• 
    

    
    

      99热精品在线国产_美女午夜性视频免费_国产精品国产高清国产av_av欧美777_自拍偷自拍亚洲精品老妇_亚洲熟女精品中文字幕_www日本黄色视频网_国产精品野战在线观看 ?

      Clinical management of myelodysplastic syndrome/myeloproliferative neoplasm overlap syndromes

      2016-10-22 05:29:00JosephClaraDavidSallmanEricPadron
      Cancer Biology & Medicine 2016年3期

      Joseph A. Clara, David A. Sallman, Eric Padron

      1Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL 33606, USA;

      2Malignant Hematology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA

      REVIEW

      Clinical management of myelodysplastic syndrome/myeloproliferative neoplasm overlap syndromes

      Joseph A. Clara1, David A. Sallman2, Eric Padron2

      1Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL 33606, USA;

      2Malignant Hematology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA

      The myelodysplastic/myeloproliferative neoplasms (MDS/MPNs) are a unique group of hematologic malignancies characterized by concomitant myelodysplastic and myeloproliferative features. According to the 2008 WHO classification, the category includes atypical chronic myeloid leukemia (aCML), chronic myelomonocytic leukemia (CMML), juvenile myelomonocytic leukemia(JMML), MDS/MPN-unclassifiable (MDS/MPN-U), and the provisional entity refractory anemia with ring sideroblasts and thrombocytosis (RARS-T). Although diagnosis currently remains based on clinicopathologic features, the incorporation of nextgeneration platforms has allowed for the recent molecular characterization of these diseases which has revealed unique and complex mutational profiles that support their distinct biology and is anticipated to soon play an integral role in diagnosis,prognostication, and treatment. Future goals of research should include the development of disease-modifying therapies, and further genetic understanding of the category will likely form the foundation of these efforts.

      Myelodysplastic syndromes; myeloproliferative neoplasms; next-generation sequencing; CMML; aCML; JMML; MDS/MPN-U

      Introduction

      The myelodysplastic/myeloproliferative neoplasms(MDS/MPNs) are a unique group of myeloid malignancies characterized by a paradoxical phenotypic presentation hallmarked by both myelodysplastic and myeloproliferative features. MDS-like features include cytopenias and dysplasia of various cell lines while MPN-like features can include constitutional symptoms (e.g. night sweats and/or weight loss), elevated blood counts as well as extramedullary infiltration. As designated by the 2008 World Health Organization (WHO) classification, the MDS/MPN group is comprised of chronic myelomonocytic leukemia (CMML),juvenile myelomonocytic leukemia (JMML), atypical chronic myeloid leukemia (aCML), and MDS/MPN unclassifiable(MDS/MPN-U)1,2. An additional provisional category is refractory anemia with ring sideroblasts and thrombocytosis(RARS-T)1,2. Diagnosis and classification remain clinicopathologic, based on laboratory, morphological, and clinical parameters2. In general, the respective disorders are identified by the predominant myeloid subset present in the peripheral blood, such as the expansion of peripheral blood monocytes in CMML and JMML, and dysplastic granulocytes in aCML. RARS-T is demarcated by thrombocytosis while MDS/MPN-U is not associated with a specific myeloid subset, but is instead identified by the presence of overlapping myeloproliferative and myelodysplastic features at presentation and not meeting criteria for other subtypes3.

      The underlying pathogenesis responsible for this group of neoplasms remains unclear as does the molecular convergence point that biologically defines the MDS/MPN category. The characteristic bone marrow phenotype involves increased cell death resulting in dysplasia and cytopenias alongside concurrent myeloid subset skewing and proliferation4. Although no molecular markers have been found to be entirely unique to disorders in the MDS/MPN category5, advances in the molecular characterization of these disorders and annotation of recurrent genetic mutations have furthered our understanding in the field (Table 1). Specifically, these analyses have revealed substantial heterogeneity and complexity of molecular defects among the MDS/MPN group and elucidated several pathways likely involved in disease pathogenesis6-9.

      Cytogenetic testing and high-resolution single polymorphism genotyping arrays (SNP-A) have detected chromosomal abnormalities in a large proportion ofMDS/MPN patients, most commonly aneuploidies (+8, +9, -7) and partial deletions (7q-, 13q-, 20q-) with a minority exhibiting reciprocal translocations involving tyrosine kinase fusion genes10,11. In addition, large regions of uniparental disomy (UPD) have been identified in around one-third of MDS/MPNs through SNP-A analysis, which are associated with mutations involving gain of function in oncogenes or loss of function in tumor suppressor genes5,10. In a mixed cohort of patients with myeloid malignancies, SNP-A detected UPD in 48% of CMML and 38% of MDS/MPN-U patients12. Deregulation of myelopoiesis-associated microRNA/miR has also been demonstrated in cases of aCML and CMML, which exhibited down-regulation of miR-10a and overexpression of miR-424, respectively13.

      Table 1 Frequencies of recurrent genetic mutations in MDS/MPNs

      Mutational spectrum of MDS/MPNs

      Most recurrent genetic mutations affect genes involved in one of four functional categories: signaling, splicing,transcription, and epigenetic14. The annotation of these mutations has provided insight regarding the dysregulated pathways that could be responsible for the paradoxical dual phenotype characteristic of MDS/MPN4. Signaling mutations, typically seen in MPNs, lead to dysregulation of proliferative and anti-apoptotic pathways14, and include mutations of growth factor receptors (CSF3R), downstream cytokine receptor signaling intermediates (JAK2,NRAS,KRAS) and negative regulators of signaling pathways(PTPN11, CBL, NF1)14-21. The most common genes mutated involved with epigenetic regulation are TET2 and ASXL1. Additional epigenetic mutations include IDH1/2, EZH2,DNMT3A, SUZ12, EED, and UTX14,22,23. Mutations of genes involved in RNA splicing are also common in MDS/MPN and are critically important in the phenotypic presentation of these patients. Specifically, the presence of SRSF2 mutation,the most common spliceosome mutation in CMML, in combination with TET2 has been found to be highly specific for monocytosis and associated with CMML while SF3B1 mutation is tightly concordant with the presence of bone marrow ringed sideroblasts in RARS-T8,9,14,24,25. The RUNX1 gene, which encodes core-binding factor alpha and plays an integral role in definitive commitment of hematopoiesis, is mutated in 15%-30% of CMML patients26. SETBP1 mutation is detected in 25% of aCML patients, and is thought to attenuate the activity of tumor suppressor phosphatase, PP2A27.

      Table 2 WHO defined diagnostic criteria for MDS/MPN classification3

      Diagnostic considerations

      Diagnostic criteria of MDS/MPNs are primarily clinicopathologic and the specific 2008 WHO criteria are listed below (Table 2)3. By definition, MDS/MPN includes phenotypic properties of both MDS and MPNs, and the potential cases hence require a discerning diagnostic evaluation to assure they do not belong to one of those respective categories28. In addition, the detection of monocytosis allows for the recognition of CMML and JMML while the diagnosis of aCML and MDS/MPN-U is more challenging due to the increased difficulty of distinguishing them from other MPNs4. A particular challenge in the diagnosis of CMML is excluding other causes of monocytosis, especially because the diagnosis does not require the presence of dysplasia and may be based solely onmonocytosis that is unlikely to be caused by a concomitant condition29. Although cytogenetic abnormalities occur in only 30% of CMML cases, recent data revealed that sequencing of only 9 genes identifies clonality in 93% of cases, and thus the incorporation of targeted next-generation sequencing (NGS) can potentially aid in clarifying cases with diagnostic uncertainty8. In addition, to the end of discerning CMML from other myeloid neoplasms with monocytosis, a monocyte subset restriction has been identified in CMML that is not seen in other causes of monocytosis including other neoplasms. This technique, which has been externally validated and shown to be highly sensitive and specific, relies on flow cytometry and thus could be incorporated into clinical practice4.

      There are several diagnoses that must be excluded in the evaluation of the potential cases of MDS/MPN. In the case of predominant monocytosis and consideration of CMML or JMML, reactive causes of monocytosis including infection,inflammatory disorders, and nutritional aberrancies, must be excluded29. Furthermore, a blast percentage greater than 20 in the peripheral blood or bone marrow, or the presence of acute myeloid leukemia (AML)-defining mutations, would confirm the diagnosis of AML29. To exclude the diagnosis of CML, BCR-ABL1 fusion gene must be tested in all cases. When eosinophilia is present, rearrangements of PDGFR and FGFR should be assessed for to exclude myeloid and lymphoid neoplasms with rearrangements of PDGFRA,PDGFRB, and FGFR4. However, we have reported on a patient with refractory CMML without eosinophilia who harbored a PDGFRB fusion gene and given the therapeutic indications should consider more generalized testing30. A general approach to diagnosis is diagramed in Figure 1.

      General prognostic and treatment considerations

      As a group, these disorders have a poor prognosis with limited treatment options. Regarding prognostication,CMML is the only MDS/MPN subtype whose prognostication has been extensively interrogated. In fact, at least 7 clinical and 2 genetic (incorporating ASXL1)prognostic models have been developed for CMML, and have all recently been validated in an analysis of an international CMML database of over 1800 patients. However, no model was demonstrated to be statistically superior, and thus, no consensus prognostic score has been established31. RARS-T has been considered an indolent disease relative to the other MDS/MPN subtypes, but emerging data from mutational annotation suggest a worse prognosis in patients who are wild-type for SF3B1 and JAK232. Although no prognostic scoring system has been developed for aCML, a worse prognosis was seen in patients with mutation of SETBP1,with a median overall survival (OS) of 22 months compared to 77 months in wild-type patients27. The lack of prognostication with regards to MDS/MPN-U is likely complicated by the category's inherent diagnostic uncertainty4.

      The majority of treatment recommendations are extrapolated from clinical trials focused on MDS or MPN patients and thus include very few overlap syndrome patients. Although several pharmacologic agents have demonstrated activity, none have shown effect on the natural history of the diseases4,5,12. Therapeutic strategies are largely directed at alleviating the predominant myelodysplastic or myeloproliferative features manifested in individual cases6. Hypomethylating agents (HMAs) such as azacitidine and decitabine are typically used when cytopenias predominate. Approaches to the management of predominant myeloproliferative symptoms include cytoreductive agents such as hydroxyurea, as well as etoposide, topotecan, arsenic,all-trans retinoic acid, and induction chemotherapy4.

      In addition to diagnostic and prognostic utility,mutational profiling also has potential treatment implications as it can identify patients who may benefit from targeted therapy, particularly given the rapid and ongoing development of targeted agents. For example, the presence of TET2 or DNMT3A mutations has been shown to predict better response to the treatment with the DNMT inhibitors/HMAs azacitidine and decitabine in a mixed cohort of patients with myeloid disease, including patients with MDS-MPN.33. In a cohort of MDS patients, mutation of TET2 was also shown to be a strong predictor of response to HMAs, particularly in patients without ASXL1 mutations34. In addition, more recent work has highlighted that differentially methylated regions (DMR) can predict response to decitabine, and utilizing an epigenetic classifier derived from methylation profiles could predict decitabine response at the time of diagnosis35. Additional DNMT inhibitors are in pre-clinical and clinical investigation and ideally will be evaluated in the context of specific biomarkers to enrich for response36. Other novel pharmacologic treatments currently under investigation include JAK inhibitors, SRC inhibitors,and MEK inhibitors4,14,37. Allogeneic stem cell transplantation (allo-SCT) is supported by data demonstrating potential for improved survival and cure.

      Figure 1 Diagnostic algorithm for the evaluation of MDS/MPN overlap syndromes. Abbreviations: AML= acute myeloid leukemia; CML = chronic myelogenous leukemia. ?: next-generation sequencing can aid in the identification of a primary cause, as mutational profiling patterns can be specific for CMML and RARS-T. ?: inv(16)/t(16;16), t(8;21) or t(15;17).

      MDS/MPN: subtypes

      CMML

      CMML is a clinically and pathologically diverse clonal hematopoietic malignancy defined by a hematologic phenotype of peripheral monocytosis and dysplasia. Although previously considered a subtype of the MDS, it was reclassified by the WHO in 2008 into the category of MDS/MPN3. The incidence of the disease is 1/100,000 adults,with a median age at diagnosis of 65-75 years and a male predominance of 1.5-3:13,38. By definition, there is an absence of the BCR-ABL1 fusion gene, as well as rearrangements of PDGFR and FGFR139. The disorder may occur secondarily inrare instances in the setting of MDS or myelofibrosis40,41.

      The pathognomonic feature of CMML is an expanded peripheral monocytosis (>1×109/L), and while myelodysplasia is also a characteristic feature, its presence is not required for diagnosis if there is persistent monocytosis(>3 months) or if a clonal cytogenetic or molecular genetic abnormality is present3. While the diagnosis is dependent on laboratory, morphological, and clinical parameters, data on the molecular profile of the disorder has expanded recently and is now incorporated into the diagnostic process. Other causes of monocytosis that should be excluded in the evaluation of CMML include pregnancy, autoimmune disorders, major depression, drugs (e.g. corticosteroids or colony stimulating factors), infection, and inflammation3.

      Peripheral monocytes present in CMML often have an abnormal morphologic appearance with bizarre nuclei and cytoplasmic granules2. At diagnosis, patients typically exhibit leukocytosis but may present a normal or slightly decreased leukocyte count with variable neutropenia3,28. Common clinical features include splenomegaly and hepatomegaly,particularly in patients with leukocytosis3. In addition,extramedullary leukemic infiltration may involve the skin and lymph nodes14.

      Clonality can be established in most cases of CMML through detection of recurrent genetic mutations which involve a heterogeneous array of pathways including signal transduction (NRAS, KRAS, CBL,JAK2), DNA methylation(DNM3TA, TET2,IDH1/2), transcriptional regulation(ETV6,RUNX1), chromatin modification (EZH2,ASXL1),and the RNA splicing machinery (SF3B1,SRSF2, ZRSR2,U2AF1)8,42. In fact, by sequencing only 9 genes, a genetic clonal event can be identified in over 90% of CMML cases43. The most frequently identified mutations include TET2(50%-60%), ASXL1 (35%-40%), SRSF2 (40%-50%), and RUNX1 (10%-15%)42,43. In addition, the co-mutation of SRSF2 and TET2 appears to be highly specific for CMML phenotype, and thus can be diagnostically useful (particularly in cases with a relative monocytosis)8,44.

      CMML exhibits a poor overall prognosis with a median OS of 20-30 months and leukemic transformation rates of 15%-20% in patients31,44-46. The majority in risk models are based on MDS studies and were formed prior to the use of HMA14. CMML is stratified into two groups, CMML-1 and CMML-2 by the current WHO classification according to the number of blasts present in the peripheral blood and bone marrow3. Patients in the CMML-2 group have been shown to have worse median survival (15 months compared to 20 months in CMML-1) and are more likely to progress to AML47. Assessment of cytogenetic abnormalities has identified a high risk group that includes the presence of trisomy 8, chromosome 7 abnormalities, or a complex karyotype48. The prognostic implication of genetic mutations has also been investigated, and incorporation of molecular data has recently led to improved prognostic discrimination in CMML. This is demonstrated by two models (Table 3)which investigated the prognostic value of integrating molecular lesions along with other clinical variables. On multivariate analysis, the mutation of ASXL1 was found by both models to be the only mutation independently predictive of survival44,45. In analysis of the largest CMML database created to date, the negative prognostic impact of ASXL1 was validated along with identifying mutation of CBL to have a negative prognostic impact31. Mutation of SETBP1 has also been confirmed to have independent prognostic value in CMML49.

      Table 3 GFM and Mayo molecular prognostic models39,40

      There have been few clinical trials specifically designed for CMML and treatment for this disease is largely targeted at the alleviation of symptoms28. The only randomized clinical trial published in CMML to date evaluated hydroxyurea versus etoposide with improved survival and response ratesin the hydroxyurea arm50. Allo-HCT remains the only potentially curative therapy. One of the largest HCT trials for CMML included 85 patients and demonstrated a 10-year OS of 40%, and identified CMML risk group (1 vs. 2), pretransplant hematocrit, cytogenetic risk category, comorbidity index, and age as factors associated with favorable outcome51. A recent retrospective study identified splenomegaly as a negative predictor of OS and event free survival following allo-HCT52.

      There is no consensus on the optimal treatment strategy for patients not eligible for transplant. Although HMAs (e.g. azacitidine/decitabine) are typically the preferred pharmacologic treatment option for patients with symptomatic cytopenias, it remains unclear if these agents are disease modifying14. In recent phase 1 and 2 trials, HMAs have resulted in response rates in CMML comparable with those in MDS53-56. The Groupe Francophone des Myelodysplasies (GFM) is running a phase 3 trial comparing decitabine +/- hydroxurea to hydroxyurea alone to address the question of the optimal first line therapy for these patients57. A recent phase 1 trial of the JAK inhibitor ruxolitinib demonstrated efficacy, particularly in patients with splenomegaly and constitutional symptoms, thus confirming the JAK/STAT pathway as a therapeutic target in CMML58. The phase 2 portion of the trial is actively accruing with a recommended phase 2 dose of 20mg PO BID. Additional cytoreductive agents that have shown efficacy at palliating proliferative or constitutional symptoms include topotecan and etoposide as single agents or in combination with cytarabine50,59,60.

      JMML

      JMML is a rare clon al hematopoietic malignancy of childhood and its key feature is proliferation of monocytic and granulocytic lineages. JMML comprises under 2%-3% of all childhood leukemias and most frequently occurs in children less than 3 years of age, with a male-to-female ratio of approximately 2:13,61. An increased risk of developing JMML is seen in children with the congenital syndromes of neurofibromatosis 1 (NF1) and Noonan syndrome which converge on RAS signaling abnormalities62. The clinical trajectory is heterogenous, resulting in a fulminant course refractory to treatment in some patients, while those with Noonan syndrome may have spontaneous resolution despite the detection of clonal hematopoiesis63. Leukemic transformation can be seen in JMML, but is much less common than in adult myeloid malignancies14. Overproduction of monocytic and granulocytic cells leads to infiltration of various organs including the spleen, liver, lung,skin, and gastrointestinal tract, with consequent substantial morbidity and mortality64. Other characteristic clinical features include fever, thrombocytopenia, and hemoglobin F elevation64,65. Clinical and laboratory findings may mimic infections such as Epstein-Barr virus (EBV), cytomegalovirus(CMV), and herpes simplex virus (HSV), which must be excluded prior to the diagnosis of JMML3.

      Over 90% of patients with JMML harbor somatic or germline mutations of PTPN11, NF1, N-RAS, K-RAS2, or CBL. All of these genes encode proteins that signal through the RAS-RAF-MAPK pathways66,67. Germline mutations of these genes have been identified in patients with congenital genetic syndromes, namely NF1 in patients with neurofibromatosis 1, and PTPN11 in patients with Noonan syndrome67. Recently, through the use of exome sequencing,secondary mutations of SETBP1 and JAK3 were identified in around 15% of patients and were associated with poorer outcomes68. Interestingly, a molecular hallmark of JMML present in most patients is marked hypersensitivity of myeloid progenitor cells in vitro to granulocyte-macrophage colony-stimulating factor (GM-CSF), which appears to augment signaling of other downstream effectors,particularly JAK/STAT, despite virtually universal dysregulation of RAS69,70.

      JMML remains an aggressive and rapidly fatal disease if patients are left untreated, with a median survival of around one year64. Leukemic transformation is not common in JMML and patients without treatment generally succumb to respiratory failure due to leukemic infiltration of the lungs64,71. Recent investigation of epigenetic abnormalities has shown CpG island hypermethylation to be strongly associated with older age, elevated fetal hemoglobin, and poorer prognosis72. In another study, methylation of RASA4 isoform-2 promoter correlated with clinical parameters predictive of poor prognosis, PTPN11 mutation, and higher risk of relapse after allo-SCT73.

      The mainstay approach to treatment is allo-SCT which leads to event-free 5-year survival in about half the patients74. Despite the substantial cure rate, relapse is a significant problem and is the major cause of treatment failure,occurring in nearly 50% of patients75. Half of patients who relapse, however, can be rescued with a second allograft76. Overall, strategies to rescue patients after relapse remain suboptimal, including the limited success of donor lymphocyte infusions (DLI)77.

      Current non-transplant alternatives are also limited and efforts are underway to target underlying mutations. Specifically, targeting the RAS pathway has had limitedsuccess so far78. The HMA azacitadine has been shown to lead to molecular and hematologic remissions in a small number of patients with JMML, and requires further investigation to determine its usefulness in the pre-transplant and non-transplant settings71,79. Investigation of treatment with MEK, JAK, and SRC inhibitors, as well as low-dose chemotherapy and HMAs, is actively ongoing80,81. Children with JMML with mutations of CBL often have spontaneous resolution of their disease and can typically be actively monitored instead of being treated with allo-SCT71,82.

      aCML

      Atypical chronic myeloid leukemia is an extremely rare BCRABL1-negative myeloid neoplasm with an estimated incidence of 1% of typical BCR-ABL1-positive CML83. The median age is over 60 years, with approximate equal male-tofemale distribution3. It is characterized by a hypercellular bone marrow which results in leukocytosis (>13×109/L)composed predominantly of morphologically dysplastic neutrophils and their precursors1,84. Additional key diagnostic requirements include absence of BCR-ABL1,PDGFR and FGFR1 rearrangements, and no significant monocytosis (<10% of leukocytes) or basophilia (<2% of leukocytes)1. Other clinical characteristics include severe anemia, thrombocytopenia, and splenomegaly85.

      Multiple recurrent mutations have been described in aCML, including NRAS, KRAS,JAK2V617F, TET2, and CBL,although none are specific to aCML19,86,87. Mutations of SETBP1 were found in 25% of aCML cases and were associated with a worse prognosis, but have also been described in other myeloid malignancies including CMML27,68. Mutations of CSF3R, the gene encoding the receptor of colony stimulating factor 3, were recently detected in about 40% of patients with aCML and 90% of those with chronic neutrophilic leukemia (CNL)88. Membrane proximal mutations (T618I) were the most common in aCML patients although truncating mutations of the cytoplasmic tail and compound mutations were also seen88. However, more recent investigations suggest proximal mutations to be highly sensitive and specific for CNL as 46 aCML patients in these studies were all negative for CSF3R mutation89,90.

      Overall prognosis is poor with the median survival of aCML patients ranging from 2 to 3 years. Leukemic transformation occurs in around 40% of cases and the remainder of patients typically succumb to bone marrow failure2,84. As with other MDS/MPNs, there is no standard treatment, and allo-SCT appears to be the only strategy that offers a potential long-term remission although data is extremely limited due to the low incidence of the disease91. The insight gleaned from molecular profiling has led to potential targeted therapies. Specifically, truncating mutations of CSF3R have been shown to result in aberrant constitutive activation of SRC kinases, and are thus predisposed to inhibition by SRC inhibitors such as dasatinib. In similar fashion, proximal mutations of CSF3R lead to JAK/STAT pathway activation predisposing to in vitro sensitivity to the JAK inhibitor ruxolitinib88. A recent case report of a patient with a membrane proximal CSF3R mutation treated with ruxolitinib showed dramatic improvement of blood counts, reduced splenomegaly, and constitutional symptoms without change in the clonal burden92. Otherwise, symptom-directed treatment includes HMAs which lead to transient improvements in some clinicopathologic features and hydroxyurea which palliates symptoms related to leukocytosis and splenomegaly with high response rates but duration of only two-to-four months93,94. Interferon has also demonstrated modest activity in a few cases95.

      MDS/MPN-U

      MDS/MPN-U is the most heterogeneous and least well characterized MDS/MPN subgroup which includes patients with features of both MDS and MPN but lacking defining characteristics of the other subtypes. Incidence of MDS/MPN-U is not well known, but it is estimated to make up around 5% of all myeloid malignancies96. Two recent series of MDS/MPN-U patients have provided insight into biological and clinical features of the disorder90,96. Median age at diagnosis is 71 years and there is a male predominance of about 2:190,96. Other common characteristics include splenomegaly, low monocyte counts, 20%-30% JAK2v617F-positivity, and non-specific cytogenetic findings90,96.

      There are no currently recognized specific cytogenetic or molecular features for MDS/MPN-U, and cytogenetic studies in cases of MDS/MPN-U mainly serve to exclude other similar disorders28. For example, cases with mutations involving SF3B1, CSF3R, T6181, or CALR are assigned to specific MDS/MPN subgroups, and are excluded from categorization as MDS/MPN-U. Despite the lack of specific mutations, molecular overlap is seen with aCML as mutations of SETBP1 can also be seen in around 10% of MDS/MPN-U cases97.

      Overall median survival of MDS/MPN-U patients varied among two series: 12.4 months vs. 21.8 months. Most clinical and pathologic features were similar among the two groups,but a higher proportion of patients in the series with inferior survival were noted to have thrombocytopenia, possibly representing a more aggressive phenotype with poorer prognosis90,96. In addition, AML-free survival was 18.9months in the Orazi group, but not reported in the other series90,96. There is currently no consensus on optimal treatment for MDS/MPN-U patients who are ineligible for allo-SCT. In the largest series of MDS/MPN-U patients to date, those treated with HMAs demonstrated a superior OS compared with patients with other non-transplant treatments (16.4 months vs. 11.5 months)96. Other pharmacologic treatments include interferon alpha,cyclosporine, thalidomide, lenalidomide, and anti-thymocyte globulin90,96.

      RARS-T

      RARS-T is a provisional entity in the WHO 2008 classification characterized by MDS features of refractory anemia with ring sideroblasts (>15% of erythroid precursors)along with thrombocytosis (>450×109/L) associated with the proliferation of large atypical megakaryocytes resembling essential thrombocythemia3,98. Median age of at presentation ranges from 71 to 75 years with an almost equal distribution among the sexes99-101. Features that support the inclusion of RARS-T into the hybrid MDS/MPN category are the commonly identified MPN-associated gene mutations(JAK2v617Fand MPL) and MDS-like demonstrated poor in vitro colony forming capacity1.

      Recent characterization of the mutational landscape in RARS-T has broadened the understanding of the underlying pathogenesis100. Mutation of the spliceosome gene SF3B1 is identified in up to 90% of patients with RARS-T, and is likely responsible for the induction of ringed sideroblasts,ineffective erythropoiesis, and anemia100,102. Mutation of JAK2V617Fis another molecular hallmark of RARS-T, as it occurs in approximately 50% of cases and is associated with significantly higher platelet counts100,101. Presence of a JAK2V617Fmutation is an important molecular characteristic of RARS-T as it is not seen in patients with RARS. Interestingly, SF3B1 wildtype patients exhibited co-mutation of ASXL1 and JAK2V61for ASXL1 and other spliceosome mutations (U2AF1/SRSF2) in the majority of cases. Combined, one of these 5 mutations is present in 99% of RARS-T cases100. Regarding prognosis, improved OS was demonstrated in patients with SF3B1 and/or JAK2V617Fmutations when compared with wildtype patients of 6.9 versus 3.3 years. Furthermore, age (≤80 years) was also identified as a predictor of survival, and these three factors were incorporated into a prognostic model, with a median survival of 1.6 vs. 8.0 years in the high and low risk groups,respectively32.

      There is little data pertaining to the treatment of RARS-T and no optimal strategy has been established. Thus,therapeutic approaches are mainly derived from management of RARS and cytoreductive strategies for MPNs. Recent reports of treatment with lenalidomide have demonstrated benefit both in regards to cytopenias and splenomegaly103. Other potential strategies include JAK2 inhibitors patients with proliferative symptoms. Spliceosome inhibitors should enter clinical investigation in the near future and can potentially exploit the haplodeficient state with the prospect of selective synthetic lethality104. In addition, luspatercept is a fusion protein (modified activin receptor IIB-IgG Fc) that inhibits SMAD2/3 signaling, and is currently undergoing placebo-controlled, phase 3 evaluation in MDS patients with ringed sideroblasts105as phase 2 data highlighted efficacy in this patient population106. There is no recommendation regarding the use of aspirin or plateletsuppressive therapy, but given the recent report of increased risk of thrombotic events in RARS-T patients with SF3B1 mutations, low-dose aspirin therapy could be considered in these and cases of JAK2 mutated patients14.

      Conclusions

      The category of MDS/MPN overlap disorders comprises a heterogenous group of diseases that share a common paradoxical phenotype of concomitant myeloid subset proliferation and bone marrow failure. Although their categorization is currently based mainly on morphology,ongoing efforts to understand the molecular characteristics of these entities continue to uncover a complex underlying molecular pathogenesis responsible for the observed hybrid phenotypes. These advances have started to augment our diagnostic and prognostic capabilities, but our ability to alter the course of the disease outside of allo-SCT remains extremely limited. Further investigation is needed in order to refine our understanding of these aggressive diseases, expand the available therapeutic armamentarium, and ultimately improve the clinical care of MDS/MPN patients. Given the rapidly expanding identification of mutations within MDS/MPNs along with the ongoing development of novel targeted therapies, future treatment will likely be designed to address these underlying pathway drivers. Due to the rarity of these disorders, however, collaborative multicenter trials will likely be required for definite answers.

      Conflict of interest statement

      No potential conflicts of interest are disclosed.

      References

      1.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 andacute leukemia: Rationale and important changes. Blood. 2009;114: 937-51.

      2.Orazi A, Germing U. The myelodysplastic/myeloproliferative neoplasms: Myeloproliferative diseases with dysplastic features. Leukemia. 2008; 22: 1308-19.

      3.Swerdlow SH, Campo E, Harris NL, Jaffe ES, Pileri SA, Stein H, et al, eds. WHO classification of tumours of haematopoietic and lymphoid tissues. Lyon, France: IARC; 2008.

      4.Padron E. Surveying the landscape of MDS/MPN research: overlap among the overlap syndromes? Hematology Am Soc Hematol Educ Program. 2015; 2015: 349-54.

      5.Zoi K, Cross NC. Molecular pathogenesis of atypical CML,CMML and MDS/MPN-unclassifiable. Int J Hematol. 2015; 101: 229-42.

      6.Sakaguchi H, Okuno Y, Muramatsu H, Yoshida K, Shiraishi Y,Takahashi M, et al. Exome sequencing identifies secondary mutations of SETBP1 and JAK3 in juvenile myelomonocytic leukemia. Nat Genet. 2013; 45: 937-41.

      7.Haferlach T, Nagata Y, Grossmann V, Okuno Y, Bacher U, Nagae G, et al. Landscape of genetic lesions in 944 patients with myelodysplastic syndromes. Leukemia. 2014; 28: 241-7.

      8.Meggendorfer M, Roller A, Haferlach T, et al. SRSF2 mutations in 275 cases with chronic myelomonocytic leukemia (CMML). Blood. 2012; 120: 3080-8.

      9.Yoshida K, Sanada M, Shiraishi Y, Nowak D, Nagata Y,Yamamoto R, et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature. 2011; 478: 64-9.

      10.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.

      11.Delhommeau F, Pisani DF, James C, Casadevall N,Constantinescu S, Vainchenker W. Oncogenic mechanisms in myeloproliferative disorders. Cell Mol Life Sci. 2006; 63: 2939-53.

      12.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 Res. 2008; 68:10349-57.

      13.Hussein K, Büsche G, Muth M, Gohring G, Kreipe H, Bok O. Expression of myelopoiesis-associated microRNA in bone marrow cells of atypical chronic myeloid leukaemia and chronic myelomonocytic leukaemia. Ann Hematol. 2011; 90: 307-13.

      14.Mughal TI, Cross NCP, Padron E, Tiu RV, Savona M, Malcovati L, 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: 1117-30.

      15.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.

      16.Savage N, George TI, Gotlib J. Myeloid neoplasms associated with eosinophilia and rearrangement of PDGFRA, PDGFRB, and FGFRL: A review. Int J Lab Hematol. 2013; 35: 491-500.

      17.Kratz CP, Niemeyer CM, Castleberry RP, Cetin M, Bergstrasser E,Emanuel PD, et al. The mutational spectrum of PTPN11 in juvenile myelomonocytic leukemia and noonan syndrome/myeloproliferative disease. Blood. 2005; 106: 2183-5.

      18.Bumm TG, Elsea C, Corbin AS, Loriaux M, Sherbenou D, Wood L, et al. Characterization of murine JAK2V617F-positive myeloproliferative disease. Cancer Res. 2006; 66: 11156-65.

      19.Grand FH, Hidalgo-Curtis CE, Ernst T, Zoi K, Zoi C, McGuire C,et al. Frequent CBL mutations associated with 11q acquired uniparental disomy in myeloproliferative neoplasms. Blood. 2009;113: 6182-92.

      20.Haferlach C, Grossmann V, Kohlmann A, Schindela S, Kern W,Schnittger S, et al. Deletion of the tumor-suppressor gene NF1 occurs in 5% of myeloid malignancies and is accompanied by a mutation in the remaining allele in half of the cases. Leukemia. 2012; 26: 834-9.

      21.Maxson JE, Gotlib J, Pollyea DA, Fleischman AG, Agarwal A, Eide CA, et al. Oncogenic CSF3R mutations in chronic neutrophilic leukemia and atypical cml. N Engl J Med. 2013; 368:1781-90.

      22.Gelsi-Boyer V, Trouplin V, Adelaide J, Bonansea J, Cervera N,Carbuccia N, et al. Mutations of polycomb-associated gene ASXL1 in myelodysplastic syndromes and chronic myelomonocytic leukaemia. Br J Haematol. 2009; 145: 788-800.

      23.Shih AH, Abdel-Wahab O, Patel JP, Levine RL. The role of mutations in epigenetic regulators in myeloid malignancies. Nat Rev Cancer. 2012; 12: 599-612.

      24.Pandit S, Zhou Y, Shiue L, Coutinho-Mansfield G, Li H, Qiu J, et al. Genome-wide analysis reveals sr protein cooperation and competition in regulated splicing. Mol Cell. 2013; 50: 223-35.

      25.Jeromin S, Haferlach T, Weissmann S, Meggendorfer M, Eder C,Nadarajah N, et al. Refractory anemia with ring sideroblasts and marked thrombocytosis (RARS-T) cases harbor mutations in SF3B1 or other spliceosome genes accompanied by JAK2V617F and ASXL1 mutations. Haematologica. 2014; 100: e125-7.

      26.Kuo MC, Liang DC, Huang CF, Shih YS, Wu JH, Lin TL, et al. Runx1 mutations are frequent in chronic myelomonocytic leukemia and mutations at the C-terminal region might predict acute myeloid leukemia transformation. Leukemia. 2009; 23: 1426-31.

      27.Piazza R, Valletta S, Winkelmann N, Redaelli S, Spinelli R, Pirola A, et al. Recurrent SETBP1 mutations in atypical chronic myeloid leukemia. Nat Genet. 2012; 45: 18-24.

      28.Tiu RV, Sekeres MA. Making sense of the myelodysplastic/myeloproliferative neoplasms overlap syndromes. Curr Opin Hematol. 2014; 21: 131-40.

      29.Hall J, Foucar K. Diagnosing myelodysplastic/myeloproliferative neoplasms: Laboratory testing strategies to exclude other disorders. Int J Lab Hematol. 2010; 32: 559-71.

      30.Bell GC, Padron E. Detection of a pdgfrb fusion in refractory cmml without eosinophilia: A case for broad spectrum tumor profiling. Leuk Res Rep. 2015; 4: 70-1.

      31.Padron E, Garcia-Manero G, Patnaik MM, Itzykson R, Lasho T,Nazha A, 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.

      32.Broseus J, Alpermann T, Wulfert M, Florensa Brichs L, Jeromin S,Lippert E, et al. Age, JAK2(V617F) and SF3B1 mutations are the main predicting factors for survival in refractory anaemia with ring sideroblasts and marked thrombocytosis. Leukemia. 2013; 27: 1826-31.

      33.Traina F, Visconte V, Elson P, Tabarroki A, Jankowska AM,Hasrouni E, et al. Impact of molecular mutations on treatment response to DNMT inhibitors in myelodysplasia and related neoplasms. Leukemia. 2014; 28: 78-87.

      34.Bejar R, Lord A, Stevenson K, Bar-Natan M, Perez-Ladaga A,Zaneveld J, et al. TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. Blood. 2014; 124: 2705-12.

      35.Meldi K, Qin T, Buchi F, Droin N, Sotzen J, Micol JB, et al. Specific molecular signatures predict decitabine response in chronic myelomonocytic leukemia. J Clin Invest. 2015; 125: 1857-72.

      36.Subramaniam D, Thombre R, Dhar A, Anant S. DNA methyltransferases: a novel target for prevention and therapy. Front Oncol. 2014; 4: 80.

      37.Eghtedar A, Verstovsek S, Estrov Z, Burger J, Cortes J, Bivins C, et al. Phase 2 study of the jak kinase inhibitor ruxolitinib in patients with refractory leukemias, including postmyeloproliferative neoplasm acute myeloid leukemia. Blood. 2012; 119: 4614-8.

      38.Rollison DE, Howlader N, Smith MT, Strom SS, Merritt WD, Ries LA, et al. Epidemiology of myelodysplastic syndromes and chronic myeloproliferative disorders in the united states, 2001-2004, using data from the naaccr and seer programs. Blood. 2008; 112: 45-52.

      39.Patnaik MM, Parikh SA, Hanson CA, Tefferi A. Chronic myelomonocytic leukaemia: A concise clinical and pathophysiological review. Br J Haematol. 2014; 165: 273-86.

      40.Wang SA, Galili N, Cerny J, Sechman E, Chen SS, Loew J, et al. Chronic myelomonocytic leukemia evolving from preexisting myelodysplasia shares many features with de novo disease. Am J Clin Pathol. 2006; 126:789-97.

      41.Boiocchi L, Espinal-Witter R, Geyer JT, Steinhilber J, Bonzheim I,Knowles DM, et al. Development of monocytosis in patients with primary myelofibrosis indicates an accelerated phase of the disease. Mod Pathol 2013; 26:204-12.

      42.Patnaik MM, Tefferi A. Cytogenetic and molecular abnormalities in chronic myelomonocytic leukemia. Blood Cancer J 2016;6:e393.

      43.Patnaik MM, Itzykson R, Lasho TL, Kosmider O, Finke CM,Hanson CA, et al. ASXL1 and SETBP1 mutations and their prognostic contribution in chronic myelomonocytic leukemia: A two-center study of 466 patients. Leukemia. 2014; 28: 2206-12.

      44.Itzykson R, Kosmider O, Renneville A, Gelsi-Boyer V,Meggendorfer M, Morabito M, et al. Prognostic score including gene mutations in chronic myelomonocytic leukemia. J Clin Oncol. 2013; 31: 2428-36.

      45.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. 2013; 27: 1504-10.

      46.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.

      47.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

      48.Such E, Cervera J, Costa D, Sole F, Vallespi T, Luno E, et al. Cytogenetic risk stratification in chronic myelomonocytic leukemia. Haematologica. 2011; 96: 375-83.

      49.Laborde RR, Patnaik MM, Lasho TL, Finke CM, Hanson CA,Knudson RA, et al. SETBP1 mutations in 415 patients with primary myelofibrosis or chronic myelomonocytic leukemia: independent prognostic impact in CMML. Leukemia. 2013; 27: 2100-2.

      50.Wattel E, Guerci A, Hecquet B, Economopoulos T, Copplestone A, Mahe B, et al. A randomized trial of hydroxyurea versus VP16 in adult chronic myelomonocytic leukemia. Groupe Fran?ais des Myélodysplasies and European CMML Group. Blood. 1996; 88: 2480-87.

      51.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. Biol Blood Marrow Transplant. 2011; 17: 908-15.

      52.Park S, Labopin M, Yakoub-Agha I, Delaunay J, Dhedin N,Deconinck E, 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: 355-64.

      53.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. J Clin Oncol. 2011; 29: 2521-7.

      54.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.

      55.Wijermans P, Lübbert M, Verhoef G, Bosly A, Ravoet C, Andre M,et al. Low-dose 5-aza-2’-deoxycytidine, a DNA hypomethylating agent, for the treatment of high-risk myelodysplastic syndrome: a multicenter phase study in elderly patients. J Clin Oncol. 2000;18: 956-62.

      56.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 2 trial. Blood. 2011; 118: 3824-31.

      57.Groupe Francophone des Myelodysplasies. Randomized phasestudy of decitabine +/- hydroxyurea (HY) versus HY in advanced proliferative CMML (GFM-DAC-CMML). In: ClinicalTrials.gov[Internet]. Bethesda (MD): National Library of Medicine (US). 2000. Available from: https://clinicaltrials.gov/ct2/show/NCT02214407 NLM Identifier: NCT02214407. [Last accessed on 2016 Apr 2]

      58.Padron E, Dezern A, Andrade-Campos M, Vaddi K, Scherle P,Zhang Q, et al. A multi-institution phase 1 trial of ruxolitinib in patients with chronic myelomonocytic leukemia (CMML). Clin Cancer Res 2016 Feb 8. doi: 10.1158/1078-0432.CCR-15-2781[Epub ahead of print].

      59.Weihrauch MR, Staib P, Seiberlich B, Hoffmann M, Diehl V,Tesch H. Phase I/ clinical study of topotecan and cytarabine in patients with myelodysplastic syndrome, chronic myelomonocytic leukemia and acute myeloid leukemia. Leuk Lymphoma. 2004; 45: 699-704.

      60.Beran M, Estey E, O’Brien S, Cortes J, Koller CA, Giles FJ, et al. Topotecan and cytarabine is an active combination regimen in myelodysplastic syndromes and chronic myelomonocytic leukemia. J Clin Oncol. 1999; 17: 2819-30.

      61.Passmore SJ, Chessells JM, Kempski H. Paediatric myelodysplastic syndromes and juvenile myelomonocytic leukaemia in the UK: A population-based study of incidence and survival. Br J Haematol. 2003; 121: 758-67.

      62.Bader-Meunier B, Tchernia G, Mielot F, Fontaine JL, Thomas C,Lyonnet S, Lavergne JM, et al. Occurrence of myeloproliferative disorder in patients with Noonan syndrome. J Pediatr. 1997; 130: 885-9.

      63.Matsuda K, Shimada A, Yoshida N, Ogawa A, Watanabe A,Yajima S, et al. Spontaneous improvement of hematologic abnormalities in patients having juvenile myelomonocytic leukemia with specific Ras mutations. Blood. 2007; 109: 5477-80.

      64.Niemeyer CM, Arico M, Basso G, Biondi A, Cantu Rajnoldi A,Creutzig U, et al. Chronic myelomonocytic leukemia in childhood: a retrospective analysis of 110 cases. European Working Group on Myelodysplastic Syndromes in Childhood(EWOG-MDS). Blood. 1997; 89: 3534-43.

      65.Weinberg RS, Leibowitz D, Weinblatt ME, Kochen J, Alter BP. Juvenile chronic myelogenous leukaemia: the only example of truly fetal (not fetal-like) erythropoiesis. Br J Haematol. 1990; 76: 307-10.

      66.Niemeyer CM, Kratz CP. Paediatric myelodysplastic syndromes and juvenile myelomonocytic leukaemia: Molecular classification and treatment options. Br J Haematol. 2008; 140: 610-24.

      67.Loh ML. Recent advances in the pathogenesis and treatment of juvenile myelomonocytic leukaemia. Br J Haematol. 2011; 152: 677-87.

      68.Makishima H, Yoshida K, Nguyen N, Przychodzen B, Sanada M,Okuno Y, et al. Somatic SETBP1 mutations in myeloid malignancies. Nat Genet. 2013; 45: 942-6.

      69.Emanuel PD, Bates LJ, Castleberry RP, Gualtieri RJ, Zuckerman KS, et al. Selective hypersensitivity to granulocyte-macrophage colony-stimulating factor by juvenile chronic myeloid leukemia hematopoietic progenitors. Blood. 1991; 77: 925-9.

      70.Kotecha N, Flores NJ, Irish JM, Simonds EF, Sakai DS,Archambeault S, et al. Single-cell profiling identifies aberrant stat5 activation in myeloid malignancies with specific clinical and biologic correlates. Cancer Cell. 2008; 14: 335-43.

      71.Locatelli F, Niemeyer CM. How I treat Juvenile Myelomonocytic Leukemia (JMML). Blood. 2015; 125: 1083-91.

      72.Olk-Batz C, Poetsch AR, Nollke P, Claus R, Zucknick M,Sandrock I, et al. Aberrant DNA methylation characterizes juvenile myelomonocytic leukemia with poor outcome. Blood. 2011; 117: 4871-80.

      73.Poetsch AR, Lipka DB, Witte T, Claus R, Nollke P, Zucknick M, et al. Rasa4 undergoes DNA hypermethylation in resistant juvenile myelomonocytic leukemia. Epigenetics. 2014; 9:1252-60.

      74.Locatelli F, N?llke P, Zecca M, Korthof E, Lanino E, Peters C, et al. Hematopoietic stem cell transplantation (HSCT) in children with juvenile myelomonocytic leukemia (JMML): results of the EWOG-MDS/EBMT trial. Blood. 2005; 105: 410-9.

      75.Woods WG, Barnard DR, Alonzo TA, Buckley JD, Kobrinsky N,Arthur DC, et al. Prospective study of 90 children requiring treatment for juvenile myelomonocytic leukemia or myelodysplastic syndrome: a report from the Children's Cancer Group. J Clin Oncol. 2002; 20: 434-40.

      76.Yoshimi A, Mohamed M, Bierings M, Urban C, Korthof E, Zecca M, et al. Second allogeneic hematopoietic stem cell transplantation (hsct) results in outcome similar to that of first hsct for patients with juvenile myelomonocytic leukemia. Leukemia. 2007; 21: 556-60.

      77.Yoshimi A, Bader P, Matthes-Martin S, Stary J, Sedlacek P,Duffner U, et al. Donor leukocyte infusion after hematopoietic stem cell transplantation in patients with juvenile myelomonocytic leukemia. Leukemia. 2005; 19: 971-7.

      78.Downward J. Targeting RAS signaling pathways in cancer therapy. Nat Rev Cancer. 2003; 3: 11-22.

      79.Furlan I, Batz C, Flotho C, Mohr B, Lubbert M, Suttorp M, et al. Intriguing response to azacitidine in a patient with juvenile myelomonocytic leukemia and monosomy 7. Blood. 2009; 113: 2867-8.

      80.Bunda S, Kang MW, Sybingco SS, Weng J, Favre H, Shin DH, et al. Inhibition of src corrects GM-CSF hypersensitivity that underlies juvenile myelomonocytic leukemia. Cancer Res. 2013;73: 2540-50.

      81.Kong G, Wunderlich M, Yang D, Ranheim EA, Young KH, Wang J, et al. Combined mek and JAK inhibition abrogates murine myeloproliferative neoplasm. J Clin Invest. 2014; 124: 2762-73.

      82.Niemeyer CM, Kang MW, Shin DH, Furlan I, Erlacher M, Bunin NJ, et al. Germline CBL mutations cause developmental abnormalities and predispose to juvenile myelomonocytic leukemia. Nat Genet. 2010; 42: 794-800.

      83.Fend F, Horn T, Koch I, Vela T, Orazi A. Atypical chronic myeloid leukemia as defined in the who classification is a JAK2 V617F negative neoplasm. Leuk Res. 2008; 32: 1931-5.

      84.Hernández JM, Del Ca?izo MC, Cuneo A, Garcia JL, GutierrezNC, Gonzalez M, et al. Clinical, hematological and cytogenetic characteristics of atypical chronic myeloid leukemia. Ann Oncol. 2000; 11: 441-4.

      85.Breccia M, Biondo F, Latagliata R, Carmosino I, Mandelli F,Alimena G, et al. Identification of risk factors in atypical chronic myeloid leukemia. Haematologica. 2006; 91: 1568-70.

      86.Reiter A, Invernizzi R, Cross NC, Cazzola M. Molecular basis of myelodysplastic/myeloproliferative neoplasms. Haematologica. 2009; 94: 1634-8.

      87.Levine RL, Loriaux M, Huntly BJ, Loh ML, Beran M, Stoffregen E,et al. The JAK2V617F activating 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.

      88.Gotlib J, Maxson JE, George TI, Tyner JW. The new genetics of chronic neutrophilic leukemia and atypical CML: Implications for diagnosis and treatment. Blood. 2013; 122: 1707-11.

      89.Pardanani A, Lasho TL, Laborde RR, Elliott M, Hanson CA,Knudson RA, et al. CSF3R T618I is a highly prevalent and specific mutation in chronic neutrophilic leukemia. Leukemia. 2013; 27: 1870-3.

      90.Wang SA, Hasserjian RP, Fox PS, Rogers HJ, Geyer JT, Chabot-Richards D, et al. Atypical chronic myeloid leukemia is clinically distinct from unclassifiable myelodysplastic/myeloproliferative neoplasms. Blood. 2014; 123: 2645-52.

      91.Koldehoff M, Beelen DW, Trenschel R, Steckel NK, Peceny R,Ditschkowski M, et al. Outcome of hematopoietic stem cell transplantation in patients with atypical chronic myeloid leukemia. Bone Marrow Transplant. 2004; 34: 1047-50.

      92.Dao KH, Solti MB, Maxson JE, Winton EF, Press RD, Druker BJ,et al. Significant clinical response to JAK1/2 inhibition in a patient with CSF3R-T618I-positive atypical chronic myeloid leukemia. Leuk Res Rep. 2014; 3:67-9.

      93.Jiang H, Wu Z, Ren LI, Tao D, Tong H. Decitabine for the treatment of atypical chronic myeloid leukemia: A report of two cases. Oncol Lett. 2016; 11:689-692.

      94.Mao L, You L, Yang M, Li Y, Ye XN, Tong HY. The first case of decitabine successfully in treatment of atypical chronic myeloid leukemia with CEBPA double mutation. Chemotherapy. 2013; 02: 114.

      95.Kurzrock R, Bueso-Ramos CE, Kantarjian H, Freireich E, Tucker SL, Siciliano M, et al. BCR rearrangement negative chronic myelogenous leukemia revisited. J Clin Oncol. 2001; 19: 2915-26.

      96.DiNardo CD, Daver N, Jain N, Pemmaraju N, Bueso-Ramos C,Yin CC, et al. Myelodysplastic/myeloproliferative neoplasms,unclassifiable (MDS/MPN, U): Natural history and clinical outcome by treatment strategy. Leukemia 2014; 28: 958-61.

      97.Meggendorfer M, Bacher U, Alpermann T, Haferlach C, Kern W,Gambacorti-Passerini C, et al. SETBP1 mutations occur in 9% of mds/mpn and in 4% of mpn cases and are strongly associated with atypical CML, monosomy 7, isochromosome i(17)(q10), asxl1 and cbl mutations. Leukemia. 2013; 27: 1852-60.

      98.Patnaik MM, Tefferi A. Refractory anemia with ring sideroblasts and rars with thrombocytosis. Am J Hematol. 2015; 90: 549-59.

      99.Broseus J, Florensa L, Zipperer E, Schnittger S, Malcovati L,Richebourg S, et al. Clinical features and course of refractory anemia with ring sideroblasts associated with marked thrombocytosis. Haematologica. 2012; 97: 1036-41.

      100.Jeromin S, Haferlach T, Weissmann S, Meggendorfer M, Eder C,Nadarajah N, et al. Refractory anemia with ring sideroblasts and marked thrombocytosis cases harbor mutations in SF3B1 or other spliceosome genes accompanied by JAK2V617F and ASXL1 mutations. Haematologica. 2015; 100: e125-7.

      101.Jeromin S, Haferlach T, Grossmann V, Alpermann T, Kowarsch A, Haferlach C, et al. High frequencies of SF3B1 and JAK2 mutations in refractory anemia with ring sideroblasts associated with marked thrombocytosis strengthen the assignment to the category of myelodysplastic/myeloproliferative neoplasms. Haematologica. 2013; 98: e15-7.

      102.Visconte V, Rogers HJ, Singh J, Barnard J, Bupathi M, Traina F, et al. Sf3b1 haploinsufficiency leads to formation of ring sideroblasts in myelodysplastic syndromes. Blood. 2012; 120: 3173-86.

      103.Taylor G, Culligan D, Vickers MA. Refractory anemia with ring sideroblasts associated with marked thrombocytosis complicated by massive splenomegaly treated with lenalidomide resulting in resolution of splenomegaly but severe and prolonged pancytopenia. Case Rep Hematol. 2013; 2013: 718480.

      104.Lee SC, Dvinge H, Kim E, Cho H, Chung YR. Therapeutic targeting of spliceosomal mutant myeloid leukemias through modulation of splicing catalysis. Plenary Session at: ASH 57th Annual Meeting and Exposition. 2015 Dec 5-8; Orlando, FL.

      105.Celgene Corporation. A study of luspatercept (ACE-536) to treat anemia due to very low, low, or intermediate risk myelodysplastic syndromes (MEDALIST). In: ClinicalTrials.gov [Internet]. Bethesda (MD): National Library of Medicine (US). 2000- [cited 2016 Apr 2]. Available from: https://clinicaltrials.gov/ct2/show/NCT02631070 NLM Identifier: NCT02631070.

      106.Giagounidis A, Platzbecker U, Germing U, Gotze K, Ottmann O,Radsak M, et al. Luspatercept treatment leads to long term increases in hemoglobin and reductions in transfusion burden in patients with low or intermediate-1 risk myelodysplastic syndromes (MDS): preliminary results from the phase 2 PACEMDS extension study. Oral presentation at: ASH 57th Annual Meeting and Exposition. 2015 Dec 5-8; Orlando, FL.

      Cite this article as: Clara JA, Sallman DA, Padron E. Clinical management of myelodysplastic syndrome/myeloproliferative neoplasm overlap syndromes. Cancer Biol Med. 2016; 13: 360-72. doi: 10.20892/j.issn.2095-3941.2016.0043

      Correspondence to: Eric Padron

      E-mail: eric.padron@moffitt.org

      April 20, 2016; accepted May 25, 2016. Available at www.cancerbiomed.org

      Copyright ? 2016 by Cancer Biology & Medicine

      宁蒗| 读书| 同德县| 韩城市| 塘沽区| 厦门市| 铜川市| 车致| 洛扎县| 乌审旗| 蓬溪县| 昆山市| 岑溪市| 光泽县| 两当县| 天台县| 石台县| 林州市| 岑巩县| 昔阳县| 新野县| 家居| 麻城市| 吐鲁番市| 漳平市| 霞浦县| 老河口市| 柯坪县| 克什克腾旗| 体育| 比如县| 鸡西市| 信丰县| 峡江县| 靖安县| 利川市| 云阳县| 竹溪县| 广州市| 吉林市| 峨边|