51
|
Shahrabi S, Khosravi A, Shahjahani M, Rahim F, Saki N. Genetics and Epigenetics of Myelodysplastic Syndromes and Response to Drug Therapy: New Insights. Oncol Rev 2016; 10:311. [PMID: 28058097 PMCID: PMC5178845 DOI: 10.4081/oncol.2016.311] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 12/06/2016] [Indexed: 12/12/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of hematologic neoplasms ocurring mostly in the elderly. The clinical outcome of MDS patients is still poor despite progress in treatment approaches. About 90% of patients harbor at least one somatic mutation. This review aimed to assess the potential of molecular abnormalities in understanding pathogenesis, prognosis, diagnosis and in guiding choice of proper therapy in MDS patients. Papers related to this topic from 2000 to 2016 in PubMed and Scopus databases were searched and studied. The most common molecular abnormalities were TET2, ASXL1 as well as molecules involved in spliceosome machinery (U2AF1, SRSF2 and SF3B1). Patients with defects in TET2 molecule show better response to treatment with azacitidine. IDH and DNMT3A mutations are associated with a good response to decitabine therapy. In addition, patients with del5q subtype harboring TP53 mutation do not show a good response to lenalidomide therapy. In general, the results of this study show that molecular abnormalities can be associated with the occurrence of a specific morphological phenotype in patients. Therefore, considering the morphology of patients, different gene profiling methods can be selected to choice the most appropriate therapeutic measure in these patients in addition to faster and more cost-effective diagnosis of molecular abnormalities.
Collapse
Affiliation(s)
- Saeid Shahrabi
- Department of Biochemistry and Hematology, Semnan University of Medical Sciences, Semnan
| | - Abbas Khosravi
- Health Research Institute, Thalassemia and Hemoglobinopathy Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz
| | - Mohammad Shahjahani
- Colestan Hospital Clinical Research Development Unit. Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fakher Rahim
- Health Research Institute, Thalassemia and Hemoglobinopathy Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz
| | - Najmaldin Saki
- Health Research Institute, Thalassemia and Hemoglobinopathy Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz
- Colestan Hospital Clinical Research Development Unit. Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
52
|
Lim CY, Zoncu R. The lysosome as a command-and-control center for cellular metabolism. J Cell Biol 2016; 214:653-64. [PMID: 27621362 PMCID: PMC5021098 DOI: 10.1083/jcb.201607005] [Citation(s) in RCA: 220] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 08/22/2016] [Indexed: 12/25/2022] Open
Abstract
Lysosomes are membrane-bound organelles found in every eukaryotic cell. They are widely known as terminal catabolic stations that rid cells of waste products and scavenge metabolic building blocks that sustain essential biosynthetic reactions during starvation. In recent years, this classical view has been dramatically expanded by the discovery of new roles of the lysosome in nutrient sensing, transcriptional regulation, and metabolic homeostasis. These discoveries have elevated the lysosome to a decision-making center involved in the control of cellular growth and survival. Here we review these recently discovered properties of the lysosome, with a focus on how lysosomal signaling pathways respond to external and internal cues and how they ultimately enable metabolic homeostasis and cellular adaptation.
Collapse
Affiliation(s)
- Chun-Yan Lim
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720 The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720
| | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720 The Paul F. Glenn Center for Aging Research at the University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
53
|
Suzuki Y, Tomita A, Nakamura F, Iriyama C, Shirahata-Adachi M, Shimada K, Akashi A, Ishikawa Y, Kaneda N, Kiyoi H. Peripheral blood cell-free DNA is an alternative tumor DNA source reflecting disease status in myelodysplastic syndromes. Cancer Sci 2016; 107:1329-37. [PMID: 27323954 PMCID: PMC5021037 DOI: 10.1111/cas.12994] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/13/2016] [Accepted: 06/17/2016] [Indexed: 12/17/2022] Open
Abstract
Genetic alterations in myelodysplastic syndromes (MDS) are critical for pathogenesis. We previously showed that peripheral blood cell-free DNA (PBcfDNA) may be more sensitive for genetic/epigenetic analyses than whole bone marrow (BM) cells and mononuclear cells in peripheral blood (PB). Here we analyzed the detailed features of PBcfDNA and its utility in genetic analyses in MDS. The plasma-PBcfDNA concentration in MDS and related diseases (N = 33) was significantly higher than that in healthy donors (N = 14; P = 0.041) and in International Prognostic Scoring System higher-risk groups than that in lower-risk groups (P = 0.034). The concentration of plasma-/serum-PBcfDNA was significantly correlated with the serum lactate dehydrogenase level (both P < 0.0001) and the blast cell count in PB (P = 0.034 and 0.025, respectively). One nanogram of PBcfDNA was sufficient for one assay of Sanger sequencing using optimized primer sets to amplify approximately 160-bp PCR products. PBcfDNA (approximately 50 ng) can also be utilized for targeted sequencing. Almost all mutations detected in BM-DNA were also detected using corresponding PBcfDNA. Analyses using serially harvested PBcfDNA from an RAEB-2 patient showed that the somatic mutations and a single nucleotide polymorphism that were detected before allogeneic transplantation were undetectable after transplantation, indicating that PBcfDNA likely comes from MDS clones that reflect the disease status. PBcfDNA may be a safer and easier alternative to obtain tumor DNA in MDS.
Collapse
Affiliation(s)
- Yasuhiro Suzuki
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Akihiro Tomita
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Fumika Nakamura
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Analytical Neurobiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Chisako Iriyama
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mizuho Shirahata-Adachi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuyuki Shimada
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Institute for Adavanced Research, Nagoya University, Nagoya, Japan
| | - Akimi Akashi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichi Ishikawa
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norio Kaneda
- Department of Analytical Neurobiology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Hitoshi Kiyoi
- Department of Hematology and Oncology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
54
|
Raza A, Ali AM, Reddy MVR, Hoffman BS, Petrone ME, Maniar M, Pinheiro RF, Coutinho DF, Fruchtman SM. Rigosertib in myelodysplastic syndromes (MDS). Expert Opin Orphan Drugs 2016. [DOI: 10.1080/21678707.2016.1213628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- A. Raza
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - A. M. Ali
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - M. V. R. Reddy
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - M. Maniar
- Onconova Therapeutics, Inc., Newtown, PA, USA
| | - R. F. Pinheiro
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - D. F. Coutinho
- Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | | |
Collapse
|
55
|
Patnaik MM, Tefferi A. Chronic myelomonocytic leukemia: 2016 update on diagnosis, risk stratification, and management. Am J Hematol 2016; 91:631-42. [PMID: 27185207 DOI: 10.1002/ajh.24396] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 04/19/2016] [Indexed: 12/15/2022]
Abstract
Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic stem cell disorder characterized by overlapping features of myelodysplastic syndromes and myeloproliferative neoplasms. Diagnosis is based on the presence of persistent (>3 months) peripheral blood monocytosis (>1 × 10(9) /L), along with bone marrow dysplasia. Clonal cytogenetic abnormalities occur in ∼20-30% of patients, while >90% have gene mutations. Mutations involving TET2 (∼60%), SRSF2 (∼50%), ASXL1 (∼40%), and RAS (∼30%) are frequent; with only ASXL1 mutations negatively impacting overall survival. Two molecularly integrated, CMML-specific prognostic models include; the Groupe Français des Myélodysplasies (GFM) and the Molecular Mayo Model (MMM). The GFM model segregates patients into 3 groups based on: age >65 years, WBC >15 × 10(9) /L, anemia, platelets <100 × 10(9) /L, and ASXL1 mutation status, with respective median survivals of 56 (low), 27.4 (intermediate), and 9.2 (high) months. The MMM is based on ASXL1 mutational status, absolute monocyte count >10 × 10(9) /L, hemoglobin <10 g/dL, platelets <100 × 109/L and circulating immature myeloid cells. This model stratifies patients into four groups; high (≥3 risk factors), intermediate-2 (2 risk factors), intermediate-1 (1 risk factor) and low (no risk factors), with median survivals of 16, 31, 59, and 97 months, respectively. Hypomethylating agents such as 5-azacitidine and decitabine are commonly used, with overall response rates of ∼30-40% and complete remission rates of ∼7-17%. Allogeneic stem cell transplant is the only potentially curative option, but is associated with significant morbidity and mortality. Individualized therapy, including epigenetic modifiers and small molecule inhibitors, are exciting prospects. Am. J. Hematol. 91:632-642, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mrinal M. Patnaik
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| | - Ayalew Tefferi
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
56
|
Aberrations identified by genomic arrays in normal karyotype CMML can be detected in 40% of patients, but do not add prognostic information to molecular mutations. Leukemia 2016; 30:2235-2238. [PMID: 27220668 DOI: 10.1038/leu.2016.158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
57
|
Awad MM. Impaired c-Met Receptor Degradation Mediated by MET Exon 14 Mutations in Non–Small-Cell Lung Cancer. J Clin Oncol 2016; 34:879-81. [DOI: 10.1200/jco.2015.64.2777] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Mark M. Awad
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
58
|
Jing Z, Li L, Wang X, Wang M, Cai Y, Jin ZI, Zhang YE. High c-Cbl expression in gliomas is associated with tumor progression and poor prognosis. Oncol Lett 2016; 11:2787-2791. [PMID: 27073553 PMCID: PMC4812512 DOI: 10.3892/ol.2016.4318] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 01/26/2016] [Indexed: 01/17/2023] Open
Abstract
Casitas B-lineage lymphoma (c-Cbl) expression has been linked to the development of several types of cancer. However, no studies on the association of c-Cbl and glioma have been published thus far. The present study examined glioma samples obtained from 136 patients treated at The First Hospital of China Medical University (Shenyang, China) from January 2007 to December 2009, and the expression levels of c-Cbl in the samples were evaluated by reverse transcription-quantitative polymerase chain reaction, immunohistochemistry and western blotting. Kaplan-Meier survival curves were generated and subjected to Cox regression analysis. The messenger RNA and protein levels of c-Cbl were observed to be upregulated in high-grade glioma, compared with low-grade glioma. A multivariate analysis revealed that the protein levels of c-Cbl were independently associated with overall survival [hazard ratio (HR)=4.923, 95% confidence interval (CI)=3.163–7.662; P<0.001]. Furthermore, the grade of the glioma (according to the World Health Organization criteria) was observed to be independent prognostic factors for progression-free survival and overall survival time (HR=8.842, 95% CI=7.827–9.989; P<0.001, and HR=10.247, 95% CI=9.009–11.655; P<0.001, respectively). Kaplan-Meier analysis and log-rank test indicated that high protein expression levels of c-Cbl were significantly associated with overall and progression-free survival (P<0.001). To the best of our knowledge, these results provide the first evidence that the overexpression of c-Cbl is correlated with advanced clinicopathological features and poor prognosis in patients with glioma.
Collapse
Affiliation(s)
- Zhitao Jing
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Long Li
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xin Wang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Minghao Wang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Ying Cai
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Z I Jin
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Y E Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
59
|
Katzav S, Schmitz ML. Mutations of c-Cbl in myeloid malignancies. Oncotarget 2016; 6:10689-96. [PMID: 26028666 PMCID: PMC4484412 DOI: 10.18632/oncotarget.3986] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/15/2015] [Indexed: 12/18/2022] Open
Abstract
Next generation sequencing has shown the frequent occurrence of point mutations in the ubiquitin E3 ligase c-Cbl in myeloid malignancies. Mouse models revealed a causal contribution of c-Cbl for the onset of such neoplasms. The point mutations typically cluster in the linker region and RING finger domain and affect both alleles by acquired uniparental disomy. The fast progress in the detection of c-Cbl mutations is contrasted by our scarce knowledge on their functional consequences. The c-Cbl protein displays several enzymatic functions by promoting the attachment of differentially composed ubiquitin chains and of the ubiquitin-like protein NEDD8 to its target proteins. In addition, c-Cbl functions as an adapter protein and undergoes phosphorylation-dependent inducible conformation changes. Studies on the impact of c-Cbl mutations on its functions as a dynamic and versatile adapter protein, its interactomes and on its various enzymatic activities are now important to allow the identification of druggable targets within the c-Cbl signaling network.
Collapse
Affiliation(s)
- Shulamit Katzav
- Developmental Biology and Cancer Research, IMRIC, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - M Lienhard Schmitz
- Institute of Biochemistry, University of Giessen, Friedrichstrasse, Giessen, Germany
| |
Collapse
|
60
|
Choi J, Polcher A, Joas A. Systematic literature review on Parkinson's disease and Childhood Leukaemia and mode of actions for pesticides. ACTA ACUST UNITED AC 2016. [DOI: 10.2903/sp.efsa.2016.en-955] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
61
|
Saeidi K. Myeloproliferative neoplasms: Current molecular biology and genetics. Crit Rev Oncol Hematol 2015; 98:375-89. [PMID: 26697989 DOI: 10.1016/j.critrevonc.2015.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Revised: 09/10/2015] [Accepted: 11/09/2015] [Indexed: 12/16/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are clonal disorders characterized by increased production of mature blood cells. Philadelphia chromosome-negative MPNs (Ph-MPNs) consist of polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). A number of stem cell derived mutations have been identified in the past 10 years. These findings showed that JAK2V617F, as a diagnostic marker involving JAK2 exon 14 with a high frequency, is the best molecular characterization of Ph-MPNs. Somatic mutations in an endoplasmic reticulum chaperone, named calreticulin (CALR), is the second most common mutation in patients with ET and PMF after JAK2 V617F mutation. Discovery of CALR mutations led to the increased molecular diagnostic of ET and PMF up to 90%. It has been shown that JAK2V617F is not the unique event in disease pathogenesis. Some other genes' location such as TET oncogene family member 2 (TET2), additional sex combs-like 1 (ASXL1), casitas B-lineage lymphoma proto-oncogene (CBL), isocitrate dehydrogenase 1/2 (IDH1/IDH2), IKAROS family zinc finger 1 (IKZF1), DNA methyltransferase 3A (DNMT3A), suppressor of cytokine signaling (SOCS), enhancer of zeste homolog 2 (EZH2), tumor protein p53 (TP53), runt-related transcription factor 1 (RUNX1) and high mobility group AT-hook 2 (HMGA2) have also identified to be involved in MPNs phenotypes. Here, current molecular biology and genetic mechanisms involved in MNPs with a focus on the aforementioned factors is presented.
Collapse
Affiliation(s)
- Kolsoum Saeidi
- Department of Medical Genetics, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
62
|
Goyama S, Schibler J, Gasilina A, Shrestha M, Lin S, Link KA, Chen J, Whitman SP, Bloomfield CD, Nicolet D, Assi SA, Ptasinska A, Heidenreich O, Bonifer C, Kitamura T, Nassar NN, Mulloy JC. UBASH3B/Sts-1-CBL axis regulates myeloid proliferation in human preleukemia induced by AML1-ETO. Leukemia 2015; 30:728-39. [PMID: 26449661 DOI: 10.1038/leu.2015.275] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/22/2015] [Accepted: 09/30/2015] [Indexed: 12/24/2022]
Abstract
The t(8;21) rearrangement, which creates the AML1-ETO fusion protein, represents the most common chromosomal translocation in acute myeloid leukemia (AML). Clinical data suggest that CBL mutations are a frequent event in t(8;21) AML, but the role of CBL in AML1-ETO-induced leukemia has not been investigated. In this study, we demonstrate that CBL mutations collaborate with AML1-ETO to expand human CD34+ cells both in vitro and in a xenograft model. CBL depletion by shRNA also promotes the growth of AML1-ETO cells, demonstrating the inhibitory function of endogenous CBL in t(8;21) AML. Mechanistically, loss of CBL function confers hyper-responsiveness to thrombopoietin and enhances STAT5/AKT/ERK/Src signaling in AML1-ETO cells. Interestingly, we found the protein tyrosine phosphatase UBASH3B/Sts-1, which is known to inhibit CBL function, is upregulated by AML1-ETO through transcriptional and miR-9-mediated regulation. UBASH3B/Sts-1 depletion induces an aberrant pattern of CBL phosphorylation and impairs proliferation in AML1-ETO cells. The growth inhibition caused by UBASH3B/Sts-1 depletion can be rescued by ectopic expression of CBL mutants, suggesting that UBASH3B/Sts-1 supports the growth of AML1-ETO cells partly through modulation of CBL function. Our study reveals a role of CBL in restricting myeloid proliferation of human AML1-ETO-induced leukemia, and identifies UBASH3B/Sts-1 as a potential target for pharmaceutical intervention.
Collapse
Affiliation(s)
- S Goyama
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.,Division of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - J Schibler
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - A Gasilina
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - M Shrestha
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - S Lin
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - K A Link
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - J Chen
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - S P Whitman
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - C D Bloomfield
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - D Nicolet
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.,Alliance for Clinical Trials in Oncology Statistics and Data Center, Mayo Clinic, Rochester, MN, USA
| | - S A Assi
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - A Ptasinska
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - O Heidenreich
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, UK
| | - C Bonifer
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - T Kitamura
- Division of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - N N Nassar
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - J C Mulloy
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
63
|
William BM, An W, Feng D, Nadeau S, Mohapatra BC, Storck MA, Band V, Band H. Fasudil, a clinically safe ROCK inhibitor, decreases disease burden in a Cbl/Cbl-b deficiency-driven murine model of myeloproliferative disorders. ACTA ACUST UNITED AC 2015; 21:218-24. [PMID: 26177294 DOI: 10.1179/1607845415y.0000000031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Mutations in Cbl or Cbl-b gene occur in 10% of myeloproliferative disorder (MPD) patients and are associated with poor prognosis. Hematopoietic Cbl/Cbl-b double knockout (DKO) leads to a disease in mice phenotypically similar to human MPDs. The aim of this study was to evaluate the anti-MPD activity of a clinically safe drug, Fasudil, identified in an in vitro kinase inhibitor as an inhibitor of proliferation of DKO mouse hematopoietic stem/progenitor cells (HSPCs). METHODS Fasudil exhibited relatively selective anti-proliferative activity against Cbl/Cbl-b DKO vs. control murine bone marrow HSPCs. We established a mouse model with uniform time of MPD onset by transplanting Cbl/Cbl-b DKO HSPCs into busulfan-conditioned NOD/SCID/gamma chain-deficient mice. Four weeks post-transplant, mice were treated with 100 mg/kg fasudil (13 mice) or water (control, 8 mice) daily by oral gavage, followed by blood cell count every 2 weeks. RESULTS By 2 weeks of treatment, total white cell and monocyte counts were significantly lower in mice treated with fasudil. We observed a trend towards improved survival in fasudil-treated mice that did not reach statistical significance. Notably, prolonged survival beyond 27 weeks was observed in two fasudil-treated mice, nearly twice the 16-week average life-span in the Cbl/Cbl-b DKO MPD model. CONCLUSIONS Our results suggest a therapeutic potential for fasudil, a clinically safe drug with promising results in vascular diseases, in the treatment of MPDs or other mutant Cbl-driven myeloid disorders.
Collapse
Affiliation(s)
- Basem M William
- a Division of Hematology and Oncology, Department of Medicine , University of Nebraska Medical Center , Omaha , USA
| | - Wei An
- b Department of Genetics, Cell Biology and Anatomy , University of Nebraska Medical Center , Omaha , USA.,c Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center , Omaha , USA
| | - Dan Feng
- c Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center , Omaha , USA
| | - Scott Nadeau
- b Department of Genetics, Cell Biology and Anatomy , University of Nebraska Medical Center , Omaha , USA.,c Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center , Omaha , USA
| | - Bhopal C Mohapatra
- c Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center , Omaha , USA.,d Department of Biochemistry and Molecular Biology , University of Nebraska Medical Center , Omaha , USA
| | - Matthew A Storck
- c Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center , Omaha , USA
| | - Vimla Band
- b Department of Genetics, Cell Biology and Anatomy , University of Nebraska Medical Center , Omaha , USA.,c Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center , Omaha , USA.,e Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center , Omaha , USA
| | - Hamid Band
- b Department of Genetics, Cell Biology and Anatomy , University of Nebraska Medical Center , Omaha , USA.,c Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center , Omaha , USA.,d Department of Biochemistry and Molecular Biology , University of Nebraska Medical Center , Omaha , USA.,e Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center , Omaha , USA
| |
Collapse
|
64
|
Gondek LP, DeZern AE. I walk the line: how to tell MDS from other bone marrow failure conditions. Curr Hematol Malig Rep 2015; 9:389-99. [PMID: 25079655 DOI: 10.1007/s11899-014-0224-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Myelodysplastic syndromes (MDS) are clonal hematopoietic stem cell disorders characterized by peripheral cytopenias and ineffective hematopoiesis. MDS is an example of an age-related malignancy and its increasing prevalence and incidence can be attributed to a greater life expectancy in developed countries. Although frequently encountered in hematology/oncology clinics, MDS may constitute a diagnostic challenge especially with equivocal bone marrow morphology. Certain syndromes of bone marrow failure (BMF) may mimic MDS and formulating a correct diagnosis is vital for adequate prognostication as well as therapeutic approaches. Metaphase karyotyping (MK) is a very important diagnostic tool and marker of prognosis and can be an indicator of response to certain therapies. Unfortunately, chromosomal abnormalities may only be found in approximately 50 % of patients with MDS. In this review, we discuss the diagnostic approaches to patients with pancytopenia with a particular focus on the growing number of somatic mutations through new molecular testing.
Collapse
Affiliation(s)
- Lukasz P Gondek
- Department of Oncology, Division of Hematological Malignancies, Johns Hopkins University, 1650 Orleans St, CRB1-290, Baltimore, MD, 21231, USA,
| | | |
Collapse
|
65
|
Zhang X, Lancet JE, Zhang L. Molecular pathology of myelodysplastic syndromes: new developments and implications for diagnosis and treatment. Leuk Lymphoma 2015; 56:3022-30. [DOI: 10.3109/10428194.2015.1037756] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
66
|
Abstract
In the last decade, genomic studies have identified multiple recurrent somatic mutations in myeloproliferative neoplasms (MPNs). Beginning with the discovery of the JAK2 V617F mutation, multiple additional mutations have been found that constitutively activate cell-signaling pathways, including MPL, CBL, and LNK. Furthermore, several classes of epigenetic modifiers have also been identified, in patients with MPN, revealing a requirement for mutations in other pathways to cooperate with JAK-STAT pathway mutations in MPN pathogenesis. Mutations in the de novo DNA methylation protein, DNMT3A, demethylation machinery, TET2 and related IDH1/2 production of oncometabolite 2-hydroxygluterate, and polycomb complex proteins EZH2 and ASXL1 have opened new pathophysiologic clues into these diseases. The prognostic relevance of these novel disease alleles remains an important area of investigation, and clinical trials are currently underway to determine if these findings represent tractable therapeutic targets, either alone, or in combination with JAK2 inhibition.
Collapse
Affiliation(s)
- Aaron D Viny
- From the Human Oncology and Pathogenesis Program and Leukemia Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, Weill Cornell Medical College, New York, NY
| | | |
Collapse
|
67
|
Dan C, Chi J, Wang L. Molecular mechanisms of the progression of myelodysplastic syndrome to secondary acute myeloid leukaemia and implication for therapy. Ann Med 2015; 47:209-17. [PMID: 25861829 DOI: 10.3109/07853890.2015.1009156] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Myelodysplastic syndrome (MDS) includes a heterogeneous group of clonal haematological stem cell disorders characterized by dysplasia, cytopenias, ineffective haematopoiesis, and an increased risk of progression to acute myeloid leukaemia (AML), which is also called secondary AML (sAML). Approximately one-third of patients with MDS will progress to sAML within a few months to a few years, and this type of transformation is more common and rapid in patients with high-risk MDS (HR-MDS). However, the precise mechanisms underlying the evolution of MDS to sAML remain unclear. Currently, chemotherapy for sAML has minimal efficacy. The only method of curing patients with sAML is allogeneic haematopoietic stem cell transplantation (Allo-HSCT). Unfortunately, only a few patients are appropriate for transplantation because this disease primarily affects older adult patients. Additionally, compared to de novo AML, sAML is more difficult to cure, and the prognosis is often worse. Therefore, it is important to clarify the molecular mechanisms of the progression of MDS to sAML and to explore the potent drugs for clinical use. This review will highlight several molecular mechanisms of the progression of MDS to sAML and new therapeutic strategies of this disease.
Collapse
Affiliation(s)
- Chunli Dan
- Department of Haematology, The First Affiliated Hospital of Chongqing Medical University , Chongqing , China
| | | | | |
Collapse
|
68
|
Liyasova MS, Ma K, Lipkowitz S. Molecular pathways: cbl proteins in tumorigenesis and antitumor immunity-opportunities for cancer treatment. Clin Cancer Res 2015; 21:1789-94. [PMID: 25477533 PMCID: PMC4401614 DOI: 10.1158/1078-0432.ccr-13-2490] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Accepted: 11/05/2014] [Indexed: 11/16/2022]
Abstract
The Cbl proteins are a family of ubiquitin ligases (E3s) that regulate signaling through many tyrosine kinase-dependent pathways. A predominant function is to negatively regulate receptor tyrosine kinase (RTK) signaling by ubiquitination of active RTKs, targeting them for trafficking to the lysosome for degradation. Also, Cbl-mediated ubiquitination can regulate signaling protein function by altered cellular localization of proteins without degradation. In addition to their role as E3s, Cbl proteins play a positive role in signaling by acting as adaptor proteins that can recruit signaling molecules to the active RTKs. Cbl-b, a second family member, negatively regulates the costimulatory pathway of CD8 T cells and also negatively regulates natural killer cell function. The different functions of Cbl proteins and their roles both in the development of cancer and the regulation of immune responses provide multiple therapeutic opportunities. Mutations in Cbl that inactivate the negative E3 function while maintaining the positive adaptor function have been described in approximately 5% of myeloid neoplasms. An improved understanding of how the signaling pathways [e.g., Fms-like tyrosine kinase 3 (Flt3), PI3K, and signal transducer and activator of transcription (Stat)] are dysregulated by these mutations in Cbl has helped to identify potential targets for therapy of myeloid neoplasms. Conversely, the loss of Cbl-b leads to increased adaptive and innate antitumor immunity, suggesting that inhibiting Cbl-b may be a means to increase antitumor immunity across a wide variety of tumors. Thus, targeting the pathways regulated by Cbl proteins may provide attractive opportunities for treating cancer.
Collapse
Affiliation(s)
- Mariya S Liyasova
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ke Ma
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stanley Lipkowitz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
69
|
Clinical and biological implications of ancestral and non-ancestral IDH1 and IDH2 mutations in myeloid neoplasms. Leukemia 2015; 29:2134-42. [PMID: 25836588 PMCID: PMC5821256 DOI: 10.1038/leu.2015.91] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 03/23/2015] [Indexed: 01/08/2023]
Abstract
Mutations in isocitrate dehydrogenase 1/2 (IDH1/2MT) are drivers of a variety of myeloid neoplasms. As they yield the same oncometabolite, D-2-hydroxyglutarate, they are often treated as equivalent, and pooled. We studied the validity of this approach and found IDH1/2 mutations in 179 of 2119 myeloid neoplasms (8%). Cross-sectionally, the frequencies of these mutations increased from lower- to higher-risk disease, thus suggesting a role in clinical progression. Variant allelic frequencies indicated that IDH1MT and IDH2MT are ancestral in up to 14/74 (19%) vs. 34/99 (34%; P=0.027) of cases, respectively, illustrating the pathogenic role of these lesions in myeloid neoplasms. IDH1/2MT was associated with poor overall survival, particularly in lower-risk myelodysplastic syndromes. Ancestral IDH1MT cases were associated with a worse prognosis than subclonal IDH1MT cases, whereas the position of IDH2MT within clonal hierarchy did not impact survival. This may relate to distinct mutational spectra with more DNMT3A and NPM1 mutations associated with IDH1MT cases, and more ASXL1, SRSF2, RUNX1, STAG2 mutations associated with IDH2MT cases. Our data demonstrate important clinical and biological differences between IDH1MT and IDH2MT myeloid neoplasms. These mutations should be considered separately as their differences could have implications for diagnosis, prognosis, and treatment with IDH1/2MT inhibitors of IDH1/2MT patients.
Collapse
|
70
|
Lutz-Nicoladoni C, Wolf D, Sopper S. Modulation of Immune Cell Functions by the E3 Ligase Cbl-b. Front Oncol 2015; 5:58. [PMID: 25815272 PMCID: PMC4356231 DOI: 10.3389/fonc.2015.00058] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 02/24/2015] [Indexed: 01/10/2023] Open
Abstract
Maintenance of immunological tolerance is a critical hallmark of the immune system. Several signaling checkpoints necessary to balance activating and inhibitory input to immune cells have been described so far, among which the E3 ligase Cbl-b appears to be a central player. Cbl-b is expressed in all leukocyte subsets and regulates several signaling pathways in T cells, NK cells, B cells, and different types of myeloid cells. In most cases, Cbl-b negatively regulates activation signals through antigen or pattern recognition receptors and co-stimulatory molecules. In line with this function, cblb-deficient immune cells display lower activation thresholds and cblb knockout mice spontaneously develop autoimmunity and are highly susceptible to experimental autoimmunity. Interestingly, genetic association studies link CBLB-polymorphisms with autoimmunity also in humans. Vice versa, the increased activation potential of cblb-deficient cells renders them more potent to fight against malignancies or infections. Accordingly, several reports have shown that cblb knockout mice reject tumors, which mainly depends on cytotoxic T and NK cells. Thus, targeting Cbl-b may be an interesting strategy to enhance anti-cancer immunity. In this review, we summarize the findings on the molecular function of Cbl-b in different cell types and illustrate the potential of Cbl-b as target for immunomodulatory therapies.
Collapse
Affiliation(s)
- Christina Lutz-Nicoladoni
- Department of Hematology and Oncology, Medical University Innsbruck , Innsbruck , Austria ; Tumor Immunology Laboratory, Tyrolean Cancer Research Institute , Innsbruck , Austria
| | - Dominik Wolf
- Medical Clinic III for Oncology, Haematology and Rheumatology, University Clinic Bonn (UKB) , Bonn , Germany
| | - Sieghart Sopper
- Department of Hematology and Oncology, Medical University Innsbruck , Innsbruck , Austria ; Tumor Immunology Laboratory, Tyrolean Cancer Research Institute , Innsbruck , Austria
| |
Collapse
|
71
|
Genetic and molecular characterization of myelodysplastic syndromes and related myeloid neoplasms. Int J Hematol 2015; 101:213-8. [PMID: 25690487 DOI: 10.1007/s12185-015-1747-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 01/18/2015] [Indexed: 10/24/2022]
Abstract
Whole exome next generation sequencing systematically applied as a discovery tool in myelodysplastic syndromes (MDS) has led to the identification of a large number of novel mutations. Despite hundreds of patients studied, mutational saturation has not been reached and it is expected that new driver mutations will be discovered in this very heterogeneous condition. Serial samples and deep sequencing of the identified alterations has allowed for a dynamic/chronologic analysis of clonal architecture and identification of a subset of ancestral and secondary molecular lesions. Chromosomal gains and losses have been incorporated into the mutational analyses because they can either cooperate with mutations or produce a functional phenocopy. In addition to the search for somatic defects in MDS, similar discovery studies have been also performed to identify germ line mutations/alterations. Clinical analysis showed applicability of multiplexed somatic mutational panels that would complement current pathomorphologic diagnosis, allow for subclassification of nosologic entities, and enhance predictive power of current prognostic algorithms. Overall, comprehensive genomic analysis in MDS has revealed a tremendous heterogeneity of somatic lesions and their combinations further enhanced by the heterogeneity of clonal architecture and chromosomal lesions.
Collapse
|
72
|
Li B, Gale RP, Xiao Z. Molecular genetics of chronic neutrophilic leukemia, chronic myelomonocytic leukemia and atypical chronic myeloid leukemia. J Hematol Oncol 2014; 7:93. [PMID: 25498990 PMCID: PMC4266232 DOI: 10.1186/s13045-014-0093-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 12/04/2014] [Indexed: 12/21/2022] Open
Abstract
According to the 2008 World Health Organization classification, chronic neutrophilic leukemia, chronic myelomonocytic leukemia and atypical chronic myeloid leukemia are rare diseases. The remarkable progress in our understanding of the molecular genetics of myeloproliferative neoplasms and myelodysplastic/myeloproliferative neoplasms has made it clear that there are some specific genetic abnormalities in these 3 rare diseases. At the same time, there is considerable overlap among these disorders at the molecular level. The various combinations of genetic abnormalities indicate a multi-step pathogenesis, which likely contributes to the marked clinical heterogeneity of these disorders. This review focuses on the current knowledge and challenges related to the molecular pathogenesis of chronic neutrophilic leukemia, chronic myelomonocytic leukemia and atypical chronic myeloid leukemia and relationships between molecular findings, clinical features and prognosis.
Collapse
Affiliation(s)
| | | | - Zhijian Xiao
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, 288 Nanjing Road, Tianjin 300020, China.
| |
Collapse
|
73
|
Zoi K, Cross NCP. Molecular pathogenesis of atypical CML, CMML and MDS/MPN-unclassifiable. Int J Hematol 2014; 101:229-42. [PMID: 25212680 DOI: 10.1007/s12185-014-1670-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 09/02/2014] [Accepted: 09/02/2014] [Indexed: 12/21/2022]
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.
Collapse
Affiliation(s)
- Katerina Zoi
- Haematology Research Laboratory, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | | |
Collapse
|
74
|
Bedside to bench in juvenile myelomonocytic leukemia: insights into leukemogenesis from a rare pediatric leukemia. Blood 2014; 124:2487-97. [PMID: 25163700 DOI: 10.1182/blood-2014-03-300319] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is a typically aggressive myeloid neoplasm of childhood that is clinically characterized by overproduction of monocytic cells that can infiltrate organs, including the spleen, liver, gastrointestinal tract, and lung. JMML is categorized as an overlap myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) by the World Health Organization and also shares some clinical and molecular features with chronic myelomonocytic leukemia, a similar disease in adults. Although the current standard of care for patients with JMML relies on allogeneic hematopoietic stem cell transplant, relapse is the most frequent cause of treatment failure. Tremendous progress has been made in defining the genomic landscape of JMML. Insights from cancer predisposition syndromes have led to the discovery of nearly 90% of driver mutations in JMML, all of which thus far converge on the Ras signaling pathway. This has improved our ability to accurately diagnose patients, develop molecular markers to measure disease burden, and choose therapeutic agents to test in clinical trials. This review emphasizes recent advances in the field, including mapping of the genomic and epigenome landscape, insights from new and existing disease models, targeted therapeutics, and future directions.
Collapse
|
75
|
Wang L, Yamaguchi S, Burstein MD, Terashima K, Chang K, Ng HK, Nakamura H, He Z, Doddapaneni H, Lewis L, Wang M, Suzuki T, Nishikawa R, Natsume A, Terasaka S, Dauser R, Whitehead W, Adekunle A, Sun J, Qiao Y, Marth G, Muzny DM, Gibbs RA, Leal SM, Wheeler DA, Lau CC. Novel somatic and germline mutations in intracranial germ cell tumours. Nature 2014; 511:241-5. [PMID: 24896186 DOI: 10.1038/nature13296] [Citation(s) in RCA: 153] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2013] [Accepted: 03/28/2014] [Indexed: 12/12/2022]
Abstract
Intracranial germ cell tumours (IGCTs) are a group of rare heterogeneous brain tumours that are clinically and histologically similar to the more common gonadal GCTs. IGCTs show great variation in their geographical and gender distribution, histological composition and treatment outcomes. The incidence of IGCTs is historically five- to eightfold greater in Japan and other East Asian countries than in Western countries, with peak incidence near the time of puberty. About half of the tumours are located in the pineal region. The male-to-female incidence ratio is approximately 3-4:1 overall, but is even higher for tumours located in the pineal region. Owing to the scarcity of tumour specimens available for research, little is currently known about this rare disease. Here we report the analysis of 62 cases by next-generation sequencing, single nucleotide polymorphism array and expression array. We find the KIT/RAS signalling pathway frequently mutated in more than 50% of IGCTs, including novel recurrent somatic mutations in KIT, its downstream mediators KRAS and NRAS, and its negative regulator CBL. Novel somatic alterations in the AKT/mTOR pathway included copy number gains of the AKT1 locus at 14q32.33 in 19% of patients, with corresponding upregulation of AKT1 expression. We identified loss-of-function mutations in BCORL1, a transcriptional co-repressor and tumour suppressor. We report significant enrichment of novel and rare germline variants in JMJD1C, which codes for a histone demethylase and is a coactivator of the androgen receptor, among Japanese IGCT patients. This study establishes a molecular foundation for understanding the biology of IGCTs and suggests potentially promising therapeutic strategies focusing on the inhibition of KIT/RAS activation and the AKT1/mTOR pathway.
Collapse
Affiliation(s)
- Linghua Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Shigeru Yamaguchi
- Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Matthew D Burstein
- 1] Structural and Computational Biology and Molecular Biophysics Program, Baylor College of Medicine, Houston, Texas 77030, USA [2] Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Keita Terashima
- 1] Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas 77030, USA [2] National Center for Child Health and Development, Tokyo, 157-8535, Japan
| | - Kyle Chang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Ho-Keung Ng
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - Hideo Nakamura
- Department of Neurosurgery, Kumamoto University, Kumamoto, 860-0862, Japan
| | - Zongxiao He
- Center for Statistical Genetics, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | - Lora Lewis
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Mark Wang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Tomonari Suzuki
- Department of Neurosurgery, Saitama Medical University, Saitama, 350-0495, Japan
| | - Ryo Nishikawa
- Department of Neurosurgery, Saitama Medical University, Saitama, 350-0495, Japan
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University, Nagoya, 466-8550, Japan
| | - Shunsuke Terasaka
- Department of Neurosurgery, Hokkaido University, Hokkaido Prefecture, 060-0808, Japan
| | - Robert Dauser
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | - William Whitehead
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Adesina Adekunle
- Department of Pathology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Jiayi Sun
- Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Yi Qiao
- Department of Biology, Boston College, Chestnut Hill, Maryland 02467, USA
| | - Gábor Marth
- Department of Biology, Boston College, Chestnut Hill, Maryland 02467, USA
| | - Donna M Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Richard A Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Suzanne M Leal
- Center for Statistical Genetics, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - David A Wheeler
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Ching C Lau
- 1] Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, Texas 77030, USA [2] Medical Scientist Training Program, Baylor College of Medicine, Houston, Texas 77030, USA [3] Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
76
|
Iacobucci I, Lonetti A, Papayannidis C, Martinelli G. Use of single nucleotide polymorphism array technology to improve the identification of chromosomal lesions in leukemia. Curr Cancer Drug Targets 2014; 13:791-810. [PMID: 23941516 PMCID: PMC4104470 DOI: 10.2174/15680096113139990089] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 04/05/2013] [Accepted: 07/01/2013] [Indexed: 12/13/2022]
Abstract
Acute leukemias are characterized by recurring chromosomal and genetic abnormalities that disrupt normal development and drive aberrant cell proliferation and survival. Identification of these abnormalities plays important role in diagnosis, risk assessment and patient classification. Until the last decade methods to detect these aberrations have included genome wide approaches, such as conventional cytogenetics, but with a low sensitivity (5-10%), or gene candidate approaches, such as fluorescent in situ hybridization, having a greater sensitivity but being limited to only known regions of the genome. Single nucleotide polymorphism (SNP) technology is a screening method that has revolutionized our way to find genetic alterations, enabling linkage and association studies between SNP genotype and disease as well as the identification of alterations in DNA content on a whole genome scale. The adoption of this approach for the study of lymphoid and myeloid leukemias contributed to the identification of novel genetic alterations, such as losses/gains/uniparental disomy not visible by cytogenetics and implicated in pathogenesis, improving risk assessment and patient classification and in some cases working as targets for tailored therapies. In this review, we reported recent advances obtained in the knowledge of the genomic complexity of chronic myeloid leukemia and acute leukemias thanks to the use of high-throughput technologies, such as SNP array.
Collapse
Affiliation(s)
- Ilaria Iacobucci
- Institute of Hematology "L. e A. Seràgnoli" Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy, Via Massarenti, 9 - 40138 Bologna, Italy.
| | | | | | | |
Collapse
|
77
|
Dasatinib targets B-lineage cells but does not provide an effective therapy for myeloproliferative disease in c-Cbl RING finger mutant mice. PLoS One 2014; 9:e94717. [PMID: 24718698 PMCID: PMC3981816 DOI: 10.1371/journal.pone.0094717] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 03/18/2014] [Indexed: 11/30/2022] Open
Abstract
This study aimed to determine whether the multi-kinase inhibitor dasatinib would provide an effective therapy for myeloproliferative diseases (MPDs) involving c-Cbl mutations. These mutations, which occur in the RING finger and linker domains, abolish the ability of c-Cbl to function as an E3 ubiquitin ligase and downregulate activated protein tyrosine kinases. Here we analyzed the effects of dasatinib in a c-Cbl RING finger mutant mouse that develops an MPD with a phenotype similar to the human MPDs. The mice are characterized by enhanced tyrosine kinase signaling resulting in an expansion of hematopoietic stem cells, multipotent progenitors and cells within the myeloid lineage. Since c-Cbl is a negative regulator of c-Kit and Src signaling we reasoned that dasatinib, which targets these kinases, would be an effective therapy. Furthermore, two recent studies showed dasatinib to be effective in inhibiting the in vitro growth of cells from leukemia patients with c-Cbl RING finger and linker domain mutations. Surprisingly we found that dasatinib did not provide an effective therapy for c-Cbl RING finger mutant mice since it did not suppress any of the hematopoietic lineages that promote MPD development. Thus we conclude that dasatinib may not be an appropriate therapy for leukemia patients with c-Cbl mutations. We did however find that dasatinib caused a marked reduction of pre-B cells and immature B cells which correlated with a loss of Src activity. This study is therefore the first to provide a detailed characterization of in vivo effects of dasatinib in a hematopoietic disorder that is driven by protein tyrosine kinases other than BCR-ABL.
Collapse
|
78
|
Abstract
We describe the development of acute myeloid leukemia (AML) in an adult with CBL syndrome caused by a heterozygous de novo germline mutation in CBL codon D390. In the AML bone marrow, the mutated CBL allele was homozygous after copy number-neutral loss-of-heterozygosity and amplified through a chromosomal gain; moreover, an inv(16)(p13q22) and, as assessed by whole-exome sequencing, 12 gene mutations (eg, in CAND1, NID2, PTPRT, DOCK6) were additionally acquired. During complete remission of the AML, in the presence of normal blood counts, the hematopoiesis stably maintained the homozygous CBL mutation, which is reminiscent of the situation in children with CBL syndrome and transient juvenile myelomonocytic leukemia. No additional mutations were identified by whole-exome sequencing in granulocytes during complete remission. The study highlights the development of AML in an adult with CBL syndrome and, more generally, in genetically aberrant but clinically inconspicuous hematopoiesis.
Collapse
|
79
|
Abstract
Higher-risk myelodysplastic syndromes (MDS) are defined by patients who fall into higher-risk group categories in the original or revised International Prognostic Scoring System. Survival for these patients is dismal, and treatment should be initiated rapidly. Standard therapies include the hypomethylating agents azacitidine and decitabine, which should be administered for a minimum of 6 cycles, and continued for as long as a patient is responding. Once a drug fails in one of these patients, further treatment options are limited, median survival is <6 months, and consideration should be given to clinical trials. Higher-risk eligible patients should be offered consultation to discuss hematopoietic stem cell transplantation close to the time of diagnosis, depending on patient goals of therapy, with consideration given to proceeding to transplantation soon after an optimal donor is located. In the interim period before transplantation, hypomethylating agent therapy, induction chemotherapy, or enrollment in a clinical trial should be considered to prevent disease progression, although the optimal pretransplantation therapy is unknown.
Collapse
|
80
|
Parikh SA, Tefferi A. Chronic myelomonocytic leukemia: 2013 update on diagnosis, risk stratification, and management. Am J Hematol 2013; 88:967-74. [PMID: 23963888 DOI: 10.1002/ajh.23574] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 08/14/2013] [Indexed: 01/20/2023]
Abstract
DISEASE OVERVIEW Chronic myelomonocytic leukemia (CMML) is a clonal hematopoietic stem cell disorder that is classified as a myelodysplastic/myeloproliferative neoplasm by the 2008 World Health Organization classification of hematopoietic tumors. It is characterized by absolute monocytosis (>1 × 10⁹/L) in the peripheral blood that persists for at least 3 months. DIAGNOSIS The diagnosis of CMML rests on a combination of morphologic, histopathologic and chromosomal abnormalities in the bone marrow. It is important to exclude other myeloproliferative neoplasms and infectious/autoimmune conditions that can cause monocytosis. RISK STRATIFICATION Several CMML-specific prognostic models incorporating novel mutations have been recently reported. The Mayo prognostic model classified CMML patients into three risk groups based on: increased absolute monocyte count, presence of circulating blasts, hemoglobin <10 gm/dL and platelets <100 × 10⁹/L. The median survival was 32 months, 18.5 months and 10 months in the low, intermediate, and high-risk groups, respectively. The Groupe Francophone des (GFM) score segregated CMML patients into three risk groups based on: age >65 years, WBC >15 × 10⁹/L, anemia, platelets <100 × 10⁹/L, and ASXL1 mutation status. After a median follow-up of 2.5 years, survival ranged from not reached in the low-risk group to 14.4 months in the high-risk group. RISK-ADAPTED THERAPY The Food and Drug Administration has approved azacitidine and decitabine for the treatment of patients with CMML. An allogeneic stem cell transplant can potentially offer a curative option to a subset of CMML patients. It is hoped that with the discovery of several novel mutations, targeted therapies will become available in the near future.
Collapse
Affiliation(s)
- Sameer A. Parikh
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| | - Ayalew Tefferi
- Division of Hematology, Department of Medicine; Mayo Clinic; Rochester Minnesota
| |
Collapse
|
81
|
Klampfl T, Milosevic JD, Puda A, Schönegger A, Bagienski K, Berg T, Harutyunyan AS, Gisslinger B, Rumi E, Malcovati L, Pietra D, Elena C, Della Porta MG, Pieri L, Guglielmelli P, Bock C, Doubek M, Dvorakova D, Suvajdzic N, Tomin D, Tosic N, Racil Z, Steurer M, Pavlovic S, Vannucchi AM, Cazzola M, Gisslinger H, Kralovics R. Complex patterns of chromosome 11 aberrations in myeloid malignancies target CBL, MLL, DDB1 and LMO2. PLoS One 2013; 8:e77819. [PMID: 24147083 PMCID: PMC3797696 DOI: 10.1371/journal.pone.0077819] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 09/04/2013] [Indexed: 11/19/2022] Open
Abstract
Exome sequencing of primary tumors identifies complex somatic mutation patterns. Assignment of relevance of individual somatic mutations is difficult and poses the next challenge for interpretation of next generation sequencing data. Here we present an approach how exome sequencing in combination with SNP microarray data may identify targets of chromosomal aberrations in myeloid malignancies. The rationale of this approach is that hotspots of chromosomal aberrations might also harbor point mutations in the target genes of deletions, gains or uniparental disomies (UPDs). Chromosome 11 is a frequent target of lesions in myeloid malignancies. Therefore, we studied chromosome 11 in a total of 813 samples from 773 individual patients with different myeloid malignancies by SNP microarrays and complemented the data with exome sequencing in selected cases exhibiting chromosome 11 defects. We found gains, losses and UPDs of chromosome 11 in 52 of the 813 samples (6.4%). Chromosome 11q UPDs frequently associated with mutations of CBL. In one patient the 11qUPD amplified somatic mutations in both CBL and the DNA repair gene DDB1. A duplication within MLL exon 3 was detected in another patient with 11qUPD. We identified several common deleted regions (CDR) on chromosome 11. One of the CDRs associated with de novo acute myeloid leukemia (P=0.013). One patient with a deletion at the LMO2 locus harbored an additional point mutation on the other allele indicating that LMO2 might be a tumor suppressor frequently targeted by 11p deletions. Our chromosome-centered analysis indicates that chromosome 11 contains a number of tumor suppressor genes and that the role of this chromosome in myeloid malignancies is more complex than previously recognized.
Collapse
Affiliation(s)
- Thorsten Klampfl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Jelena D. Milosevic
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Ana Puda
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Andreas Schönegger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Klaudia Bagienski
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Tiina Berg
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Ashot S. Harutyunyan
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Bettina Gisslinger
- Division of Hematology and Blood Coagulation, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Elisa Rumi
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Luca Malcovati
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Daniela Pietra
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Chiara Elena
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Matteo Giovanni Della Porta
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Lisa Pieri
- Section of Hematology, University of Florence, Florence, Italy
| | | | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Michael Doubek
- Department of Internal Medicine Hematology and Oncology, University Hospital Brno, Masaryk University Brno, Brno, Czech Republic, Czech Republic
- CEITEC - Central European Institute of Technology, Masaryk University Brno, Brno, Czech Republic
| | - Dana Dvorakova
- Department of Internal Medicine Hematology and Oncology, University Hospital Brno, Masaryk University Brno, Brno, Czech Republic, Czech Republic
- CEITEC - Central European Institute of Technology, Masaryk University Brno, Brno, Czech Republic
| | - Nada Suvajdzic
- Clinic of Hematology, Clinical Center of Serbia, University of Belgrade, School of Medicine, Belgrade, Serbia
| | - Dragica Tomin
- Clinic of Hematology, Clinical Center of Serbia, University of Belgrade, School of Medicine, Belgrade, Serbia
| | - Natasa Tosic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | - Zdenek Racil
- Department of Internal Medicine Hematology and Oncology, University Hospital Brno, Masaryk University Brno, Brno, Czech Republic, Czech Republic
- CEITEC - Central European Institute of Technology, Masaryk University Brno, Brno, Czech Republic
| | - Michael Steurer
- Division of Hematology and Oncology, Innsbruck University Hospital, Innsbruck, Austria
| | - Sonja Pavlovic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade, Serbia
| | | | - Mario Cazzola
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Heinz Gisslinger
- Division of Hematology and Blood Coagulation, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Robert Kralovics
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Division of Hematology and Blood Coagulation, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
- * E-mail:
| |
Collapse
|
82
|
Itzykson R, Fenaux P, Solary E. Chronic myelomonocytic leukemia: myelodysplastic or myeloproliferative? Best Pract Res Clin Haematol 2013; 26:387-400. [PMID: 24507815 DOI: 10.1016/j.beha.2013.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Chronic myelomonocytic leukemia (CMML) is a clonal disease of the hematopoietic stem cell that provokes a stable increase in peripheral blood monocyte count. The World Health Organisation classification appropriately underlines that the disease combines dysplastic and proliferative features. The percentage of blast cells in the blood and bone marrow distinguishes CMML-1 from CMML-2. The disease is usually diagnosed after the age of 50, with a strong male predominance. Inconstant and non-specific cytogenetic aberrations have a negative prognostic impact. Recurrent gene mutations affect mainly the TET2, SRSF2, and ASXL1 genes. Median survival is 3 years, with patients dying from progression to AML (20-30%) or from cytopenias. ASXL1 is the only gene whose mutation predicts outcome and can be included within a prognostic score. Allogeneic stem cell transplantation is possibly curative but rarely feasible. Hydroxyurea, which is the conventional cytoreductive agent, is used in myeloproliferative forms, and demethylating agents could be efficient in the most aggressive forms of the disease.
Collapse
Affiliation(s)
| | - Pierre Fenaux
- Hôpital Avicenne, Service d'hématologie clinique, Paris 13 university, 125 rue de Stalingrad, 93009 Bobigny, France.
| | - Eric Solary
- Inserm UMR 1009, Institut Gustave Roussy, 14 rue Edouard Vaillant, 94805 Villejuif cedex, France.
| |
Collapse
|
83
|
Traina F, Visconte V, Elson P, Tabarroki A, Jankowska AM, Hasrouni E, Sugimoto Y, Szpurka H, Makishima H, O'Keefe CL, Sekeres MA, Advani AS, Kalaycio M, Copelan EA, Saunthararajah Y, Olalla Saad ST, Maciejewski JP, Tiu RV. Impact of molecular mutations on treatment response to DNMT inhibitors in myelodysplasia and related neoplasms. Leukemia 2013; 28:78-87. [PMID: 24045501 DOI: 10.1038/leu.2013.269] [Citation(s) in RCA: 244] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/23/2013] [Accepted: 08/29/2013] [Indexed: 01/28/2023]
Abstract
We hypothesized that specific molecular mutations are important biomarkers for response to DNA methyltransferase inhibitors (DNMT inhibitors) and may have prognostic value in patients with myelodysplastic syndromes (MDS). Mutational analysis was performed in 92 patients with MDS and related disorders who received 5-azacytidine (n=55), decitabine (n=26) or both (n=11). Mutational status was correlated with overall response rate (ORR), progression-free survival (PFS) and overall survival (OS) by univariate and multivariate analysis. Risk stratification models were created. TET2, DNMT3A, IDH1/IDH2, ASXL1, CBL, RAS and SF3B1 mutations were found in 18, 9, 8, 26, 3, 2 and 13% of patients, respectively. In multivariate analysis, TET2(MUT) and/or DNMT3A(MUT) (P=0.03), platelets > or = 100 × 10(9)/l (P=0.007) and WBC<3.0 × 10(9)/l (P=0.03) were independent predictors of better response. TET2(MUT) and/or DNMT3A(MUT) (P=0.04) status was also independently prognostic for improved PFS, as were good or intermediate cytogenetic risk (P<0.0001), age<60 (P=0.0001), treatment with both 5-azacytidine and decitabine (P=0.02) and hemoglobin > or = 10 g/dl (P=0.01). Better OS was associated with ASXL1(WT) (P=0.008) and SF3B1(MUT) (P=0.01), and, similar to PFS, cytogenetic risk (P=0.0002), age (P=0.02) and hemoglobin (P=0.04). These data support the role of molecular mutations as predictive biomarkers for response and survival in MDS patients treated with DNMT inhibitors.
Collapse
Affiliation(s)
- F Traina
- 1] Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA [2] Hematology and Hemotherapy Center - INCT do Sangue, University of Campinas - UNICAMP, Campinas, SP, Brazil [3] Department of Internal Medicine, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, SP, Brazil
| | - V Visconte
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - P Elson
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - A Tabarroki
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - A M Jankowska
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - E Hasrouni
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Y Sugimoto
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - H Szpurka
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - H Makishima
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - C L O'Keefe
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - M A Sekeres
- Department of Hematologic Oncology and Blood Disorders, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - A S Advani
- Department of Hematologic Oncology and Blood Disorders, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - M Kalaycio
- Department of Hematologic Oncology and Blood Disorders, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - E A Copelan
- Department of Hematologic Oncology and Blood Disorders, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Y Saunthararajah
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - S T Olalla Saad
- Hematology and Hemotherapy Center - INCT do Sangue, University of Campinas - UNICAMP, Campinas, SP, Brazil
| | - J P Maciejewski
- 1] Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA [2] Department of Hematologic Oncology and Blood Disorders, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - R V Tiu
- 1] Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA [2] Department of Hematologic Oncology and Blood Disorders, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
84
|
Diagnosis and treatment of primary myelodysplastic syndromes in adults: recommendations from the European LeukemiaNet. Blood 2013; 122:2943-64. [PMID: 23980065 DOI: 10.1182/blood-2013-03-492884] [Citation(s) in RCA: 510] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Within the myelodysplastic syndrome (MDS) work package of the European LeukemiaNet, an Expert Panel was selected according to the framework elements of the National Institutes of Health Consensus Development Program. A systematic review of the literature was performed that included indexed original papers, indexed reviews and educational papers, and abstracts of conference proceedings. Guidelines were developed on the basis of a list of patient- and therapy-oriented questions, and recommendations were formulated and ranked according to the supporting level of evidence. MDSs should be classified according to the 2008 World Health Organization criteria. An accurate risk assessment requires the evaluation of not only disease-related factors but also of those related to extrahematologic comorbidity. The assessment of individual risk enables the identification of fit patients with a poor prognosis who are candidates for up-front intensive treatments, primarily allogeneic stem cell transplantation. A high proportion of MDS patients are not eligible for potentially curative treatment because of advanced age and/or clinically relevant comorbidities and poor performance status. In these patients, the therapeutic intervention is aimed at preventing cytopenia-related morbidity and preserving quality of life. A number of new agents are being developed for which the available evidence is not sufficient to recommend routine use. The inclusion of patients into prospective clinical trials is strongly recommended.
Collapse
|
85
|
Otrock ZK, Tiu RV, Maciejewski JP, Sekeres MA. The need for additional genetic markers for myelodysplastic syndrome stratification: what does the future hold for prognostication? Expert Rev Hematol 2013; 6:59-68. [PMID: 23373781 DOI: 10.1586/ehm.12.67] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Myelodysplastic syndromes (MDS) constitute a heterogeneous group of clonal hematopoietic disorders. Metaphase cytogenetics has been the gold standard for genetic testing in MDS, but it detects clonal cytogenetic abnormalities in only 50% of cases. New karyotyping tests include FISH, array-based comparative genomic hybridization and single-nucleotide polymorphism arrays. These techniques have increased the detected genetic abnormalities in MDS, many of which confer prognostic significance to overall and leukemia-free survival. This has eventually increased our understanding of MDS genetics. With the help of new technologies, we anticipate that the existing prognostic scoring systems will incorporate mutational data into their parameters. This review discusses the progress in MDS diagnosis through the use of array-based technologies. The authors also discuss the recently investigated genetic mutations in MDS and revisit the MDS classification and prognostic scoring systems.
Collapse
Affiliation(s)
- Zaher K Otrock
- Leukemia Program, Cleveland Clinic Taussig Cancer Institute, OH 44195, USA
| | | | | | | |
Collapse
|
86
|
Itzykson R, Kosmider O, Renneville A, Gelsi-Boyer V, Meggendorfer M, Morabito M, Berthon C, Adès L, Fenaux P, Beyne-Rauzy O, Vey N, Braun T, Haferlach T, Dreyfus F, Cross NC, Preudhomme C, Bernard OA, Fontenay M, Vainchenker W, Schnittger S, Birnbaum D, Droin N, Solary E. Prognostic Score Including Gene Mutations in Chronic Myelomonocytic Leukemia. J Clin Oncol 2013; 31:2428-36. [DOI: 10.1200/jco.2012.47.3314] [Citation(s) in RCA: 406] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose Several prognostic scoring systems have been proposed for chronic myelomonocytic leukemia (CMML), a disease in which some gene mutations—including ASXL1—have been associated with poor prognosis in univariable analyses. We developed and validated a prognostic score for overall survival (OS) based on mutational status and standard clinical variables. Patients and Methods We genotyped ASXL1 and up to 18 other genes including epigenetic (TET2, EZH2, IDH1, IDH2, DNMT3A), splicing (SF3B1, SRSF2, ZRSF2, U2AF1), transcription (RUNX1, NPM1, TP53), and signaling (NRAS, KRAS, CBL, JAK2, FLT3) regulators in 312 patients with CMML. Genotypes and clinical variables were included in a multivariable Cox model of OS validated by bootstrapping. A scoring system was developed using regression coefficients from this model. Results ASXL1 mutations (P < .0001) and, to a lesser extent, SRSF2 (P = .03), CBL (P = .003), and IDH2 (P = .03) mutations predicted inferior OS in univariable analysis. The retained independent prognostic factors included ASXL1 mutations, age older than 65 years, WBC count greater than 15 ×109/L, platelet count less than 100 ×109/L, and anemia (hemoglobin < 10 g/dL in female patients, < 11g/dL in male patients). The resulting five-parameter prognostic score delineated three groups of patients with median OS not reached, 38.5 months, and 14.4 months, respectively (P < .0001), and was validated in an independent cohort of 165 patients (P < .0001). Conclusion A new prognostic score including ASXL1 status, age, hemoglobin, WBC, and platelet counts defines three groups of CMML patients with distinct outcomes. Based on concordance analysis, this score appears more discriminative than those based solely on clinical parameters.
Collapse
Affiliation(s)
- Raphaël Itzykson
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Olivier Kosmider
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Aline Renneville
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Véronique Gelsi-Boyer
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Manja Meggendorfer
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Margot Morabito
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Céline Berthon
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Lionel Adès
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Pierre Fenaux
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Odile Beyne-Rauzy
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Norbert Vey
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Thorsten Braun
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Torsten Haferlach
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - François Dreyfus
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Nicholas C.P. Cross
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Claude Preudhomme
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Olivier A. Bernard
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Michaela Fontenay
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - William Vainchenker
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Susanne Schnittger
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Daniel Birnbaum
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Nathalie Droin
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| | - Eric Solary
- Raphaël Itzykson, Margot Morabito, Olivier A. Bernard, William Vainchenker, Nathalie Droin, Eric Solary, Institut Gustave Roussy, Villejuif; Raphaël Itzykson, Margot Morabito, William Vainchenker, Nathalie Droin, Eric Solary, Université Paris-Sud, Le Kremlin-Bicêtre, Orsay; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Assistance Publique–Hôpitaux de Paris, Hôpital Cochin; Olivier Kosmider, François Dreyfus, Michaela Fontenay, Université Paris Descartes; Olivier Kosmider, Michaela Fontenay,
| |
Collapse
|
87
|
Javadi M, Richmond TD, Huang K, Barber DL. CBL linker region and RING finger mutations lead to enhanced granulocyte-macrophage colony-stimulating factor (GM-CSF) signaling via elevated levels of JAK2 and LYN. J Biol Chem 2013; 288:19459-70. [PMID: 23696637 DOI: 10.1074/jbc.m113.475087] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is characterized by hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF). SHP2, NF-1, KRAS, and NRAS are mutated in JMML patients, leading to aberrant regulation of RAS signaling. A subset of JMML patients harbor CBL mutations associated with 11q acquired uniparental disomy. Many of these mutations are in the linker region and the RING finger of CBL, leading to a loss of E3 ligase activity. We investigated the mechanism by which CBL-Y371H, a linker region mutant, and CBL-C384R, a RING finger mutant, lead to enhanced GM-CSF signaling. Expression of CBL mutants in the TF-1 cell line resulted in enhanced survival in the absence of GM-CSF. Cells expressing CBL mutations displayed increased phosphorylation of GM-CSF receptor βc subunit in response to stimulation, although expression of total GM-CSFR βc was lower. This suggested enhanced kinase activity downstream of GM-CSFR. JAK2 and LYN kinase expression is elevated in CBL-Y371H and CBL-C384R mutant cells, resulting in enhanced phosphorylation of CBL and S6 in response to GM-CSF stimulation. Incubation with the JAK2 inhibitor, TG101348, abolished the increased phosphorylation of GM-CSFR βc in cells expressing CBL mutants, whereas treatment with the SRC kinase inhibitor dasatinib resulted in equalization of GM-CSFR βc phosphorylation signal between wild type CBL and CBL mutant samples. Dasatinib treatment inhibited the elevated phosphorylation of CBL-Y371H and CBL-C384R mutants. Our study indicates that CBL linker and RING finger mutants lead to enhanced GM-CSF signaling due to elevated kinase expression, which can be blocked using small molecule inhibitors targeting specific downstream pathways.
Collapse
Affiliation(s)
- Mojib Javadi
- Ontario Cancer Institute, Campbell Family Cancer Research Institute, Toronto, Ontario M5G 2M9, Canada
| | | | | | | |
Collapse
|
88
|
Grossmann V, Roller A, Klein HU, Weissmann S, Kern W, Haferlach C, Dugas M, Haferlach T, Schnittger S, Kohlmann A. Robustness of amplicon deep sequencing underlines its utility in clinical applications. J Mol Diagn 2013; 15:473-84. [PMID: 23680131 DOI: 10.1016/j.jmoldx.2013.03.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 02/08/2013] [Accepted: 03/01/2013] [Indexed: 12/16/2022] Open
Abstract
We investigated the robustness of amplicon deep sequencing to study its utility in routine clinical applications offering patient-specific individualized assays for molecular disease characterization and monitoring. Amplicons were designed targeting RUNX1, CEBPA, CBL, NRAS, KRAS, DNMT3A, EZH2, and TP53 using different PCR amplification strategies and Roche GS FLX Titanium and Illumina MiSeq sequencing platforms. Thirty-three patients with leukemia were selected as an exemplary cohort representing heterogeneous cancer specimens. Both standard two-primer amplification and four-primer microfluidics PCRs yielded highly linear characteristics in detecting molecular alterations in series of dilution experiments. By fitting a linear mixed-effects model to the logarithmized data, a slope β of -1.000 (95% CI, ±0.046) was obtained for two-primer assays and of -0.998 (95% CI, ±0.105) was obtained for four-primer assays, which represented a near-perfect decrease of the mutation load. Furthermore, data are presented on technical precision, limit of detection, and occurrence of small subclones in TP53- and RUNX1-mutated patients to identify clonal disease progression and residual disease. We demonstrate that, depending on the local sequence context for each amplicon, the limit of detection of the assay cannot be lower than a range of 0.25% to 3.5%. In conclusion, amplicon deep sequencing enabled the assessment of mutations in a highly robust manner and across a broad range of relative frequencies of mutations. This assay detects residual disease or identifies mutations with predictive relevance to direct treatment strategies.
Collapse
|
89
|
Fiskus W, Verstovsek S, Manshouri T, Smith JE, Peth K, Abhyankar S, McGuirk J, Bhalla KN. Dual PI3K/AKT/mTOR inhibitor BEZ235 synergistically enhances the activity of JAK2 inhibitor against cultured and primary human myeloproliferative neoplasm cells. Mol Cancer Ther 2013; 12:577-88. [PMID: 23445613 DOI: 10.1158/1535-7163.mct-12-0862] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Hemopoietic progenitor cells (HPC) from myeloproliferative neoplasms (MPN) such as myelofibrosis commonly express mutant JAK2-V617F or other mutations that are associated with increased activities of JAK-STAT5/3, RAS/RAF/MAPK, and PI3K/AKT/mTOR pathways. This confers proliferative and survival advantage on the MPN HPCs. Treatment with JAK tyrosine kinase inhibitor (TKI), for example, TG101209, TG101348 (SAR302503), or INCB018424 (ruxolitinib), inhibits mutant JAK2-mediated signaling. Although effective in reducing constitutional symptoms and splenomegaly, treatment with JAK-TKI does not ameliorate myelofibrosis or significantly improve survival of patients with advanced myelofibrosis. Here, we show that treatment with the dual phosphoinositide-3-kinase (PI3K)/AKT and mTOR inhibitor BEZ235 attenuated PI3K/AKT and mTOR signaling, as well as induced cell-cycle growth arrest and apoptosis of the cultured human JAK2-V617F-expressing HEL92.1.7 (HEL), UKE1 cells, and primary CD34+ myelofibrosis (MF)-MPN cells. Treatment with BEZ235 also induced significant apoptosis of the JAK2-TKI resistant HEL/TGR cells that were selected for resistance against JAK-TKI. Cotreatment with BEZ235 and JAK2-TKI (TG101209 and SAR302503) synergistically induced lethal activity against the cultured and primary CD34+ MPN cells while relatively sparing the normal CD34+ HPCs. These findings create a compelling rationale to determine the in vivo activity of dual PI3K/mTOR inhibitors in combination with JAK inhibitors against myelofibrosis HPCs.
Collapse
Affiliation(s)
- Warren Fiskus
- The University of Kansas Cancer Center, Kansas City, KS, USA
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Mallo M, del Rey M, Ibáñez M, Calasanz MJ, Arenillas L, Larráyoz MJ, Pedro C, Jerez A, Maciejewski J, Costa D, Nomdedeu M, Diez-Campelo M, Lumbreras E, González-Martínez T, Marugán I, Such E, Cervera J, Cigudosa JC, Álvarez S, Florensa L, Hernández JM, Solé F. Response to lenalidomide in myelodysplastic syndromes with del(5q): influence of cytogenetics and mutations. Br J Haematol 2013; 162:74-86. [DOI: 10.1111/bjh.12354] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 03/12/2013] [Indexed: 12/15/2022]
Affiliation(s)
| | - Mónica del Rey
- Servicio de Hematología; Centro de Investigación del Cáncer; IBSAL (Instituto de Biomedicina de Salamanca) y IBMCC; Universidad de Salamanca; Salamanca; Spain
| | - Mariam Ibáñez
- Servicio de Hematología; Hospital Universitario La Fe; Valencia; Spain
| | - Mª José Calasanz
- Departamento de Genética; Universidad de Navarra; Pamplona; Spain
| | - Leonor Arenillas
- Laboratori de Citogenètica Molecular; Laboratori de Citologia Hematològica; Servei de Patologia; Hospital del Mar; GRETNHE; IMIM (Hospital del Mar Research Institute); Barcelona; Spain
| | - Mª José Larráyoz
- Departamento de Genética; Universidad de Navarra; Pamplona; Spain
| | - Carmen Pedro
- Servei d'Hematologia Clínica; Hospital del Mar; GRETNHE; IMIM (Hospital del Mar Research Institute); Barcelona; Spain
| | - Andrés Jerez
- Department of Translational Hematology and Oncology Research; Taussig Cancer Institute; Cleveland Clinic; Cleveland; OH; USA
| | - Jaroslaw Maciejewski
- Department of Translational Hematology and Oncology Research; Taussig Cancer Institute; Cleveland Clinic; Cleveland; OH; USA
| | - Dolors Costa
- Servei d'Hematopatologia; Hospital Clínic; Barcelona; Spain
| | | | - María Diez-Campelo
- Servicio de Hematología; Centro de Investigación del Cáncer; IBSAL (Instituto de Biomedicina de Salamanca) y IBMCC; Universidad de Salamanca; Salamanca; Spain
| | - Eva Lumbreras
- Servicio de Hematología; Centro de Investigación del Cáncer; IBSAL (Instituto de Biomedicina de Salamanca) y IBMCC; Universidad de Salamanca; Salamanca; Spain
| | - Teresa González-Martínez
- Citoxenética-oncohematolóxica; Fundación Pública Galega de Medicina Xenómica Hospital Clínico Universitario; Santiago de Compostela; Spain
| | - Isabel Marugán
- Servicio de Hematología y Oncología Médica; Hospital Clínico Universitario de Valencia; Valencia; Spain
| | - Esperanza Such
- Servicio de Hematología; Hospital Universitario La Fe; Valencia; Spain
| | - José Cervera
- Servicio de Hematología; Hospital Universitario La Fe; Valencia; Spain
| | - Juan C. Cigudosa
- Grupo de Citogenética Molecular; Centro Nacional de Investigaciones Oncológicas; Madrid; Spain
| | - Sara Álvarez
- Grupo de Citogenética Molecular; Centro Nacional de Investigaciones Oncológicas; Madrid; Spain
| | - Lourdes Florensa
- Laboratori de Citogenètica Molecular; Laboratori de Citologia Hematològica; Servei de Patologia; Hospital del Mar; GRETNHE; IMIM (Hospital del Mar Research Institute); Barcelona; Spain
| | - Jesús Mª Hernández
- Servicio de Hematología; Centro de Investigación del Cáncer; IBSAL (Instituto de Biomedicina de Salamanca) y IBMCC; Universidad de Salamanca; Salamanca; Spain
| | | |
Collapse
|
91
|
Prithviraj GK, Mathew BM, Komrokji RK, List AF, Padron E. Chronic myelomonocytic leukemia: a review of the molecular biology, prognostic models and treatment. Int J Hematol Oncol 2013. [DOI: 10.2217/ijh.13.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Chronic myelomonocytic leukemia (CMML) is a genetically heterogeneous hematologic neoplasm that manifests with features of both a myelodysplastic syndrome and a myeloproliferative neoplasm. Recent advances in the characterization of recurrent genetic markers have resulted in a better understanding of the leukemia-initiating events and have distinguished CMML from other clonal hematopoietic malignancies. Although these mutations may lead to CMML-specific therapies in the relatively near future, the current state of therapy for CMML is based on treatments designed for the myelodysplastic syndromes. Here we review the recurrent genetic mutations and, if known, their clinical significance. We also review the treatment and available CMML-specific prognostic models and novel therapies moving forward.
Collapse
Affiliation(s)
- Gopi K Prithviraj
- H Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Blessy M Mathew
- H Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Rami K Komrokji
- H Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Alan F List
- H Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA
| | - Eric Padron
- H Lee Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| |
Collapse
|
92
|
Nadeau S, An W, Palermo N, Feng D, Ahmad G, Dong L, Borgstahl GEO, Natarajan A, Naramura M, Band V, Band H. Oncogenic Signaling by Leukemia-Associated Mutant Cbl Proteins. ACTA ACUST UNITED AC 2013; Suppl 6. [PMID: 23997989 DOI: 10.4172/2161-1009.s6-001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Members of the Cbl protein family (Cbl, Cbl-b, and Cbl-c) are E3 ubiquitin ligases that have emerged as critical negative regulators of protein tyrosine kinase (PTK) signaling. This function reflects their ability to directly interact with activated PTKs and to target them as well as their associated signaling components for ubiquitination. Given the critical roles of PTK signaling in driving oncogenesis, recent studies in animal models and genetic analyses in human cancer have firmly established that Cbl proteins function as tumor suppressors. Missense mutations or small in-frame deletions within the regions of Cbl protein that are essential for its E3 activity have been identified in nearly 5% of leukemia patients with myelodysplastic/myeloproliferative disorders. Based on evidence from cell culture studies, in vivo models and clinical data, we discuss the potential signaling mechanisms of mutant Cbl-driven oncogenesis. Mechanistic insights into oncogenic Cbl mutants and associated animal models are likely to enhance our understanding of normal hematopoietic stem cell homeostasis and provide avenues for targeted therapy of mutant Cbl-driven cancers.
Collapse
Affiliation(s)
- Scott Nadeau
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 985950 Nebraska Medical Center Omaha, NE 68198-5950, USA ; Departments of Genetics, Cell Biology & Anatomy, University of Nebraska Medical Center, 985950 Nebraska Medical Center Omaha, NE 68198-5950, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Itzykson R, Droin N, Solary E. Current insights in the cellular and molecular biology of chronic myelomonocytic leukemia. Int J Hematol Oncol 2012. [DOI: 10.2217/ijh.12.19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
SUMMARY Chronic myelomonocytic leukemia is a rare clonal myeloid disorder most often seen in the elderly that remains a virtually incurable disease. Chronic myelomonocytic leukemia has long been considered as a myelodysplastic syndrome by diagnostic classifications, but recent insights in the cellular and molecular biology of the disease has refined its identity. The malignant clone was shown to generate myeloid-derived suppressive cells that may contribute to disease expansion, whereas the role of progenitor hypersensitivity to granulomonocyte colony-stimulating factor probably defines two distinct subgroups. At least one gene mutation can now be identified in almost all the patients. The most frequently mutated genes are TET2, SRSF2 and ASXL1, with a frequent combination of mutations in the first two genes, whereas ASXL1 mutations define a poor prognostic subgroup of patients. A number of additional mutations have been identified that confer to the disease its phenotype specificity; for example, mutations in RUNX1 induce thrombocytopenia, those in SF3B1 can be associated with anemia, and those in signaling molecules including NRAS, KRAS, CBL, JAK2 and FLT3, characterize the proliferative forms of the disease. Based on these recent observations, new working models on disease pathogenesis are proposed and may serve as a basis for the search for alternative and more efficient therapeutic approaches.
Collapse
Affiliation(s)
- Raphaël Itzykson
- Inserm UMR 1009, Institut Gustave Roussy 114, Rue Edouard Vaillant, 94805 Villejuif, France
- IFR54 (IRCIV), Institut Gustave Roussy, Villejuif, France
- Faculty of Medicine, University Paris-Sud, Le Kremlin-Bicêtre, France
| | - Nathalie Droin
- Inserm UMR 1009, Institut Gustave Roussy 114, Rue Edouard Vaillant, 94805 Villejuif, France
- IFR54 (IRCIV), Institut Gustave Roussy, Villejuif, France
- Faculty of Medicine, University Paris-Sud, Le Kremlin-Bicêtre, France
| | - Eric Solary
- Inserm UMR 1009, Institut Gustave Roussy 114, Rue Edouard Vaillant, 94805 Villejuif, France
| |
Collapse
|
94
|
Flt3 inhibitor AC220 is a potent therapy in a mouse model of myeloproliferative disease driven by enhanced wild-type Flt3 signaling. Blood 2012; 120:4049-57. [DOI: 10.1182/blood-2012-06-436675] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Abstract
High levels of expression of wild-type Flt3 characterize many hematopoietic proliferative diseases and neoplasms, providing a potential therapeutic target. Using the c-Cbl RING finger mutant mouse as a model of a myeloproliferative disease (MPD) driven by wild-type Flt3, in the present study, we show that treatment with the Flt3 kinase inhibitor AC220 blocks MPD development by targeting Flt3+ multipotent progenitors (MPPs). We found that daily administration of AC220 caused a marked reduction in Flt3 expression, induction of quiescence, and a significant loss of MPPs within 4 days. Unexpectedly, a robust Flt3 ligand–associated proliferative recovery response soon followed, preventing further loss of MPPs. However, continued AC220 treatment limited MPP recovery and maintained reduced, steady-state levels of cycling MPPs that express low levels of Flt3. Therefore, a finely tuned balance between the opposing forces of AC220 and Flt3 ligand production was established; whereas the Flt3 ligand blunted the inhibitory effects of AC220, the disease was held in remission for as long as therapy was continued. The net effect is a potent therapy indicating that patients with c-Cbl mutations, or those with similarly enhanced Flt3 signaling, may respond well to AC220 even after the induction of high levels of Flt3 ligand.
Collapse
|
95
|
Loh ML, Mullighan CG. Advances in the genetics of high-risk childhood B-progenitor acute lymphoblastic leukemia and juvenile myelomonocytic leukemia: implications for therapy. Clin Cancer Res 2012; 18:2754-67. [PMID: 22589484 DOI: 10.1158/1078-0432.ccr-11-1936] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hematologic malignancies of childhood comprise the most common childhood cancers. These neoplasms derive from the pathologic clonal expansion of an abnormal cancer-initiating cell and span a diverse spectrum of phenotypes, including acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), myeloproliferative neoplasms (MPN), and myelodysplastic syndromes (MDS). Expansion of immature lymphoid or myeloid blasts with suppression of normal hematopoiesis is the hallmark of ALL and AML, whereas MPN is associated with proliferation of 1 or more lineages that retain the ability to differentiate, and MDS is characterized by abnormal hematopoiesis and cytopenias. The outcomes for children with the most common childhood cancer, B-progenitor ALL (B-ALL), in general, is quite favorable, in contrast to children affected by myeloid malignancies. The advent of highly sensitive genomic technologies reveals the remarkable genetic complexity of multiple subsets of high-risk B-progenitor ALL, in contrast to a somewhat simpler model of myeloid neoplasms, although a number of recently discovered alterations displayed by both types of malignancies may lead to common therapeutic approaches. This review outlines recent advances in our understanding of the genetic underpinnings of high-risk B-ALL and juvenile myelomonocytic leukemia, an overlap MPN/MDS found exclusively in children, and we also discuss novel therapeutic approaches that are currently being tested in clinical trials. Recent insights into the clonal heterogeneity of leukemic samples and the implications for diagnostic and therapeutic approaches are also discussed.
Collapse
Affiliation(s)
- Mignon L Loh
- Department of Pediatrics and the Helen Diller Comprehensive Cancer Center, Benioff Children's Hospital, University of California, San Francisco, San Francisco, California, USA
| | | |
Collapse
|
96
|
Mohapatra B, Ahmad G, Nadeau S, Zutshi N, An W, Scheffe S, Dong L, Feng D, Goetz B, Arya P, Bailey TA, Palermo N, Borgstahl GEO, Natarajan A, Raja SM, Naramura M, Band V, Band H. Protein tyrosine kinase regulation by ubiquitination: critical roles of Cbl-family ubiquitin ligases. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1833:122-39. [PMID: 23085373 DOI: 10.1016/j.bbamcr.2012.10.010] [Citation(s) in RCA: 171] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 10/05/2012] [Accepted: 10/08/2012] [Indexed: 12/20/2022]
Abstract
Protein tyrosine kinases (PTKs) coordinate a broad spectrum of cellular responses to extracellular stimuli and cell-cell interactions during development, tissue homeostasis, and responses to environmental challenges. Thus, an understanding of the regulatory mechanisms that ensure physiological PTK function and potential aberrations of these regulatory processes during diseases such as cancer are of broad interest in biology and medicine. Aside from the expected role of phospho-tyrosine phosphatases, recent studies have revealed a critical role of covalent modification of activated PTKs with ubiquitin as a critical mechanism of their negative regulation. Members of the Cbl protein family (Cbl, Cbl-b and Cbl-c in mammals) have emerged as dominant "activated PTK-selective" ubiquitin ligases. Structural, biochemical and cell biological studies have established that Cbl protein-dependent ubiquitination targets activated PTKs for degradation either by facilitating their endocytic sorting into lysosomes or by promoting their proteasomal degradation. This mechanism also targets PTK signaling intermediates that become associated with Cbl proteins in a PTK activation-dependent manner. Cellular and animal studies have established that the relatively broadly expressed mammalian Cbl family members Cbl and Cbl-b play key physiological roles, including their critical functions to prevent the transition of normal immune responses into autoimmune disease and as tumor suppressors; the latter function has received validation from human studies linking mutations in Cbl to human leukemia. These newer insights together with embryonic lethality seen in mice with a combined deletion of Cbl and Cbl-b genes suggest an unappreciated role of the Cbl family proteins, and by implication the ubiquitin-dependent control of activated PTKs, in stem/progenitor cell maintenance. Future studies of existing and emerging animal models and their various cell lineages should help test the broader implications of the evolutionarily-conserved Cbl family protein-mediated, ubiquitin-dependent, negative regulation of activated PTKs in physiology and disease.
Collapse
Affiliation(s)
- Bhopal Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Mohapatra B, Ahmad G, Nadeau S, Zutshi N, An W, Scheffe S, Dong L, Feng D, Goetz B, Arya P, Bailey TA, Palermo N, Borgstahl GEO, Natarajan A, Raja SM, Naramura M, Band V, Band H. Protein tyrosine kinase regulation by ubiquitination: critical roles of Cbl-family ubiquitin ligases. BIOCHIMICA ET BIOPHYSICA ACTA 2012. [PMID: 23085373 DOI: 10.1016/j.bbamcr] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Protein tyrosine kinases (PTKs) coordinate a broad spectrum of cellular responses to extracellular stimuli and cell-cell interactions during development, tissue homeostasis, and responses to environmental challenges. Thus, an understanding of the regulatory mechanisms that ensure physiological PTK function and potential aberrations of these regulatory processes during diseases such as cancer are of broad interest in biology and medicine. Aside from the expected role of phospho-tyrosine phosphatases, recent studies have revealed a critical role of covalent modification of activated PTKs with ubiquitin as a critical mechanism of their negative regulation. Members of the Cbl protein family (Cbl, Cbl-b and Cbl-c in mammals) have emerged as dominant "activated PTK-selective" ubiquitin ligases. Structural, biochemical and cell biological studies have established that Cbl protein-dependent ubiquitination targets activated PTKs for degradation either by facilitating their endocytic sorting into lysosomes or by promoting their proteasomal degradation. This mechanism also targets PTK signaling intermediates that become associated with Cbl proteins in a PTK activation-dependent manner. Cellular and animal studies have established that the relatively broadly expressed mammalian Cbl family members Cbl and Cbl-b play key physiological roles, including their critical functions to prevent the transition of normal immune responses into autoimmune disease and as tumor suppressors; the latter function has received validation from human studies linking mutations in Cbl to human leukemia. These newer insights together with embryonic lethality seen in mice with a combined deletion of Cbl and Cbl-b genes suggest an unappreciated role of the Cbl family proteins, and by implication the ubiquitin-dependent control of activated PTKs, in stem/progenitor cell maintenance. Future studies of existing and emerging animal models and their various cell lineages should help test the broader implications of the evolutionarily-conserved Cbl family protein-mediated, ubiquitin-dependent, negative regulation of activated PTKs in physiology and disease.
Collapse
Affiliation(s)
- Bhopal Mohapatra
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Coenen EA, Driessen EMC, Zwaan CM, Stary J, Baruchel A, de Haas V, de Bont ESJM, Reinhardt D, Kaspers GJL, Arentsen-Peters STCJM, Meyer C, Marschalek R, Pieters R, Stam RW, van den Heuvel-Eibrink MM. CBL mutations do not frequently occur in paediatric acute myeloid leukaemia. Br J Haematol 2012; 159:577-84. [PMID: 23025505 DOI: 10.1111/bjh.12068] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 08/27/2012] [Indexed: 11/26/2022]
Abstract
RAS-pathway mutations, causing a proliferative advantage, occur in acute myeloid leukaemia (AML) and MLL-rearranged leukaemia. Recently, mutations in the Casitas B lineage lymphoma (CBL) gene were reported to be involved in RAS-pathway activation in various myeloid malignancies, but their role in paediatric AML is still unknown. We performed mutation analysis of 283 newly diagnosed and 33 relapsed paediatric AML cases. Only two mutant cases (0·7%) were identified in the newly diagnosed paediatric AML samples, of which one was MLL-rearranged. Both mutant cases showed CBL mRNA expression in the range of the non-mutated cases. Phosphorylated extracellular signal-regulated kinase (pERK) was not correlated with CBL protein expression (n = 11). In conclusion, we report a very low CBL mutation frequency in paediatric AML, which, together with the lack of difference in protein and mRNA expression, illustrates the limited role of CBL in paediatric AML.
Collapse
Affiliation(s)
- Eva A Coenen
- Department of Paediatric Oncology/Haematology, Erasmus MC/Sophia Children's Hospital, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Current Outlook on Molecular Pathogenesis and Treatment of Myeloproliferative Neoplasms. Mol Diagn Ther 2012; 16:269-83. [DOI: 10.1007/s40291-012-0006-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
100
|
Tibes R, Bogenberger JM, Geyer HL, Mesa RA. JAK2 inhibitors in the treatment of myeloproliferative neoplasms. Expert Opin Investig Drugs 2012; 21:1755-74. [PMID: 22991927 DOI: 10.1517/13543784.2012.721352] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Dysregulation of JAK-STAT signaling is a pathogenetic hallmark of myeloproliferative neoplasms (MPNs) arising from several distinct molecular aberrations, including mutations in JAK2, the thrombopoietin receptor (MPL), mutations in negative regulators of JAK-STAT signaling, such as lymphocyte-specific adapter protein (SH2B3), and epigenetic dysregulation as seen with Suppressor of Cytokine Signaling (SOCS) proteins. In addition, growth factor/cytokine stimulatory events activate JAK-STAT signaling independent of mutations. AREAS COVERED The various mutations and molecular events activating JAK-STAT signaling in MPNs are reviewed. Detailed inhibitory kinase profiles of the currently developed JAK inhibitors are presented. Clinical trial results for currently developed JAK targeting agents are comprehensively summarized. The limitations of JAK-STAT targeting in MPNs, as well as potential rational combination therapies with JAK2 inhibitors, are discussed. EXPERT OPINION Aberrant JAK-STAT signaling is an underlying theme in the pathogenesis of MPNs. While JAK2 inhibitors are active in JAK2V617F and wild-type JAK2 MPNs, JAK2V617F mutation-specific or JAK2-selective inhibitors may possess unique clinical attributes. Complimentary targeting of parallel pathways operating in MPNs may offer novel therapeutic approaches in combination with JAK inhibition. Understanding the intricacies of JAK-STAT pathway activation, including growth factor/cytokine-driven signaling, will open new avenues for therapeutic intervention at known and novel molecular vulnerabilities of MPNs.
Collapse
Affiliation(s)
- Raoul Tibes
- Mayo Clinic, Hematology, 200 First Street SW, Rochester, MN 55905, USA.
| | | | | | | |
Collapse
|