print 13 June 2017]
.
Blood
.
doi:10.1182/blood-2017-04-779579
.
16.
McCullough
KB
,
Patnaik
MM
.
Chronic myelomonocytic leukemia: a genetic and clinical update
.
Curr Hematol Malig Rep
.
2015
;
10
(
3
):
292
-
302
.
17.
Grignano
E
,
Mekinian
A
,
Braun
T
, et al
;
GFM, SNFMI and CRI
.
Autoimmune and inflammatory diseases associated with chronic myelomonocytic leukemia: a series of 26 cases and literature review
.
Leuk Res
.
2016
;
47
:
136
-
141
.
18.
Bennett
JM
,
Catovsky
D
,
Daniel
MT
, et al
;
Proposals by the French-American-British Cooperative Leukaemia Group
.
The chronic myeloid leukaemias: guidelines for distinguishing chronic granulocytic, atypical chronic myeloid, and chronic myelomonocytic leukaemia
.
Br J Haematol
.
1994
;
87
(
4
):
746
-
754
.
19.
Qubaja
M
,
Marmey
B
,
Le Tourneau
A
, et al
.
The detection of CD14 and CD16 in paraffin-embedded bone marrow biopsies is useful for the diagnosis of chronic myelomonocytic leukemia
.
Virchows Arch
.
2009
;
454
(
4
):
411
-
419
.
20.
Naresh
KN
,
Pavlu
J
.
Plasmacytoid dendritic cell nodules in bone marrow biopsies of chronic myelomonocytic leukemia
.
Am J Hematol
.
2010
;
85
(
11
):
893
.
21.
Ji
P
,
Peterson
LC
.
Plasmacytoid dendritic cells in chronic myelomonocytic leukemia
.
Blood
.
2014
;
123
(
21
):
3220
.
22.
Ball
M
,
List
AF
,
Padron
E
.
When clinical heterogeneity exceeds genetic heterogeneity: thinking outside the genomic box in chronic myelomonocytic leukemia
.
Blood
.
2016
;
128
(
20
):
2381
-
2387
.
23.
Droin
N
,
Jacquel
A
,
Hendra
JB
, et al
.
Alpha-defensins secreted by dysplastic granulocytes inhibit the differentiation of monocytes in chronic myelomonocytic leukemia
.
Blood
.
2010
;
115
(
1
):
78
-
88
.
24.
Goasguen
JE
,
Bennett
JM
,
Bain
BJ
,
Vallespi
T
,
Brunning
R
,
Mufti
GJ
;
International Working Group on Morphology of Myelodysplastic Syndrome
.
Morphological evaluation of monocytes and their precursors
.
Haematologica
.
2009
;
94
(
7
):
994
-
997
.
25.
Worsley
A
,
Oscier
DG
,
Stevens
J
, et al
.
Prognostic features of chronic myelomonocytic leukaemia: a modified Bournemouth score gives the best prediction of survival
.
Br J Haematol
.
1988
;
68
(
1
):
17
-
21
.
26.
Onida
F
,
Kantarjian
HM
,
Smith
TL
, et al
.
Prognostic factors and scoring systems in chronic myelomonocytic leukemia: a retrospective analysis of 213 patients
.
Blood
.
2002
;
99
(
3
):
840
-
849
.
27.
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
.
Leuk Res
.
2002
;
26
(
9
):
821
-
824
.
28.
Germing
U
,
Kündgen
A
,
Gattermann
N
.
Risk assessment in chronic myelomonocytic leukemia (CMML)
.
Leuk Lymphoma
.
2004
;
45
(
7
):
1311
-
1318
.
29.
Beran
M
,
Wen
S
,
Shen
Y
, et al
.
Prognostic factors and risk assessment in chronic myelomonocytic leukemia: validation study of the M.D. Anderson Prognostic Scoring System
.
Leuk Lymphoma
.
2007
;
48
(
6
):
1150
-
1160
.
30.
Kantarjian
H
,
O’Brien
S
,
Ravandi
F
, et al
.
Proposal for a new risk model in myelodysplastic syndrome that accounts for events not considered in the original International Prognostic Scoring System
.
Cancer
.
2008
;
113
(
6
):
1351
-
1361
.
31.
Such
E
,
Germing
U
,
Malcovati
L
, et al
.
Development and validation of a prognostic scoring system for patients with chronic myelomonocytic leukemia
.
Blood
.
2013
;
121
(
15
):
3005
-
3015
.
32.
Wassie
EA
,
Itzykson
R
,
Lasho
TL
, et al
.
Molecular and prognostic correlates of cytogenetic abnormalities in chronic myelomonocytic leukemia: a Mayo Clinic-French Consortium Study
.
Am J Hematol
.
2014
;
89
(
12
):
1111
-
1115
.
33.
Greenberg
P
,
Cox
C
,
LeBeau
MM
, et al
.
International scoring system for evaluating prognosis in myelodysplastic syndromes
.
Blood
.
1997
;
89
(
6
):
2079
-
2088
.
34.
Patnaik
MM
,
Itzykson
R
,
Lasho
TL
, et al
.
ASXL1 and SETBP1 mutations and their prognostic contribution in chronic myelomonocytic leukemia: a two-center study of 466 patients
.
Leukemia
.
2014
;
28
(
11
):
2206
-
2212
.
35.
Padron
E
,
Abdel-Wahab
O
.
Importance of genetics in the clinical management of chronic myelomonocytic leukemia
.
J Clin Oncol
.
2013
;
31
(
19
):
2374
-
2376
.
36.
Padron
E
,
Garcia-Manero
G
,
Patnaik
MM
, et al
.
An international data set for CMML validates prognostic scoring systems and demonstrates a need for novel prognostication strategies
.
Blood Cancer J
.
2015
;
5
:
e333
.
37.
Uziel
L
,
Fabrizi
I
,
Zighetti
ML
, et al
.
A favourable effect of recombinant human erythropoietin in three cases of leukaemic transformation from chronic myelomonocytic leukaemia
.
Br J Haematol
.
1992
;
80
(
2
):
260
-
262
.
38.
Kerridge
I
,
Spencer
A
,
Azzi
A
,
Seldon
M
.
Response to erythropoietin in chronic myelomonocytic leukaemia
.
Intern Med J
.
2001
;
31
(
6
):
371
-
372
.
39.
Xicoy
B
,
Germing
U
,
Jimenez
MJ
, et al
.
Response to erythropoietic-stimulating agents in patients with chronic myelomonocytic leukemia
.
Eur J Haematol
.
2016
;
97
(
1
):
33
-
38
.
40.
Antony-Debré
I
,
Bluteau
D
,
Itzykson
R
, et al
.
MYH10 protein expression in platelets as a biomarker of RUNX1 and FLI1 alterations
.
Blood
.
2012
;
120
(
13
):
2719
-
2722
.
41.
Shiba
N
,
Hasegawa
D
,
Park
MJ
, et al
.
CBL mutation in chronic myelomonocytic leukemia secondary to familial platelet disorder with propensity to develop acute myeloid leukemia (FPD/AML)
.
Blood
.
2012
;
119
(
11
):
2612
-
2614
.
42.
Zahid
MF
,
Barraco
D
,
Lasho
TL
, et al
.
Spectrum of autoimmune diseases and systemic inflammatory syndromes in patients with chronic myelomonocytic leukemia
.
Leuk Lymphoma
.
2017
;
58
(
6
):
1488
-
1493
.
43.
Peker
D
,
Padron
E
,
Bennett
JM
, et al
.
A close association of autoimmune-mediated processes and autoimmune disorders with chronic myelomonocytic leukemia: observation from a single institution
.
Acta Haematol
.
2015
;
133
(
2
):
249
-
256
.
44.
Hadjadj
J
,
Michel
M
,
Chauveheid
MP
,
Godeau
B
,
Papo
T
,
Sacre
K
.
Immune thrombocytopenia in chronic myelomonocytic leukemia
.
Eur J Haematol
.
2014
;
93
(
6
):
521
-
526
.
45.
Ramadan
H
,
Duong
VH
,
Al Ali
N
, et al
.
Eltrombopag use in patients with chronic myelomonocytic leukemia (CMML): a cautionary tale
.
Clin Lymphoma Myeloma Leuk
.
2016
;
16
(
Suppl
):
S64
-
S66
.
46.
Solal-Celigny
P
,
Desaint
B
,
Herrera
A
, et al
.
Chronic myelomonocytic leukemia according to FAB classification: analysis of 35 cases
.
Blood
.
1984
;
63
(
3
):
634
-
638
.
47.
Tefferi
A
,
Hoagland
HC
,
Therneau
TM
,
Pierre
RV
.
Chronic myelomonocytic leukemia: natural history and prognostic determinants
.
Mayo Clin Proc
.
1989
;
64
(
10
):
1246
-
1254
.
48.
Maeda
T
,
Ashie
T
,
Kikuiri
K
,
Ishiyama
N
,
Takakura
M
,
Ise
T
.
Chronic myelomonocytic leukemia with polyneuropathy and IgA-paraprotein
.
Jpn J Med
.
1989
;
28
(
6
):
709
-
716
.
49.
Niscola
P
,
Siniscalchi
A
,
Tendas
A
, et al
.
Chronic myelomonocytic leukemia coexisting with monoclonal gammopathy: concomitant response to azacitidine of both disorders
.
Ann Hematol
.
2015
;
94
(
10
):
1753
-
1754
.
50.
Hagihara
M
,
Inoue
M
,
Kodama
K
,
Uchida
T
,
Hua
J
.
Simultaneous manifestation of chronic myelomonocytic leukemia and multiple myeloma during treatment by prednisolone and eltrombopag for immune-mediated thrombocytopenic purpura
. Case Rep Hematol.
2016
;2016:4342820.
51.
Ferrara
F
,
Del Vecchio
L
,
Antonolfi
I
,
Mele
G
,
Rametta
V
,
Cimino
R
.
Chronic lymphocytic leukemia coexisting with chronic myelomonocytic leukemia
.
Haematologica
.
1992
;
77
(
2
):
171
-
173
.
52.
Gaulier
A
,
Jary-Bourguignat
L
,
Serna
R
,
Pulik
M
,
Davi
F
,
Raphaël
M
.
Occurrence of angioimmunoblastic T cell lymphoma in a patient with chronic myelomonocytic leukemia features
.
Leuk Lymphoma
.
2000
;
40
(
1-2
):
197
-
204
.
53.
Quivoron
C
,
Couronné
L
,
Della Valle
V
, et al
.
TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis
.
Cancer Cell
.
2011
;
20
(
1
):
25
-
38
.
54.
Lemonnier
F
,
Couronné
L
,
Parrens
M
, et al
.
Recurrent TET2 mutations in peripheral T-cell lymphomas correlate with TFH-like features and adverse clinical parameters
.
Blood
.
2012
;
120
(
7
):
1466
-
1469
.
55.
Solary
E
,
Bernard
OA
,
Tefferi
A
,
Fuks
F
,
Vainchenker
W
.
The Ten-Eleven Translocation-2 (TET2) gene in hematopoiesis and hematopoietic diseases
.
Leukemia
.
2014
;
28
(
3
):
485
-
496
.
56.
Mekinian
A
,
Grignano
E
,
Braun
T
, et al
.
Systemic inflammatory and autoimmune manifestations associated with myelodysplastic syndromes and chronic myelomonocytic leukaemia: a French multicentre retrospective study
.
Rheumatology (Oxford)
.
2016
;
55
(
2
):
291
-
300
.
57.
Braun
T
,
Fenaux
P
.
Myelodysplastic syndromes (MDS) and autoimmune disorders (AD): cause or consequence?
Best Pract Res Clin Haematol
.
2013
;
26
(
4
):
327
-
336
.
58.
Grivennikov
SI
,
Greten
FR
,
Karin
M
.
Immunity, inflammation, and cancer
.
Cell
.
2010
;
140
(
6
):
883
-
899
.
59.
Kristinsson
SY
,
Björkholm
M
,
Hultcrantz
M
,
Derolf
ÅR
,
Landgren
O
,
Goldin
LR
.
Chronic immune stimulation might act as a trigger for the development of acute myeloid leukemia or myelodysplastic syndromes
.
J Clin Oncol
.
2011
;
29
(
21
):
2897
-
2903
.
60.
Elbæk
MV
,
Sørensen
AL
,
Hasselbalch
HC
.
Chronic inflammation and autoimmunity as risk factors for the development of chronic myelomonocytic leukemia?
Leuk Lymphoma
.
2016
;
57
(
8
):
1793
-
1799
.
61.
Hamidou
MA
,
Boumalassa
A
,
Larroche
C
,
El Kouri
D
,
Blétry
O
,
Grolleau
JY
.
Systemic medium-sized vessel vasculitis associated with chronic myelomonocytic leukemia
.
Semin Arthritis Rheum
.
2001
;
31
(
2
):
119
-
126
.
62.
Fraison
JB
,
Mekinian
A
,
Grignano
E
, et al
.
Efficacy of azacitidine in autoimmune and inflammatory disorders associated with myelodysplastic syndromes and chronic myelomonocytic leukemia
.
Leuk Res
.
2016
;
43
:
13
-
17
.
63.
Patnaik
MM
,
Wassie
EA
,
Padron
E
, et al
.
Chronic myelomonocytic leukemia in younger patients: molecular and cytogenetic predictors of survival and treatment outcome
.
Blood Cancer J
.
2015
;
5
:
e270
.
64.
West
RR
,
Hsu
AP
,
Holland
SM
,
Cuellar-Rodriguez
J
,
Hickstein
DD
.
Acquired ASXL1 mutations are common in patients with inherited GATA2 mutations and correlate with myeloid transformation
.
Haematologica
.
2014
;
99
(
2
):
276
-
281
.
65.
Perez Botero
J
,
Oliveira
JL
,
Chen
D
, et al
.
ASXL1 mutated chronic myelomonocytic leukemia in a patient with familial thrombocytopenia secondary to germline mutation in ANKRD26
.
Blood Cancer J
.
2015
;
5
:
e315
.
66.
Polprasert
C
,
Schulze
I
,
Sekeres
MA
, et al
.
Inherited and somatic defects in DDX41 in myeloid neoplasms
.
Cancer Cell
.
2015
;
27
(
5
):
658
-
670
.
67.
Micol
JB
,
Abdel-Wahab
O
.
Collaborating constitutive and somatic genetic events in myeloid malignancies: ASXL1 mutations in patients with germline GATA2 mutations
.
Haematologica
.
2014
;
99
(
2
):
201
-
203
.
68.
Takahashi
K
,
Pemmaraju
N
,
Strati
P
, et al
.
Clinical characteristics and outcomes of therapy-related chronic myelomonocytic leukemia
.
Blood
.
2013
;
122
(
16
):
2807
-
2811, quiz 2920
.
69.
Gillis
NK
,
Ball
M
,
Zhang
Q
, et al
.
Clonal haemopoiesis and therapy-related myeloid malignancies in elderly patients: a proof-of-concept, case-control study
.
Lancet Oncol
.
2017
;
18
(
1
):
112
-
121
.
70.
Symeonidis
A
,
van Biezen
A
,
de Wreede
L
, et al
;
Chronic Malignancies Working Party of the European Group for Blood and Marrow Transplantation
.
Achievement of complete remission predicts outcome of allogeneic haematopoietic stem cell transplantation in patients with chronic myelomonocytic leukaemia. A study of the Chronic Malignancies Working Party of the European Group for Blood and Marrow Transplantation
.
Br J Haematol
.
2015
;
171
(
2
):
239
-
246
.
71.
Itonaga
H
,
Iwanaga
M
,
Aoki
K
, et al
.
Impacts of graft-versus-host disease on outcomes after allogeneic hematopoietic stem cell transplantation for chronic myelomonocytic leukemia: a nationwide retrospective study
.
Leuk Res
.
2016
;
41
:
48
-
55
.
72.
Mittal
P
,
Saliba
RM
,
Giralt
SA
, et al
.
Allogeneic transplantation: a therapeutic option for myelofibrosis, chronic myelomonocytic leukemia and Philadelphia-negative/BCR-ABL-negative chronic myelogenous leukemia
.
Bone Marrow Transplant
.
2004
;
33
(
10
):
1005
-
1009
.
73.
Kerbauy
DM
,
Chyou
F
,
Gooley
T
, et al
.
Allogeneic hematopoietic cell transplantation for chronic myelomonocytic leukemia
.
Biol Blood Marrow Transplant
.
2005
;
11
(
9
):
713
-
720
.
74.
Elliott
MA
,
Tefferi
A
,
Hogan
WJ
, et al
.
Allogeneic stem cell transplantation and donor lymphocyte infusions for chronic myelomonocytic leukemia
.
Bone Marrow Transplant
.
2006
;
37
(
11
):
1003
-
1008
.
75.
Eissa
H
,
Gooley
TA
,
Sorror
ML
, et al
.
Allogeneic hematopoietic cell transplantation for chronic myelomonocytic leukemia: relapse-free survival is determined by karyotype and comorbidities
.
Biol Blood Marrow Transplant
.
2011
;
17
(
6
):
908
-
915
.
76.
Park
S
,
Labopin
M
,
Yakoub-Agha
I
, et al
.
Allogeneic stem cell transplantation for chronic myelomonocytic leukemia: a report from the Societe Francaise de Greffe de Moelle et de Therapie Cellulaire
.
Eur J Haematol
.
2013
;
90
(
5
):
355
-
364
.
77.
Kongtim
P
,
Popat
U
,
Jimenez
A
, et al
.
Treatment with hypomethylating agents before allogeneic stem cell transplant improves progression-free survival for patients with chronic myelomonocytic leukemia
.
Biol Blood Marrow Transplant
.
2016
;
22
(
1
):
47
-
53
.
78.
de Witte
T
,
Bowen
D
,
Robin
M
, et al
.
Allogeneic hematopoietic stem cell transplantation for MDS and CMML: recommendations from an international expert panel
.
Blood
.
2017
;
129
(
13
):
1753
-
1762
.
79.
Robin
M
,
Fenaux
P
.
Hypomethylating agents as bridging therapy before allogeneic hematopoietic stem cell transplantation in patients with chronic myelomonocytic leukemia?
Biol Blood Marrow Transplant
.
2016
;
22
(
1
):
1
-
2
.
80.
Kantarjian
H
,
Oki
Y
,
Garcia-Manero
G
, 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
(
1
):
52
-
57
.
81.
Liu
HD
,
Ahn
KW
,
Hu
ZH
, et al
.
Allogeneic hematopoietic cell transplantation for adult chronic myelomonocytic leukemia
.
Biol Blood Marrow Transplant
.
2017
;
23
(
5
):
767
-
775
.
82.
Caye
A
,
Strullu
M
,
Guidez
F
, et al
.
Juvenile myelomonocytic leukemia displays mutations in components of the RAS pathway and the PRC2 network
.
Nat Genet
.
2015
;
47
(
11
):
1334
-
1340
.
83.
Stieglitz
E
,
Taylor-Weiner
AN
,
Chang
TY
, et al
.
The genomic landscape of juvenile myelomonocytic leukemia
.
Nat Genet
.
2015
;
47
(
11
):
1326
-
1333
.
84.
Kotecha
N
,
Flores
NJ
,
Irish
JM
, et al
.
Single-cell profiling identifies aberrant STAT5 activation in myeloid malignancies with specific clinical and biologic correlates
.
Cancer Cell
.
2008
;
14
(
4
):
335
-
343
.
85.
Padron
E
,
Painter
JS
,
Kunigal
S
, et al
.
GM-CSF-dependent pSTAT5 sensitivity is a feature with therapeutic potential in chronic myelomonocytic leukemia
.
Blood
.
2013
;
121
(
25
):
5068
-
5077
.
86.
Itzykson
R
,
Kosmider
O
,
Renneville
A
, et al
.
Clonal architecture of chronic myelomonocytic leukemias
.
Blood
.
2013
;
121
(
12
):
2186
-
2198
.
87.
Sagaster
V
,
Ohler
L
,
Berer
A
, et al
.
High spontaneous colony growth in chronic myelomonocytic leukemia correlates with increased disease activity and is a novel prognostic factor for predicting short survival
.
Ann Hematol
.
2004
;
83
(
1
):
9
-
13
.
88.
Geissler
K
,
Jäger
E
,
Barna
A
, et al
.
Chronic myelomonocytic leukemia patients with RAS pathway mutations show high in vitro myeloid colony formation in the absence of exogenous growth factors
.
Leukemia
.
2016
;
30
(
11
):
2280
-
2281
.
89.
Solary
E
.
Unplugging JAK/STAT in chronic myelomonocytic leukemia
.
Clin Cancer Res
.
2016
;
22
(
15
):
3707
-
3709
.
90.
Padron
E
,
Dezern
A
,
Andrade-Campos
M
, et al
;
Myelodysplastic Syndrome Clinical Research Consortium
.
A multi-institution phase I trial of ruxolitinib in patients with chronic myelomonocytic leukemia (CMML)
.
Clin Cancer Res
.
2016
;
22
(
15
):
3746
-
3754
.
91.
Steensma
DP
,
Tefferi
A
,
Li
CY
.
Splenic histopathological patterns in chronic myelomonocytic leukemia with clinical correlations: reinforcement of the heterogeneity of the syndrome
.
Leuk Res
.
2003
;
27
(
9
):
775
-
782
.
92.
O’Connell
DM
,
Fagan
WA
,
Skinner
SM
,
Pulitzer
DR
,
Devillez
RL
.
Cutaneous involvement in chronic myelomonocytic leukemia
.
Int J Dermatol
.
1994
;
33
(
9
):
628
-
631
.
93.
Mathew
RA
,
Bennett
JM
,
Liu
JJ
, et al
. Cutaneous manifestations in CMML: indication of disease acceleration or transformation to AML and review of the literature. Leuk Res.
2012
;36(1):72-80.
94.
Hunter
AE
,
Russell
NH
,
Ellis
I
.
Extramedullary haemopoiesis as a cause of mediastinal lymphadenopathy in chronic myelomonocytic leukaemia
.
Clin Lab Haematol
.
1990
;
12
(
4
):
465
-
469
.
95.
Derlin
T
,
Clauditz
TS
,
Bannas
P
.
18F-FDG PET/CT for staging and detection of extramedullary organ involvement in chronic myelomonocytic leukemia
.
Clin Nucl Med
.
2014
;
39
(
9
):
811
-
812
.
96.
Vural
F
,
Ozcan
MA
,
Ozsan
GH
, et al
.
Gingival involvement in a patient with CD56+ chronic myelomonocytic leukemia
.
Leuk Lymphoma
.
2004
;
45
(
2
):
415
-
418
.
97.
Hyams
ES
,
Gupta
R
,
Melamed
J
,
Taneja
SS
,
Shah
O
.
Renal involvement by chronic myelomonocytic leukemia requiring nephroureterectomy
.
Rev Urol
.
2009
;
11
(
1
):
33
-
37
.
98.
Bradford
CR
,
Smith
SR
,
Wallis
JP
.
Pericardial extramedullary haemopoiesis in chronic myelomonocytic leukaemia
.
J Clin Pathol
.
1993
;
46
(
7
):
674
-
675
.
99.
Wolff-Kormann
PG
,
Hasenfratz
BC
,
Heinrich
B
,
Kormann
B
.
Successful treatment with hydroxyurea of ocular involvement in chronic myelomonocytic leukemia
.
N Engl J Med
.
1991
;
325
(
3
):
209
-
210
.
100.
Chevalier
X
,
Hermine
O
,
Rahmouni
A
, et al
.
Bilateral tumoral infiltration of the Achilles tendons revealing chronic myelomonocytic leukemia
.
Arthritis Rheum
.
1993
;
36
(
6
):
852
-
856
.
101.
Hiçsönmez
G
,
Cetin
M
,
Tunç
B
,
Tuncer
AM
,
Gümrük
F
,
Yenicesu
I
.
Dramatic resolution of pleural effusion in children with chronic myelomonocytic leukemia following short-course high-dose methylprednisolone
.
Leuk Lymphoma
.
1998
;
29
(
5-6
):
617
-
623
.
102.
Costa
R
,
Abdulhaq
H
,
Haq
B
, et al
.
Activity of azacitidine in chronic myelomonocytic leukemia
.
Cancer
.
2011
;
117
(
12
):
2690
-
2696
.
103.
Subari
S
,
Patnaik
M
,
Alfakara
D
, et al
.
Patients with therapy-related CMML have shorter median overall survival than those with de novo CMML: Mayo Clinic long-term follow-up experience
.
Clin Lymphoma Myeloma Leuk
.
2015
;
15
(
9
):
546
-
549
.
104.
Adès
L
,
Sekeres
MA
,
Wolfromm
A
, et al
.
Predictive factors of response and survival among chronic myelomonocytic leukemia patients treated with azacitidine
.
Leuk Res
.
2013
;
37
(
6
):
609
-
613
.
105.
Wattel
E
,
Guerci
A
,
Hecquet
B
, et al
;
Groupe Français des Myélodysplasies and European CMML Group
.
A randomized trial of hydroxyurea versus VP16 in adult chronic myelomonocytic leukemia
.
Blood
.
1996
;
88
(
7
):
2480
-
2487
.
106.
Braun
T
,
Itzykson
R
,
Renneville
A
, et al
;
Groupe Francophone des Myélodysplasies
.
Molecular predictors of response to decitabine in advanced chronic myelomonocytic leukemia: a phase 2 trial
.
Blood
.
2011
;
118
(
14
):
3824
-
3831
.
107.
Ghanem
H
,
Garcia-Manero
G
,
Faderl
S
, et al
.
Outcomes of patients with myelodysplatic syndrome and chronic myelomonocytic leukemia post clofarabine failure
.
Ther Adv Hematol
.
2014
;
5
(
2
):
29
-
34
.
108.
Fenaux
P
,
Mufti
GJ
,
Hellstrom-Lindberg
E
, et al
;
International Vidaza High-Risk MDS Survival Study Group
.
Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study
.
Lancet Oncol
.
2009
;
10
(
3
):
223
-
232
.
109.
Cheson
BD
,
Greenberg
PL
,
Bennett
JM
, et al
.
Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia
.
Blood
.
2006
;
108
(
2
):
419
-
425
.
110.
Pleyer
L
,
Germing
U
,
Sperr
WR
, et al
.
Azacitidine in CMML: matched-pair analyses of daily-life patients reveal modest effects on clinical course and survival
.
Leuk Res
.
2014
;
38
(
4
):
475
-
483
.
111.
Drummond
MW
,
Pocock
C
,
Boissinot
M
, et al
.
A multi-centre phase 2 study of azacitidine in chronic myelomonocytic leukaemia
.
Leukemia
.
2014
;
28
(
7
):
1570
-
1572
.
112.
Tantravahi
SK
,
Szankasi
P
,
Khorashad
JS
, et al
.
A phase II study of the efficacy, safety, and determinants of response to 5-azacitidine (Vidaza®) in patients with chronic myelomonocytic leukemia
.
Leuk Lymphoma
.
2016
;
57
(
10
):
2441
-
2444
.
113.
Harel
S
,
Cherait
A
,
Berthon
C
, et al
.
Outcome of patients with high risk myelodysplastic syndrome (MDS) and advanced chronic myelomonocytic leukemia (CMML) treated with decitabine after azacitidine failure
.
Leuk Res
.
2015
;
39
(
5
):
501
-
504
.
114.
Meldi
K
,
Qin
T
,
Buchi
F
, et al
.
Specific molecular signatures predict decitabine response in chronic myelomonocytic leukemia
.
J Clin Invest
.
2015
;
125
(
5
):
1857
-
1872
.
115.
van der Helm
LH
,
Alhan
C
,
Wijermans
PW
, et al
.
Platelet doubling after the first azacitidine cycle is a promising predictor for response in myelodysplastic syndromes (MDS), chronic myelomonocytic leukaemia (CMML) and acute myeloid leukaemia (AML) patients in the Dutch azacitidine compassionate named patient programme
.
Br J Haematol
.
2011
;
155
(
5
):
599
-
606
.
116.
Aucagne
R
,
Droin
N
,
Paggetti
J
, et al
.
Transcription intermediary factor 1γ is a tumor suppressor in mouse and human chronic myelomonocytic leukemia
.
J Clin Invest
.
2011
;
121
(
6
):
2361
-
2370
.
117.
Duchmann
M
,
Braun
T
,
Micol
JB
, et al
.
Validation of response assessment according to international consortium for MDS/MPN criteria in chronic myelomonocytic leukemia treated with hypomethylating agents
.
Blood Cancer J
.
2017
;
7
(
5
):
e562
.
118.
Beran
M
,
Estey
E
,
O’Brien
SM
, et al
.
Results of topotecan single-agent therapy in patients with myelodysplastic syndromes and chronic myelomonocytic leukemia
.
Leuk Lymphoma
.
1998
;
31
(
5-6
):
521
-
531
.
119.
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
.
1996
;
10
(
7
):
1164
-
1167
.
120.
Catalano
L
,
Majolino
I
,
Musto
P
, et al
.
Alpha interferon in the treatment of chronic myelomonocytic leukemia
.
Haematologica
.
1989
;
74
(
6
):
577
-
581
.
121.
Issa
JP
,
Roboz
G
,
Rizzieri
D
, et al
.
Safety and tolerability of guadecitabine (SGI-110) in patients with myelodysplastic syndrome and acute myeloid leukaemia: a multicentre, randomised, dose-escalation phase 1 study
.
Lancet Oncol
.
2015
;
16
(
9
):
1099
-
1110
.
122.
Yang
H
,
Bueso-Ramos
C
,
DiNardo
C
, et al
.
Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents
.
Leukemia
.
2014
;
28
(
6
):
1280
-
1288
.
123.
Ibañez
M
,
Such
E
,
Cervera
J
, et al
.
Screening for IDH mutations in chronic myelomonocytic leukemia
.
Leuk Lymphoma
.
2013
;
54
(
2
):
406
-
407
.
124.
Mondesir
J
,
Willekens
C
,
Touat
M
,
de Botton
S
.
IDH1 and IDH2 mutations as novel therapeutic targets: current perspectives
.
J Blood Med
.
2016
;
7
:
171
-
180
.
125.
Janin
M
,
Mylonas
E
,
Saada
V
, et al
.
Serum 2-hydroxyglutarate production in IDH1- and IDH2-mutated de novo acute myeloid leukemia: a study by the Acute Leukemia French Association group
.
J Clin Oncol
.
2014
;
32
(
4
):
297
-
305
.
126.
Frankel
AE
,
Lilly
M
,
Kreitman
R
, et al
.
Diphtheria toxin fused to granulocyte-macrophage colony-stimulating factor is toxic to blasts from patients with juvenile myelomonocytic leukemia and chronic myelomonocytic leukemia
.
Blood
.
1998
;
92
(
11
):
4279
-
4286
.
127.
Asati
V
,
Mahapatra
DK
,
Bharti
SK
.
K-Ras and its inhibitors towards personalized cancer treatment: pharmacological and structural perspectives
.
Eur J Med Chem
.
2017
;
125
:
299
-
314
.
128.
Akutagawa
J
,
Huang
TQ
,
Epstein
I
, et al
.
Targeting the PI3K/Akt pathway in murine MDS/MPN driven by hyperactive Ras
.
Leukemia
.
2016
;
30
(
6
):
1335
-
1343
.
129.
Lyubynska
N
,
Gorman
MF
,
Lauchle
JO
, et al
.
A MEK inhibitor abrogates myeloproliferative disease in Kras mutant mice
.
Sci Transl Med
.
2011
;
3
(
76
):
76ra27
.
130.
Chang
T
,
Krisman
K
,
Theobald
EH
, et al
.
Sustained MEK inhibition abrogates myeloproliferative disease in Nf1 mutant mice
.
J Clin Invest
.
2013
;
123
(
1
):
335
-
339
.
131.
Geissler
K
,
Jäger
E
,
Barna
A
, et al
.
In vitro and in vivo effects of JAK2 inhibition in chronic myelomonocytic leukemia
.
Eur J Haematol
.
2016
;
97
(
6
):
562
-
567
.
132.
Komrokji
RS
,
Seymour
JF
,
Roberts
AW
, et al
Results of a phase 2 study of pacritinib (SB1518), a JAK2/JAK2(V617F) inhibitor, in patients with myelofibrosis. Blood.
2015
;125(17):2649-2655.
133.
Mesa
RA
,
Vannucchi
AM
,
Mead
A
, et al
.
Pacritinib versus best available therapy for the treatment of myelofibrosis irrespective of baseline cytopenias (PERSIST-1): an international, randomised, phase 3 trial
.
Lancet Haematol
.
2017
;
4
(
5
):
e225
-
e236
.
134.
Martinelli
G
,
Oehler
VG
,
Papayannidis
C
, et al
.
Treatment with PF-04449913, an oral smoothened antagonist, in patients with myeloid malignancies: a phase 1 safety and pharmacokinetics study
.
Lancet Haematol
.
2015
;
2
(
8
):
e339
-
e346
.
135.
Frolova
O
,
Benito
J
,
Brooks
C
, et al
.
SL-401 and SL-501, targeted therapeutics directed at the interleukin-3 receptor, inhibit the growth of leukaemic cells and stem cells in advanced phase chronic myeloid leukaemia
.
Br J Haematol
.
2014
;
166
(
6
):
862
-
874
.
136.
Kim
E
,
Ilagan
JO
,
Liang
Y
, et al
.
SRSF2 mutations contribute to myelodysplasia by mutant-specific effects on exon recognition
.
Cancer Cell
.
2015
;
27
(
5
):
617
-
630
.
137.
Lee
SC
,
Dvinge
H
,
Kim
E
, et al
.
Modulation of splicing catalysis for therapeutic targeting of leukemia with mutations in genes encoding spliceosomal proteins
.
Nat Med
.
2016
;
22
(
6
):
672
-
678
.
138.
Dussiot
M
,
Maciel
TT
,
Fricot
A
, et al
.
An activin receptor IIA ligand trap corrects ineffective erythropoiesis in β-thalassemia
.
Nat Med
.
2014
;
20
(
4
):
398
-
407
.
139.
Tosolini
M
,
Pont
F
,
Bétous
D
, et al
.
Human monocyte recognition of adenosine-based cyclic dinucleotides unveils the A2a Gαs protein-coupled receptor tonic inhibition of mitochondrially induced cell death
.
Mol Cell Biol
.
2015
;
35
(
2
):
479
-
495
.
140.
Romano
M
,
Della Porta
MG
,
Gallì
A
, et al
.
Antitumour activity of trabectedin in myelodysplastic/myeloproliferative neoplasms
.
Br J Cancer
.
2017
;
116
(
3
):
335
-
343
.
141.
Itzykson
R
,
Solary
E
.
An evolutionary perspective on chronic myelomonocytic leukemia
.
Leukemia
.
2013
;
27
(
7
):
1441
-
1450
.
142.
Reynaud
D
,
Pietras
E
,
Barry-Holson
K
, et al
.
IL-6 controls leukemic multipotent progenitor cell fate and contributes to chronic myelogenous leukemia development
.
Cancer Cell
.
2011
;
20
(
5
):
661
-
673
.
143.
Welner
RS
,
Amabile
G
,
Bararia
D
, et al
.
Treatment of chronic myelogenous leukemia by blocking cytokine alterations found in normal stem and progenitor cells
.
Cancer Cell
.
2015
;
27
(
5
):
671
-
681
.
144.
Zhang
Q
,
Zhao
K
,
Shen
Q
, et al
.
Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL-6
.
Nature
.
2015
;
525
(
7569
):
389
-
393
.
145.
Zambetti
NA
,
Ping
Z
,
Chen
S
, et al
.
Mesenchymal inflammation drives genotoxic stress in hematopoietic stem cells and predicts disease evolution in human pre-leukemia
.
Cell Stem Cell
.
2016
;
19
(
5
):
613
-
627
.
146.
Theilgaard-Mönch
K
.
Gut microbiota sustains hematopoiesis
.
Blood
.
2017
;
129
(
6
):
662
-
663
.
147.
Patnaik
MM
,
Barraco
D
,
Lasho
TL
, et al
.
DNMT3A mutations are associated with inferior overall and leukemia-free survival in chronic myelomonocytic leukemia
.
Am J Hematol
.
2017
;
92
(
1
):
56
-
61
.
Sign in via your Institution