Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
Filter
1.
Ann Hematol ; 103(6): 2059-2072, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38662207

ABSTRACT

Myelodysplastic syndrome (MDS) is well known to be complicated by systemic inflammatory autoimmune disease (SIADs). However, it remains unclear how the prognosis after allogenic hematopoietic stem cell transplantation (allo-HSCT) in patients with MDS is impacted by SIADs that occur before allo-HSCT. Therefore, we hypothesized that SIADs before allo-HSCT may be a risk factor for negative outcomes after allo-HSCT in patients with MDS. We conducted a single-center, retrospective, observational study of sixty-nine patients with MDS or chronic myelomonocytic leukemia who underwent their first allo-HCT. Fourteen of the patients had SIADs before allo-HSCT. In multivariate analysis, the presence of SIADs before allo-HSCT was an independent risk factor for overall survival (HR, 3.36, 95% confidence interval: 1.34-8.42, p = 0.009). Endothelial dysfunction syndrome was identified in five of 14 patients with SIADs who required immunosuppressive therapy or intensive chemotherapy, and notably, all patients with uncontrollable SIADs at allo-HSCT developed serious endothelial dysfunction syndrome and died in the early phase after allo-HSCT. The development of SIADs in the context of MDS is thought to reflect the degree of dysfunction of hematopoietic cells in MDS and suggests a higher risk of disease progression. In addition, MDS patients with SIADs before allo-HSCT are considered to be at higher risk of endothelial dysfunction syndrome because of preexisting vascular endothelial dysfunction due to SIADs. In conclusion, SIADs before allo-HSCT constitute an independent risk factor for death in MDS patients undergoing allo-HSCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myelomonocytic, Chronic , Myelodysplastic Syndromes , Humans , Female , Male , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/therapy , Myelodysplastic Syndromes/therapy , Myelodysplastic Syndromes/mortality , Myelodysplastic Syndromes/complications , Middle Aged , Hematopoietic Stem Cell Transplantation/adverse effects , Retrospective Studies , Risk Factors , Adult , Aged , Autoimmune Diseases/mortality , Autoimmune Diseases/therapy , Transplantation, Homologous/adverse effects , Allografts , Survival Rate
2.
Hematology ; 27(1): 367-378, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35306971

ABSTRACT

PURPOSE: The prognostic role of TET2 and/or ASXL1 mutations which are common gene mutations in chronic myelomonocytic leukemia (CMML) remains controversial. Therefore, we conducted this meta-analysis to evaluate the prognostic efficacy of ASXL1 and TET2 mutations in CMML population. METHODS: PubMed, Cochrane and Embase for relevant research were employed to identify 16 studies. Overall survival rate (OS) with hazard ratios (HRs) was used for analysis, and each individual HR was applied to calculate the combined HR. RESULTS: The total HR of OS was 0.74, 95% CI = 0.61 - 0.91, P = 0.005, compared with CMML patients without TET2 mutations (TET2MT), and the total HR of OS was 1.56, 95% CI = 1.34 - 1.80, P = 0.000, compared with CMML patients without ASXL1 mutation (ASXL1WT), indicating that TET2MT and ASXL1WT were favorable for prognosis of CMML. According to whether the gene is mutated or not, the acute transformation rate of disease and mortality rate were further considered for assessment. Compared with the CMML patients with TET2MT and ASXL1WT, the HR of patients with in both TET2MT and ASXL1MT was 1.51 (95% CI = 1.14 - 1.99; P = 0.004), the HR of patients with neither TET2MT nor ASXL1MT was 1.49 (95%CI = 1.12 - 1.98; P = 0.007), and the HR of TET2WT and ASXL1MT patients was 1.88 (95%CI = 1.21 - 2.94; P = 0.005). CONCLUSION: Presence of TET2MT and ASXL1WT genotype was the most beneficial for the survival of CMML patients.


Subject(s)
DNA-Binding Proteins/genetics , Dioxygenases/genetics , Leukemia, Myelomonocytic, Chronic/mortality , Repressor Proteins/genetics , Humans , Leukemia, Myelomonocytic, Chronic/genetics , Mutation , Prognosis
3.
Am J Hematol ; 97(3): 352-372, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34985762

ABSTRACT

DISEASE OVERVIEW: Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic stem cell disorder with overlapping features of myelodysplastic syndromes and myeloproliferative neoplasms, with an inherent risk for leukemic transformation (~15% over 3-5 years). DIAGNOSIS: Diagnosis is based on the presence of sustained (>3 months) peripheral blood monocytosis (≥1 × 109 /L; monocytes ≥10%), usually with accompanying bone marrow dysplasia. Clonal cytogenetic abnormalities occur in ~30% of patients, while >90% have somatic gene mutations. Mutations involving TET2 (~60%), SRSF2 (~50%), ASXL1 (~40%), and the oncogenic RAS pathway (~30%) are frequent, while the presence of ASXL1 and DNMT3A mutations and the absence of TET2 mutations negatively impact overall survival. RISK-STRATIFICATION: Molecularly integrated prognostic models include the Groupe Français des Myélodysplasies, Mayo Molecular Model (MMM), and the CMML specific prognostic model. Risk factors incorporated into the MMM include presence of truncating ASXL1 mutations, absolute monocyte count >10 × 109 /L, hemoglobin <10 g/dL, platelet count <100 × 109 /L, and the presence of circulating immature myeloid cells. The MMM stratifies CMML patients into four groups: high (≥3 risk factors), intermediate-2 (2 risk factors), intermediate-1 (1 risk factor), and low (no risk factors), with median survivals of 16, 31, 59, and 97 months, respectively. RISK-ADAPTED THERAPY: Hypomethylating agents such as 5-azacitidine and decitabine are commonly used, with overall response rates of ~40%-50% and complete remission rates of ~7%-17%; with no impact on mutational allele burdens. Allogeneic stem cell transplant is the only potentially curative option but is associated with significant morbidity and mortality.


Subject(s)
Chromosome Aberrations , Leukemia, Myelomonocytic, Chronic , Neoplasm Proteins/genetics , Disease-Free Survival , Humans , Leukemia, Myelomonocytic, Chronic/diagnosis , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/therapy , Risk Assessment , Risk Factors , Survivors
4.
Int J Hematol ; 115(1): 21-32, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34449040

ABSTRACT

We evaluated the mutational landscape of chronic myelomonocytic leukemia (CMML) and its potential clinical significance. We analyzed 47 samples with a panel of 112 genes using next-generation sequencing. Forty-five of the 47 patients (95.74%) had at least one mutation identified, with an average of 3.7 (range 0-9) per patient. The most common mutation was NRAS, followed by ASXL1, TET2, SRSF2, RUNX1, KRAS, and SETBP1. Patients 60 years and older more frequently had mutations in TET2 (56% vs. 9.09%, P = 0.001) and ASXL1 (48% vs. 18.18%, P = 0.031) than patients younger than 60 years. Median overall survival (OS) in patients with CMML was 22.0 months (95% CI 19.7-24.3 months). ASXL1 (18 vs. 22 months, P = 0.012), RUNX1 (17 vs. 22 months, P = 0.001), and SETBP1 (20 vs. 27 months, P = 0.032) mutations predicted inferior OS. However, only RUNX1 mutation was significantly associated with inferior acute myeloid leukemia (AML)-free survival. Our data showed that mutation profile differed significantly between CMML patients aged 60 years and older versus those younger than 60 years, and some of these mutations impact the progression and prognosis of the disease to a certain extent.


Subject(s)
DNA-Binding Proteins/genetics , Dioxygenases/genetics , GTP Phosphohydrolases/genetics , Genetic Association Studies , Leukemia, Myelomonocytic, Chronic/genetics , Membrane Proteins/genetics , Mutation , Repressor Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Aging/genetics , Carrier Proteins/genetics , Core Binding Factor Alpha 2 Subunit/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myelomonocytic, Chronic/mortality , Male , Middle Aged , Nuclear Proteins/genetics , Survival Rate , Young Adult
5.
Clin. transl. oncol. (Print) ; 23(9): 1731-1742, sept. 2021. graf
Article in English | IBECS | ID: ibc-222172

ABSTRACT

Chronic myelomonocytic leukemia (CMML) is a hematologic malignancy that overlaps with myeloproliferative neoplasms (MPN) and myelodysplastic syndromes (MDS) and tends to transform into acute myeloid leukemia (AML). Among cases of CMML, > 90% have gene mutations, primarily involving TET2 (~ 60%), ASXL1 (~ 40%), SRSF2 (~ 50%), and the RAS pathways (~ 30%). These gene mutations are associated with both the clinical phenotypes and the prognosis of CMML, special CMML variants and pre-phases of CMML. Cytogenetic abnormalities and the size of genome are also associated with prognosis. Meanwhile, cases with ASXL1, DNMT3A, NRAS, SETBP1, CBL and RUNX1 mutations may have inferior prognoses, but only ASXL1 mutations were confirmed to be independent predictors of the patient outcome and were included in three prognostic models. Novel treatment targets related to the various gene mutations are emerging. Therefore, this review provides new insights to explore the correlations among gene mutations, clinical phenotypes, prognosis, and novel drugs in CMML (AU)


Subject(s)
Humans , Leukemia, Myelomonocytic, Chronic/drug therapy , Leukemia, Myelomonocytic, Chronic/genetics , Membrane Proteins/genetics , Antineoplastic Agents/therapeutic use , Epigenesis, Genetic , Mutation/genetics , DNA Methylation/genetics , Methyltransferases/genetics , Dioxygenases/genetics , Epigenomics , Leukemia, Myelomonocytic, Chronic/mortality , Phenotype , Prognosis
6.
Acta Haematol ; 144(6): 649-659, 2021.
Article in English | MEDLINE | ID: mdl-34233332

ABSTRACT

INTRODUCTION: This study aimed to identify genetic predictors of treatment response and survival in patients with myeloid neoplasms treated with hypomethylating agents (HMAs). METHODS: We performed next-generation sequencing on bone marrow aspiration samples of 59 patients diagnosed with acute myeloid leukemia (AML), myelodysplastic syndrome with excess blasts-2, or chronic myelomonocytic leukemia and treated with decitabine or azacitidine as a frontline therapy. RESULTS: A single gene with the most common mutations was TP53 (14 of 59 patients), and mutations in RAS pathway-related genes including KRAS, NRAS, FLT3, PTPN11, CBL, and KIT were found in 28.8% of patients. The overall response rate to HMAs was 33.9%. Predictive factors for a poor response were an age >75 years (p = 0.007), 3 or more gene mutations (p = 0.004), mutations in RAS pathway-related genes (p = 0.033), and a mutated NRAS gene (p = 0.042). An age >75 years (hazard ratio 2.946), diagnosis of AML (hazard ratio 2.915), and mutations in NRAS (hazard ratio 4.440) were identified as poor prognostic factors for survival. CONCLUSION: In conclusion, mutations in RAS pathway-related genes were predictors of a poor response to HMAs. Particularly, mutated NRAS was associated with inferior survival rates.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myelomonocytic, Chronic/genetics , Myelodysplastic Syndromes/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction/genetics , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/therapeutic use , Decitabine/therapeutic use , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Leukemia, Myelomonocytic, Chronic/drug therapy , Leukemia, Myelomonocytic, Chronic/mortality , Male , Middle Aged , Mutation , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/mortality , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins c-kit/genetics , Survival Rate , Tumor Suppressor Protein p53/genetics , fms-Like Tyrosine Kinase 3/genetics
7.
Clin Cancer Res ; 27(22): 6095-6105, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34253584

ABSTRACT

PURPOSE: Chronic myelomonocytic leukemia (CMML) is a rare leukemia characterized by peripheral monocytosis with no disease-modifying therapies. CMML cells are uniquely hypersensitive to granulocyte-macrophage colony-stimulating factor (GM-CSF) and robustly engraft in immunocompromised mice that secrete human cytokines. To leverage these unique biological features, we conducted an integrated human and murine study evaluating ruxolitinib, a JAK1/2 inhibitor that potently downregulates intracellular GM-CSF signaling. PATIENTS AND METHODS: A total of 50 patients with WHO-defined CMML were enrolled in this open-label, multi-institution phase I/II clinical study, with a ruxolitinib dose of 20 mg twice daily studied in phase II. In parallel, 49 patient-derived xenografts (PDX) derived from 13 study participants were generated and randomized to receive ruxolitinib or vehicle control. RESULTS: The most common grade 3/4 treatment-related toxicities observed were anemia (10%) and thrombocytopenia (6%). The clinical overall response rate was 38% by Myelodysplastic Syndrome/Myeloproliferative Neoplasm (MDS/MPN) International Working Group (IWG) criteria and 43% of patients with baseline splenomegaly achieved a spleen response. Profiling of cytokine levels and somatic mutations at baseline failed to identify predictive biomarkers. PDX models derived from screening samples of study participants recapitulated responses seen in humans, particularly spleen responses, and corroborated ruxolitinib's clinical efficacy in a randomized murine study not feasible in human trials. CONCLUSIONS: Ruxolitinib demonstrated clinical efficacy and an acceptable adverse event profile in patients with CMML, identifying a potential novel therapeutic in this rare malignancy. Furthermore, this study demonstrates proof of concept that PDX modeling can recapitulate responses of patients treated on clinical trial and represents a novel correlative study that corroborates clinical efficacy seen in humans.See related commentary by Shastri and Adrianzen-Herrera, p. 6069.


Subject(s)
Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Leukemia, Myelomonocytic, Chronic/drug therapy , Nitriles/pharmacology , Nitriles/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Adult , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor , Clinical Trials as Topic , Cytokines/blood , Cytokines/genetics , Cytokines/metabolism , Drug Evaluation, Preclinical , Female , Humans , Leukemia, Myelomonocytic, Chronic/diagnosis , Leukemia, Myelomonocytic, Chronic/etiology , Leukemia, Myelomonocytic, Chronic/mortality , Male , Mice , Middle Aged , Mutation , Prognosis , Treatment Outcome
8.
Curr Hematol Malig Rep ; 16(3): 286-303, 2021 06.
Article in English | MEDLINE | ID: mdl-33945086

ABSTRACT

PURPOSE OF REVIEW: Monocytosis is a distinct but non-specific manifestation of various physiologic and pathologic conditions. Among hematopoietic stem cell neoplasms, depending on the criteria used for disease classification, monocytosis may be a consistent and integral component of diseases such as chronic myelomonocytic leukemia or acute myeloid leukemia with monocytic differentiation, or it may represent an inconsistent finding that often provides a clue to the underlying genetic changes driving the neoplasm. The purpose of this review is to provide the readers with a laboratory-based approach to neoplastic monocytosis. RECENT FINDINGS: In-depth elucidation of the genomic landscape of myeloid neoplasms within the past few years has broadened our understanding of monocytosis and its implications for diagnosis and prognosis. Genetic findings also shed light on potential disease response - or lack thereof - to various therapeutic agents used in the setting of myeloid neoplasms. In this review, we provide our approach to diagnose neoplastic monocytosis in the context of case-based studies while incorporating the most recent literature on this topic.


Subject(s)
Leukemia, Myeloid/diagnosis , Leukemia, Myelomonocytic, Chronic/diagnosis , Age Factors , Biomarkers , Bone Marrow/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Management , Disease Progression , Disease Susceptibility , Genetic Predisposition to Disease , Genetic Variation , Humans , Leukemia, Myeloid/etiology , Leukemia, Myeloid/mortality , Leukemia, Myelomonocytic, Chronic/etiology , Leukemia, Myelomonocytic, Chronic/mortality , Monocytes/metabolism , Monocytes/pathology
9.
Eur J Haematol ; 107(2): 265-274, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33998054

ABSTRACT

In older patients with chronic myelomonocytic leukaemia (CMML) and limited life expectancy due to age and or comorbidities, it is particularly important to consider the risk of transformation for individualised treatment decisions. There is limited information on potential differences between younger and older CMML patients regarding the cumulative risk of transformation as well as haematological, molecular and biologic characteristics. We analysed data from the Austrian Biodatabase for CMML (ABCMML) to compare these parameters in 518 CMML patients. Categorisation of patients into 3 age-related groups: <60 years, 60-79 years and ≥80 years, showed a significantly lower risk of transformation at higher age by competing risk analysis, with a 4-year risk of 39%, 23% and 13%, respectively (P < .0001). The lower probability of transformation was associated with a lower percentage of blast cells in the peripheral blood (PB) of older patients. Furthermore, we provide a simple score based on age, PB blasts and platelet counts that allowed us to define subgroups of CMML patients with a different cumulative transformation risk, including a low-risk group with a transformation risk of only 5%. Our findings may facilitate reasonable treatment decisions in elderly patients with CMML.


Subject(s)
Health Impact Assessment , Leukemia, Myelomonocytic, Chronic/epidemiology , Leukemia, Myelomonocytic, Chronic/pathology , Adult , Age Factors , Aged , Aged, 80 and over , Biomarkers, Tumor , Comorbidity , Disease Progression , Disease Susceptibility , Female , Humans , Leukemia, Myelomonocytic, Chronic/etiology , Leukemia, Myelomonocytic, Chronic/mortality , Male , Middle Aged , Odds Ratio , Prognosis
10.
Clin Transl Oncol ; 23(9): 1731-1742, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33861431

ABSTRACT

Chronic myelomonocytic leukemia (CMML) is a hematologic malignancy that overlaps with myeloproliferative neoplasms (MPN) and myelodysplastic syndromes (MDS) and tends to transform into acute myeloid leukemia (AML). Among cases of CMML, > 90% have gene mutations, primarily involving TET2 (~ 60%), ASXL1 (~ 40%), SRSF2 (~ 50%), and the RAS pathways (~ 30%). These gene mutations are associated with both the clinical phenotypes and the prognosis of CMML, special CMML variants and pre-phases of CMML. Cytogenetic abnormalities and the size of genome are also associated with prognosis. Meanwhile, cases with ASXL1, DNMT3A, NRAS, SETBP1, CBL and RUNX1 mutations may have inferior prognoses, but only ASXL1 mutations were confirmed to be independent predictors of the patient outcome and were included in three prognostic models. Novel treatment targets related to the various gene mutations are emerging. Therefore, this review provides new insights to explore the correlations among gene mutations, clinical phenotypes, prognosis, and novel drugs in CMML.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myelomonocytic, Chronic/drug therapy , Leukemia, Myelomonocytic, Chronic/genetics , Mutation , Carrier Proteins/genetics , Chromosome Aberrations , Core Binding Factor Alpha 2 Subunit/genetics , DNA Methylation , DNA Methyltransferase 3A/genetics , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Epigenesis, Genetic , Epigenetic Repression , GTP Phosphohydrolases/genetics , Genes, ras , Genome Size , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myelomonocytic, Chronic/mortality , Membrane Proteins/genetics , Myelodysplastic Syndromes/genetics , Nuclear Proteins/genetics , Phenotype , Prognosis , Proto-Oncogene Proteins c-cbl/genetics , Repressor Proteins/genetics , Serine-Arginine Splicing Factors/genetics , Signal Transduction/genetics
11.
Cancer Med ; 10(5): 1715-1725, 2021 03.
Article in English | MEDLINE | ID: mdl-33559357

ABSTRACT

Chronic myelomonocytic leukemia (CMML) is a rare disease of elderly people characterized by the presence of sustained peripheral blood monocytosis, overlapping features of myeloproliferation, and myelodysplasia. We present a large retrospective study of 156 CMML patients in China. Mean age at diagnosis was 68 years old (range 23-91). According to the CMML-specific prognostic scoring system (CPSS), 10 patients (8.3%) were low risk, 27 patients (22.5%) were intermediate-1 risk, 72 patients (60%) were intermediate-2 risk, and 11 patients (9.2%) were high risk. A total of 90 patients (57.7%) received hypomethylating agents (HMAs) treatment, 19 patients (12.2%) received chemotherapy and 47 patients (30.1%) received the best supportive care. Seventeen patients (10.9%) underwent allogeneic hematopoietic stem cell transplantation (allo-SCT) after HMAs treatment or chemotherapy. With a median follow-up of 35.3 months, overall response rate (ORR) was 69.5% in the HMAs ± chemotherapy group, 79.5% in the HMAs monotherapy group, 60.0% in the HMAs + chemotherapy group, and 37.5% in the chemotherapy group. HMAs monotherapy group had prolonged OS compared with the chemotherapy group (23.57 months vs. 11.73 months; p = 0.035). Patients who achieved ORR had prolonged OS (25.83 months vs. 8.00 months; p < 0.001) and LFS (20.53 months vs. 6.80 months; p < 0.001) compared with those not achieved ORR in the HMA ± chemotherapy group. By univariate analysis, only higher hemoglobulin (≥80 g/L) and lower serum LDH levels (<300 U/L) predicted for better OS and LFS. By multivariate analysis, only Hb ≥ 80 g/L predicted for prolonged OS, Hb ≥ 80 g/L, and monocytes < 3 × 109/L predicted for prolonged LFS. In summary, our study highlights the benefit of HMAs therapy in CMML, but we still need to develop novel therapeutics to achieve better outcomes.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myelomonocytic, Chronic/drug therapy , Adult , Aged , Aged, 80 and over , China , Hematopoietic Stem Cell Transplantation/statistics & numerical data , Humans , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/mortality , Middle Aged , Multivariate Analysis , Retrospective Studies , Risk , Time Factors , Transplantation, Homologous/statistics & numerical data , Treatment Outcome , Young Adult
12.
Lancet Haematol ; 8(2): e135-e148, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33513373

ABSTRACT

BACKGROUND: Approval of hypomethylating agents in patients with chronic myelomonocytic leukaemia is based on trials done in patients with myelodysplastic syndromes. We aimed to investigate whether hypomethylating agents provide a benefit in subgroups of patients with chronic myelomonocytic leukaemia compared with other treatments. METHODS: For this retrospective cohort study, data were retrieved between Nov 30, 2017, and Jan 5, 2019, from 38 centres in the USA and Europe. We included non-selected, consecutive patients diagnosed with chronic myelomonocytic leukaemia, who received chronic myelomonocytic leukaemia-directed therapy. Patients with acute myeloid leukaemia according to 2016 WHO criteria at initial diagnosis (ie, ≥20% blasts in the bone marrow or peripheral blood) or with unavailability of treatment data were excluded. Outcomes assessed included overall survival, time to next treatment, and time to transformation to acute myeloid leukaemia. Analyses were adjusted by age, sex, platelet count, and Chronic myelomonocytic leukaemia-Specific Prognostic Scoring System (CPSS). Patients were grouped by first received treatment with either hydroxyurea, hypomethylating agents, or intensive chemotherapy, and stratified by risk according to blast count, French-American-British subtype, CPSS, WHO 2016 subtype, and the eligibility criteria of the DACOTA trial (NCT02214407). FINDINGS: 949 patients diagnosed with chronic myelomonocytic leukaemia between April 13, 1981, and Oct 26, 2018, were included. Median follow-up was 23·4 months (IQR 11·5-42·3) from diagnosis and 16·2 months (6·6-31·6) from start of first-line treatment. 412 (43%) of 949 patients received hypomethylating agents as first treatment, 391 (41%) hydroxyurea, and 83 (9%) intensive chemotherapy. Adjusted median overall survival for patients treated with hydroxyurea versus hypomethylating agents was 15·6 months (95% CI 13·1-17·3) versus 20·7 months (17·9-23·4); hazard ratio (HR) 1·39 (1·17-1·65; p=0·0002) and 14·0 months (9·8-17·2) versus 20·7 months (17·9-23·4; HR 1·55 [1·16-2·05]; p=0·0027) for those treated with intensive chemotherapy versus hypomethylating agents. In patients with myeloproliferative chronic myelomonocytic leukaemia (myeloproliferative CMML), median overall survival was 12·6 months (10·7-15·0) versus 17·6 months (14·8-21·5; HR 1·38 [1·12-1·70]; p=0·0027) for patients treated with hydroxyurea versus hypomethylating agents, and 12·3 months (8·4-16·6) versus 17·6 months (14·8-21·5; HR 1·44 [1·02-2·03]; p=0·040) for intensive chemotherapy versus hypomethylating agents. Hypomethylating agents did not confer an overall survival advantage for patients classified as having lower-risk disease (ie, myelodysplastic chronic myelomonocytic leukaemia with <10% blasts, CMML-0, or lower-risk CPSS). INTERPRETATION: These data suggest hypomethylating agents as the preferred therapy for patients with higher-risk chronic myelomonocytic leukaemia and those with myeloproliferative CMML. Our findings also suggest that CPSS is a valuable tool to identify patients who are most likely to benefit from hypomethylating agents. Further evidence from prospective cohorts would be desirable. FUNDING: The Austrian Group for Medical Tumor Therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myelomonocytic, Chronic/drug therapy , Aged , Azacitidine/therapeutic use , Female , Humans , Hydroxyurea/therapeutic use , Kaplan-Meier Estimate , Leukemia, Myelomonocytic, Chronic/diagnosis , Leukemia, Myelomonocytic, Chronic/mortality , Male , Middle Aged , Prognosis , Proportional Hazards Models , Retrospective Studies , Treatment Outcome
13.
Eur J Haematol ; 106(5): 627-633, 2021 May.
Article in English | MEDLINE | ID: mdl-33432601

ABSTRACT

BACKGROUND: Myelomonocytic skewing is considered as a key pathophysiologic phenomenon in chronic myelomonocytic leukemia (CMML), but its prevalence and potential correlation with phenotypic, genotypic, and clinical features are poorly defined. METHODS: Skewed differentiation toward the myelomonocytic over erythroid commitment as indicated by an inverse ratio of myelomonocytic/erythroid colonies was investigated in 146 patients with CMML by semisolid in vitro cultures. RESULTS: There was a high prevalence of myelomonocytic skewing in patients with CMML (120/146, 82%); whereas, this phenomenon was rare in normal individuals (1/98, 1%). Patients with CMML with myelomonocytic skewing had higher white blood cell and peripheral blast cell counts, and lower platelet values. The number of mutations in genes of the epigenetic and/or splicing category was higher in CMML patients with as compared with patients without skewing. Patients with myelomonocytic skewing had more frequently mutations in RASopathy genes and higher growth factor independent myeloid colony formation. Interestingly, the lack of myelomonocytic skewing discriminated patients with CMML with a particularly favorable prognosis (60 vs 19 months, P = .003) and a minimal risk of transformation. CONCLUSION: Myelomonocytic skewing as determined by semisolid cultures can discriminate subgroups of patients with CMML with a different phenotype, a different genotype, and a different prognosis.


Subject(s)
Leukemia, Myelomonocytic, Chronic/diagnosis , Leukemia, Myelomonocytic, Chronic/mortality , Leukocyte Count , Leukocytes, Mononuclear/pathology , Myeloid Cells/pathology , Biomarkers , Biomarkers, Tumor , Disease Progression , Female , Humans , Kaplan-Meier Estimate , Leukemia, Myelomonocytic, Chronic/etiology , Leukocytes, Mononuclear/metabolism , Male , Mutation , Myeloid Cells/metabolism , Phenotype , Prognosis , Signal Transduction , ras Proteins/genetics , ras Proteins/metabolism
16.
Int J Mol Sci ; 21(17)2020 Aug 22.
Article in English | MEDLINE | ID: mdl-32842710

ABSTRACT

We have originally reported that colony-forming units granulocyte/macrophage (CFU-GM) formation is an in vitro feature of chronic myelomonocytic leukemia (CMML) and a strong predictor for short survival. Elucidation of the molecular basis underlying this in vitro phenomenon could be helpful to define molecular features that predict inferior outcome in patients. We studied the correlation between the mutational landscape and spontaneous colony formation in 164 samples from 125 CMML patients. As compared to wildtype samples, spontaneous in vitro CFU-GM formation was significantly increased in samples containing mutations in NRAS, CBL and EZH2 that were confirmed as independent stimulatory factors by multiple regression analysis. Inducible expression of mutated RAS but not JAK2 was able to induce growth factor independence of Ba/F3 cells. Whereas high colony CFU-GM growth was a strong unfavorable parameter for survival (p < 0.00001) and time to transformation (p = 0.01390), no single mutated gene had the power to significantly predict for both outcome parameters. A composite molecular parameter including NRAS/CBL/EZH2, however, was predictive for inferior survival (p = 0.00059) as well as for increased risk of transformation (p = 0.01429). In conclusion, we show that the composite molecular profile NRAS/CBL/EZH2 derived from its impact on spontaneous in vitro myeloid colony formation improves the predictive power over single molecular parameters in patients with CMML.


Subject(s)
Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/pathology , Adult , Aged , Aged, 80 and over , Animals , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/genetics , Female , GTP Phosphohydrolases/genetics , Gene Expression Regulation, Leukemic , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Janus Kinase 2/genetics , Male , Membrane Proteins/genetics , Mice , Middle Aged , Mutation , Prognosis , Proto-Oncogene Proteins c-cbl/genetics , Tumor Stem Cell Assay , ras Proteins/genetics
17.
Leukemia ; 34(11): 2951-2963, 2020 11.
Article in English | MEDLINE | ID: mdl-32576961

ABSTRACT

To establish novel and effective treatment combinations for chronic myelomonocytic leukemia (CMML) preclinically, we hypothesized that supplementation of CMML cells with the human oncogene Meningioma 1 (MN1) promotes expansion and serial transplantability in mice, while maintaining the functional dependencies of these cells on their original genetic profile. Using lentiviral expression of MN1 for oncogenic supplementation and transplanting transduced primary mononuclear CMML cells into immunocompromised mice, we established three serially transplantable CMML-PDX models with disease-related gene mutations that recapitulate the disease in vivo. Ectopic MN1 expression was confirmed to enhance the proliferation of CMML cells, which otherwise did not engraft upon secondary transplantation. Furthermore, MN1-supplemented CMML cells were serially transplantable into recipient mice up to 5 generations. This robust engraftment enabled an in vivo RNA interference screening targeting CMML-related mutated genes including NRAS, confirming that their functional relevance is preserved in the presence of MN1. The novel combination treatment with azacitidine and the MEK-inhibitor trametinib additively inhibited ERK-phosphorylation and thus depleted the signal from mutated NRAS. The combination treatment significantly prolonged survival of CMML mice compared to single-agent treatment. Thus, we identified the combination of azacitidine and trametinib as an effective treatment in NRAS-mutated CMML and propose its clinical development.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Evaluation, Preclinical , Leukemia, Myelomonocytic, Chronic/drug therapy , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/pharmacology , Clonal Evolution , Disease Models, Animal , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , Drug Synergism , Female , GTP Phosphohydrolases/genetics , Humans , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/pathology , Membrane Proteins/genetics , Mice , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , RNA, Small Interfering/genetics , Receptor, Notch1/genetics , Xenograft Model Antitumor Assays/methods
18.
Best Pract Res Clin Haematol ; 33(2): 101138, 2020 06.
Article in English | MEDLINE | ID: mdl-32460980

ABSTRACT

Chronic Myelomonocytic Leukemias are frequently diagnosed in older adults. Their prognosis is heterogeneous, but several prognostic factors can identify patients with an expected survival of a few years only, including among younger patients eligible for allogeneic stem cell transplantation. Based on the retrospective data available, we discuss how to identify CMML patients for whom curative therapy must be envisaged. We emphasize that, although transplantation remains the only path to cure in CMML, it can be envisaged in only a minority of patients. Despite increased donor availability, its potential remains limited by significant rates of mortality caused both by the procedure and by post-transplantation relapses. We review the options available to bridge patients to transplant, the management of transplantation itself (choice of donor, graft source and condition regimen), and finally the potential for post-transplantation interventions. Our review underscores the need for further prospective studies of allogeneic stem cell transplantation in CMML.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/therapy , Humans , Transplantation, Homologous
19.
Int J Mol Sci ; 21(8)2020 Apr 24.
Article in English | MEDLINE | ID: mdl-32344757

ABSTRACT

Although the RAS-pathway has been implicated as an important driver in the pathogenesis of chronic myelomonocytic leukemia (CMML) a comprehensive study including molecular and functional analyses in patients with progression and transformation has not been performed. A close correlation between RASopathy gene mutations and spontaneous in vitro myeloid colony (CFU-GM) growth in CMML has been described. Molecular and/or functional analyses were performed in three cohorts of 337 CMML patients: in patients without (A, n = 236) and with (B, n = 61) progression/transformation during follow-up, and in patients already transformed at the time of sampling (C, n = 40 + 26 who were before in B). The frequencies of RAS-pathway mutations (variant allele frequency ≥ 20%) in cohorts A, B, and C were 30%, 47%, and 71% (p < 0.0001), and of high colony growth (≥20/105 peripheral blood mononuclear cells) 31%, 44%, and 80% (p < 0.0001), respectively. Increases in allele burden of RAS-pathway mutations and in numbers of spontaneously formed CFU-GM before and after transformation could be shown in individual patients. Finally, the presence of mutations in RASopathy genes as well as the presence of high colony growth prior to transformation was significantly associated with an increased risk of acute myeloid leukemia (AML) development. Together, RAS-pathway mutations in CMML correlate with an augmented autonomous expansion of neoplastic precursor cells and indicate an increased risk of AML development which may be relevant for targeted treatment strategies.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/metabolism , Mutation , Signal Transduction , ras Proteins/genetics , ras Proteins/metabolism , Cytogenetic Analysis , Disease Progression , Gene Expression Regulation, Leukemic , Humans , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/pathology , Neoplasm Staging , Neoplastic Stem Cells/metabolism , Prognosis , Retrospective Studies
20.
Am J Hematol ; 95(1): 97-115, 2020 01.
Article in English | MEDLINE | ID: mdl-31736132

ABSTRACT

DISEASE OVERVIEW: Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic stem cell disorder with overlapping features of myelodysplastic syndromes and myeloproliferative neoplasms, with an inherent risk for leukemic transformation (~15% over 3-5 years). DIAGNOSIS: Diagnosis is based on the presence of sustained (>3 months) peripheral blood monocytosis (≥1 × 109 /L; monocytes ≥10%), along with bone marrow dysplasia. Clonal cytogenetic abnormalities occur in ~ 30% of patients, while >90% have gene mutations. Mutations involving TET2 (~60%), SRSF2 (~50%), ASXL1 (~40%) and the oncogenic RAS pathway (~30%) are frequent; while the presence of ASXL1 and DNMT3A mutations and the absence of TET2 mutations negatively impact over-all survival. RISK STRATIFICATION: Molecularly integrated prognostic models include; the Groupe Français des Myélodysplasies (GFM), Mayo Molecular Model (MMM) and the CMML specific prognostic model (CPSS-Mol). Risk factors incorporated into the MMM include presence of nonsense or frameshift ASXL1 mutations, absolute monocyte count>10 × 109 /L, hemoglobin <10 g/dL, platelet count <100 × 109 /L and the presence of circulating immature myeloid cells. The MMM stratifies CMML patients into four groups; high (≥3 risk factors), intermediate-2 (2 risk factors), intermediate-1 (1 risk factor) and low (no risk factors), with median survivals of 16, 31, 59 and 97 months, respectively. RISK-ADAPTED THERAPY: Hypomethylating agents such as 5-azacitidine and decitabine are commonly used, with overall response rates of ~40%-50% and complete remission rates of ~7%-17%; with no impact on mutational allele burdens. Allogeneic stem cell transplant is the only potentially curative option, but is associated with significant morbidity and mortality.


Subject(s)
Disease Management , Leukemia, Myelomonocytic, Chronic/diagnosis , Aged , Antimetabolites, Antineoplastic/therapeutic use , Female , Humans , Leukemia, Myelomonocytic, Chronic/mortality , Leukemia, Myelomonocytic, Chronic/therapy , Male , Middle Aged , Remission Induction/methods , Risk Assessment , Stem Cell Transplantation/methods , Survival Analysis , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...