112 results on '"Leukemia, Myelomonocytic, Chronic metabolism"'
Search Results
2. Targeting MCL1-driven anti-apoptotic pathways overcomes blast progression after hypomethylating agent failure in chronic myelomonocytic leukemia.
- Author
-
Montalban-Bravo G, Thongon N, Rodriguez-Sevilla JJ, Ma F, Ganan-Gomez I, Yang H, Kim YJ, Adema V, Wildeman B, Tanaka T, Darbaniyan F, Al-Atrash G, Dwyer K, Loghavi S, Kanagal-Shamanna R, Song X, Zhang J, Takahashi K, Kantarjian H, Garcia-Manero G, and Colla S
- Subjects
- Humans, Animals, Mice, Signal Transduction drug effects, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells drug effects, Disease Progression, Sulfonamides pharmacology, Sulfonamides therapeutic use, NF-kappa B metabolism, DNA Methylation drug effects, DNA Methylation genetics, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Blast Crisis pathology, Blast Crisis drug therapy, Blast Crisis genetics, Blast Crisis metabolism, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic pathology, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Myeloid Cell Leukemia Sequence 1 Protein genetics, Myeloid Cell Leukemia Sequence 1 Protein antagonists & inhibitors, Apoptosis drug effects, Mutation genetics
- Abstract
RAS pathway mutations, which are present in 30% of patients with chronic myelomonocytic leukemia (CMML) at diagnosis, confer a high risk of resistance to and progression after hypomethylating agent (HMA) therapy, the current standard of care for the disease. Here, using single-cell, multi-omics technologies, we seek to dissect the biological mechanisms underlying the initiation and progression of RAS pathway-mutated CMML. We identify that RAS pathway mutations induce transcriptional reprogramming of hematopoietic stem and progenitor cells (HSPCs) and downstream monocytic populations in response to cell-intrinsic and -extrinsic inflammatory signaling that also impair the functions of immune cells. HSPCs expand at disease progression after therapy with HMA or the BCL2 inhibitor venetoclax and rely on the NF-κB pathway effector MCL1 to maintain survival. Our study has implications for the development of therapies to improve the survival of patients with RAS pathway-mutated CMML., Competing Interests: Declaration of interests G.M.-B. declares research support from Rigel Pharmaceuticals, IFM Therapeutics, and Takeda Oncology. K.T. declares support from Symbio Pharmaceuticals, Novartis, Celgene/BMS, and GSK, and honoraria from Mission Bio and Illumina. H.K. declares research support from and an advisory role at Actinium and research support from AbbVie, Agio, Amgen, Ariad, Astex, Bristol Myers Squibb, Cyclacel, Daiichi-Sankyo, Immunogen, Jazz Pharma, Novartis, and Pfizer. G.G-M. declares research support from and an advisory role at Bristol Myers Squibb, Astex, and Helsinn, and research support from Amphivena, Novartis, AbbVie, H3 Biomedicine, Onconova, and Merck. S.C. declares research support from Amgen., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
3. Contribution of mutant HSC clones to immature and mature cells in MDS and CMML, and variations with AZA therapy.
- Author
-
Schnegg-Kaufmann AS, Thoms JAI, Bhuyan GS, Hampton HR, Vaughan L, Rutherford K, Kakadia PM, Lee HM, Johansson EMV, Failes TW, Arndt GM, Koval J, Lindeman R, Warburton P, Rodriguez-Meira A, Mead AJ, Unnikrishnan A, Davidson S, Polizzotto MN, Hertzberg M, Papaemmanuil E, Bohlander SK, Faridani OR, Jolly CJ, Zanini F, and Pimanda JE
- Subjects
- Humans, Hematopoietic Stem Cells metabolism, Monocytes, Clone Cells, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes genetics
- Abstract
Myelodysplastic neoplasms (MDSs) and chronic myelomonocytic leukemia (CMML) are clonal disorders driven by progressively acquired somatic mutations in hematopoietic stem cells (HSCs). Hypomethylating agents (HMAs) can modify the clinical course of MDS and CMML. Clinical improvement does not require eradication of mutated cells and may be related to improved differentiation capacity of mutated HSCs. However, in patients with established disease it is unclear whether (1) HSCs with multiple mutations progress through differentiation with comparable frequency to their less mutated counterparts or (2) improvements in peripheral blood counts following HMA therapy are driven by residual wild-type HSCs or by clones with particular combinations of mutations. To address these questions, the somatic mutations of individual stem cells, progenitors (common myeloid progenitors, granulocyte monocyte progenitors, and megakaryocyte erythroid progenitors), and matched circulating hematopoietic cells (monocytes, neutrophils, and naïve B cells) in MDS and CMML were characterized via high-throughput single-cell genotyping, followed by bulk analysis in immature and mature cells before and after AZA treatment. The mutational burden was similar throughout differentiation, with even the most mutated stem and progenitor clones maintaining their capacity to differentiate to mature cell types in vivo. Increased contributions from productive mutant progenitors appear to underlie improved hematopoiesis in MDS following HMA therapy., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
4. Role of the bone marrow immune microenvironment in chronic myelomonocytic leukemia pathogenesis: novel mechanisms and insights into clonal propagation.
- Author
-
Mangaonkar AA and Patnaik MM
- Subjects
- Bone Marrow metabolism, Disease Progression, Humans, Monocytes metabolism, Tumor Microenvironment, Leukemia, Myeloid, Acute genetics, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Juvenile
- Abstract
Recent studies in chronic myelomonocytic leukemia (CMML) involving clonal dendritic cell (DC) aggregates and association with systemic immune dysregulation have highlighted novel and potentially targetable pathways of disease progression. CMML DC aggregates are populated by heterogeneous cell types such as CD123
+ plasmacytoid dendritic cells (pDCs), CD11c + myeloid-derived DCs (mDCs), myeloid-derived suppressor cells (MDSCs), monocytes, and associate with an immune checkpoint called indoleamine 2,3-dioxygenase (IDO). Systemically, these IDO + DC aggregates are associated with immune tolerance marked by regulatory T cell expansion, likely mediated by aberrant DC-T cell interactions occurring within the bone marrow (BM) microenvironment. Somatic mutational events in CMML such as ASXL1 and NRAS mutations cooperate to induce T cell exhaustion and contribute toward disease progression to acute myeloid leukemia (AML). In this review, we explore the role of aging-induced alterations in the BM immune microenvironment, aberrant innate immune and proinflammatory signaling, and the adaptive immune system in CMML.- Published
- 2022
- Full Text
- View/download PDF
5. Cooperation between KDM6B overexpression and TET2 deficiency in the pathogenesis of chronic myelomonocytic leukemia.
- Author
-
Wei Y, Kanagal-Shamanna R, Zheng H, Bao N, Lockyer PP, Class CA, Darbaniyan F, Lu Y, Lin K, Yang H, Montalban-Bravo G, Ganan-Gomez I, Soltysiak KA, Do KA, Colla S, and Garcia-Manero G
- Subjects
- Animals, Gene Expression Profiling, Genome, Humans, Loss of Function Mutation, Mice, Mutation, Proto-Oncogene Proteins genetics, DNA-Binding Proteins deficiency, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Dioxygenases deficiency, Dioxygenases genetics, Dioxygenases metabolism, Jumonji Domain-Containing Histone Demethylases biosynthesis, Jumonji Domain-Containing Histone Demethylases genetics, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Juvenile genetics, Leukemia, Myelomonocytic, Juvenile metabolism
- Abstract
Loss-of-function TET2 mutations are recurrent somatic lesions in chronic myelomonocytic leukemia (CMML). KDM6B encodes a histone demethylase involved in innate immune regulation that is overexpressed in CMML. We conducted genomic and transcriptomic analyses in treatment naïve CMML patients and observed that the patients carrying both TET2 mutations and KDM6B overexpression constituted 18% of the cohort and 42% of patients with TET2 mutations. We therefore hypothesized that KDM6B overexpression cooperated with TET2 deficiency in CMML pathogenesis. We developed a double-lesion mouse model with both aberrations, and discovered that the mice exhibited a more prominent CMML-like phenotype than mice with either Tet2 deficiency or KDM6B overexpression alone. The phenotype includes monocytosis, anemia, splenomegaly, and increased frequencies and repopulating activity of bone marrow (BM) hematopoietic stem and progenitor cells (HSPCs). Significant transcriptional alterations were identified in double-lesion mice, which were associated with activation of proinflammatory signals and repression of signals maintaining genome stability. Finally, KDM6B inhibitor reduced BM repopulating activity of double-lesion mice and tumor burden in mice transplanted with BM-HSPCs from CMML patients with TET2 mutations. These data indicate that TET2 deficiency and KDM6B overexpression cooperate in CMML pathogenesis of and that KDM6B could serve as a potential therapeutic target in this disease., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2022
- Full Text
- View/download PDF
6. Age-related diseases of inflammation in myelodysplastic syndrome and chronic myelomonocytic leukemia.
- Author
-
Weeks LD, Marinac CR, Redd R, Abel G, Lin A, Agrawal M, Stone RM, Schrag D, and Ebert BL
- Subjects
- Aged, Aged, 80 and over, Aging genetics, Aging metabolism, Aging pathology, Female, Humans, Inflammation genetics, Inflammation metabolism, Inflammation pathology, Male, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells pathology, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Mutation, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes metabolism, Myelodysplastic Syndromes pathology
- Published
- 2022
- Full Text
- View/download PDF
7. Hemophagocytosis arising during disease progression of chronic myelomonocytic leukemia.
- Author
-
O'Brien O, Sibai H, and Chang H
- Subjects
- Aged, Disease Progression, Humans, Male, Leukemia, Myelomonocytic, Chronic complications, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Lymphohistiocytosis, Hemophagocytic etiology, Lymphohistiocytosis, Hemophagocytic metabolism, Lymphohistiocytosis, Hemophagocytic pathology
- Published
- 2022
- Full Text
- View/download PDF
8. Asxl1 C-terminal mutation perturbs neutrophil differentiation in zebrafish.
- Author
-
Fang X, Xu S, Zhang Y, Xu J, Huang Z, Liu W, Wang S, Yen K, and Zhang W
- Subjects
- Animals, Cell Differentiation, Embryo, Nonmammalian metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Neutrophils metabolism, Phenotype, Repressor Proteins genetics, Zebrafish, Zebrafish Proteins genetics, Embryo, Nonmammalian pathology, Leukemia, Myeloid, Acute pathology, Leukemia, Myelomonocytic, Chronic pathology, Mutation, Neutrophils pathology, Repressor Proteins metabolism, Zebrafish Proteins metabolism
- Abstract
ASXL1 is one of the most frequently mutated genes in malignant myeloid diseases. In patients with myeloid malignancies, ASXL1 mutations are usually heterozygous frameshift or nonsense mutations leading to C-terminal truncation. Current disease models have predominantly total loss of ASXL1 or overexpressed C-terminal truncations. These models cannot fully recapitulate leukemogenesis and disease progression. We generated an endogenous C-terminal-truncated Asxl1 mutant in zebrafish that mimics human myeloid malignancies. At the embryonic stage, neutrophil differentiation was explicitly blocked. At 6 months, mutants initially exhibited a myelodysplastic syndrome-like phenotype with neutrophilic dysplasia. At 1 year, about 13% of mutants further acquired the phenotype of monocytosis, which mimics chronic myelomonocytic leukemia, or increased progenitors, which mimics acute myeloid leukemia. These features are comparable to myeloid malignancy progression in humans. Furthermore, transcriptome analysis, inhibitor treatment, and rescue assays indicated that asxl1-induced neutrophilic dysplasia was associated with reduced expression of bmi1a, a subunit of polycomb repressive complex 1 and a reported myeloid leukemia-associated gene. Our model demonstrated that neutrophilic dysplasia caused by asxl1 mutation is a foundation for the progression of myeloid malignancies, and illustrated a possible effect of the Asxl1-Bmi1a axis on regulating neutrophil development., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
9. Cytokine-like protein 1-induced survival of monocytes suggests a combined strategy targeting MCL1 and MAPK in CMML.
- Author
-
Sevin M, Debeurme F, Laplane L, Badel S, Morabito M, Newman HL, Torres-Martin M, Yang Q, Badaoui B, Wagner-Ballon O, Saada V, Sélimoglu-Buet D, Kraus-Berthier L, Banquet S, Derreal A, Fenaux P, Itzykson R, Braun T, Etienne G, Berthon C, Thépot S, Kepp O, Kroemer G, Padron E, Figueroa ME, Droin N, and Solary E
- Subjects
- Adult, Aged, Aged, 80 and over, Cell Survival drug effects, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Humans, Male, Middle Aged, Myeloid Cell Leukemia Sequence 1 Protein metabolism, Xenograft Model Antitumor Assays, Blood Proteins metabolism, Cytokines metabolism, Extracellular Signal-Regulated MAP Kinases antagonists & inhibitors, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, MAP Kinase Signaling System drug effects, Monocytes metabolism, Monocytes pathology, Myeloid Cell Leukemia Sequence 1 Protein antagonists & inhibitors, Protein Kinase Inhibitors pharmacology
- Abstract
Mouse models of chronic myeloid malignancies suggest that targeting mature cells of the malignant clone disrupts feedback loops that promote disease expansion. Here, we show that in chronic myelomonocytic leukemia (CMML), monocytes that accumulate in the peripheral blood show a decreased propensity to die by apoptosis. BH3 profiling demonstrates their addiction to myeloid cell leukemia-1 (MCL1), which can be targeted with the small molecule inhibitor S63845. RNA sequencing and DNA methylation pattern analysis both point to the implication of the mitogen-activated protein kinase (MAPK) pathway in the resistance of CMML monocytes to death and reveal an autocrine pathway in which the secreted cytokine-like protein 1 (CYTL1) promotes extracellular signal-regulated kinase (ERK) activation through C-C chemokine receptor type 2 (CCR2). Combined MAPK and MCL1 inhibition restores apoptosis of monocytes from patients with CMML and reduces the expansion of patient-derived xenografts in mice. These results show that the combined inhibition of MCL1 and MAPK is a promising approach to slow down CMML progression by inducing leukemic monocyte apoptosis., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
10. Genomic Landscape and Risk Stratification in Chronic Myelomonocytic Leukemia.
- Author
-
Hunter A and Padron E
- Subjects
- Biomarkers, Tumor, Chromosome Aberrations, DNA Methylation, Epigenesis, Genetic, Genetic Association Studies, Humans, Leukemia, Myelomonocytic, Chronic metabolism, Mutation, Phenotype, Prognosis, RNA Splicing, Risk Assessment, Risk Factors, Signal Transduction, Genetic Predisposition to Disease, Genomics methods, Leukemia, Myelomonocytic, Chronic diagnosis, Leukemia, Myelomonocytic, Chronic genetics
- Abstract
Purpose: The advent of next-generation sequencing has allowed for the annotation of a vast array of recurrent somatic mutations across human malignancies, ushering in a new era of precision oncology. Chronic myelomonocytic leukemia is recognized as a myelodysplastic/myeloproliferative neoplasm and displays heterogenous clinical and genetic features. Herein, we review what is currently understood regarding the genomic landscape of this disease and discuss how somatic mutations have impacted current risk stratification methods., Recent Findings: Genomic studies in chronic myelomonocytic leukemia have identified a characteristic spectrum of cytogenetic and molecular abnormalities. Chromosomal abnormalities are detected in ~30% of patients and somatic gene mutations in up to 90% of patients, most commonly in TET2, SRSF2, and ASXL1. While cytogenetic abnormalities have long been known to impact the prognosis of myeloid neoplasms, recent studies have identified that somatic mutations impact prognosis independent of cytogenetic and clinical variables. This is best exemplified by mutations in ASXL1, which have been uniformly associated with inferior survival. These findings have led to the development of three molecularly inspired prognostic models, in an attempt to more accurately prognosticate in the disease. Our understanding of the genomic landscape of chronic myelomonocytic leukemia continues to evolve, with somatic mutations demonstrating an expanding role in diagnosis, risk stratification, and therapeutic decision-making. Given these findings, molecular profiling by next-generation sequencing should be considered standard of care in all patients.
- Published
- 2021
- Full Text
- View/download PDF
11. RAS mutations drive proliferative chronic myelomonocytic leukemia via a KMT2A-PLK1 axis.
- Author
-
Carr RM, Vorobyev D, Lasho T, Marks DL, Tolosa EJ, Vedder A, Almada LL, Yurcheko A, Padioleau I, Alver B, Coltro G, Binder M, Safgren SL, Horn I, You X, Solary E, Balasis ME, Berger K, Hiebert J, Witzig T, Buradkar A, Graf T, Valent P, Mangaonkar AA, Robertson KD, Howard MT, Kaufmann SH, Pin C, Fernandez-Zapico ME, Geissler K, Droin N, Padron E, Zhang J, Nikolaev S, and Patnaik MM
- Subjects
- Animals, Cell Cycle Proteins metabolism, GTP Phosphohydrolases metabolism, Gene Expression Profiling methods, Gene Expression Regulation, Leukemic, Histone-Lysine N-Methyltransferase metabolism, Kaplan-Meier Estimate, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic therapy, Membrane Proteins metabolism, Mice, Inbred C57BL, Mice, Knockout, Mice, Transgenic, Myeloid-Lymphoid Leukemia Protein metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins metabolism, Signal Transduction genetics, Stem Cell Transplantation methods, Transplantation, Homologous, Exome Sequencing methods, Xenograft Model Antitumor Assays methods, Polo-Like Kinase 1, Mice, Cell Cycle Proteins genetics, GTP Phosphohydrolases genetics, Histone-Lysine N-Methyltransferase genetics, Leukemia, Myelomonocytic, Chronic genetics, Membrane Proteins genetics, Mutation, Myeloid-Lymphoid Leukemia Protein genetics, Protein Serine-Threonine Kinases genetics, Proto-Oncogene Proteins genetics
- Abstract
Proliferative chronic myelomonocytic leukemia (pCMML), an aggressive CMML subtype, is associated with dismal outcomes. RAS pathway mutations, mainly NRAS
G12D , define the pCMML phenotype as demonstrated by our exome sequencing, progenitor colony assays and a Vav-Cre-NrasG12D mouse model. Further, these mutations promote CMML transformation to acute myeloid leukemia. Using a multiomics platform and biochemical and molecular studies we show that in pCMML RAS pathway mutations are associated with a unique gene expression profile enriched in mitotic kinases such as polo-like kinase 1 (PLK1). PLK1 transcript levels are shown to be regulated by an unmutated lysine methyl-transferase (KMT2A) resulting in increased promoter monomethylation of lysine 4 of histone 3. Pharmacologic inhibition of PLK1 in RAS mutant patient-derived xenografts, demonstrates the utility of personalized biomarker-driven therapeutics in pCMML.- Published
- 2021
- Full Text
- View/download PDF
12. Mast cell differentiation of leukemic blasts in diverse myeloid neoplasms: A potential pre-myelomastocytic leukemia condition.
- Author
-
Panda D, Chatterjee G, Khanka T, Ghogale S, Badrinath Y, Deshpande N, Sardana R, Chaturvedi A, Rajpal S, Shetty D, Patkar NV, Gujral S, Subramanian PG, and Tembhare PR
- Subjects
- Adolescent, Adult, Aged, Antigens, CD metabolism, Bone Marrow metabolism, Bone Marrow pathology, Child, Female, Hematologic Neoplasms metabolism, Hematologic Neoplasms pathology, Humans, Immunophenotyping methods, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Male, Mast Cells metabolism, Mastocytosis, Systemic metabolism, Mastocytosis, Systemic pathology, Middle Aged, Myeloproliferative Disorders metabolism, Myeloproliferative Disorders pathology, Primary Myelofibrosis metabolism, Cell Differentiation physiology, Mast Cells pathology, Primary Myelofibrosis pathology
- Abstract
Introduction: Myeloid neoplasm with blasts showing mast cell (MC)-differentiation and MC-component less than 10% of all nucleated cells but not fulfilling the criteria for systemic mastocytosis with associated hematological neoplasm (SM-AHN) or myelomastocytic leukemia (MML) has not been described in the literature. Herein, we report a study of diverse myeloid malignancies with blasts showing MC-differentiation but not meeting the criteria for SM-AHN or MML. We also evaluated the utility of flow-cytometric immunophenotyping (FCI) in the characterization of immature-MCs (iMCs)., Methods: We identified nine patients of myeloid neoplasms and studied their morphological, FCI, immunohistochemistry, cytogenetic and molecular characteristics. We also compared the immunophenotypic features of MCs from patient samples with control samples., Results: The study included patients with newly-diagnosed acute myeloid leukemia (n = 4), chronic myelomonocytic leukemia (n = 1), and chronic myeloid leukemia on follow-up (n = 4) showing MC differentiation in leukemic-blasts. These patients had mildly increased MCs (range, 0.5%-3%) in bone-marrow morphology, including immature-forms and did not meet the criteria for either SM-AHN or MML. On FCI, iMCs were positive for bright-CD117, heterogeneous-CD34, dim-to-negative-HLADR, and moderate-CD203c expression. Expression-levels of CD123 and CD38 were higher (p < 0.001) but CD33 and CD45 were lower in iMCs compared to mature-MC from control samples (p = 0.019 and p = 0.0037)., Conclusion: We reported a rare finding of MC differentiation of leukemic blasts in diverse myeloid neoplasms and proposed it as a potential pre-myelomastocytic leukemia condition. We described the distinct immunophenotypic signature of immature-MCs using commonly used markers and highlighted the utility of FCI for the diagnosis of this entity., (© 2020 International Clinical Cytometry Society.)
- Published
- 2021
- Full Text
- View/download PDF
13. Chronic myelomonocytic leukemia and blastic plasmacytoid dendritic cell neoplasm. A case report and systematic review.
- Author
-
Espasa A, Sorigue M, Tapia G, Cabezon M, Vergara S, Raya M, Navarro JT, Junca J, Zamora L, and Xicoy B
- Subjects
- Aged, 80 and over, CD4 Antigens metabolism, CD56 Antigen metabolism, Dendritic Cells metabolism, Flow Cytometry methods, Humans, Leukemia, Myelomonocytic, Chronic metabolism, Male, Myeloproliferative Disorders metabolism, Myeloproliferative Disorders pathology, Skin Neoplasms metabolism, Skin Neoplasms pathology, Dendritic Cells pathology, Leukemia, Myelomonocytic, Chronic pathology
- Published
- 2021
- Full Text
- View/download PDF
14. Cut-like homeobox 1 (CUX1) tumor suppressor gene haploinsufficiency induces apoptosis evasion to sustain myeloid leukemia.
- Author
-
Supper E, Rudat S, Iyer V, Droop A, Wong K, Spinella JF, Thomas P, Sauvageau G, Adams DJ, and Wong CC
- Subjects
- Animals, Apoptosis drug effects, CASP8 and FADD-Like Apoptosis Regulating Protein genetics, Cell Cycle drug effects, Cell Cycle genetics, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation genetics, Cell Survival genetics, Chromatin Immunoprecipitation, Dipeptides pharmacology, Gene Expression Regulation, Neoplastic drug effects, Gene Ontology, Genes, Tumor Suppressor, Hematopoietic Stem Cells metabolism, Homeodomain Proteins genetics, Humans, Indoles pharmacology, Kaplan-Meier Estimate, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute pathology, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Mutation, Nuclear Proteins deficiency, Nuclear Proteins genetics, Promoter Regions, Genetic, Protein Array Analysis, Repressor Proteins deficiency, Repressor Proteins genetics, fms-Like Tyrosine Kinase 3 genetics, fms-Like Tyrosine Kinase 3 metabolism, Apoptosis genetics, CASP8 and FADD-Like Apoptosis Regulating Protein metabolism, Gene Expression Regulation, Neoplastic genetics, Haploinsufficiency, Homeodomain Proteins metabolism, Leukemia, Myeloid, Acute metabolism, Nuclear Proteins metabolism, Repressor Proteins metabolism
- Abstract
While oncogenes promote tumorigenesis, they also induce deleterious cellular stresses, such as apoptosis, that cancer cells must combat by coopting adaptive responses. Whether tumor suppressor gene haploinsufficiency leads to such phenomena and their mechanistic basis is unclear. Here, we demonstrate that elevated levels of the anti-apoptotic factor, CASP8 and FADD-like apoptosis regulator (CFLAR), promotes apoptosis evasion in acute myeloid leukemia (AML) cells haploinsufficient for the cut-like homeobox 1 (CUX1) transcription factor, whose loss is associated with dismal clinical prognosis. Genome-wide CRISPR/Cas9 screening identifies CFLAR as a selective, acquired vulnerability in CUX1-deficient AML, which can be mimicked therapeutically using inhibitor of apoptosis (IAP) antagonists in murine and human AML cells. Mechanistically, CUX1 deficiency directly alleviates CUX1 repression of the CFLAR promoter to drive CFLAR expression and leukemia survival. These data establish how haploinsufficiency of a tumor suppressor is sufficient to induce advantageous anti-apoptosis cell survival pathways and concurrently nominate CFLAR as potential therapeutic target in these poor-prognosis leukemias.
- Published
- 2021
- Full Text
- View/download PDF
15. Prognostic value of monocyte subset distribution in chronic myelomonocytic leukemia: results of a multicenter study.
- Author
-
Jestin M, Tarfi S, Duchmann M, Badaoui B, Freynet N, Tran Quang V, Sloma I, Droin N, Morabito M, Leclerc M, Maury S, Fenaux P, Solary E, Selimoglu-Buet D, and Wagner-Ballon O
- Subjects
- Humans, Leukemia, Myelomonocytic, Chronic classification, Leukemia, Myelomonocytic, Chronic metabolism, Prognosis, Survival Rate, Leukemia, Myelomonocytic, Chronic pathology, Monocytes pathology
- Published
- 2021
- Full Text
- View/download PDF
16. Type I interferon upregulation and deregulation of genes involved in monopoiesis in chronic myelomonocytic leukemia.
- Author
-
Montalban-Bravo G, Darbaniyan F, Kanagal-Shamanna R, Ganan-Gomez I, Class CA, Sasaki K, Naqvi K, Wei Y, Yang H, Soltysiak KA, Chien KS, Bueso-Ramos C, Do KA, Kantarjian H, and Garcia-Manero G
- Subjects
- Female, Humans, Male, Middle Aged, Gene Expression Regulation, Neoplastic drug effects, Interferon Type I administration & dosage, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Myelopoiesis drug effects, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, Up-Regulation drug effects
- Abstract
Chronic myelomonocytic leukemia (CMML) is characterized by myelomonocytic bias and monocytic proliferation. Whether cell-intrinsic innate immune or inflammatory upregulation mediate disease pathogenesis and phenotype or whether the degree of aberrant monocytic differentiation influences outcomes remains unclear. We compared the transcriptomic features of bone marrow CD34+ cells from 19 patients with CMML and compared to healthy individuals. A total of 1495 genes had significantly differential expression in CMML (q<0.05, fold change>2), including 1271 genes that were significantly upregulated and 224 that were significantly downregulated in CMML. Top upregulated genes were associated with interferon (IFN) alpha and beta signaling, chemokine receptors, IFN gamma, G protein-coupled receptor ligand signaling, and genes involved in immunomodulatory interactions between lymphoid and non-lymphoid cells. Additionally, 6 gene sets were differentially upregulated and 139 were significantly downregulated in patients with myeloproliferative compared to myelodysplastic CMML. A total of 23 genes involved in regulation of monopoiesis were upregulated in CMML compared to healthy controls. We developed a prediction model using Cox regression including 3 of these genes, which differentiated patients into two prognostic subsets with distinct survival outcomes. This data warrants further evaluation of the roles and therapeutic potential of type I IFN signaling and monopoiesis in CMML., (Copyright © 2021. Published by Elsevier Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
17. L1 drives HSC aging and affects prognosis of chronic myelomonocytic leukemia.
- Author
-
Wang Y, Zheng JP, Luo Y, Wang J, Xu L, Wang J, Sedivy JM, Song Z, Wang H, and Ju Z
- Subjects
- Animals, Hematopoietic Stem Cells pathology, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic pathology, Mice, Mice, Knockout, Cellular Senescence, Hematopoietic Stem Cells metabolism, Leukemia, Myelomonocytic, Chronic metabolism, Retroelements
- Published
- 2020
- Full Text
- View/download PDF
18. Phase 1 study of lenzilumab, a recombinant anti-human GM-CSF antibody, for chronic myelomonocytic leukemia.
- Author
-
Patnaik MM, Sallman DA, Mangaonkar AA, Heuer R, Hirvela J, Zblewski D, Al-Kali A, Binder M, Balasis ME, Newman H, Letson C, Kruer TL, Gangat N, Komrokji RS, Tefferi A, Lo A, Shih T, Durrant C, List AF, and Padron E
- Subjects
- Aged, Aged, 80 and over, Antibodies, Monoclonal administration & dosage, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal pharmacokinetics, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Agents, Immunological adverse effects, Antineoplastic Agents, Immunological pharmacokinetics, Chemotherapy, Adjuvant, Dose-Response Relationship, Drug, Drug-Related Side Effects and Adverse Reactions blood, Drug-Related Side Effects and Adverse Reactions immunology, Female, Granulocyte-Macrophage Colony-Stimulating Factor immunology, Humans, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic therapy, Male, Maximum Tolerated Dose, Middle Aged, Recombinant Proteins administration & dosage, Recombinant Proteins adverse effects, Recombinant Proteins pharmacokinetics, Treatment Outcome, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized pharmacokinetics, Antineoplastic Agents, Immunological administration & dosage, Leukemia, Myelomonocytic, Chronic drug therapy
- Published
- 2020
- Full Text
- View/download PDF
19. Concordance among hematopathologists in classifying blasts plus promonocytes: A bone marrow pathology group study.
- Author
-
Foucar K, Hsi ED, Wang SA, Rogers HJ, Hasserjian RP, Bagg A, George TI, Bassett RL Jr, Peterson LC, Morice WG 2nd, Arber DA, Orazi A, and Bueso-Ramos CE
- Subjects
- Adult, Female, Humans, Male, Middle Aged, Blast Crisis classification, Blast Crisis metabolism, Blast Crisis pathology, Bone Marrow metabolism, Bone Marrow pathology, Leukemia, Myelogenous, Chronic, BCR-ABL Positive classification, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Leukemia, Myelomonocytic, Chronic classification, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Monocyte-Macrophage Precursor Cells classification, Monocyte-Macrophage Precursor Cells metabolism, Monocyte-Macrophage Precursor Cells pathology
- Abstract
Enumeration of blasts and promonocytes is essential for World Health Organization (WHO) classification of myelomonocytic neoplasms. The accuracy of distinguishing blasts, promonocytes and monocytes, including normal vs abnormal monocytes, remains controversial. The objective of this analysis is to assess concordances between experienced hematopathologists in classifying cells as blasts, promonocytes, and monocytes according to WHO criteria. Each of 11 hematopathologists assessed glass slides from 20 patients [12 with chronic myelomonocytic leukemia (CMML) and 8 with acute myeloid leukemia (AML)] including blood and BM aspirate smears, and limited nonspecific esterase (NSE) stains. All cases were blindly reviewed. Fleiss' extension of Cohen's kappa for multiple raters was used on these variables, separately for peripheral blood (PB) and bone marrow (BM). Spearman's rank correlation was used to assess correlations between each pair of hematopathologists for each measurement. For the classification based on the sum of blasts and promonocytes in the BM, Fleiss' kappa was estimated as 0.744. For PB, categorizing patients according to the sum of blasts and promonocytes, Fleiss' kappa was estimated as 0.949. Distinction of abnormal monocytes from normal monocytes in PB did not achieve a good concordance and showed strong evidence of differences between hematopathologists (P < .0001). The hematopathologists achieved a good concordance rate of 74% in CMML vs AML classification and a high k rate, confirming that criteria for defining the blasts equivalents (blasts plus promonocytes) could be applied consistently. Identification of monocyte subtypes (abnormal vs normal) was not concordant. Our results support the practice of combining blasts/promonocytes into a single category., (© 2020 John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
20. Myelodysplastic/myeloproliferative neoplasms - Justified inclusion as unique biological entities.
- Author
-
Patnaik MM
- Subjects
- Humans, Leukemia, Myelomonocytic, Chronic classification, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Juvenile classification, Leukemia, Myelomonocytic, Juvenile genetics, Leukemia, Myelomonocytic, Juvenile metabolism, Myelodysplastic Syndromes classification, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes metabolism, Neoplasm Proteins genetics, Neoplasm Proteins metabolism
- Abstract
Competing Interests: Declaration of competing interest The author has served on the advisory board for StemLine Pharmaceuticals.
- Published
- 2020
- Full Text
- View/download PDF
21. Oligo-monocytic CMML and other pre-CMML states: Clinical impact, prognostication and management.
- Author
-
Valent P
- Subjects
- Clonal Hematopoiesis, Diagnosis, Differential, Humans, Prognosis, Leukemia, Myelomonocytic, Chronic diagnosis, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic therapy, Mutation, Myelodysplastic Syndromes diagnosis, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes metabolism, Myelodysplastic Syndromes therapy
- Abstract
Chronic myelomonocytic leukemia (CMML) is defined by myelodysplasia, pathologic accumulation of monocytes and a substantial risk to transform to secondary acute myeloid leukemia (sAML). In recent years, minimal diagnostic criteria for classical CMML and CMML-variants were proposed. Moreover, potential pre-stages of CMML and interface conditions have been postulated. Oligomonocytic CMML is a condition where the absolute peripheral blood monocyte count does not reach a diagnostic level but all other criteria for CMML are fulfilled. Among potential pre-stages of CMML, clonal and non-clonal conditions have been described, including idiopathic monocytosis (IMUS) and clonal monocytosis of unknown significance (CMUS). Patients with myelodysplastic syndromes (MDS), clonal cytopenia of unknown significance (CCUS), clonal hematopoiesis of indeterminate potential (CHIP) and idiopathic cytopenia of undetermined significance (ICUS) may also progress to CMML. The current article provides an overview of pre-CMML conditions and oligomonocytic CMML, with special reference to diagnostic criteria, differential diagnoses, clinical outcomes and management., Competing Interests: Declaration of competing interest P.V. has no conflict of interest in this study. Conflicts of interest (past 3 years) outside the study are as follows: Research grant: Celgene, Incyte, Pfizer; Honoraria (Advisory Board, Speaker): Celgene, Novartis, Incyte, Pfizer, Deciphera., (Copyright © 2019 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
22. Correlation of RAS-Pathway Mutations and Spontaneous Myeloid Colony Growth with Progression and Transformation in Chronic Myelomonocytic Leukemia-A Retrospective Analysis in 337 Patients.
- Author
-
Geissler K, Jäger E, Barna A, Gurbisz M, Graf T, Graf E, Nösslinger T, Pfeilstöcker M, Tüchler H, Sliwa T, Keil F, Geissler C, Heibl S, Thaler J, Machherndl-Spandl S, Zach O, Weltermann A, Bettelheim P, Stauder R, Zebisch A, Sill H, Schwarzinger I, Schneeweiss B, Öhler L, Ulsperger E, Kusec R, Germing U, Sperr WR, Knöbl P, Jäger U, Hörmann G, and Valent P
- Subjects
- 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, 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
- 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 (≥20/10
5 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.- Published
- 2020
- Full Text
- View/download PDF
23. Transcriptomic analysis implicates necroptosis in disease progression and prognosis in myelodysplastic syndromes.
- Author
-
Montalban-Bravo G, Class CA, Ganan-Gomez I, Kanagal-Shamanna R, Sasaki K, Richard-Carpentier G, Naqvi K, Wei Y, Yang H, Soltysiak KA, Chien K, Bueso-Ramos C, Do KA, Kantarjian H, and Garcia-Manero G
- Subjects
- Aged, Antigens, CD34 metabolism, Bone Marrow metabolism, Disease Progression, Female, Humans, Inflammation, Leukemia, Myelomonocytic, Chronic diagnosis, Leukemia, Myelomonocytic, Chronic metabolism, Male, Middle Aged, Myelodysplastic Syndromes metabolism, Phenotype, Prognosis, Protein Kinases metabolism, Receptor-Interacting Protein Serine-Threonine Kinases metabolism, Myelodysplastic Syndromes diagnosis, Necroptosis, Transcriptome
- Abstract
Myelodysplastic syndromes (MDS) are characterized by ineffective hematopoiesis and cytopenias due to uncontrolled programmed cell death. The presence of pro-inflammatory cytokines and constitutive activation of innate immunity signals in MDS cells suggest inflammatory cell death, such as necroptosis, may be responsible for disease phenotype. We evaluated 64 bone marrow samples from 55 patients with MDS or chronic myelomonocytic leukemia (CMML) obtained prior to (n = 46) or after (n = 18) therapy with hypomethylating agents (HMAs). RNA from sorted bone marrow CD34+ cells was isolated and subject to amplification and RNA-Seq. Compared with healthy controls, expression levels of MLKL (CMML: 2.09 log2FC, p = 0.0013; MDS: 1.89 log2FC, p = 0.003), but not RIPK1 or RIPK3, were significantly upregulated. Higher expression levels of MLKL were associated with lower hemoglobin levels at diagnosis (-0.19 log2FC per 1 g/dL increase of Hgb, p = 0.03). Significant reduction in MLKL levels was observed after HMA therapy (-1.06 log2FC, p = 0.05) particularly among nonresponders (-2.89 log2FC, p = 0.06). Higher RIPK1 expression was associated with shorter survival (HR 1.92, 95% CI 1.00-3.67, p = 0.049 by Cox proportional hazards). This data provides further support for a role of necroptosis in MDS, and potentially response to HMAs and prognosis. This data also indicate that RIPK1/RIPK3/MLKL are potential therapeutic targets in MDS.
- Published
- 2020
- Full Text
- View/download PDF
24. The clinical and pathological panoply of systemic mastocytosis.
- Author
-
Radia DH, Green A, Oni C, and Moonim M
- Subjects
- Adult, Humans, Leukemia, Myelomonocytic, Chronic diagnosis, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Mastocytosis, Systemic diagnosis, Mastocytosis, Systemic metabolism, Mastocytosis, Systemic pathology
- Abstract
Mastocytosis is a rare disease with varied presentation, myriad symptomatology and variable prognosis. Most patients present with cutaneous disease and mediator-related symptomatology with a small subset having systemic disease (systemic mastocytosis, SM). A subset of the latter develops synchronous or metachronous haematologic neoplasms (SM-AHN), most commonly chronic myelomonocytic leukaemia (CMML). Advanced systemic mastocytosis (ASM) is seen in a relatively small number of patients and is usually associated with organ dysfunction, and may present with hepatosplenomegaly, lymphadenopathy and ascites with progression to leukaemic transformation (mast cell leukaemia/acute myeloid leukaemia) occurring in a few patients. This paper discusses the clinical and pathologic features of the entire spectrum of SM in adults., (© 2019 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
25. Genetic and epigenetic factors interacting with clonal hematopoiesis resulting in chronic myelomonocytic leukemia.
- Author
-
Carr RM and Patnaik MM
- Subjects
- Humans, Mutation, Epigenesis, Genetic, Epistasis, Genetic, Gene Expression Regulation, Leukemic, Hematopoiesis, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells pathology, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes metabolism, Myelodysplastic Syndromes pathology, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics
- Abstract
Purpose of Review: Since 2016, the WHO has recognized the significant phenotypic heterogeneity of chronic myelomonocytic leukemia (CMML) as a myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) overlap disease. Although sharing many somatic mutations with MDS and MPN, the purpose of this review is to put recent biological findings of CMML in the context of evolutionary theory, highlighting it as a distinct evolutionary trajectory occurring in the context of clonal hematopoiesis., Recent Findings: Clonal hematopoiesis of indeterminate potential (CHIP), with a mutational spectrum and prevalence correlated with age, has been defined. Enriched in DNMT3A, TET2, and ASXL1 mutations, clonal evolution can progress into various evolutionary trajectories including CMML. Impact of founder mutations (primarily TET2) on increased hematopoietic stem cell fitness has been well characterized. Epistatic interactions between mutations and epigenetic events have been explored, both in CMML and its pediatric counterpart juvenile myelomonocytic leukemia, including CMML transformation to acute myeloid leukemia. Together, these findings have contributed significantly toward CMML evolutionary dynamics., Summary: Despite relatively few 'driver' mutations in CMML, evolutionary development of chronic leukemia remains incompletely understood. Recent studies have shed light on the importance of studying epigenetic consequences of mutations and epistasis between key mutations to better understand clonal architecture and evolutionary dynamics.
- Published
- 2020
- Full Text
- View/download PDF
26. Biology and prognostic impact of clonal plasmacytoid dendritic cells in chronic myelomonocytic leukemia.
- Author
-
Lucas N, Duchmann M, Rameau P, Noël F, Michea P, Saada V, Kosmider O, Pierron G, Fernandez-Zapico ME, Howard MT, King RL, Niyongere S, Diop MK, Fenaux P, Itzykson R, Willekens C, Ribrag V, Fontenay M, Padron E, Soumelis V, Droin N, Patnaik MM, and Solary E
- Subjects
- Aged, Aged, 80 and over, Antigens, CD metabolism, Biomarkers metabolism, Bone Marrow metabolism, Bone Marrow pathology, Dendritic Cells metabolism, Female, Humans, Leukemia, Myelomonocytic, Chronic metabolism, Male, Membrane Proteins metabolism, Prognosis, T-Lymphocytes, Regulatory metabolism, T-Lymphocytes, Regulatory pathology, Dendritic Cells pathology, Leukemia, Myelomonocytic, Chronic pathology
- Abstract
Islands of CD123
high cells have been commonly described in the bone marrow of patients with chronic myelomonocytic leukemia (CMML). Using a multiparameter flow cytometry assay, we detected an excess of CD123+ mononucleated cells that are lineage-negative, CD45+ , CD11c- , CD33- , HLA-DR+ , BDCA-2+ , BDCA-4+ in the bone marrow of 32/159 (20%) patients. Conventional and electron microscopy, flow cytometry detection of cell surface markers, gene expression analyses, and the ability to synthesize interferon alpha in response to Toll-like receptor agonists identified these cells as bona fide plasmacytoid dendritic cells (pDCs). Whole-exome sequencing of sorted monocytes and pDCs identified somatic mutations in genes of the oncogenic RAS pathway in the two cell types of every patient. CD34+ cells could generate high amount of pDCs in the absence of FMS-like tyrosine kinase 3-ligand (FLT3L). Finally, an excess of pDCs correlates with regulatory T cell accumulation and an increased risk of acute leukemia transformation. These results demonstrate the FLT3L-independent accumulation of clonal pDCs in the bone marrow of CMML patients with mutations affecting the RAS pathway, which is associated with a higher risk of disease progression.- Published
- 2019
- Full Text
- View/download PDF
27. Chronic myelomonocytic leukaemia followed by blastic plasmacytoid dendritic cell neoplasm.
- Author
-
Lebecque B, Vial JP, Pigneux A, and Lechevalier N
- Subjects
- Aged, Female, Humans, Male, Antigens, CD metabolism, Azacitidine administration & dosage, Dendritic Cells metabolism, Dendritic Cells pathology, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Neoplasm Proteins metabolism
- Published
- 2019
- Full Text
- View/download PDF
28. Dynamic gene regulation by nuclear colony-stimulating factor 1 receptor in human monocytes and macrophages.
- Author
-
Bencheikh L, Diop MK, Rivière J, Imanci A, Pierron G, Souquere S, Naimo A, Morabito M, Dussiot M, De Leeuw F, Lobry C, Solary E, and Droin N
- Subjects
- CRISPR-Cas Systems, Cell Membrane chemistry, Cell Membrane drug effects, Cell Membrane metabolism, Chromatin chemistry, Chromatin drug effects, Chromatin metabolism, Early Growth Response Protein 1 genetics, Early Growth Response Protein 1 metabolism, Fluorescent Dyes chemistry, Gene Editing, HEK293 Cells, Histones genetics, Histones metabolism, Humans, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Macrophage Colony-Stimulating Factor metabolism, Macrophages cytology, Macrophages metabolism, Maleimides chemistry, Primary Cell Culture, Protein Binding, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor antagonists & inhibitors, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Signal Transduction, THP-1 Cells, YY1 Transcription Factor genetics, YY1 Transcription Factor metabolism, ets-Domain Protein Elk-1 genetics, ets-Domain Protein Elk-1 metabolism, Gene Expression Regulation, Leukemia, Myelomonocytic, Chronic genetics, Macrophage Colony-Stimulating Factor pharmacology, Macrophages drug effects, Receptors, Granulocyte-Macrophage Colony-Stimulating Factor genetics
- Abstract
Despite their location at the cell surface, several receptor tyrosine kinases (RTK) are also found in the nucleus, as either intracellular domains or full length proteins. However, their potential nuclear functions remain poorly understood. Here we find that a fraction of full length Colony Stimulating Factor-1 Receptor (CSF-1R), an RTK involved in monocyte/macrophage generation, migrates to the nucleus upon CSF-1 stimulation in human primary monocytes. Chromatin-immunoprecipitation identifies the preferential recruitment of CSF-1R to intergenic regions, where it co-localizes with H3K4me1 and interacts with the transcription factor EGR1. When monocytes are differentiated into macrophages with CSF-1, CSF-1R is redirected to transcription starting sites, colocalizes with H3K4me3, and interacts with ELK and YY1 transcription factors. CSF-1R expression and chromatin recruitment is modulated by small molecule CSF-1R inhibitors and altered in monocytes from chronic myelomonocytic leukemia patients. Unraveling this dynamic non-canonical CSF-1R function suggests new avenues to explore the poorly understood functions of this receptor and its ligands.
- Published
- 2019
- Full Text
- View/download PDF
29. GATA2 hypomorphism induces chronic myelomonocytic leukemia in mice.
- Author
-
Harada N, Hasegawa A, Hirano I, Yamamoto M, and Shimizu R
- Subjects
- Age Factors, Animals, Biomarkers, Disease Models, Animal, GATA2 Transcription Factor metabolism, Gene Expression, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Leukocyte Count, Leukocytosis genetics, Leukocytosis metabolism, Leukocytosis pathology, Mice, Mice, Knockout, Monocytes, RNA, Messenger, GATA2 Transcription Factor genetics, Genetic Predisposition to Disease, Leukemia, Myelomonocytic, Chronic genetics, Polymorphism, Genetic
- Abstract
The transcription factor GATA2 regulates normal hematopoiesis, particularly in- stem cell maintenance and myeloid differentiation. Various heteroallelic GATA2 gene mutations are associated with a variety of hematological neoplasms, including myelodysplastic syndromes and leukemias. Here, we report that impaired GATA2 expression induces myelodysplastic and myeloproliferative neoplasm development in elderly animals, and this neoplasm resembles chronic myelomonocytic leukemia in humans. GATA2 hypomorphic mutant (G2
f GN / fGN ) mice that were generated by the germline insertion of a neocassette into the Gata2 gene locus avoided the early embryonic lethality observed in Gata2-null mice. However, adult G2f GN / fGN mice suffered from exacerbated leukocytosis concomitant with progressive anemia and thrombocytopenia and eventually developed massive granulomonocytosis accompanied by trilineage dysplasia. The reconstitution activity of G2f GN / fGN mouse stem cells was impaired. Furthermore, G2f GN / fGN progenitors showed myeloid lineage-biased proliferation and differentiation. Myeloid progenitor accumulation started at a younger age in G2f GN / fGN mice and appeared to worsen with age. G2f GN / fGN mice showed increased expression of transcripts encoding cytokine receptors, such as macrophage colony-stimulating factor receptor and interleukin-6 receptor, in granulocyte-monocyte progenitors. This increased expression could be correlated with the hypersensitive granulomonocytic proliferation reaction when the mice were exposed to lipopolysaccharide. Taken together, these observations indicate that GATA2 hypomorphism leads to a hyperreactive defense response to infections, and this reaction is attributed to a unique intrinsic cell defect in the regulation of myeloid expansion that increases the risk of hematological neoplasm transformation., (© 2019 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.)- Published
- 2019
- Full Text
- View/download PDF
30. A miR-150/TET3 pathway regulates the generation of mouse and human non-classical monocyte subset.
- Author
-
Selimoglu-Buet D, Rivière J, Ghamlouch H, Bencheikh L, Lacout C, Morabito M, Diop M, Meurice G, Breckler M, Chauveau A, Debord C, Debeurme F, Itzykson R, Chapuis N, Willekens C, Wagner-Ballon O, Bernard OA, Droin N, and Solary E
- Subjects
- Animals, DNA Methylation, Down-Regulation, Female, Gene Expression Regulation, Leukemic, Humans, K562 Cells, Leukemia, Myelomonocytic, Chronic metabolism, Male, Mice, Mice, Knockout, Primary Cell Culture, Promoter Regions, Genetic, DNA-Binding Proteins metabolism, Dioxygenases metabolism, Leukemia, Myelomonocytic, Chronic immunology, MicroRNAs metabolism, Monocytes metabolism, Proto-Oncogene Proteins metabolism
- Abstract
Non-classical monocyte subsets may derive from classical monocyte differentiation and the proportion of each subset is tightly controlled. Deregulation of this repartition is observed in diverse human diseases, including chronic myelomonocytic leukemia (CMML) in which non-classical monocyte numbers are significantly decreased relative to healthy controls. Here, we identify a down-regulation of hsa-miR-150 through methylation of a lineage-specific promoter in CMML monocytes. Mir150 knock-out mice demonstrate a cell-autonomous defect in non-classical monocytes. Our pulldown experiments point to Ten-Eleven-Translocation-3 (TET3) mRNA as a hsa-miR-150 target in classical human monocytes. We show that Tet3 knockout mice generate an increased number of non-classical monocytes. Our results identify the miR-150/TET3 axis as being involved in the generation of non-classical monocytes.
- Published
- 2018
- Full Text
- View/download PDF
31. Multicenter validation of the flow measurement of classical monocyte fraction for chronic myelomonocytic leukemia diagnosis.
- Author
-
Tarfi S, Harrivel V, Dumezy F, Guy J, Roussel M, Mimoun A, Fenaux P, Chapuis N, Solary E, Selimoglu-Buet D, and Wagner-Ballon O
- Subjects
- Aged, Aged, 80 and over, Biomarkers, Female, France, Humans, Immunophenotyping, Male, Middle Aged, ROC Curve, Reproducibility of Results, Sensitivity and Specificity, Flow Cytometry methods, Leukemia, Myelomonocytic, Chronic diagnosis, Leukemia, Myelomonocytic, Chronic metabolism, Monocytes metabolism
- Abstract
Peripheral blood monocytes include three subsets defined by CD14 and CD16 surface markers. An increase in the CD14
++ CD16- classical monocyte fraction ≥ 94% of the total monocytes was proposed to rapidly and efficiently distinguish chronic myelomonocytic leukemia from reactive monocytosis. The robustness of this assay required a multicenter validation. The flow cytometry assay designed to quantify peripheral blood monocyte subsets was implemented by multiple diagnosis laboratories in France. A nationwide survey was performed to evaluate its performance. All the 48 French laboratories answered the questionnaire, revealing that 63% use this assay routinely. Central blind reanalysis of 329 cytometry files collected from five laboratories demonstrated an excellent correlation in classical monocyte fraction measurement (r = 0.93; p < 0.0001). The cutoff value of 94% classical monocytes being the critical readout for diagnosis, we then compared 115 patients with classical monocytes ≥ 94% and 214 patients with a fraction < 94% between initial analysis and reanalysis. An agreement was obtained in 311 files. Finally, an overt diagnosis, available for 86 files, confirmed a good sensitivity (93.6%) and specificity (89.7%). This survey demonstrates the robustness of the flow assay with limited variability of classical monocyte percentage between centers, validates the 94% cutoff value, and confirms its sensitivity and specificity.- Published
- 2018
- Full Text
- View/download PDF
32. Serotonin receptor type 1B constitutes a therapeutic target for MDS and CMML.
- Author
-
Banús-Mulet A, Etxabe A, Cornet-Masana JM, Torrente MÁ, Lara-Castillo MC, Palomo L, Nomdedeu M, Díaz-Beyá M, Solé F, Nomdedeu B, Esteve J, and Risueño RM
- Subjects
- Adult, Cell Line, Tumor, Female, Humans, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic metabolism, Male, Middle Aged, Myelodysplastic Syndromes drug therapy, Myelodysplastic Syndromes metabolism, Receptor, Serotonin, 5-HT1B metabolism, Leukemia, Myelomonocytic, Chronic pathology, Myelodysplastic Syndromes pathology, Receptor, Serotonin, 5-HT1B drug effects, Serotonin Antagonists therapeutic use
- Abstract
Myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML) are chronic myeloid clonal neoplasms. To date, the only potentially curative therapy for these disorders remains allogeneic hematopoietic progenitor cell transplantation (HCT), although patient eligibility is limited due to high morbimortality associated with this procedure coupled with advanced age of most patients. Dopamine receptors (DRs) and serotonin receptors type 1 (HTR1s) were identified as cancer stem cell therapeutic targets in acute myeloid leukemia. Given their close pathophysiologic relationship, expression of HTR1s and DRs was interrogated in MDS and CMML. Both receptors were differentially expressed in patient samples compared to healthy donors. Treatment with HTR1B antagonists reduced cell viability. HTR1 antagonists showed a synergistic cytotoxic effect with currently approved hypomethylating agents in AML cells. Our results suggest that HTR1B constitutes a novel therapeutic target for MDS and CMML. Due to its druggability, the clinical development of new regimens based on this target is promising.
- Published
- 2018
- Full Text
- View/download PDF
33. Clinical Utility of Classical and Nonclassical Monocyte Percentage in the Diagnosis of Chronic Myelomonocytic Leukemia.
- Author
-
Hudson CA, Burack WR, Leary PC, and Bennett JM
- Subjects
- Biomarkers metabolism, Bone Marrow metabolism, Diagnosis, Differential, Flow Cytometry, Humans, Leukemia, Myelomonocytic, Chronic metabolism, Leukocyte Count, Monocytes classification, Sensitivity and Specificity, Leukemia, Myelomonocytic, Chronic diagnosis, Monocytes metabolism
- Abstract
Objectives: To determine if a clinically applicable flow cytometry methodology could identify chronic myelomonocytic leukemia (CMML) cases., Methods: Monocyte subset screening (CD14/CD16 expression) was performed on 68 blood and 25 bone marrow specimens with a monocytosis and/or flagged as possible CMML. Fifty thousand total events were obtained per case. Cases were categorized as CMML, atypical chronic myeloid leukemia (aCML), or non-CMML + non-aCML by clinicopathologic diagnosis., Results: The methodology differentiated blood and bone marrow CMML cases from non-CMML + non-aCML but not three aCML cases in the clinical setting. Furthermore, a decreased percentage of nonclassical monocytes (CD14dimCD16+) showed better sensitivity than the previously described approach that relied on increased percentage of classical monocytes (CD14brightCD16-)., Conclusions: Quantification of monocyte subsets is useful in clinical practice as a diagnostic marker of CMML in blood and bone marrow specimens. The percentage of nonclassical monocytes should be included in analysis of monocyte subsets.
- Published
- 2018
- Full Text
- View/download PDF
34. CBL mutation and MEFV single-nucleotide variant are important genetic predictors of tumor reduction in glucocorticoid-treated patients with chronic myelomonocytic leukemia.
- Author
-
Watanabe J, Sato K, Osawa Y, Horiuchi T, Kato S, Hikota-Saga R, Maekawa T, Yamamura T, Kobayashi A, Kobayashi S, and Kimura F
- Subjects
- Cytokines metabolism, Forecasting, Glucocorticoids pharmacology, Granulocyte-Macrophage Colony-Stimulating Factor, Humans, Inflammation Mediators metabolism, Leukemia, Myelomonocytic, Chronic metabolism, Methylprednisolone pharmacology, Phosphorylation genetics, STAT5 Transcription Factor metabolism, Signal Transduction, Tumor Cells, Cultured, Glucocorticoids administration & dosage, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic pathology, Loss of Function Mutation genetics, Methylprednisolone administration & dosage, Polymorphism, Single Nucleotide genetics, Proto-Oncogene Proteins c-cbl genetics, Pyrin genetics
- Abstract
Glucocorticoid (GC) therapy occasionally relieves tumor-related fever and promotes tumor reduction in patients with chronic myelomonocytic leukemia (CMML). A mutation analysis of 24 patients with CMML revealed the relationship of GC effectiveness, defined as a monocyte reduction of > 50% within 3 days of methylprednisolone administration, with the MEFV single-nucleotide variant (SNV) and CBL mutation. Lipopolysaccharide-stimulated monocytes harboring MEFV E148Q produced greater amounts of IL-1β and TNF-α than did wild-type monocytes; this was effectively suppressed by GC. Primary CMML cells harboring the MEFV SNV and CBL mutation, and the myelomonocytic leukemia cell line GDM-1, harboring the CBL mutation, were both more significantly suppressed than non-mutated cells following GC treatment in the presence of GM-CSF. A loss-of-function CBL mutation prolonged STAT5 phosphorylation after GM-CSF stimulation, which was rapidly terminated in both patient samples and GDM-1 cells. In conclusion, GC therapy effectively treats CMML cells harboring the MEFV SNV and CBL mutation by reducing inflammatory cytokine production and terminating prolonged STAT5 phosphorylation in the GM-CSF signaling pathway.
- Published
- 2018
- Full Text
- View/download PDF
35. Somatic SETBP1 mutations in myeloid neoplasms.
- Author
-
Makishima H
- Subjects
- Animals, Chromosomes, Human, Pair 7 genetics, Core Binding Factor Alpha 2 Subunit genetics, Core Binding Factor Alpha 2 Subunit metabolism, Humans, Mice, Proto-Oncogene Proteins c-myb genetics, Proto-Oncogene Proteins c-myb metabolism, Repressor Proteins genetics, Repressor Proteins metabolism, Carrier Proteins genetics, Carrier Proteins metabolism, Chromosome Deletion, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute therapy, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative genetics, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative metabolism, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative mortality, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative therapy, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic mortality, Leukemia, Myelomonocytic, Chronic therapy, Leukemia, Myelomonocytic, Juvenile, Nuclear Proteins genetics, Nuclear Proteins metabolism
- Abstract
SETBP1 is a SET-binding protein regulating self-renewal potential through HOXA-protein activation. Somatic SETBP1 mutations were identified by whole exome sequencing in several phenotypes of myelodysplastic/myeloproliferative neoplasms (MDS/MPN), including atypical chronic myeloid leukemia, chronic myelomonocytic leukemia, and juvenile myelomonocytic leukemia as well as in secondary acute myeloid leukemia (sAML). Surprisingly, its recurrent somatic activated mutations are located at the identical positions of germline mutations reported in congenital Schinzel-Giedion syndrome. In general, somatic SETBP1 mutations have a significant clinical impact on the outcome as poor prognostic factor, due to downstream HOXA-pathway as well as associated aggressive types of chromosomal defects (-7/del(7q) and i(17q)), which is consistent with wild-type SETBP1 activation in aggressive types of acute myeloid leukemia and leukemic evolution. Biologically, mutant SETBP1 attenuates RUNX1 and activates MYB. The studies of mouse models confirmed biological significance of SETBP1 mutations in myeloid leukemogenesis, particularly associated with ASXL1 mutations. SETBP1 is a major oncogene in myeloid neoplasms, which cooperates with various genetic events and causes distinct phenotypes of MDS/MPN and sAML.
- Published
- 2017
- Full Text
- View/download PDF
36. In vitro and in vivo effects of JAK2 inhibition in chronic myelomonocytic leukemia.
- Author
-
Geissler K, Jäger E, Barna A, Sliwa T, Knöbl P, Schwarzinger I, Gisslinger H, and Valent P
- Subjects
- Aged, Aged, 80 and over, Antineoplastic Agents therapeutic use, Cell Proliferation drug effects, Colony-Forming Units Assay, Dose-Response Relationship, Drug, Female, Granulocyte-Macrophage Progenitor Cells drug effects, Humans, Janus Kinase 2 genetics, Leukemia, Myelomonocytic, Chronic diagnosis, Leukemia, Myelomonocytic, Chronic drug therapy, Male, Middle Aged, Mutation, Nitriles, Protein Kinase Inhibitors therapeutic use, Pyrazoles pharmacology, Pyrazoles therapeutic use, Pyrimidines, Tomography, X-Ray Computed, Antineoplastic Agents pharmacology, Janus Kinase 2 antagonists & inhibitors, Leukemia, Myelomonocytic, Chronic metabolism, Protein Kinase Inhibitors pharmacology
- Abstract
In chronic myelomonocytic leukemia (CMML), colony-forming units granulocyte/macrophage (CFU-GM), which grow in vitro in the absence of exogenous growth factors, arise from the abnormal clone that is responsible for the overproduction of granulomonocytic cells. Previous in vitro findings including ours suggest that divergent molecular aberrations in CMML seem to converge within the GM-CSF signaling pathway. As JAK2 is a sentinel kinase in this pathway, JAK2 inhibition may be an attractive treatment approach in CMML. We investigated the in vitro effects of the specific JAK2 inhibitor TG101209 on the autonomous CFU-GM formation from peripheral blood mononuclear cells of patients with CMML. TG101209 was found to either block or strongly inhibit spontaneous CFU-GM growth in all 10 patients tested. This inhibitory effect was dose dependent and significantly more pronounced as compared to the inhibitory effect on stimulated CFU-GM growth from normal individuals. In a CMML patient with splenomegaly, who was treated with the JAK1/2 inhibitor ruxolitinib off label, we can demonstrate a spleen response and the disappearance of constitutional symptoms which was associated with a decrease in autonomous CFU-GM formation ex vivo. Pharmacological JAK2 inhibition may be an interesting approach to be systematically studied in patients with CMML., (© 2016 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
37. A phase I trial of the aurora kinase inhibitor, ENMD-2076, in patients with relapsed or refractory acute myeloid leukemia or chronic myelomonocytic leukemia.
- Author
-
Yee KW, Chen HW, Hedley DW, Chow S, Brandwein J, Schuh AC, Schimmer AD, Gupta V, Sanfelice D, Johnson T, Le LW, Arnott J, Bray MR, Sidor C, and Minden MD
- Subjects
- Adult, Aged, Aged, 80 and over, Aurora Kinases antagonists & inhibitors, Drug Resistance, Neoplasm, Extracellular Signal-Regulated MAP Kinases metabolism, Female, Humans, Leukemia, Myeloid, Acute metabolism, Leukemia, Myelomonocytic, Chronic metabolism, Male, Maximum Tolerated Dose, Middle Aged, Proto-Oncogene Proteins c-akt metabolism, Proto-Oncogene Proteins c-kit antagonists & inhibitors, Recurrence, Ribosomal Protein S6 Kinases metabolism, STAT5 Transcription Factor metabolism, Treatment Outcome, Vascular Endothelial Growth Factor Receptor-2 antagonists & inhibitors, fms-Like Tyrosine Kinase 3 antagonists & inhibitors, Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myelomonocytic, Chronic drug therapy, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Pyrazoles adverse effects, Pyrazoles pharmacokinetics, Pyrazoles pharmacology, Pyrazoles therapeutic use, Pyrimidines adverse effects, Pyrimidines pharmacokinetics, Pyrimidines pharmacology, Pyrimidines therapeutic use
- Abstract
ENMD-2076 is a novel, orally-active molecule that inhibits Aurora A kinase, as well as c-Kit, FLT3 and VEGFR2. A phase I study was conducted to determine the maximum tolerated dose (MTD), recommended phase 2 dose (RP2D) and toxicities of ENMD-2076 in patients with acute myeloid leukemia (AML) and chronic myelomonocytic leukemia (CMML). Patients received escalating doses of ENMD-2076 administered orally daily [225 mg (n = 7), 375 mg (n = 6), 325 mg (n = 9), or 275 mg (n = 5)]. Twenty-seven patients were treated (26 AML; 1 CMML-2). The most common non-hematological toxicities of any grade, regardless of association with drug, were fatigue, diarrhea, dysphonia, dyspnea, hypertension, constipation, and abdominal pain. Dose-limiting toxicities (DLTs) consisted of grade 3 fatigue, grade 3 typhilitis, grade 3 syncope and grade 3 QTc prolongation). Of the 16 evaluable patients, one patient achieved a complete remission with incomplete count recovery (CRi), three experienced a morphologic leukemia-free state (MLFS) with a major hematologic improvement in platelets (HI-P), and 5 other patients had a reduction in marrow blast percentage (i.e. 11-65 %). The RP2D in this patient population is 225 mg orally once daily.
- Published
- 2016
- Full Text
- View/download PDF
38. CD33 is frequently expressed in cases of myelodysplastic syndrome and chronic myelomonocytic leukemia with elevated blast count.
- Author
-
Sanford D, Garcia-Manero G, Jorgensen J, Konoplev S, Pierce S, Cortes J, Kantarjian H, and Ravandi F
- Subjects
- Cell Count, Flow Cytometry, Humans, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Leukemia, Myelomonocytic, Chronic pathology, Myelodysplastic Syndromes pathology, Myeloid Progenitor Cells pathology, Leukemia, Myelomonocytic, Chronic metabolism, Myelodysplastic Syndromes metabolism, Myeloid Progenitor Cells metabolism, Sialic Acid Binding Ig-like Lectin 3 analysis
- Published
- 2016
- Full Text
- View/download PDF
39. A Multi-Institution Phase I Trial of Ruxolitinib in Patients with Chronic Myelomonocytic Leukemia (CMML).
- Author
-
Padron E, Dezern A, Andrade-Campos M, Vaddi K, Scherle P, Zhang Q, Ma Y, Balasis ME, Tinsley S, Ramadan H, Zimmerman C, Steensma DP, Roboz GJ, Lancet JE, List AF, Sekeres MA, and Komrokji RS
- Subjects
- Aged, Aged, 80 and over, Antineoplastic Agents administration & dosage, Antineoplastic Agents adverse effects, Biomarkers, Cytokines metabolism, Female, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Humans, Inflammation Mediators, Leukemia, Myelomonocytic, Chronic diagnosis, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic mortality, Male, Middle Aged, Mutation, Nitriles, Protein Kinase Inhibitors administration & dosage, Protein Kinase Inhibitors adverse effects, Pyrazoles administration & dosage, Pyrazoles adverse effects, Pyrimidines, STAT5 Transcription Factor metabolism, Treatment Outcome, Antineoplastic Agents therapeutic use, Leukemia, Myelomonocytic, Chronic drug therapy, Protein Kinase Inhibitors therapeutic use, Pyrazoles therapeutic use
- Abstract
Purpose: To conduct a phase I clinical trial exploring the safety and efficacy of ruxolitinib, a JAK1/2 inhibitor, for chronic myelomonocytic leukemia (CMML)., Experimental Design: Patients with CMML-1 were included without regard to previous therapy. Key exclusion criteria included an absolute neutrophil count (ANC) <0.25 × 10(3) cells/dL and a platelet count <35 × 10(3) cells/dL. Four cohorts were enrolled using a "rolling six" study design, with doses ranging from 5 to 20 mg twice daily of ruxolitinib in 5-mg dose escalations., Results: Between March 2013 and January 2015, 20 patients were enrolled and treated with ruxolitinib. Seventy percent of patients had the proliferative subtype and 47% had higher risk disease by the Global MD Anderson Scoring System. Eight patients (42%) received a prior hypomethylating agent. No dose-limiting toxicities for ruxolitinib were identified. One subject had grade (G)3 thrombocytopenia with no other drug-associated G3 or G4 adverse events. The mean duration of therapy was 122 days (range, 28-409 days). Four had hematologic improvement and one patient had a partial response per 2006 International Working Group (IWG) criteria. Five of 9 patients with splenomegaly had a reduction in spleen size. Ten of 11 patients with reported disease-related symptoms had clinically meaningful or complete resolution. When combining IWG and spleen responses, a total response rate of 35% (n = 7) was identified. Correlative analysis demonstrated a reduction in inflammatory cytokines and GM-CSF-dependent STAT5 phosphorylation., Conclusions: The recommended phase II dose of ruxolitinib is 20 mg twice daily. We demonstrate that ruxolitinib has promising activity in CMML with particular benefit in those with disease-related B symptoms that warrants further study. Clin Cancer Res; 22(15); 3746-54. ©2016 AACRSee related commentary by Solary, p. 3707., Competing Interests: of Potential Conflicts of Interest E. Padron reports receiving commercial research grants from Cell Therapeutics and Incyte Corporation, speakers bureau honoraria from Novartis, and is a consultant/advisory board member for Cell Therapeutics. K. Vaddi has ownership interest (including patents) in Incyte Corporation. S. Tinsley reports receiving speakers bureau honoraria from Incyte Corporation. D.P. Steensma is a consultant/advisory board member for Amgen, Celgene, Incyte Corporation, and Novartis. R.S. Komrokji reports receiving speakers bureau honoraria from and is a consultant/advisory board member for Incyte Corporation. No potential conflicts of interest were disclosed by the other authors., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
40. Unplugging JAK/STAT in Chronic Myelomonocytic Leukemia.
- Author
-
Solary E
- Subjects
- Humans, Nitriles, Pyrimidines, Signal Transduction drug effects, Treatment Outcome, Antinematodal Agents therapeutic use, Granulocyte-Macrophage Colony-Stimulating Factor metabolism, Janus Kinases metabolism, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic metabolism, Pyrazoles therapeutic use, STAT Transcription Factors metabolism
- Abstract
The proliferative component of chronic myelomonocytic leukemia, related to an increased sensitivity of myeloid progenitors to granulocyte macrophage-colony stimulating factor, suggests dedicated therapeutic approaches. In this issue, ruxolitinib, a JAK1 and -2 inhibitory drug, is shown to induce objective responses in chronic myelomonocytic leukemia patients. Clin Cancer Res; 22(15); 3707-9. ©2016 AACRSee related article by Padron et al., p. 3746., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
41. Double minute chromosomes in acute myeloid leukemia, myelodysplastic syndromes, and chronic myelomonocytic leukemia are associated with micronuclei, MYC or MLL amplification, and complex karyotype.
- Author
-
Huh YO, Tang G, Talwalkar SS, Khoury JD, Ohanian M, Bueso-Ramos CE, and Abruzzo LV
- Subjects
- Adult, Aged, Aged, 80 and over, Chromosome Aberrations, Female, Gene Amplification, Humans, Karyotyping methods, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute mortality, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic mortality, Male, Middle Aged, Myelodysplastic Syndromes metabolism, Myelodysplastic Syndromes mortality, Proto-Oncogene Proteins c-myc metabolism, Young Adult, Histone-Lysine N-Methyltransferase genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myelomonocytic, Chronic genetics, Micronuclei, Chromosome-Defective classification, Myelodysplastic Syndromes genetics, Myeloid-Lymphoid Leukemia Protein genetics, Proto-Oncogene Proteins c-myc genetics
- Abstract
Double minute chromosomes (dmin) are small, paired chromatin bodies that lack a centromere and represent a form of extrachromosomal gene amplification. Dmin are rare in myeloid neoplasms and are generally associated with a poor prognosis. Most studies of dmin in myeloid neoplasms are case reports or small series. In the current study, we present the clinicopathologic and cytogenetic features of 22 patients with myeloid neoplasms harboring dmin. These neoplasms included acute myeloid leukemia (AML) (n = 18), myelodysplastic syndrome (MDS) (n = 3), and chronic myelomonocytic leukemia (CMML) (n = 1). The AML cases consisted of AML with myelodysplasia-related changes (n = 13) and therapy-related AML (n = 5). Dmin were detected in initial pre-therapy samples in 14 patients with AML or CMML; they were acquired during the disease course in 8 patients who had AML or MDS. The presence of dmin was associated with micronuclei (18/18; 100%), complex karyotype (17/22; 77.3%), and amplification of MYC (12/16; 75%) or MLL (4/16; 25%). Immunohistochemical staining for MYC performed on bone marrow core biopsy or clot sections revealed increased MYC protein in all 19 cases tested. Except for one patient, most patients failed to respond to risk-adapted chemotherapies. At last follow up, all patients had died of disease after a median of 5 months following dmin detection. In conclusion, dmin in myeloid neoplasms commonly harbor MYC or MLL gene amplification and manifest as micronuclei within leukemic blasts. Dmin are often associated with myelodysplasia or therapy-related disease, and complex karyotypes., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
42. Cytogenetic and molecular abnormalities in chronic myelomonocytic leukemia.
- Author
-
Patnaik MM and Tefferi A
- Subjects
- Animals, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Chromatin genetics, Chromatin metabolism, Chromosomal Proteins, Non-Histone chemistry, Chromosomal Proteins, Non-Histone metabolism, DNA Damage, DNA Methylation, Epigenesis, Genetic, Gene Expression Regulation, Leukemic, Histones metabolism, Humans, Leukemia, Myelomonocytic, Chronic diagnosis, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic mortality, Mutation, Prognosis, Protein Multimerization, Signal Transduction, Spliceosomes genetics, Spliceosomes metabolism, Transcription Factors metabolism, Cohesins, Chromosome Aberrations, Genetic Association Studies, Genetic Predisposition to Disease, Genetic Variation, Leukemia, Myelomonocytic, Chronic genetics
- Abstract
Chronic myelomonocytic leukemia (CMML) is a clonal stem cell disorder associated with peripheral blood monocytosis and an inherent tendency to transform to acute myeloid leukemia. CMML has overlapping features of myelodysplastic syndromes and myeloproliferative neoplasms. Clonal cytogenetic changes are seen in ~30%, whereas gene mutations are seen in >90% of patients. Common cytogenetic abnormalities include; trisomy 8, -Y, -7/del(7q), trisomy 21 and del(20q), with the Mayo-French risk stratification effectively risk stratifying patients based on cytogenetic abnormalities. Gene mutations frequently involve epigenetic regulators (TET2 ~60%), modulators of chromatin (ASXL1 ~40%), spliceosome components (SRSF2 ~50%), transcription factors (RUNX1 ~15%) and signal pathways (RAS ~30%, CBL ~15%). Of these, thus far, only nonsense and frameshift ASXL1 mutations have been shown to negatively impact overall survival. This has resulted in the development of contemporary, molecularly integrated (inclusive of ASXL1 mutations) CMML prognostic models, including Molecular Mayo Model and the Groupe Français des Myélodysplasies model. Better understanding of the prevalent genetic and epigenetic dysregulation has resulted in emerging targeted treatment options for some patients. The development of an integrated (cytogenetic and molecular) prognostic model along with CMML-specific response assessment criteria are much needed future goals.
- Published
- 2016
- Full Text
- View/download PDF
43. Specific molecular signatures predict decitabine response in chronic myelomonocytic leukemia.
- Author
-
Meldi K, Qin T, Buchi F, Droin N, Sotzen J, Micol JB, Selimoglu-Buet D, Masala E, Allione B, Gioia D, Poloni A, Lunghi M, Solary E, Abdel-Wahab O, Santini V, and Figueroa ME
- Subjects
- Aged, Aged, 80 and over, Antimetabolites, Antineoplastic pharmacology, Azacitidine pharmacology, Azacitidine therapeutic use, Bone Marrow pathology, DNA Methylation drug effects, DNA Mutational Analysis, DNA, Intergenic genetics, Decitabine, Enhancer Elements, Genetic genetics, Female, Humans, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Male, Middle Aged, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, Platelet Factor 4 biosynthesis, Platelet Factor 4 genetics, Platelet Factor 4 physiology, Treatment Outcome, beta-Thromboglobulin biosynthesis, beta-Thromboglobulin genetics, beta-Thromboglobulin physiology, Antimetabolites, Antineoplastic therapeutic use, Azacitidine analogs & derivatives, Drug Resistance, Neoplasm genetics, Gene Expression Profiling, Gene Expression Regulation, Neoplastic genetics, Genes, Neoplasm, Leukemia, Myelomonocytic, Chronic drug therapy
- Abstract
Myelodysplastic syndromes and chronic myelomonocytic leukemia (CMML) are characterized by mutations in genes encoding epigenetic modifiers and aberrant DNA methylation. DNA methyltransferase inhibitors (DMTis) are used to treat these disorders, but response is highly variable, with few means to predict which patients will benefit. Here, we examined baseline differences in mutations, DNA methylation, and gene expression in 40 CMML patients who were responsive or resistant to decitabine (DAC) in order to develop a molecular means of predicting response at diagnosis. While somatic mutations did not differentiate responders from nonresponders, we identified 167 differentially methylated regions (DMRs) of DNA at baseline that distinguished responders from nonresponders using next-generation sequencing. These DMRs were primarily localized to nonpromoter regions and overlapped with distal regulatory enhancers. Using the methylation profiles, we developed an epigenetic classifier that accurately predicted DAC response at the time of diagnosis. Transcriptional analysis revealed differences in gene expression at diagnosis between responders and nonresponders. In responders, the upregulated genes included those that are associated with the cell cycle, potentially contributing to effective DAC incorporation. Treatment with CXCL4 and CXCL7, which were overexpressed in nonresponders, blocked DAC effects in isolated normal CD34+ and primary CMML cells, suggesting that their upregulation contributes to primary DAC resistance.
- Published
- 2015
- Full Text
- View/download PDF
44. Hypomethylation and up-regulation of PD-1 in T cells by azacytidine in MDS/AML patients: A rationale for combined targeting of PD-1 and DNA methylation.
- Author
-
Ørskov AD, Treppendahl MB, Skovbo A, Holm MS, Friis LS, Hokland M, and Grønbæk K
- Subjects
- Aged, Aged, 80 and over, Blood Cells drug effects, Blood Cells metabolism, Drug Resistance, Female, Gene Expression Regulation drug effects, Humans, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Lymphocyte Activation, Male, Middle Aged, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes metabolism, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, Neoplasm Proteins physiology, Programmed Cell Death 1 Receptor biosynthesis, Programmed Cell Death 1 Receptor physiology, Promoter Regions, Genetic drug effects, RNA, Messenger biosynthesis, RNA, Messenger genetics, RNA, Neoplasm biosynthesis, RNA, Neoplasm genetics, T-Lymphocyte Subsets drug effects, Antimetabolites pharmacology, Azacitidine pharmacology, DNA Methylation drug effects, Myelodysplastic Syndromes drug therapy, Programmed Cell Death 1 Receptor genetics, Promoter Regions, Genetic genetics, T-Lymphocyte Subsets metabolism
- Abstract
The hypomethylating agents (HMAs) are standard therapy for patients with higher-risk myelodysplastic syndrome (MDS); however, the majority of the patients will lose their response to HMAs over time due to unknown mechanisms. It has recently been shown that T cell expression of the immunoinhibitory receptor PD-1 is regulated by DNA methylation. In 12 of 27 patients (44%) PD-1 promoter demethylation was observed in sorted peripheral blood T cells isolated over consecutive cycles of treatment with 5-azacytidine (5-aza). The PD-1 promoter demethylation correlated with an increase in PD-1 expression. Moreover, demethylation of the PD-1 promoter correlated with a significantly worse overall response rate (8% vs. 60%, p = 0.014), and a trend towards a shorter overall survival (p = 0.11) was observed. A significantly higher baseline methylation level of the PD-1 promoter was observed in T cells of non-responding patients compared to healthy controls (p = 0.023). Accordingly, in addition to their beneficial function, HMAs induce PD-1 expression on T cells in the MDS microenvironment, thereby likely hampering the immune response against the MDS blasts. Thus, we suggest that activation of the PD-1 checkpoint during HMA treatment can be a possible resistance mechanism, which may be overcome by combination therapy with a PD-1 pathway inhibitor.
- Published
- 2015
- Full Text
- View/download PDF
45. Molecular pathogenesis of atypical CML, CMML and MDS/MPN-unclassifiable.
- Author
-
Zoi K and Cross NC
- Subjects
- Animals, Cytogenetic Analysis, DNA Damage, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Humans, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative metabolism, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative pathology, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Mutation, Myelodysplastic-Myeloproliferative Diseases classification, Myelodysplastic-Myeloproliferative Diseases metabolism, Myelodysplastic-Myeloproliferative Diseases pathology, RNA Splicing, Signal Transduction, Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative genetics, Leukemia, Myelomonocytic, Chronic genetics, Myelodysplastic-Myeloproliferative Diseases genetics
- Abstract
According to the 2008 WHO classification, the category of myelodysplastic/myeloproliferative neoplasms (MDS/MPN) includes atypical chronic myeloid leukaemia (aCML), chronic myelomonocytic leukaemia (CMML), MDS/MPN-unclassifiable (MDS/MPN-U), juvenile myelomonocytic leukaemia (JMML) and a "provisional" entity, refractory anaemia with ring sideroblasts and thrombocytosis (RARS-T). The remarkable progress in our understanding of the somatic pathogenesis of MDS/MPN has made it clear that there is considerable overlap among these diseases at the molecular level, as well as layers of unexpected complexity. Deregulation of signalling plays an important role in many cases, and is clearly linked to more highly proliferative disease. Other mutations affect a range of other essential, interrelated cellular mechanisms, including epigenetic regulation, RNA splicing, transcription, and DNA damage response. The various combinations of mutations indicate a multi-step pathogenesis, which likely contributes to the marked clinical heterogeneity of these disorders. The delineation of complex clonal architectures may serve as the cornerstone for the identification of novel therapeutic targets and lead to better patient outcomes. This review summarizes some of the current knowledge of molecular pathogenetic lesions in the MDS/MPN subtypes that are seen in adults: atypical CML, CMML and MDS/MPN-U.
- Published
- 2015
- Full Text
- View/download PDF
46. Multitarget effects of quercetin in leukemia.
- Author
-
Maso V, Calgarotto AK, Franchi GC Jr, Nowill AE, Filho PL, Vassallo J, and Saad ST
- Subjects
- Animals, Biomarkers, Tumor metabolism, Blotting, Western, Cell Proliferation, Female, Humans, Immunoenzyme Techniques, Leukemia, Myelomonocytic, Chronic metabolism, Membrane Potential, Mitochondrial drug effects, Mice, Mice, Inbred NOD, Reactive Oxygen Species metabolism, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antioxidants pharmacology, Apoptosis drug effects, Autophagy drug effects, Cell Cycle Checkpoints drug effects, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic pathology, Quercetin pharmacology
- Abstract
This study proposes to investigate quercetin antitumor efficacy in vitro and in vivo, using the P39 cell line as a model. The experimental design comprised leukemic cells or xenografts of P39 cells, treated in vitro or in vivo, respectively, with quercetin; apoptosis, cell-cycle and autophagy activation were then evaluated. Quercetin caused pronounced apoptosis in P39 leukemia cells, followed by Bcl-2, Bcl-xL, Mcl-1 downregulation, Bax upregulation, and mitochondrial translocation, triggering cytochrome c release and caspases activation. Quercetin also induced the expression of FasL protein. Furthermore, our results demonstrated an antioxidant activity of quercetin. Quercetin treatment resulted in an increased cell arrest in G1 phase of the cell cycle, with pronounced decrease in CDK2, CDK6, cyclin D, cyclin E, and cyclin A proteins, decreased Rb phosphorylation and increased p21 and p27 expression. Quercetin induced autophagosome formation in the P39 cell line. Autophagy inhibition induced by quercetin with chloroquine triggered apoptosis but did not alter quercetin modulation in the G1 phase. P39 cell treatment with a combination of quercetin and selective inhibitors of ERK1/2 and/or JNK (PD184352 or SP600125, respectively), significantly decreased cells in G1 phase, this treatment, however, did not change the apoptotic cell number. Furthermore, in vivo administration of quercetin significantly reduced tumor volume in P39 xenografts and confirmed in vitro results regarding apoptosis, autophagy, and cell-cycle arrest. The antitumor activity of quercetin both in vitro and in vivo revealed in this study, point to quercetin as an attractive antitumor agent for hematologic malignancies., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
47. inv(2)(p23q13)/RAN-binding protein 2 (RANBP2)-ALK fusion gene in myeloid leukemia that developed in an elderly woman.
- Author
-
Maesako Y, Izumi K, Okamori S, Takeoka K, Kishimori C, Okumura A, Honjo G, Akasaka T, and Ohno H
- Subjects
- Aged, Anaplastic Lymphoma Kinase, Antineoplastic Agents administration & dosage, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Azacitidine administration & dosage, Azacitidine therapeutic use, Cytarabine administration & dosage, Cytarabine therapeutic use, Daunorubicin administration & dosage, Daunorubicin therapeutic use, Diagnosis, Differential, Female, Humans, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myelomonocytic, Chronic diagnosis, Leukemia, Myelomonocytic, Chronic drug therapy, Leukemia, Myelomonocytic, Chronic metabolism, Molecular Chaperones metabolism, Nuclear Pore Complex Proteins metabolism, Oncogene Proteins, Fusion metabolism, Receptor Protein-Tyrosine Kinases metabolism, Treatment Outcome, Chromosome Inversion, Chromosomes, Human, Pair 2, Leukemia, Myelomonocytic, Chronic genetics, Molecular Chaperones genetics, Nuclear Pore Complex Proteins genetics, Oncogene Proteins, Fusion genetics, Receptor Protein-Tyrosine Kinases genetics
- Abstract
A 75-year-old woman presented with marked leukocytosis; the white cell count was 143.6 × 10³/μL with 38.6 % monocytes and 13.6 % immature granulocytes, including blasts. Bone marrow (BM) aspirate smears showed >90 % cellularity with hyperplasia of myeloid-lineage cells, 14.6 % monocytes, and 32.1 % blasts. The granulocyte series showed a range of dysplastic morphologies. The rate of peroxidase positivity was 51.5 %. CD36+ cells with monocytic differentiation comprised 64.6 % mononuclear cells. Metaphase spreads obtained from the BM revealed an aneuploid karyotype with -7 and a submetacentric marker chromosome derived from chromosome 2, which was determined to be inv(2)(p23q13) by fluorescence in situ hybridization using the Vysis ALK probe. RAN-binding protein 2 (RANBP2)-ALK fusion mRNA was confirmed by reverse transcriptase-mediated polymerase chain reaction and nucleotide sequencing. High-sensitivity anti-ALK immunohistochemistry of a BM biopsy specimen demonstrated nuclear membrane staining of leukemia cells. As the leukemia showed features of chronic myelomonocytic leukemia, the patient was treated with standard daunorubicin-cytarabine followed by azacitidine, leading to the durable suppression of leukemia progression. These findings suggest that inv(2)(p23q13)/RABBP2-ALK defines a small subset of myeloid leukemia characterized by differentiation to monocytes and sharing features of myelodysplastic syndrome/myeloproliferative neoplasm.
- Published
- 2014
- Full Text
- View/download PDF
48. Chronic myelomonocytic leukemia monocytes uniformly display a population of monocytes with CD11c underexpression.
- Author
-
Sojitra P, Gandhi P, Fitting P, Kini AR, Alkan S, Velankar MM, and Venkataraman G
- Subjects
- Antigens, CD blood, Biomarkers metabolism, Bone Marrow Cells metabolism, Bone Marrow Cells pathology, Female, HLA-DR Antigens blood, Humans, Leukemia, Myelogenous, Chronic, BCR-ABL Positive complications, Leukemia, Myelogenous, Chronic, BCR-ABL Positive metabolism, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Leukemia, Myelomonocytic, Chronic metabolism, Leukocytosis complications, Leukocytosis metabolism, Leukocytosis pathology, Male, Monocytes metabolism, Neoplasm Staging, CD11c Antigen blood, Leukemia, Myelomonocytic, Chronic pathology, Monocytes pathology
- Abstract
Objectives: To examine the utility of CD11c expression on monocytes in normal controls and patients with chronic myelomonocytic leukemia (CMML) (n = 23) with flow cytometric immunophenotyping., Methods: Twenty-three CMML samples and 10 control bone marrows submitted for lymphoma staging without evidence of disease were examined., Results: Monocytes in CMML samples ranged from 4% to 35%. Expression of at least one aberrant monocytic marker was found on the monocytes in 18 (82%) of 22 evaluable cases. The most common aberrancy was underexpression of CD11c (n = 15), while none of the bone marrow controls showed underexpression of CD11c., Conclusions: A distinct heterogeneous population of monocytic cells with underexpression of CD11c was identified in all these cases. CD11c underexpression was independent of other aberrancies, including HLA-DR underexpression (n = 14), aberrant CD56 expression (n = 11), and underexpression of CD33, CD38, and CD14 (n = 6, 5, and 5, respectively), supporting the utility of CD11c expression status on monocytes in establishing a CMML diagnosis.
- Published
- 2013
- Full Text
- View/download PDF
49. A role for miR-142-3p in colony-stimulating factor 1-induced monocyte differentiation into macrophages.
- Author
-
Lagrange B, Martin RZ, Droin N, Aucagne R, Paggetti J, Largeot A, Itzykson R, Solary E, Delva L, and Bastie JN
- Subjects
- Antigens, CD genetics, Antigens, CD metabolism, Antigens, Differentiation, Myelomonocytic genetics, Antigens, Differentiation, Myelomonocytic metabolism, Cell Differentiation drug effects, Cell Differentiation genetics, Cell Line, Tumor, Down-Regulation drug effects, Early Growth Response Protein 1 genetics, Early Growth Response Protein 1 metabolism, Early Growth Response Protein 2 genetics, Early Growth Response Protein 2 metabolism, GPI-Linked Proteins genetics, GPI-Linked Proteins metabolism, Humans, K562 Cells, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Macrophages cytology, Macrophages metabolism, MicroRNAs biosynthesis, MicroRNAs metabolism, Monocytes cytology, Monocytes metabolism, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism, Receptors, IgG genetics, Receptors, IgG metabolism, Repressor Proteins genetics, Repressor Proteins metabolism, Up-Regulation drug effects, Macrophage Colony-Stimulating Factor pharmacology, Macrophages drug effects, Macrophages physiology, MicroRNAs genetics, Monocytes drug effects, Monocytes physiology
- Abstract
The differentiation of human peripheral blood monocytes into macrophages can be reproduced ex vivo by culturing the cells in the presence of colony-stimulating factor 1 (CSF1). Using microarray profiling to explore the role of microRNAs (miRNAs), we identified a dramatic decrease in the expression of the hematopoietic specific miR-142-3p. Up- and down-regulation of this miRNA in primary human monocytes altered CSF1-induced differentiation of monocytes, as demonstrated by changes in the expression of the cell surface markers CD16 and CD163. One of the genes whose expression is repressed by miR-142-3p encodes the transcription factor Early Growth Response 2 (Egr2). In turn, Egr2 associated with its co-repressor NGFI-A (Nerve Growth Factor-Induced gene-A) binding protein 2 (NAB2) binds to the pre-miR-142-3p promoter to negatively regulate its expression. Interestingly, the expression of miR-142-3p is abnormally low in monocytes from patients with the most proliferative forms of chronic myelomonocytic leukemia (CMML), and miR-142-3p re-expression in CMML dysplastic monocytes can improve their differentiation potential. Altogether, miR-142-3p which functions in a molecular circuitry with Egr2 is an actor of CSF1-induced differentiation of human monocytes whose expression could be altered in CMML., (Copyright © 2013 Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
50. Atypical generalized eruptive histiocytosis clonally related to chronic myelomonocytic leukemia with loss of Y chromosome.
- Author
-
Shon W, Peters MS, Reed KB, Ketterling RP, Dogan A, and Gibson LE
- Subjects
- Aged, 80 and over, Antigens, CD biosynthesis, Antigens, CD genetics, Gene Expression Regulation, Leukemic genetics, Histiocytosis, Langerhans-Cell genetics, Histiocytosis, Langerhans-Cell metabolism, Histiocytosis, Langerhans-Cell pathology, Humans, Male, Neoplasm Proteins biosynthesis, Neoplasm Proteins genetics, S100 Proteins biosynthesis, S100 Proteins genetics, Blast Crisis genetics, Blast Crisis metabolism, Blast Crisis pathology, Chromosome Deletion, Chromosomes, Human, Y, Head and Neck Neoplasms genetics, Head and Neck Neoplasms metabolism, Head and Neck Neoplasms pathology, Leukemia, Myelomonocytic, Chronic genetics, Leukemia, Myelomonocytic, Chronic metabolism, Leukemia, Myelomonocytic, Chronic pathology, Skin Neoplasms genetics, Skin Neoplasms metabolism, Skin Neoplasms pathology
- Abstract
Generalized eruptive histiocytosis, described in 1963 by Winklemann and Muller, is a reactive, self-healing form of non-Langerhans histiocytosis. Rare cases of atypical generalized eruptive histiocytosis have been reported in patients with hematopoietic malignancy, but the biological relationship between the two disorders is not known. We report an 84-year-old man with chronic myelomonocytic leukemia who presented with coalescing erythematous papules and plaques on the posterior neck, ear and lower lip, followed by development of blast crisis. Skin biopsy revealed a thick band-like dermal infiltrate of cells that exhibited morphologic features of macrophages or histiocytes and prominent elastolytic phagocytosis. These cells demonstrated a mature immunophenotype, expressing CD14 and CD68, with partial expression of CD13 but not CD1a, CD43, CD56, CD123, Langerin, or S-100 protein. Karyotype and fluorescence in situ hybridization analyses showed loss of the Y chromosome in bone marrow and skin specimens, providing evidence of a clonal relationship between the cutaneous eruption and the underlying chronic myelomonocytic leukemia. The presence of the same clone in skin and bone marrow specimens from our patient supports the possibility that atypical generalized eruptive histiocytosis is a marker for underlying hematopoietic malignancy. Discovery of additional cases may shed further light on the pathogenesis of this rare entity., (© 2013 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.)
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.