451
|
Bose P, Verstovsek S. Mutational profiling in myelofibrosis: implications for management. Int J Hematol 2020; 111:192-199. [PMID: 31630335 PMCID: PMC11951241 DOI: 10.1007/s12185-019-02758-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 09/27/2019] [Indexed: 02/06/2023]
Abstract
Mutational profiling, usually by targeted next-generation sequencing, is increasingly performed on patients with myeloproliferative neoplasm-associated myelofibrosis (MF), whether primary (PMF) or post-polycythemia vera/essential thrombocythemia (post-PV/ET MF). "Driver" mutations in JAK2, MPL and indels in CALR underlie the vast majority of cases of PMF and post-ET MF; the remainder (≈ 10%) lack identifiable driver mutations, but other clonal markers are usually detectable. Nearly all patients with post-PV MF carry activating JAK2 mutations. In both PMF and post-ET MF, type 1/-like CALR mutations confer a favorable prognosis. Since both type 1/-like and type 2/-like CALR mutations have essentially the same functional consequence, this is a subject of intense research. Additional, "non-driver" mutations, mostly affecting genes encoding epigenetic modifiers or spliceosome components, e.g., ASXL1, EZH2, TET2, DNMT3A, SRSF2 and U2AF1, are frequently found; some of these are associated with inferior survival and have been incorporated into prognostic models. Some mutations, e.g., IDH1/2, are relatively infrequent in chronic phase but are substantially more common in blast phase, and are now therapeutically targetable. While mutational information does not currently influence choice of drug therapy in chronic-phase MF, the presence of a "high molecular risk" genotype is now routinely taken into account for transplant decision-making.
Collapse
Affiliation(s)
- Prithviraj Bose
- Department of Leukemia, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 428, Houston, TX, 77030, USA
| | - Srdan Verstovsek
- Department of Leukemia, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 428, Houston, TX, 77030, USA.
| |
Collapse
|
452
|
Zhang Y, Zhou Y, Wang Y, Teng G, Li D, Wang Y, Du C, Chen Y, Zhang H, Li Y, Fu L, Chen K, Bai J. Thrombosis among 1537 patients with JAK2 V617F -mutated myeloproliferative neoplasms: Risk factors and development of a predictive model. Cancer Med 2020; 9:2096-2105. [PMID: 31994332 PMCID: PMC7064115 DOI: 10.1002/cam4.2886] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/20/2019] [Accepted: 01/14/2020] [Indexed: 12/11/2022] Open
Abstract
To explore the risk factors of thrombosis in patients with JAK2V617F‐mutated myeloproliferative neoplasms (MPNs), a cohort of 1537 Chinese patients with JAK2V617F‐mutated MPN was retrospectively analyzed. The Kaplan‐Meier method and multivariate Cox analysis were used to study the risk factors of thrombosis in patients with JAK2V617F‐mutated MPN. Among the 1537 MPN patients, 931, 468, and 138 had polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), respectively. The median follow‐up time was 7 years (range 1‐47), and 12.8% of patients (197/1537) died during this period. A total of 16.8% (259/1399) of PV and ET patients had secondary myelofibrosis, and 2.5% (38/1537) of patients developed acute myeloid leukemia (AML). Thrombotic events occurred in 43.9% (675/1537) of patients, among which 91.4% (617/675) were arterial thrombosis and 16.6% (112/675) were venous thrombosis. The number of thrombotic events in PV, ET, and PMF patients was 439 (47.2%), 197 (42.1%) and 39 (28.2%), respectively. The multivariate analysis indicated that age ≥60 years old, HCT ≥48%, at least one cardiovascular risk factor, a history of thrombosis, and JAK2V617F allele burden (V617F%) ≥50% are risk factors for thrombosis in JAK2V617F‐mutated MPN. According to the results of the multivariate analysis, a risk model of thrombosis was established and comprised low‐risk (0 points), intermediate‐risk (1 points) and high‐risk (≥2 points) groups, among which the incidence of thrombosis was 9.1%, 33.7% and 72.9%. For elderly patients with JAK2V617F‐mutated MPN and a history of thrombosis, reducing the V617F%, controlling HCT and preventing cardiovascular risk factors are necessary measures to prevent thrombosis.
Collapse
Affiliation(s)
- Yuhui Zhang
- Department of Hematology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yuan Zhou
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yingshao Wang
- Department of Hematology, The Second Hospital of Tianjin Medical University, Tianjin, China.,State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Guangshuai Teng
- Department of Hematology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Dapeng Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yan Wang
- Department of Hematology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Chenxiao Du
- Department of Hematology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yafang Chen
- Department of Hematology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Huiqin Zhang
- Department of Hematology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yanqi Li
- Department of Hematology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Lixia Fu
- Department of Hematology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Kangyin Chen
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jie Bai
- Department of Hematology, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
453
|
Pedersen RK, Andersen M, Knudsen TA, Sajid Z, Gudmand-Hoeyer J, Dam MJB, Skov V, Kjaer L, Ellervik C, Larsen TS, Hansen D, Pallisgaard N, Hasselbalch HC, Ottesen JT. Data-driven analysis of JAK2V617F kinetics during interferon-alpha2 treatment of patients with polycythemia vera and related neoplasms. Cancer Med 2020; 9:2039-2051. [PMID: 31991066 PMCID: PMC7064092 DOI: 10.1002/cam4.2741] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 12/22/2022] Open
Abstract
Treatment with PEGylated interferon-alpha2 (IFN) of patients with essential thrombocythemia and polycythemia vera induces major molecular remissions with a reduction in the JAK2V617F allele burden to undetectable levels in a subset of patients. A favorable response to IFN has been argued to depend upon the tumor burden, implying that institution of treatment with IFN should be as early as possible after the diagnosis. However, evidence for this statement is not available. We present a thorough analysis of unique serial JAK2V617F measurements in 66 IFN-treated patients and in 6 untreated patients. Without IFN treatment, the JAK2V617F allele burden increased exponentially with a period of doubling of 1.4 year. During monotherapy with IFN, the JAK2V617F allele burden decreased mono- or bi-exponentially for 33 responders of which 28 patients satisfied both descriptions. Bi-exponential description improved the fits in 19 cases being associated with late JAK2V617F responses. The decay of the JAK2V617F allele burden during IFN treatment was estimated to have half-lives of 1.6 year for the monoexponential response and 1.0 year in the long term for the bi-exponential response. In conclusion, through data-driven analysis of the JAK2V617F allele burden, we provide novel information regarding the JAK2V617F kinetics during IFN-treatment, arguing for early intervention.
Collapse
Affiliation(s)
- Rasmus K Pedersen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Morten Andersen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Trine A Knudsen
- Department of Haematology, Zealand University Hospital, Roskilde, Denmark
| | - Zamra Sajid
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | | | - Marc J B Dam
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Vibe Skov
- Department of Haematology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjaer
- Department of Haematology, Zealand University Hospital, Roskilde, Denmark
| | - Christina Ellervik
- Department of Production, Research, and Innovation, Region Zealand, Sorø, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pathology, Harvard Medical School, Boston, FL, USA.,Department of Laboratory Medicine, Boston Children's Hospital, Boston, FL, USA
| | - Thomas S Larsen
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Dennis Hansen
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Niels Pallisgaard
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Hans C Hasselbalch
- Department of Haematology, Zealand University Hospital, Roskilde, Denmark
| | - Johnny T Ottesen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| |
Collapse
|
454
|
Fenerich BA, Fernandes JC, Rodrigues Alves APN, Coelho-Silva JL, Scopim-Ribeiro R, Scheucher PS, Eide CA, Tognon CE, Druker BJ, Rego EM, Machado-Neto JA, Traina F. NT157 has antineoplastic effects and inhibits IRS1/2 and STAT3/5 in JAK2 V617F-positive myeloproliferative neoplasm cells. Signal Transduct Target Ther 2020; 5:5. [PMID: 32296029 PMCID: PMC6978524 DOI: 10.1038/s41392-019-0102-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 11/15/2019] [Accepted: 11/24/2019] [Indexed: 12/25/2022] Open
Abstract
Recent data indicate that IGF1R/IRS signaling is a potential therapeutic target in BCR-ABL1-negative myeloproliferative neoplasms (MPN); in this pathway, IRS2 is involved in the malignant transformation induced by JAK2V617F, and upregulation of IGF1R signaling induces the MPN phenotype. NT157, a synthetic compound designed as an IGF1R-IRS1/2 inhibitor, has been shown to induce antineoplastic effects in solid tumors. Herein, we aimed to characterize the molecular and cellular effects of NT157 in JAK2V617F-positive MPN cell lines (HEL and SET2) and primary patient hematopoietic cells. In JAK2V617F cell lines, NT157 decreased cell viability, clonogenicity, and cell proliferation, resulting in increases in apoptosis and cell cycle arrest in the G2/M phase (p < 0.05). NT157 treatment inhibited IRS1/2, JAK2/STAT, and NFκB signaling, and it activated the AP-1 complex, downregulated four oncogenes (CCND1, MYB, WT1, and NFKB1), and upregulated three apoptotic-related genes (CDKN1A, FOS, and JUN) (p < 0.05). NT157 induced genotoxic stress in a JAK2/STAT-independent manner. NT157 inhibited erythropoietin-independent colony formation in cells from polycythemia vera patients (p < 0.05). These findings further elucidate the mechanism of NT157 action in a MPN context and suggest that targeting IRS1/2 proteins may represent a promising therapeutic strategy for MPN.
Collapse
Affiliation(s)
- Bruna Alves Fenerich
- Department of Medical Images, Hematology, and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
- Center for Cell-Based Therapy, Sao Paulo Research Foundation, Ribeirão Preto, São Paulo, Brazil
| | - Jaqueline Cristina Fernandes
- Department of Medical Images, Hematology, and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
- Center for Cell-Based Therapy, Sao Paulo Research Foundation, Ribeirão Preto, São Paulo, Brazil
| | - Ana Paula Nunes Rodrigues Alves
- Department of Medical Images, Hematology, and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
- Center for Cell-Based Therapy, Sao Paulo Research Foundation, Ribeirão Preto, São Paulo, Brazil
| | - Juan Luiz Coelho-Silva
- Department of Medical Images, Hematology, and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
- Center for Cell-Based Therapy, Sao Paulo Research Foundation, Ribeirão Preto, São Paulo, Brazil
| | - Renata Scopim-Ribeiro
- Department of Medical Images, Hematology, and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
- Center for Cell-Based Therapy, Sao Paulo Research Foundation, Ribeirão Preto, São Paulo, Brazil
| | - Priscila Santos Scheucher
- Department of Medical Images, Hematology, and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
| | - Christopher A Eide
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Howard Hughes Medical Institute, Portland, OR, USA
| | - Cristina E Tognon
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Howard Hughes Medical Institute, Portland, OR, USA
| | - Brian J Druker
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Howard Hughes Medical Institute, Portland, OR, USA
| | - Eduardo Magalhães Rego
- Department of Medical Images, Hematology, and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
- Center for Cell-Based Therapy, Sao Paulo Research Foundation, Ribeirão Preto, São Paulo, Brazil
- Department of Internal Medicine, University of São Paulo Medical School, São Paulo, Brazil
| | - João Agostinho Machado-Neto
- Department of Medical Images, Hematology, and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil
- Department of Pharmacology, Institute of Biomedical Sciences of the University of São Paulo, São Paulo, Brazil
| | - Fabiola Traina
- Department of Medical Images, Hematology, and Clinical Oncology, University of São Paulo at Ribeirão Preto Medical School, Ribeirão Preto, São Paulo, Brazil.
- Center for Cell-Based Therapy, Sao Paulo Research Foundation, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
455
|
Aguilera-Diaz A, Vazquez I, Ariceta B, Mañú A, Blasco-Iturri Z, Palomino-Echeverría S, Larrayoz MJ, García-Sanz R, Prieto-Conde MI, del Carmen Chillón M, Alfonso-Pierola A, Prosper F, Fernandez-Mercado M, Calasanz MJ. Assessment of the clinical utility of four NGS panels in myeloid malignancies. Suggestions for NGS panel choice or design. PLoS One 2020; 15:e0227986. [PMID: 31978184 PMCID: PMC6980571 DOI: 10.1371/journal.pone.0227986] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 01/04/2020] [Indexed: 12/17/2022] Open
Abstract
The diagnosis of myeloid neoplasms (MN) has significantly evolved through the last few decades. Next Generation Sequencing (NGS) is gradually becoming an essential tool to help clinicians with disease management. To this end, most specialized genetic laboratories have implemented NGS panels targeting a number of different genes relevant to MN. The aim of the present study is to evaluate the performance of four different targeted NGS gene panels based on their technical features and clinical utility. A total of 32 patient bone marrow samples were accrued and sequenced with 3 commercially available panels and 1 custom panel. Variants were classified by two geneticists based on their clinical relevance in MN. There was a difference in panel’s depth of coverage. We found 11 discordant clinically relevant variants between panels, with a trend to miss long insertions. Our data show that there is a high risk of finding different mutations depending on the panel of choice, due both to the panel design and the data analysis method. Of note, CEBPA, CALR and FLT3 genes, remains challenging the use of NGS for diagnosis of MN in compliance with current guidelines. Therefore, conventional molecular testing might need to be kept in place for the correct diagnosis of MN for now.
Collapse
Affiliation(s)
- Almudena Aguilera-Diaz
- Advanced Genomics Laboratory, Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Iria Vazquez
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
| | - Beñat Ariceta
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
| | - Amagoia Mañú
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
| | - Zuriñe Blasco-Iturri
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
| | | | - María José Larrayoz
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
| | - Ramón García-Sanz
- Hematology Department, University Hospital of Salamanca, IBSAL and CIBERONC, Salamanca, Spain
| | | | | | - Ana Alfonso-Pierola
- Hematology Department, Clinica Universidad de Navarra (CUN), Pamplona, Spain
| | - Felipe Prosper
- Advanced Genomics Laboratory, Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Hematology Department, Clinica Universidad de Navarra (CUN), Pamplona, Spain
| | - Marta Fernandez-Mercado
- Advanced Genomics Laboratory, Hemato-Oncology, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
- Biomedical Engineering Department, School of Engineering, University of Navarra, San Sebastian, Spain
- * E-mail: ,
| | - María José Calasanz
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Hematological Diseases Laboratory, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
- Scientific Co-Director of CIMA LAB Diagnostics, CIMA LAB Diagnostics, University of Navarra, Pamplona, Spain
| |
Collapse
|
456
|
Fabre MA, McKerrell T, Zwiebel M, Vijayabaskar MS, Park N, Wells PM, Rad R, Deloukas P, Small K, Steves CJ, Vassiliou GS. Concordance for clonal hematopoiesis is limited in elderly twins. Blood 2020; 135:269-273. [PMID: 31697828 PMCID: PMC6978156 DOI: 10.1182/blood.2019001807] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 10/06/2019] [Indexed: 12/14/2022] Open
Abstract
Although acquisition of leukemia-associated somatic mutations by 1 or more hematopoietic stem cells is inevitable with advancing age, its consequences are highly variable, ranging from clinically silent clonal hematopoiesis (CH) to leukemic progression. To investigate the influence of heritable factors on CH, we performed deep targeted sequencing of blood DNA from 52 monozygotic (MZ) and 27 dizygotic (DZ) twin pairs (aged 70-99 years). Using this highly sensitive approach, we identified CH (variant allele frequency ≥0.5%) in 62% of individuals. We did not observe higher concordance for CH within MZ twin pairs as compared with that within DZ twin pairs, or to that expected by chance. However, we did identify 2 MZ pairs in which both twins harbored identical rare somatic mutations, suggesting a shared cell of origin. Finally, in 3 MZ twin pairs harboring mutations in the same driver genes, serial blood samples taken 4 to 5 years apart showed substantial twin-to-twin variability in clonal trajectories. Our findings propose that the inherited genome does not exert a dominant influence on the behavior of adult CH and provide evidence that CH mutations may be acquired in utero.
Collapse
Affiliation(s)
- Margarete A Fabre
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, Cambridge University Hospitals National Health Service (NHS) Trust, Cambridge, United Kingdom
| | - Thomas McKerrell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, Cambridge University Hospitals National Health Service (NHS) Trust, Cambridge, United Kingdom
| | - Maximillian Zwiebel
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- German Consortium for Translational Cancer Research (DKTK), Partnering Site Munich, Munich, Germany
| | - M S Vijayabaskar
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, Cambridge, United Kingdom
| | - Naomi Park
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Philippa M Wells
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, United Kingdom; and
| | - Roland Rad
- German Consortium for Translational Cancer Research (DKTK), Partnering Site Munich, Munich, Germany
| | - Panagiotis Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Kerrin Small
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, United Kingdom; and
| | - Claire J Steves
- Department of Twin Research and Genetic Epidemiology, School of Life Course Sciences, King's College London, London, United Kingdom; and
| | - George S Vassiliou
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, Cambridge, United Kingdom
- Department of Haematology, Cambridge University Hospitals National Health Service (NHS) Trust, Cambridge, United Kingdom
| |
Collapse
|
457
|
Yoon SY, Jeong SY, Kim C, Lee MY, Kim J, Kim KH, Lee N, Won JH. Philadelphia+ Chronic Myeloid Leukemia with CALR Mutation: A Case Report and Literature Review. Cancer Res Treat 2020; 52:987-991. [PMID: 32019282 PMCID: PMC7373853 DOI: 10.4143/crt.2019.703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/13/2020] [Indexed: 01/12/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are classified as chronic myeloid leukemia (CML) and Philadelphia chromosome-negative MPN. In MPN cases, the presence of a BCR-ABL1 translocation with a coexisting mutation is exceptionally rare. Herein, we report the first documented patient with CML harboring CALR mutation in Korea. A 33-year-old woman was referred to our hospital in February 2015 with splenomegaly, leukocytosis, and thrombocytosis. She was diagnosed with CML and started receiving nilotinib. In October 2015, a major molecular response was observed, but thrombocytosis persisted. A repeat bone marrow (BM) examination revealed no specific findings. However, as thrombocytosis worsened, we changed nilotinib to dasatinib. In May 2019, owing to persistent thrombocytosis, we repeated the BM examination and found CALR mutation (15.97%) on the MPN–next generation sequencing (NGS) test. We then retrospectively performed repeat MPN-NGS testing using the BM aspirate sample obtained in 2015 and found CALR mutation (10.64%).
Collapse
Affiliation(s)
- Seug Yun Yoon
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Sun Young Jeong
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Changgon Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Min-Young Lee
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Jieun Kim
- Department of Laboratory Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Kyoung-Ha Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Namsu Lee
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Jong-Ho Won
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Korea
| |
Collapse
|
458
|
Mutant Calreticulin in the Myeloproliferative Neoplasms. Hemasphere 2020; 4:e333. [PMID: 32382708 PMCID: PMC7000472 DOI: 10.1097/hs9.0000000000000333] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/03/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022] Open
Abstract
Mutations in the gene for calreticulin (CALR) were identified in the myeloproliferative neoplasms (MPNs) essential thrombocythaemia (ET) and primary myelofibrosis (MF) in 2013; in combination with previously described mutations in JAK2 and MPL, driver mutations have now been described for the majority of MPN patients. In subsequent years, researchers have begun to unravel the mechanisms by which mutant CALR drives transformation and to understand their clinical implications. Mutant CALR activates the thrombopoietin receptor (MPL), causing constitutive activation of Janus kinase 2 (JAK2) signaling and cytokine independent growth in vitro. Mouse models show increased numbers of hematopoietic stem cells (HSCs) and overproduction of megakaryocytic lineage cells with associated thrombocytosis. In the clinic, detection of CALR mutations has been embedded in World Health Organization and other international diagnostic guidelines. Distinct clinical and laboratory associations of CALR mutations have been identified together with their prognostic significance, with CALR mutant patients showing increased overall survival. The discovery and subsequent study of CALR mutations have illuminated novel aspects of megakaryopoiesis and raised the possibility of new therapeutic approaches.
Collapse
|
459
|
Fujino T, Kitamura T. ASXL1 mutation in clonal hematopoiesis. Exp Hematol 2020; 83:74-84. [PMID: 31945396 DOI: 10.1016/j.exphem.2020.01.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/04/2020] [Accepted: 01/06/2020] [Indexed: 12/13/2022]
Abstract
Recent advances in DNA sequencing technologies have enhanced our knowledge about several diseases. Coupled with easy accessibility to blood samples, hematology plays a leading role in understanding the process of carcinogenesis. Clonal hematopoiesis (CH) with somatic mutations is observed in at least 10% of people over 65 years of age, without apparent hematologic disorders. CH is associated with increased risk of hematologic malignancies, which is indicative of a pre-malignant condition. Therefore, a better understanding of CH will help elucidate the mechanism of multi-step tumorigenesis in the hematopoietic system. Somatic mutations of ASXL1 are frequently detected in CH and myeloid malignancies. Although ASXL1 does not have any catalytic activity, it is involved in multiple histone modifications including H3K4me3, H3K27me3, and H2AK119Ub, suggesting its function as a scaffolding protein. Most ASXL1 mutations detected in CH and myeloid malignancies are frameshift or nonsense mutations of the last exon, generating a C-terminally truncated protein. Deletion of Asxl1 or expression of mutant ASXL1 in mice alters histone modifications and facilitates aberrant gene expression, resulting in myeloid transformation. On the contrary, these mice exhibit impaired functioning of hematopoietic stem cells (HSCs), suggesting the negative effects of ASXL1 mutations on stem cell function. Thus, how ASXL1 mutations induce a clonal advantage of hematopoietic cells and subsequent CH development has not been elucidated. Here, we have reviewed the current literature that enhances our understanding of ASXL1, including its mutational landscape, function, and involvement of its mutation in pathogenesis of CH and myeloid malignancies. Finally, we discuss the potential causes of CH harboring ASXL1 mutations with our latest knowledge.
Collapse
Affiliation(s)
- Takeshi Fujino
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Toshio Kitamura
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan.
| |
Collapse
|
460
|
Takenaka K. Progress in elucidation of molecular pathophysiology and its application in therapeutic decision-making for myeloproliferative neoplasms. Int J Hematol 2020; 111:180-181. [PMID: 31907735 DOI: 10.1007/s12185-019-02812-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 12/25/2019] [Indexed: 01/05/2023]
Affiliation(s)
- Katsuto Takenaka
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, 791-0295, Japan.
| |
Collapse
|
461
|
Holmström MO, Cordua S, Skov V, Kjær L, Pallisgaard N, Ellervik C, Hasselbalch HC, Andersen MH. Evidence of immune elimination, immuno-editing and immune escape in patients with hematological cancer. Cancer Immunol Immunother 2020; 69:315-324. [PMID: 31915854 DOI: 10.1007/s00262-019-02473-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/31/2019] [Indexed: 10/25/2022]
Abstract
There is mounting evidence that the immune system can spontaneously clear malignant lesions before they manifest as overt cancer, albeit this activity has been difficult to demonstrate in humans. The calreticulin (CALR) exon 9 mutations are driver mutations in patients with chronic myeloproliferative neoplasms (MPN), which are chronic blood cancers. The CALR mutations generate a neo-antigen that is recognized by patient T cells, and T cells isolated from a patient with a CALR-mutation can recognize and kill autologous CALR-mutant cells. Surprisingly, healthy individuals display frequent and strong T cell responses to the CALR neo-antigens too. Furthermore, healthy individuals display immune responses to all parts of the mutant CALR epitope, and the CALR neo-epitope specific responses are memory T cell responses. These data suggest that although healthy individuals might acquire a CALR mutation, the mutant cells can be eliminated by the immune system. Additionally, a small fraction of healthy individuals harbor a CALR exon 9 mutation. Four healthy individuals carrying CALR mutations underwent a full medical examination including a bone marrow biopsy after a median follow up of 6.2 years. None of these patients displayed any signs of CALR-mutant MPN. Additionally, all healthy individuals displayed strong CALR neo-epitope specific T cell responses suggesting that these healthy individuals retained their CALR-mutant cells in the editing stage for several years. Thus, we suggest that CALR-mutant MPN could be a disease model of cancer immuno-editing, as we have demonstrated that CALR-mutant MPN displays all three stages described in the theory of cancer immuno-editing.
Collapse
Affiliation(s)
- Morten Orebo Holmström
- National Center for Cancer Immune Therapy, Herlev Hospital, Borgmester Ib Juuls Vej 25C, 5. Sal, 2730, Herlev, Denmark.
| | - Sabrina Cordua
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Niels Pallisgaard
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Christina Ellervik
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Production, Research, and Innovation, Region Zealand, Sorø, Denmark.,Department of Laboratory Medicine, Harvard Medical School, Boston Children's Hospital, Boston, USA
| | | | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Herlev Hospital, Borgmester Ib Juuls Vej 25C, 5. Sal, 2730, Herlev, Denmark.,Institute for Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
462
|
Single-cell analysis based dissection of clonality in myelofibrosis. Nat Commun 2020; 11:73. [PMID: 31911629 PMCID: PMC6946829 DOI: 10.1038/s41467-019-13892-x] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 11/28/2019] [Indexed: 12/29/2022] Open
Abstract
Cancer development is an evolutionary genomic process with parallels to Darwinian selection. It requires acquisition of multiple somatic mutations that collectively cause a malignant phenotype and continuous clonal evolution is often linked to tumor progression. Here, we show the clonal evolution structure in 15 myelofibrosis (MF) patients while receiving treatment with JAK inhibitors (mean follow-up 3.9 years). Whole-exome sequencing at multiple time points reveal acquisition of somatic mutations and copy number aberrations over time. While JAK inhibition therapy does not seem to create a clear evolutionary bottleneck, we observe a more complex clonal architecture over time, and appearance of unrelated clones. Disease progression associates with increased genetic heterogeneity and gain of RAS/RTK pathway mutations. Clonal diversity results in clone-specific expansion within different myeloid cell lineages. Single-cell genotyping of circulating CD34 + progenitor cells allows the reconstruction of MF phylogeny demonstrating loss of heterozygosity and parallel evolution as recurrent events. Myelofibrosis is a myeloproliferative neoplasm. Here, the authors show the clonal evolution of myelofibrosis during JAK inhibitor therapy, revealing how the treatment results in an increase in clonal complexity and a gain of RAS pathway mutations.
Collapse
|
463
|
Modifiable Lifestyle and Medical Risk Factors Associated With Myeloproliferative Neoplasms. Hemasphere 2020; 4:e327. [PMID: 32072143 PMCID: PMC7000482 DOI: 10.1097/hs9.0000000000000327] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 11/17/2019] [Accepted: 11/19/2019] [Indexed: 11/30/2022] Open
Abstract
Despite the identification of acquired genetic mutations associated with Myeloproliferative Neoplasms (MPNs) there is a paucity of information relating to modifiable risk factors that may lead to these mutations. The MOSAICC Study was an exploratory case-control study of polycythemia vera (PV), essential thrombocythemia (ET), and Myelofibrosis (MF). MPN patients and population controls (identified by General Practitioners) and non-blood relative/friend controls were recruited from 2 large UK centers. Participants completed a telephone-based questionnaire analyzed by unconditional logistic regression analysis adjusting for potential confounders. Risk factors for MPNs identified included increasing childhood household density [odds ratio (OR) 2.55, 95% confidence interval (CI) 1.16–5.62], low childhood socioeconomic status (OR 2.30, 95%CI 1.02–5.18) and high pack years smoking (OR 2.19, 95%CI 1.03–4.66) and current smoking restricted to JAK2 positive PV cases (OR 3.73, 95%CI 1.06–13.15). Obesity was linked with ET (OR 2.59, 95%CI 1.02–6.58) confirming results in previous cohort studies. Receipt of multiple CT scans was associated with a strongly increased risk of MPN although with wide confidence intervals (OR 5.38, 95%CI 1.67–17.3). Alcohol intake was inversely associated with risk of PV (OR 0.41, 95%CI 0.19–0.92) and ET (OR 0.48, 95%CI 0.24–0.98). The associations with childhood household density, high pack years smoking and alcohol were also seen in multivariate analysis. This is the largest case control study in MPNs to date and confirms the previously reported associations with obesity and cigarette smoking from cohort studies in addition to novel associations. In particular, the role of smoking and JAK2 mutation cases merits further evaluation.
Collapse
|
464
|
Craver BM, Nguyen TK, Nguyen J, Nguyen H, Huynh C, Morse SJ, Fleischman AG. The SMAC mimetic LCL-161 selectively targets JAK2 V617F mutant cells. Exp Hematol Oncol 2020; 9:1. [PMID: 31908904 PMCID: PMC6941266 DOI: 10.1186/s40164-019-0157-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 12/24/2019] [Indexed: 12/19/2022] Open
Abstract
Background Evasion from programmed cell death is a hallmark of cancer and can be achieved in cancer cells by overexpression of inhibitor of apoptosis proteins (IAPs). Second mitochondria-derived activator of caspases (SMAC) directly bind to IAPs and promote apoptosis; thus, SMAC mimetics have been investigated in a variety of cancer types. particularly in diseases with high inflammation and NFĸB activation. Given that elevated TNFα levels and NFĸB activation is a characteristic feature of myeloproliferative neoplasms (MPN), we investigated the effect of the SMAC mimetic LCL-161 on MPN cell survival in vitro and disease development in vivo. Methods To investigate the effect of the SMAC mimetic LCL-161 in vitro, we utilized murine and human cell lines to perform cell viability assays as well as primary bone marrow from mice or humans with JAK2V617F-driven MPN to interrogate myeloid colony formation. To elucidate the effect of the SMAC mimetic LCL-161 in vivo, we treated a JAK2V617F-driven mouse model of MPN with LCL-161 then assessed blood counts, splenomegaly, and myelofibrosis. Results We found that JAK2V617F-mutated cells are hypersensitive to the SMAC mimetic LCL-161 in the absence of exogenous TNFα. JAK2 kinase activity and NFĸB activation is required for JAK2V617F-mediated sensitivity to LCL-161, as JAK or NFĸB inhibitors diminished the differential sensitivity of JAK2V617F mutant cells to IAP inhibition. Finally, LCL-161 reduces splenomegaly and may reduce fibrosis in a mouse model of JAK2V617F-driven MPN. Conclusion LCL-161 may be therapeutically useful in MPN, in particular when exogenous TNFα signaling is blocked. NFĸB activation is a characteristic feature of JAK2V617F mutant cells and this sensitizes them to SMAC mimetic induced killing even in the absence of TNFα. However, when exogenous TNFα is added, NFĸB is activated in both mutant and wild-type cells, abolishing the differential sensitivity. Moreover, JAK kinase activity is required for the differential sensitivity of JAK2V617F mutant cells, suggesting that the addition of JAK2 inhibitors to SMAC mimetics would detract from the ability of SMAC mimetics to selectively target JAK2V617F mutant cells. Instead, combination therapy with other agents that reduce inflammatory cytokines but preserve JAK2 signaling in mutant cells may be a more beneficial combination therapy in MPN.
Collapse
Affiliation(s)
- Brianna M Craver
- 1Department of Biological Chemistry, University of California, Irvine, CA USA
| | - Thanh Kim Nguyen
- 1Department of Biological Chemistry, University of California, Irvine, CA USA
| | - Jenny Nguyen
- 1Department of Biological Chemistry, University of California, Irvine, CA USA
| | - Hellen Nguyen
- 1Department of Biological Chemistry, University of California, Irvine, CA USA
| | - Christy Huynh
- 1Department of Biological Chemistry, University of California, Irvine, CA USA
| | - Sarah J Morse
- 1Department of Biological Chemistry, University of California, Irvine, CA USA
| | - Angela G Fleischman
- 1Department of Biological Chemistry, University of California, Irvine, CA USA.,2Division of Hematology/Oncology, Department of Medicine, University of California, 839 Health Sciences Road, Irvine, CA 92697 USA.,3Chao Family Comprehensive Cancer Center, University of California, Irvine, CA USA
| |
Collapse
|
465
|
Liu C, Hu R, Du Z, Abecasis M, Wang C. Atypical myeloproliferative neoplasm with concurrent BCR-ABL1 fusion and CALR mutation: A case report and literature review. Medicine (Baltimore) 2020; 99:e18811. [PMID: 32000382 PMCID: PMC7004640 DOI: 10.1097/md.0000000000018811] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
RATIONALE Concurrent calreticulin (CALR) mutation and BCR-ABL1 fusion are extremely rare in chronic myelogenous leukemia; to date, only 12 cases have been reported. PATIENT CONCERNS A 57-year-old male who had an 11-year history of essential thrombocytosis presented to our hospital with leukocytosis and marked splenomegaly for 3 months. DIAGNOSES Chronic myelogenous leukemia with myeloid fibrosis arising on the background of essential thrombocytosis harboring both BCR-ABL1 fusion and type-1 like CALR mutation. INTERVENTIONS Imatinib was started at 300 mg daily and increased to 400 mg daily after 3 months; interferon was added after 12 months. OUTCOMES Partial cytogenetic response was achieved after 3 months of imatinib therapy and complete cytogenetic response was achieved after 1 year of treatment. However, CALR mutation was still present with a stable mutational allele burden. LESSONS In this case report and review of additional 12 cases with simultaneous presence of CALR-mutation and BCR-ABL1 fusion, we highlighted the importance of integrating clinical, morphological, and molecular genetic data for classifying atypical myeloid neoplasms.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Calreticulin/genetics
- Fusion Proteins, bcr-abl/genetics
- Genes, abl
- Humans
- Imatinib Mesylate/pharmacology
- Imatinib Mesylate/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Male
- Middle Aged
- Protein-Tyrosine Kinases/antagonists & inhibitors
- Thrombocythemia, Essential/complications
- Thrombocythemia, Essential/genetics
Collapse
Affiliation(s)
- Chunshui Liu
- Department of Hematology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Ruiping Hu
- Department of Hematology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhonghua Du
- Department of Hematology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Manuel Abecasis
- Department of Hematology, Instituto Português de Oncologia de Francisco Gentil, Lisbon, Portugal
| | - Cong Wang
- Department of Hematology, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
466
|
Chronic Myeloid Neoplasms. Genomic Med 2020. [DOI: 10.1007/978-3-030-22922-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
467
|
Primignani M. Thrombophilia and Primary Budd–Chiari Syndrome. BUDD-CHIARI SYNDROME 2020:57-71. [DOI: 10.1007/978-981-32-9232-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
468
|
Jang MA, Choi CW. Recent insights regarding the molecular basis of myeloproliferative neoplasms. Korean J Intern Med 2020; 35:1-11. [PMID: 31778606 PMCID: PMC6960053 DOI: 10.3904/kjim.2019.317] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are a heterogeneous group of clonal disorders characterized by the overproduction of mature blood cells that have an increased risk of thrombosis and progression to acute myeloid leukemia. Next-generation sequencing studies have provided key insights regarding the molecular mechanisms of MPNs. MPN driver mutations in genes associated with the JAK-STAT pathway include JAK2 V617F, JAK2 exon 12 mutations and mutations in MPL, CALR, and CSF3R. Cooperating driver genes are also frequently detected and also mutated in other myeloid neoplasms; these driver genes are involved in epigenetic methylation, messenger RNA splicing, transcription regulation, and signal transduction. In addition, other genetic factors such as germline predisposition, order of mutation acquisition, and variant allele frequency also influence disease initiation and progression. This review summarizes the current understanding of the genetic basis of MPN, and demonstrates how molecular pathophysiology can improve both our understanding of MPN heterogeneity and clinical practice.
Collapse
Affiliation(s)
- Mi-Ae Jang
- Department of Laboratory Medicine and Genetics, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Chul Won Choi
- Division of Oncology and Hematology, Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
- Correspondence to Chul Won Choi, M.D. Division of Oncology and Hematology, Department of Internal Medicine, Korea University Guro Hospital, 148 Gurodong-ro, Guro-gu, Seoul 08308, Korea Tel: +82-2-2626-3058 Fax: +82-2-862-6453 E-mail:
| |
Collapse
|
469
|
Belcic Mikic T, Pajic T, Sever M. CALR mutations in a cohort of JAK2 V617F negative patients with suspected myeloproliferative neoplasms. Sci Rep 2019; 9:19838. [PMID: 31882869 PMCID: PMC6934448 DOI: 10.1038/s41598-019-56236-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/05/2019] [Indexed: 02/07/2023] Open
Abstract
Suspicion of myeloproliferative neoplasms (MPNs) and especially essential thrombocythemia (ET) in primary care is often based solely on blood counts, with patients referred to a haematologist without a thorough evaluation. We retrospectively assessed the role of calreticulin gene (CALR) mutations in the diagnosis of MPN in this population. We studied CALR mutations in 524 JAK2 V617F-negative patients with suspected MPN. Uncommon CALR mutations were confirmed by Sanger sequencing and searched for in the COSMIC or HGMD database. Mutations were defined as frameshift or non-frameshift mutations. CALR mutations were detected in 23 patients (23/524 = 4.4%). Four mutations detected in our study were newly identified mutations. Non-frameshift mutations were detected in two patients. Most patients (380/524 = 72.5%) were diagnosed with secondary conditions leading to blood count abnormalities such as iron deficiency, inflammatory and infectious diseases, malignancy and hyposplenism. Nine patients (9/23 = 39%) were retrospectively diagnosed with ET based on CALR mutation confirmation. Two patients with non-frameshift CALR mutations were diagnosed with reactive thrombocytosis and MPN unclassifiable, respectively. Our study showed that CALR mutations are important, non-invasive diagnostic indicators of ET and can aid in its diagnosis. Moreover, the type of CALR mutation must be accurately defined, as non-frameshift mutations may not be associated with ET. Finally, CALR mutation detection should be reserved for patients with high suspicion of clonal haematological disease.
Collapse
Affiliation(s)
- Tanja Belcic Mikic
- Department of Haematology, University Medical Centre Ljubljana, Zaloska 7, 1000, Ljubljana, Slovenia. .,Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000, Ljubljana, Slovenia.
| | - Tadej Pajic
- Department of Haematology, University Medical Centre Ljubljana, Zaloska 7, 1000, Ljubljana, Slovenia.,Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia
| | - Matjaz Sever
- Department of Haematology, University Medical Centre Ljubljana, Zaloska 7, 1000, Ljubljana, Slovenia.,Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000, Ljubljana, Slovenia
| |
Collapse
|
470
|
Shide K. The role of driver mutations in myeloproliferative neoplasms: insights from mouse models. Int J Hematol 2019; 111:206-216. [PMID: 31865539 DOI: 10.1007/s12185-019-02803-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 01/11/2023]
Abstract
High frequency of JAK2V617F or CALR exon 9 mutations is a main molecular feature of myeloproliferative neoplasms (MPNs). Analysis of mouse models driven by these mutations suggests that they are a direct cause of MPNs and that the expression levels of the mutated genes define the disease phenotype. The function of MPN-initiating cells has also been elucidated by these mouse models. Such mouse models also play an important role in modeling disease to investigate the effects and action mechanisms of therapeutic drugs, such as JAK2 inhibitors and interferon α, against MPNs. The mutation landscape of hematological tumors has already been clarified by next-generation sequencing technology, and the importance of functional analysis of mutant genes in vivo should increase further in the future.
Collapse
Affiliation(s)
- Kotaro Shide
- Department of Gastroenterology and Hematology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki, 889-1692, Japan.
| |
Collapse
|
471
|
Tetu M, El Hachami K, Marty C. [Thrombopoietin receptor and mutant calreticulin : the pathogenic interaction leading to myeloproliferative neoplasms]. Med Sci (Paris) 2019; 35:901-903. [PMID: 31845884 DOI: 10.1051/medsci/2019176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Maude Tetu
- M1 Biologie Santé, Université Paris-Saclay, 91405 Orsay, France
| | | | - Caroline Marty
- Inserm, UMR1170, Gustave Roussy, Villejuif, France - Université Paris-Saclay, UMR 1170, Gustave Roussy, Villejuif, France - Gustave Roussy, UMR 1170, Villejuif, France
| |
Collapse
|
472
|
The role of calreticulin mutations in myeloproliferative neoplasms. Int J Hematol 2019; 111:200-205. [DOI: 10.1007/s12185-019-02800-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022]
|
473
|
KDEL Receptors Are Differentially Regulated to Maintain the ER Proteome under Calcium Deficiency. Cell Rep 2019; 25:1829-1840.e6. [PMID: 30428351 DOI: 10.1016/j.celrep.2018.10.055] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/17/2018] [Accepted: 10/12/2018] [Indexed: 12/21/2022] Open
Abstract
Retention of critical endoplasmic reticulum (ER) luminal proteins needed to carry out diverse functions (e.g., protein synthesis and folding, lipid metabolism) is mediated through a carboxy-terminal ER retention sequence (ERS) and its interaction with KDEL receptors. Here, we demonstrate that depleting ER calcium causes mass departure of ERS-containing proteins from cells by overwhelming KDEL receptors. In addition, we provide evidence that KDELR2 and KDELR3, but not KDELR1, are unfolded protein response (UPR) genes upregulated as an adaptive response to counteract the loss of ERS-containing proteins, suggesting previously unknown isoform-specific functions of the KDEL receptors. Overall, our findings establish that decreases in ER calcium change the composition of the ER luminal proteome and secretome, which can impact cellular functions and cell viability. The redistribution of the ER proteome from inside the cell to the outside has implications for dissecting the complex relationship of ER homeostasis with diverse disease pathologies.
Collapse
|
474
|
Shallis RM, Zeidan AM. Myelodysplastic/myeloproliferative neoplasm, unclassifiable (MDS/MPN-U): More than just a "catch-all" term? Best Pract Res Clin Haematol 2019; 33:101132. [PMID: 32460977 DOI: 10.1016/j.beha.2019.101132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 11/29/2019] [Accepted: 12/02/2019] [Indexed: 12/17/2022]
Abstract
The clinicopathology of MDS and MPN are not mutually exclusive and for this reason the category of myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN) exists. Several sub-entities have been included under the MDS/MPN umbrella, including MDS/MPN-unclassifiable (MDS/MPN-U) for those cases whose morphologic and clinical phenotype do not meet criteria to be classified as any other MDS/MPN sub-entity. Though potentially regarded as a wastebasket diagnosis, since its integration into myeloid disease classification, MDS/MPN-U has been refined with increasing understanding of the mutational and genomic events that drive particular clinicopathologic phenotypes, even within MDS/MPN-U. The prototypical example is the identification of SF3B1 mutations and its durable association with MDS/MPN with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T), an entity previously buried within, but now a separate category outside of MDS/MPN-U. Continued and enhanced study of those entities under MDS/MPN-U, a perhaps provisional category itself, is likely to progressively identify commonality between many "unclassifiables" to establish a new classifiable diagnosis.
Collapse
Affiliation(s)
- Rory M Shallis
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA; Yale Cancer Center, New Haven, USA.
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, New Haven, USA; Yale Cancer Center, New Haven, USA
| |
Collapse
|
475
|
The Expression of Myeloproliferative Neoplasm-Associated Calreticulin Variants Depends on the Functionality of ER-Associated Degradation. Cancers (Basel) 2019; 11:cancers11121921. [PMID: 31810292 PMCID: PMC6966542 DOI: 10.3390/cancers11121921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 11/26/2019] [Accepted: 11/28/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Mutations in CALR observed in myeloproliferative neoplasms (MPN) were recently shown to be pathogenic via their interaction with MPL and the subsequent activation of the Janus Kinase – Signal Transducer and Activator of Transcription (JAK-STAT) pathway. However, little is known on the impact of those variant CALR proteins on endoplasmic reticulum (ER) homeostasis. Methods: The impact of the expression of Wild Type (WT) or mutant CALR on ER homeostasis was assessed by quantifying the expression level of Unfolded Protein Response (UPR) target genes, splicing of X-box Binding Protein 1 (XBP1), and the expression level of endogenous lectins. Pharmacological and molecular (siRNA) screens were used to identify mechanisms involved in CALR mutant proteins degradation. Coimmunoprecipitations were performed to define more precisely actors involved in CALR proteins disposal. Results: We showed that the expression of CALR mutants alters neither ER homeostasis nor the sensitivity of hematopoietic cells towards ER stress-induced apoptosis. In contrast, the expression of CALR variants is generally low because of a combination of secretion and protein degradation mechanisms mostly mediated through the ER-Associated Degradation (ERAD)-proteasome pathway. Moreover, we identified a specific ERAD network involved in the degradation of CALR variants. Conclusions: We propose that this ERAD network could be considered as a potential therapeutic target for selectively inhibiting CALR mutant-dependent proliferation associated with MPN, and therefore attenuate the associated pathogenic outcomes.
Collapse
|
476
|
Veenstra C, Bruce D, Timbs A, Hamblin A. Application of Genomics to Clinical Practice in Haematological Malignancy. CURRENT GENETIC MEDICINE REPORTS 2019. [DOI: 10.1007/s40142-019-00179-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Purpose of Review
The usual abundance of fresh cells and high-quality DNA derived from bone marrow aspirate and peripheral blood mean haematological malignancies are at the forefront of the application of genomics to malignancy. This review evaluates where genomics is routinely used in clinical care and where opportunities for further application exist.
Recent Findings
The 2016 revision of the WHO classification of tumours of haematopoietic and lymphoid tissues increased the number of disease entities defined by, or whose diagnosis was strongly supported by, a specific genetic change. Increasingly combinations of mutations rather than individual lesions are being used to genomically classify heterogeneous disorders to inform prognosis and direct treatment. Furthermore, the role of different genetic aberrations as markers of measurable residual disease is being evaluated in clinical trials to allow intensification/de-intensification of treatment as appropriate and early detection of relapse.
Summary
Implementation of broader sequencing technologies such as whole exome/genome sequencing coupled with continuing developments in genomic technology to improve turn-around-times are likely to further reinforce the centrality of genomics in the management of haematological malignancies.
Collapse
|
477
|
Liu P, Zhao L, Loos F, Marty C, Xie W, Martins I, Lachkar S, Qu B, Waeckel-Énée E, Plo I, Vainchenker W, Perez F, Rodriguez D, López-Otin C, van Endert P, Zitvogel L, Kepp O, Kroemer G. Immunosuppression by Mutated Calreticulin Released from Malignant Cells. Mol Cell 2019; 77:748-760.e9. [PMID: 31785928 DOI: 10.1016/j.molcel.2019.11.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 09/23/2019] [Accepted: 11/06/2019] [Indexed: 12/11/2022]
Abstract
Mutations affecting exon 9 of the CALR gene lead to the generation of a C-terminally modified calreticulin (CALR) protein that lacks the KDEL endoplasmic reticulum (ER) retention signal and consequently mislocalizes outside of the ER where it activates the thrombopoietin receptor in a cell-autonomous fashion, thus driving myeloproliferative diseases. Here, we used the retention using selective hooks (RUSH) assay to monitor the trafficking of CALR. We found that exon-9-mutated CALR was released from cells in response to the biotin-mediated detachment from its ER-localized hook, in vitro and in vivo. Cellular CALR release was confirmed in suitable mouse models bearing exon-9-mutated hematopoietic systems or tumors. Extracellular CALR mediated immunomodulatory effects and inhibited the phagocytosis of dying cancer cells by dendritic cells (DC), thereby suppressing antineoplastic immune responses elicited by chemotherapeutic agents or by PD-1 blockade. Altogether, our results demonstrate paracrine immunosuppressive effects for exon-9-mutated CALR.
Collapse
Affiliation(s)
- Peng Liu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM UMR 1138, Paris, France; Sorbonne Université, Paris, France; Université of Paris, Paris, France
| | - Liwei Zhao
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM UMR 1138, Paris, France; Sorbonne Université, Paris, France; Université of Paris, Paris, France; Université Paris-Saclay, Villejuif, France
| | - Friedemann Loos
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM UMR 1138, Paris, France; Sorbonne Université, Paris, France; Université of Paris, Paris, France
| | - Caroline Marty
- Université Paris-Saclay, Villejuif, France; INSERM, UMR 1170, Villejuif, France; Gustave Roussy Comprehensive Cancer Center, Villejuif, France
| | - Wei Xie
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM UMR 1138, Paris, France; Sorbonne Université, Paris, France; Université of Paris, Paris, France
| | - Isabelle Martins
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM UMR 1138, Paris, France; Sorbonne Université, Paris, France; Université of Paris, Paris, France
| | - Sylvie Lachkar
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM UMR 1138, Paris, France; Sorbonne Université, Paris, France; Université of Paris, Paris, France
| | - Bo Qu
- Gustave Roussy Comprehensive Cancer Center, Villejuif, France; INSERM, U1015, Villejuif, France; Center of Clinical Investigations, CIC1428, Villejuif, France
| | - Emmanuelle Waeckel-Énée
- Université of Paris, Paris, France; INSERM, U1151, Paris, France; CNRS UMR8253, Paris, France
| | - Isabelle Plo
- Université Paris-Saclay, Villejuif, France; INSERM, UMR 1170, Villejuif, France; Gustave Roussy Comprehensive Cancer Center, Villejuif, France
| | - William Vainchenker
- Université Paris-Saclay, Villejuif, France; INSERM, UMR 1170, Villejuif, France; Gustave Roussy Comprehensive Cancer Center, Villejuif, France
| | - Franck Perez
- Cell Biology and Cancer Unit, Institut Curie, PSL Research University, CNRS, Paris, France
| | - David Rodriguez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Carlos López-Otin
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM UMR 1138, Paris, France; Sorbonne Université, Paris, France; Université of Paris, Paris, France; Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Universidad de Oviedo, 33006 Oviedo, Spain
| | - Peter van Endert
- Université of Paris, Paris, France; INSERM, U1151, Paris, France; CNRS UMR8253, Paris, France
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Center, Villejuif, France; INSERM, U1015, Villejuif, France; Center of Clinical Investigations, CIC1428, Villejuif, France
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM UMR 1138, Paris, France; Sorbonne Université, Paris, France; Université of Paris, Paris, France.
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France; Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, INSERM UMR 1138, Paris, France; Sorbonne Université, Paris, France; Université of Paris, Paris, France; Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China; Karolinska Institutet, Department of Women's and Children's Health, Stockholm, Sweden; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
478
|
Rao TN, Hansen N, Hilfiker J, Rai S, Majewska JM, Leković D, Gezer D, Andina N, Galli S, Cassel T, Geier F, Delezie J, Nienhold R, Hao-Shen H, Beisel C, Di Palma S, Dimeloe S, Trebicka J, Wolf D, Gassmann M, Fan TWM, Lane AN, Handschin C, Dirnhofer S, Kröger N, Hess C, Radimerski T, Koschmieder S, Čokić VP, Skoda RC. JAK2-mutant hematopoietic cells display metabolic alterations that can be targeted to treat myeloproliferative neoplasms. Blood 2019; 134:1832-1846. [PMID: 31511238 PMCID: PMC6872961 DOI: 10.1182/blood.2019000162] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/17/2019] [Indexed: 12/20/2022] Open
Abstract
Increased energy requirement and metabolic reprogramming are hallmarks of cancer cells. We show that metabolic alterations in hematopoietic cells are fundamental to the pathogenesis of mutant JAK2-driven myeloproliferative neoplasms (MPNs). We found that expression of mutant JAK2 augmented and subverted metabolic activity of MPN cells, resulting in systemic metabolic changes in vivo, including hypoglycemia, adipose tissue atrophy, and early mortality. Hypoglycemia in MPN mouse models correlated with hyperactive erythropoiesis and was due to a combination of elevated glycolysis and increased oxidative phosphorylation. Modulating nutrient supply through high-fat diet improved survival, whereas high-glucose diet augmented the MPN phenotype. Transcriptomic and metabolomic analyses identified numerous metabolic nodes in JAK2-mutant hematopoietic stem and progenitor cells that were altered in comparison with wild-type controls. We studied the consequences of elevated levels of Pfkfb3, a key regulatory enzyme of glycolysis, and found that pharmacological inhibition of Pfkfb3 with the small molecule 3PO reversed hypoglycemia and reduced hematopoietic manifestations of MPNs. These effects were additive with the JAK1/2 inhibitor ruxolitinib in vivo and in vitro. Inhibition of glycolysis by 3PO altered the redox homeostasis, leading to accumulation of reactive oxygen species and augmented apoptosis rate. Our findings reveal the contribution of metabolic alterations to the pathogenesis of MPNs and suggest that metabolic dependencies of mutant cells represent vulnerabilities that can be targeted for treating MPNs.
Collapse
Affiliation(s)
- Tata Nageswara Rao
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Nils Hansen
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Julian Hilfiker
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Shivam Rai
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Julia-Magdalena Majewska
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Danijela Leković
- Clinic of Hematology, Clinical Center of Serbia, Belgrade, Serbia
| | - Deniz Gezer
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Nicola Andina
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Serena Galli
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Teresa Cassel
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Florian Geier
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | | | - Ronny Nienhold
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Hui Hao-Shen
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Christian Beisel
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Serena Di Palma
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Sarah Dimeloe
- Immunobiology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Jonel Trebicka
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- European Foundation for the Study of Chronic Liver Failure, Barcelona, Spain
- Department of Gastroenterology, Odense Hospital, University of Southern Denmark, Odense, Denmark
- Institute for Bioengineering of Catalonia, Barcelona, Spain
| | - Dominik Wolf
- Internal Medicine V, Department of Hematology and Oncology, Medical University Innsbruck, Innsbruck, Austria
- Medical Clinic III for Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Teresa W-M Fan
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky, Lexington, KY
| | - Andrew N Lane
- Center for Environmental and Systems Biochemistry, Department of Toxicology and Cancer Biology and Markey Cancer Center, University of Kentucky, Lexington, KY
| | | | - Stefan Dirnhofer
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Hospital Eppendorf, Hamburg, Germany
| | - Christoph Hess
- Immunobiology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Thomas Radimerski
- Disease Area Oncology, Novartis Institutes for Biomedical Research, Basel, Switzerland; and
| | - Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaseology, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany
| | - Vladan P Čokić
- Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Radek C Skoda
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
479
|
Chammas P, Mocavini I, Di Croce L. Engaging chromatin: PRC2 structure meets function. Br J Cancer 2019; 122:315-328. [PMID: 31708574 PMCID: PMC7000746 DOI: 10.1038/s41416-019-0615-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/24/2019] [Indexed: 01/01/2023] Open
Abstract
Polycomb repressive complex 2 (PRC2) is a key epigenetic multiprotein complex involved in the regulation of gene expression in metazoans. PRC2 is formed by a tetrameric core that endows the complex with histone methyltransferase activity, allowing it to mono-, di- and tri-methylate histone H3 on lysine 27 (H3K27me1/2/3); H3K27me3 is a hallmark of facultative heterochromatin. The core complex of PRC2 is bound by several associated factors that are responsible for modulating its targeting specificity and enzymatic activity. Depletion and/or mutation of the subunits of this complex can result in severe developmental defects, or even lethality. Furthermore, mutations of these proteins in somatic cells can be drivers of tumorigenesis, by altering the transcriptional regulation of key tumour suppressors or oncogenes. In this review, we present the latest results from structural studies that have characterised PRC2 composition and function. We compare this information with data and literature for both gain-of function and loss-of-function missense mutations in cancers to provide an overview of the impact of these mutations on PRC2 activity.
Collapse
Affiliation(s)
- Paul Chammas
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, Barcelona, 08003, Spain
| | - Ivano Mocavini
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, Barcelona, 08003, Spain
| | - Luciano Di Croce
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology (BIST), Dr. Aiguader 88, Barcelona, 08003, Spain. .,Universitat Pompeu Fabra (UPF), Barcelona, Spain. .,ICREA, Pg Lluis Companys 23, Barcelona, 08010, Spain.
| |
Collapse
|
480
|
Cottin L, Riou J, Orvain C, Ianotto JC, Boyer F, Renard M, Truchan‐Graczyk M, Murati A, Jouanneau‐Courville R, Allangba O, Mansier O, Burroni B, Rousselet MC, Quintin‐Roué I, Martin A, Sadot‐Lebouvier S, Delneste Y, Chrétien J, Hunault‐Berger M, Blanchet O, Lippert E, Ugo V, Luque Paz D. Sequential mutational evaluation of CALR ‐mutated myeloproliferative neoplasms with thrombocytosis reveals an association between CALR allele burden evolution and disease progression. Br J Haematol 2019; 188:935-944. [DOI: 10.1111/bjh.16276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/25/2019] [Indexed: 12/13/2022]
|
481
|
Kokubun H, Jin H, Aoe T. Pathogenic Effects of Impaired Retrieval between the Endoplasmic Reticulum and Golgi Complex. Int J Mol Sci 2019; 20:ijms20225614. [PMID: 31717602 PMCID: PMC6888596 DOI: 10.3390/ijms20225614] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/31/2019] [Accepted: 11/07/2019] [Indexed: 12/15/2022] Open
Abstract
Cellular activities, such as growth and secretion, are dependent on correct protein folding and intracellular protein transport. Injury, like ischemia, malnutrition, and invasion of toxic substances, affect the folding environment in the endoplasmic reticulum (ER). The ER senses this information, following which cells adapt their response to varied situations through the unfolded protein response. Activation of the KDEL receptor, resulting from the secretion from the ER of chaperones containing the KDEL sequence, plays an important role in this adaptation. The KDEL receptor was initially shown to be necessary for the retention of KDEL sequence-containing proteins in the ER. However, it has become clear that the activated KDEL receptor also regulates bidirectional transport between the ER and the Golgi complex, as well as from the Golgi to the secretory pathway. In addition, it has been suggested that the signal for KDEL receptor activation may also affect several other cellular activities. In this review, we discuss KDEL receptor-mediated bidirectional transport and signaling and describe disease models and human diseases related to KDEL receptor dysfunction.
Collapse
Affiliation(s)
- Hiroshi Kokubun
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Hisayo Jin
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Tomohiko Aoe
- Department of Medicine, Pain Center, Chiba Medical Center, Teikyo University, Ichihara 299-0111, Japan
- Correspondence: ; Tel.: +81-436-62-1211
| |
Collapse
|
482
|
Padmanabhan A, Connelly-Smith L, Aqui N, Balogun RA, Klingel R, Meyer E, Pham HP, Schneiderman J, Witt V, Wu Y, Zantek ND, Dunbar NM, Schwartz GEJ. Guidelines on the Use of Therapeutic Apheresis in Clinical Practice - Evidence-Based Approach from the Writing Committee of the American Society for Apheresis: The Eighth Special Issue. J Clin Apher 2019; 34:171-354. [PMID: 31180581 DOI: 10.1002/jca.21705] [Citation(s) in RCA: 864] [Impact Index Per Article: 144.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The American Society for Apheresis (ASFA) Journal of Clinical Apheresis (JCA) Special Issue Writing Committee is charged with reviewing, updating and categorizing indications for the evidence-based use of therapeutic apheresis (TA) in human disease. Since the 2007 JCA Special Issue (Fourth Edition), the committee has incorporated systematic review and evidence-based approaches in the grading and categorization of apheresis indications. This Eighth Edition of the JCA Special Issue continues to maintain this methodology and rigor in order to make recommendations on the use of apheresis in a wide variety of diseases/conditions. The JCA Eighth Edition, like its predecessor, continues to apply the category and grading system definitions in fact sheets. The general layout and concept of a fact sheet that was introduced in the Fourth Edition, has largely been maintained in this edition. Each fact sheet succinctly summarizes the evidence for the use of TA in a specific disease entity or medical condition. The Eighth Edition comprises 84 fact sheets for relevant diseases and medical conditions, with 157 graded and categorized indications and/or TA modalities. The Eighth Edition of the JCA Special Issue seeks to continue to serve as a key resource that guides the utilization of TA in the treatment of human disease.
Collapse
Affiliation(s)
- Anand Padmanabhan
- Medical Sciences Institute & Blood Research Institute, Versiti & Department of Pathology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Laura Connelly-Smith
- Department of Medicine, Seattle Cancer Care Alliance & University of Washington, Seattle, Washington
| | - Nicole Aqui
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rasheed A Balogun
- Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Reinhard Klingel
- Apheresis Research Institute, Cologne, Germany & First Department of Internal Medicine, University of Mainz, Mainz, Germany
| | - Erin Meyer
- Department of Hematology/Oncology/BMT/Pathology, Nationwide Children's Hospital, Columbus, Ohio
| | - Huy P Pham
- Department of Pathology, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Jennifer Schneiderman
- Department of Pediatric Hematology/Oncology/Neuro-oncology/Stem Cell Transplant, Ann & Robert H. Lurie Children's Hospital of Chicago, Northwestern University, Chicago, Illinois
| | - Volker Witt
- Department for Pediatrics, St. Anna Kinderspital, Medical University of Vienna, Vienna, Austria
| | - Yanyun Wu
- Bloodworks NW & Department of Laboratory Medicine, University of Washington, Seattle, Washington, Yale University School of Medicine, New Haven, Connecticut
| | - Nicole D Zantek
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Nancy M Dunbar
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire
| | | |
Collapse
|
483
|
Accetta R, Elli L, Libera L, Siracusa C, Cassavia F, Orsini F, Orlandi L, Passamonti F, Casalone R, Pallotti F. Analysis of three screening methods for the detection of calreticulin gene mutations. Int J Lab Hematol 2019; 42:e76-e79. [PMID: 31693300 DOI: 10.1111/ijlh.13117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/01/2019] [Accepted: 10/09/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Raffaella Accetta
- SSD Laboratorio Analisi-SMEL Specializzato in Citogenetica e Genetica Medica, ASST Settelaghi-Ospedale di Circolo-Fondazione Macchi, Varese, Italy
| | - Lorenzo Elli
- SSD Laboratorio Analisi-SMEL Specializzato in Citogenetica e Genetica Medica, ASST Settelaghi-Ospedale di Circolo-Fondazione Macchi, Varese, Italy
| | - Laura Libera
- UO Ematologia, ASST Settelaghi- Ospedale di Circolo-Fondazione Macchi, Varese, Italy
| | - Claudia Siracusa
- UO Ematologia, ASST Settelaghi- Ospedale di Circolo-Fondazione Macchi, Varese, Italy
| | | | | | | | - Francesco Passamonti
- UO Ematologia, ASST Settelaghi- Ospedale di Circolo-Fondazione Macchi, Varese, Italy.,Dipartimento di Medicina e Chirurgia, Università degli Studi dell'Insubria, Varese, Italy
| | - Rosario Casalone
- SSD Laboratorio Analisi-SMEL Specializzato in Citogenetica e Genetica Medica, ASST Settelaghi-Ospedale di Circolo-Fondazione Macchi, Varese, Italy
| | - Francesco Pallotti
- SSD Laboratorio Analisi-SMEL Specializzato in Citogenetica e Genetica Medica, ASST Settelaghi-Ospedale di Circolo-Fondazione Macchi, Varese, Italy.,Dipartimento di Medicina e Chirurgia, Università degli Studi dell'Insubria, Varese, Italy
| |
Collapse
|
484
|
McPherson S, Greenfield G, Andersen C, Grinfeld J, Hasselbalch HC, Nangalia J, Mills KI, McMullin MF. Methylation age as a correlate for allele burden, disease status, and clinical response in myeloproliferative neoplasm patients treated with vorinostat. Exp Hematol 2019; 79:26-34. [PMID: 31563618 DOI: 10.1016/j.exphem.2019.09.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 09/20/2019] [Accepted: 09/22/2019] [Indexed: 01/05/2023]
Abstract
The myeloproliferative neoplasms (MPNs) are a heterogeneous group of clonal neoplastic disorders. Driver mutations in JAK2, CALR, and MPL genes have been identified in the majority of cases. Alongside these, an increasing number of genes are repeatedly identified as mutated in MPN. These, including ASXL1, TET2, DMNT3A, and EZH2, have key roles in epigenetic regulation. Dysregulation of epigenetic processes is therefore a key feature of MPN. Vorinostat is a pan histone deacetylase inhibitor (HDACi) that has been investigated in MPN. DNA methylation (DNAm) is a well-defined epigenetic mechanism of transcription modification. It is known to be affected by ageing, lifestyle, and disease. Epigenetic ageing signatures have been previously described allowing calculation of a methylation age (MA). In this study we examined the effect of vorinostat on MA in MPN cell lines and in patients with polycythaemia vera (PV) and essential thrombocythaemia (ET) treated with vorinostat as part of a clinical trial. An older MA was observed in patients with a higher JAK2 V617F allele burden and those with a longer duration of disease. PV patients had a MA older than that predicted whilst MA was younger than predicted in ET. Treatment with vorinostat resulted in a younger MA in PV patients and older MA in ET patients, in both cases a trend towards the normal chronological age. When MA change was compared against response, nonresponse was associated with a younger than predicted MA in ET patients and a higher than predicted MA in PV patients. The link between MA and JAK2 mutant allele burden implies that allele burden has a role not only in clinical phenotype and disease evolution in MPN patients, but also in the overall methylation landscape of the mutated cells.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Cell Line, Tumor
- DNA Methylation/drug effects
- DNA Methylation/genetics
- DNA, Neoplasm/genetics
- DNA, Neoplasm/metabolism
- Epigenesis, Genetic/drug effects
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Male
- Middle Aged
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Polycythemia Vera/drug therapy
- Polycythemia Vera/genetics
- Polycythemia Vera/metabolism
- Polycythemia Vera/pathology
- Thrombocythemia, Essential/drug therapy
- Thrombocythemia, Essential/genetics
- Thrombocythemia, Essential/metabolism
- Thrombocythemia, Essential/pathology
- Vorinostat/administration & dosage
Collapse
Affiliation(s)
- Suzanne McPherson
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Graeme Greenfield
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom.
| | | | - Jacob Grinfeld
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | | | - Jyoti Nangalia
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Ken I Mills
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom
| | - Mary F McMullin
- Blood Cancer Research Group, Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, United Kingdom; Centre for Medical Education, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
485
|
Cuthbert D, Stein BL. Polycythemia Vera-Associated Complications: Pathogenesis, Clinical Manifestations, And Effects On Outcomes. J Blood Med 2019; 10:359-371. [PMID: 31695542 PMCID: PMC6805785 DOI: 10.2147/jbm.s189922] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/18/2019] [Indexed: 12/13/2022] Open
Abstract
Polycythemia vera is a Philadelphia-negative chronic myeloproliferative neoplasm, characterized by erythrocytosis, which is unique, compared to essential thrombocytosis and primary myelofibrosis. Though longevity can usually be expected, vascular morbidity is associated with this condition, as well as a propensity to evolve into myelofibrosis (post-PV MF) and acute myeloid leukemia. In addition, patients can have a pronounced symptom burden. Herein, contributors to the symptomatic burden, as well as the thrombotic and transformative tendencies are reviewed. From a symptom perspective, some are explained by cytokine release, others by microvascular complications, whereas certain symptoms can herald disease evolution. Thrombosis has multifactorial contributors, including but not limited to gender, and inflammatory stress; investigators have recently hypothesized that microparticles and Neutrophil Extracellular Trap Formations may add to thrombotic burden. Finally, we examine the progression to post-PV MF as well as leukemic transformation, highlighting well-established risk factors including age and leukocytosis, certain treatments, and the presence of “non-driver” mutations.
Collapse
Affiliation(s)
- Danielle Cuthbert
- McGaw Medical Center of Northwestern University, Department of Internal Medicine, Chicago, IL 60611, USA
| | - Brady Lee Stein
- Northwestern University Feinberg School of Medicine, Division of Hematology/Oncology, Department of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
486
|
Hsu CC, Chen YJ, Huang CE, Wu YY, Wang MC, Pei SN, Liao CK, Lu CH, Chen PT, Tsou HY, Li CP, Chuang WH, Chuang CK, Yang CY, Lai YH, Lin YH, Chen CC. Molecular heterogeneity unravelled by single-cell transcriptomics in patients with essential thrombocythaemia. Br J Haematol 2019; 188:707-722. [PMID: 31610612 DOI: 10.1111/bjh.16225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/01/2019] [Indexed: 12/11/2022]
Abstract
Significant phenotypic heterogeneity exists in patients with all subtypes of myeloproliferative neoplasms (MPN), including essential thrombocythaemia (ET). Single-cell RNA sequencing (scRNA-Seq) holds the promise of unravelling the biology of MPN at an unprecedented level of resolution. Herein we employed this approach to dissect the transcriptomes in the CD34+ cells from the peripheral blood of seven previously untreated ET patients and one healthy adult. The mutational profiles in these patients were as follows: JAK2 V617F in two, CALR in three (one type I and two type II) and triple-negative (TN) in two. Our results reveal substantial heterogeneity within this enrolled cohort of patients. Activation of JAK/STAT signalling was recognized in discrepant progenitor lineages among different samples. Significantly disparate molecular profiling was identified in the comparison between ET patients and the control, between patients with different driver mutations (JAK2 V617F and CALR exon 9 indel), and even between patients harbouring the same driver. Intra-individual clonal diversity was also found in the CD34+ progenitor population of a patient, possibly indicating the presence of multiple clones in this case. Estimation of subpopulation size based on cellular immunophenotyping suggested differentiation bias in all analysed samples. Furthermore, combining the transcriptomic information with data from targeted sequencing enabled us to unravel key somatic mutations that are molecularly relevant. To conclude, we demonstrated that scRNA-Seq extended our knowledge of clonal diversity and inter-individual heterogeneity in patients with ET. The obtained information could potentially leapfrog our efforts in the elucidation of the pathogenesis of the disease.
Collapse
Affiliation(s)
- Chia-Chen Hsu
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ying-Ju Chen
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Cih-En Huang
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan.,College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Yu-Ying Wu
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ming-Chung Wang
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Sung-Nan Pei
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan.,College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.,Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chun-Kai Liao
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chang-Hsien Lu
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan.,College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Ping-Tsung Chen
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan.,College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Hsing-Yi Tsou
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chian-Pei Li
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Wei-Hsuan Chuang
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | | | - Cheng-Yu Yang
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Yi-Hua Lai
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yi-Hsuan Lin
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chih-Cheng Chen
- Division of Haematology and Oncology, Department of Medicine, Chang Gung Memorial Hospital, Chiayi, Taiwan.,College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| |
Collapse
|
487
|
Chapeau EA, Mandon E, Gill J, Romanet V, Ebel N, Powajbo V, Andraos-Rey R, Qian Z, Kininis M, Zumstein-Mecker S, Ito M, Hynes NE, Tiedt R, Hofmann F, Eshkind L, Bockamp E, Kinzel B, Mueller M, Murakami M, Baffert F, Radimerski T. A conditional inducible JAK2V617F transgenic mouse model reveals myeloproliferative disease that is reversible upon switching off transgene expression. PLoS One 2019; 14:e0221635. [PMID: 31600213 PMCID: PMC6786561 DOI: 10.1371/journal.pone.0221635] [Citation(s) in RCA: 5] [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: 04/10/2019] [Accepted: 08/12/2019] [Indexed: 11/19/2022] Open
Abstract
Aberrant activation of the JAK/STAT pathway is thought to be the critical event in the pathogenesis of the chronic myeloproliferative neoplasms, polycythemia vera, essential thrombocythemia and primary myelofibrosis. The most frequent genetic alteration in these pathologies is the activating JAK2V617F mutation, and expression of the mutant gene in mouse models was shown to cause a phenotype resembling the human diseases. Given the body of genetic evidence, it has come as a sobering finding that JAK inhibitor therapy only modestly suppresses the JAK2V617F allele burden, despite showing clear benefits in terms of reducing splenomegaly and constitutional symptoms in patients. To gain a better understanding if JAK2V617F is required for maintenance of myeloproliferative disease once it has evolved, we generated a conditional inducible transgenic JAK2V617F mouse model using the SCL-tTA-2S tet-off system. Our model corroborates that expression of JAK2V617F in hematopoietic stem and progenitor cells recapitulates key hallmarks of human myeloproliferative neoplasms, and exhibits gender differences in disease manifestation. The disease was found to be transplantable, and importantly, reversible when transgenic JAK2V617F expression was switched off. Our results indicate that mutant JAK2V617F-specific inhibitors should result in profound disease modification by disabling the myeloproliferative clone bearing mutant JAK2.
Collapse
Affiliation(s)
- Emilie A. Chapeau
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
- * E-mail:
| | - Emeline Mandon
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Jason Gill
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Vincent Romanet
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Nicolas Ebel
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Violetta Powajbo
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Rita Andraos-Rey
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Zhiyan Qian
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Miltos Kininis
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Moriko Ito
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Nancy E. Hynes
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Ralph Tiedt
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Francesco Hofmann
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Leonid Eshkind
- Institute for Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Ernesto Bockamp
- Institute for Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Bernd Kinzel
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Matthias Mueller
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Masato Murakami
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Fabienne Baffert
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Thomas Radimerski
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
488
|
Porto-Soares MA, de Oliveira RD, Cortopassi GM, Machado-Neto JA, Palma LC, Figueiredo-Pontes LLD. Clinical and molecular profile of a Brazilian cohort of patients with classical BCR-ABL1-negative myeloproliferative neoplasms. Hematol Transfus Cell Ther 2019; 42:238-244. [PMID: 31676277 PMCID: PMC7417460 DOI: 10.1016/j.htct.2019.07.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/29/2022] Open
Abstract
Background The classical BCR-ABL1-negative myeloproliferative neoplasms (MPNs) are Polycythemia Vera (PV), Essential Thrombocythemia (ET) and Primary Myelofibrosis (PMF). In developing countries, there are few reports that truly reveal the clinical setting of these patients. Therefore, we aimed to characterize a single center MPN population with a special focus on the correct diagnosis based on the recent review of the WHO criteria for the diagnosis of myeloid neoplasms. Methods This retrospective study analyzed data from medical records of patients with classical BCR-ABL1-negative MPNs diagnosed from January 1997 to October 2017 and followed at the University Hospital of Ribeirão Preto Medical School. Results A total of 162 patients were assessed, 61 with PV, 50 with ET, and 51 with PMF. The mutational status analysis revealed that 113 (69.3%) harbored the JAK2V617F mutation, 23 (14.1%), the CALR mutation, and 12 (7.4%) had a triple-negative status. None of the patients were found to have mutations on the thrombopoietin receptor gene (MPL), including some ET and PMF patients who were not tested. Among the PV patients, 57 (93.5%) were positive for the JAK2V617F mutation, one (1.6%) presented an in-frame deletion JAK2 exon 12 mutation and one (1.6%) presented a missense JAK2 exon 9 mutation, not previously described. The overall survival was lower in the triple-negative patients with PMF, when compared to the JAK2V617F or CALR-mutated (p = 0.002). Conclusion The frequency of somatic mutations and survival in our cohort, stratified according to the respective disease, was consistent with the literature data, despite some limitations. Further prospective epidemiological studies of MPN cohorts are encouraged in developing countries.
Collapse
Affiliation(s)
- Moysés Antonio Porto-Soares
- Faculdade de Medicina de Ribeirão Preto e Centro de Terapia Celular, Universidade de São Paulo, FMRP-USP, Ribeirão Preto, SP, Brazil; Hospital Beneficência Portuguesa de São Paulo, São Paulo, SP, Brazil
| | - Rafael Daltro de Oliveira
- Faculdade de Medicina de Ribeirão Preto e Centro de Terapia Celular, Universidade de São Paulo, FMRP-USP, Ribeirão Preto, SP, Brazil
| | - Gabriel Macedo Cortopassi
- Faculdade de Medicina de Ribeirão Preto e Centro de Terapia Celular, Universidade de São Paulo, FMRP-USP, Ribeirão Preto, SP, Brazil
| | - João Agostinho Machado-Neto
- Faculdade de Medicina de Ribeirão Preto e Centro de Terapia Celular, Universidade de São Paulo, FMRP-USP, Ribeirão Preto, SP, Brazil; Instituto de Ciências Biomédicas, Universidade de São Paulo, ICB USP, São Paulo, SP, Brazil
| | - Leonardo Carvalho Palma
- Faculdade de Medicina de Ribeirão Preto e Centro de Terapia Celular, Universidade de São Paulo, FMRP-USP, Ribeirão Preto, SP, Brazil
| | - Lorena Lobo de Figueiredo-Pontes
- Faculdade de Medicina de Ribeirão Preto e Centro de Terapia Celular, Universidade de São Paulo, FMRP-USP, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
489
|
Maze D, Kazi S, Gupta V, Malinowski AK, Fazelzad R, Shah PS, Shehata N. Association of Treatments for Myeloproliferative Neoplasms During Pregnancy With Birth Rates and Maternal Outcomes: A Systematic Review and Meta-analysis. JAMA Netw Open 2019; 2:e1912666. [PMID: 31584685 PMCID: PMC6784750 DOI: 10.1001/jamanetworkopen.2019.12666] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/16/2019] [Indexed: 12/15/2022] Open
Abstract
Importance Myeloproliferative neoplasms (MPNs) are increasingly being identified in women of childbearing potential. Pregnancy in women with MPNs is associated with maternal thrombosis, hemorrhage, and placental dysfunction leading to fetal growth restriction or loss. Objective To evaluate the association between the use of aspirin, heparin, interferon, or combinations and live birth rate and adverse maternal outcomes in pregnant women with MPNs. Data Sources Systematic searches of MEDLINE, Embase, Cochrane Database of Systematic Reviews, Cochrane Central Register of Controlled Trials, and the MEDLINE Epub Ahead of Print and In-Process and Other Non-Indexed Citations from inception to July 19, 2018, with no language restrictions, was conducted. Key search terms included myeloproliferative disorders, polycythemia vera, essential thrombocythemia, and primary myelofibrosis. Study Selection A study was eligible if it included pregnant patients with MPNs; interventions included aspirin, heparin, and/or interferon; there was a comparison group in which patients did not receive the intervention; the study reported on at least 1 of the study outcomes; and it was a randomized, case-control, or cohort study or series of at least 10 pregnancies. Data were extracted in duplicate; 0.5% of identified studies met selection criteria. Data Extraction and Synthesis The review was conducted according to Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines and reported in accordance with Meta-analysis of Observational Studies in Epidemiology (MOOSE) guidelines. Data were pooled using the Mantel-Haenszel approach. Main Outcomes and Measures Outcomes were the number of live births and maternal complications, specifically, arterial or venous thrombosis, hemorrhage, and preeclampsia. Results Twenty-two studies reporting on 1210 pregnancies were included. The live birth rate was 71.3% (95% CI, 65.1%-77.6%). Use of aspirin (11 studies, 227 patients; unadjusted odds ratio, 8.6; 95% CI, 4.0-18.1) and interferon (6 studies, 90 patients; unadjusted odds ratio, 9.7; 95% CI, 2.3-41.0) were associated with higher odds of live birth. Addition of heparin to aspirin was not associated with higher odds of live birth (6 studies, 96 patients; unadjusted odds ratio, 2.1; 95% CI, 0.5-9.0). The most common adverse maternal event was preeclampsia, with an incidence of 3.1% (95% CI, 1.7%-4.5%). Conclusions and Relevance Most studies reported on pregnancy with essential thrombocythemia. Few studies reported on pregnancy with polycythemia vera and none with myelofibrosis met the inclusion criteria. Most studies were retrospective and early pregnancy losses may have been underreported. Moderate-quality evidence suggests that aspirin or interferon is associated with higher odds of live birth in pregnant women with MPN.
Collapse
Affiliation(s)
- Dawn Maze
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Sajida Kazi
- University of Toronto, Toronto, Ontario, Canada
- Division of Hematology, University Health Network, Toronto, Ontario, Canada
| | - Vikas Gupta
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Ann Kinga Malinowski
- University of Toronto, Toronto, Ontario, Canada
- Division of Maternal–Fetal Medicine, Department of Obstetrics & Gynaecology, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Rouhi Fazelzad
- Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada
- University of Toronto, Toronto, Ontario, Canada
| | - Prakesh S. Shah
- University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Nadine Shehata
- University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| |
Collapse
|
490
|
TERT and JAK2 polymorphisms define genetic predisposition to myeloproliferative neoplasms in Japanese patients. Int J Hematol 2019; 110:690-698. [PMID: 31571131 DOI: 10.1007/s12185-019-02742-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 09/19/2019] [Accepted: 09/19/2019] [Indexed: 01/14/2023]
Abstract
Myeloproliferative neoplasms (MPNs), including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are often characterized by specific somatic mutations in any of the three genes: JAK2, CALR, or MPL. A single nucleotide polymorphism (SNP), rs2736100, in the reverse transcriptase gene (TERT) and a germline JAK2 46/1 haplotype have been associated with MPNs in North American and European patients. We examined 201 Japanese MPN patients, including 52 with PV, 131 with ET, and 18 with PMF, as well as 366 control individuals for TERT rs2736100 and JAK2 rs10974944, a tagging SNP of the 46/1 haplotype. Furthermore, correlations between the JAK2 V617F allele burden at diagnosis and TERT rs2736100 or JAK2 rs10974944 were evaluated using a digital PCR assay for accurate quantitation. The JAK2 46/1 haplotype, but not the TERT rs2736100 SNP, was correlated to the JAK2 V617F mutant allele burden in JAK2 V617F-positive MPN patients. In conclusion, we demonstrated that both TERT rs2736100_C and JAK2 46/1 haplotype are predisposing factors for MPNs in Japanese patients. While TERT rs2736100_C tended to have a more general, non-specific effect on all MPNs, the JAK2 46/1 haplotype was essentially predisposed to the JAK2 V617F-positive MPNs.
Collapse
|
491
|
Guy A, James C. [Pathogenesis of thrombosis in JAK2V617F myeloproliferative neoplasms]. Med Sci (Paris) 2019; 35:651-658. [PMID: 31532377 DOI: 10.1051/medsci/2019133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BCR-ABL negative myeloproliferative neoplasms are acquired hematologic diseases characterized by blood cell proliferation and that include polycythemia vera (PV), essential thrombocytemia (ET) and primary myelofibrosis (PMF). Occurring of venous and arterial thrombosis is the main complication of these diseases. Risk factors for thrombosis are individuals older than 60 and history of thrombosis. The mechanisms leading to thrombosis are complex and involve several blood compartments, plasmatic factors and endothelial cells. Over the last years, new actors of thrombosis have been discovered.
Collapse
Affiliation(s)
- Alexandre Guy
- CHU de Bordeaux, Laboratoire d'hématologie, 1, avenue de Magellan, F-33600, Pessac, France. - Univ. Bordeaux, Inserm, UMR 1034, Biologie des maladies cardio-vasculaires, 1, avenue de Magellan, F-33600, Pessac, France
| | - Chloé James
- CHU de Bordeaux, Laboratoire d'hématologie, 1, avenue de Magellan, F-33600, Pessac, France. - Univ. Bordeaux, Inserm, UMR 1034, Biologie des maladies cardio-vasculaires, 1, avenue de Magellan, F-33600, Pessac, France
| |
Collapse
|
492
|
Eran Z, Zingariello M, Bochicchio MT, Bardelli C, Migliaccio AR. Novel strategies for the treatment of myelofibrosis driven by recent advances in understanding the role of the microenvironment in its etiology. F1000Res 2019; 8:F1000 Faculty Rev-1662. [PMID: 31583083 PMCID: PMC6758840 DOI: 10.12688/f1000research.18581.1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/12/2019] [Indexed: 12/12/2022] Open
Abstract
Myelofibrosis is the advanced stage of the Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs), characterized by systemic inflammation, hematopoietic failure in the bone marrow, and development of extramedullary hematopoiesis, mainly in the spleen. The only potentially curative therapy for this disease is hematopoietic stem cell transplantation, an option that may be offered only to those patients with a compatible donor and with an age and functional status that may face its toxicity. By contrast, with the Philadelphia-positive MPNs that can be dramatically modified by inhibitors of the novel BCR-ABL fusion-protein generated by its genetic lesion, the identification of the molecular lesions that lead to the development of myelofibrosis has not yet translated into a treatment that can modify the natural history of the disease. Therefore, the cure of myelofibrosis remains an unmet clinical need. However, the excitement raised by the discovery of the genetic lesions has inspired additional studies aimed at elucidating the mechanisms driving these neoplasms towards their final stage. These studies have generated the feeling that the cure of myelofibrosis will require targeting both the malignant stem cell clone and its supportive microenvironment. We will summarize here some of the biochemical alterations recently identified in MPNs and the novel therapeutic approaches currently under investigation inspired by these discoveries.
Collapse
Affiliation(s)
- Zimran Eran
- Department of Hematology, Hadassah University Center, Jerusalem, Israel
| | - Maria Zingariello
- Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine, University Campus Bio-Medico, Rome, Italy
| | - Maria Teresa Bochicchio
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.), IRCCS, Meldola (FC), Italy
| | - Claudio Bardelli
- Dipartimento di Scienze Biomediche e NeuroMotorie, Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| | - Anna Rita Migliaccio
- Dipartimento di Scienze Biomediche e NeuroMotorie, Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| |
Collapse
|
493
|
Schieber M, Crispino JD, Stein B. Myelofibrosis in 2019: moving beyond JAK2 inhibition. Blood Cancer J 2019; 9:74. [PMID: 31511492 PMCID: PMC6739355 DOI: 10.1038/s41408-019-0236-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/26/2019] [Accepted: 03/15/2019] [Indexed: 02/08/2023] Open
Abstract
Myelofibrosis (MF) is a myeloproliferative neoplasm characterized by ineffective clonal hematopoiesis, splenomegaly, bone marrow fibrosis, and the propensity for transformation to acute myeloid leukemia. The discovery of mutations in JAK2, CALR, and MPL have uncovered activated JAK-STAT signaling as a primary driver of MF, supporting a rationale for JAK inhibition. However, JAK inhibition alone is insufficient for long-term remission and offers modest, if any, disease-modifying effects. Given this, there is great interest in identifying mechanisms that cooperate with JAK-STAT signaling to predict disease progression and rationally guide the development of novel therapies. This review outlines the latest discoveries in the biology of MF, discusses current clinical management of patients with MF, and summarizes the ongoing clinical trials that hope to change the landscape of MF treatment.
Collapse
Affiliation(s)
- Michael Schieber
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - John D Crispino
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Brady Stein
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
494
|
Wang JC, Shi G, Wong C, Gotlieb V, Ramachandran P, Chen H. Quantification of IGF-1 receptor is useful in the differential diagnosis of essential thrombocytosis from reactive thrombocytosis. Eur J Haematol 2019; 103:573-577. [PMID: 31479555 DOI: 10.1111/ejh.13323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/29/2019] [Accepted: 08/30/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND To make a definite diagnosis of essential thrombocytosis (ET) from reactive thrombocytosis (RT), the most reliable criteria are the presence of driver mutations, namely JAK2, CALR, or MPL gene mutations. In the absence of these driver mutations, so-called triple-negative ET, the differential diagnosis could be difficult. Although bone marrow biopsy could be helpful, it may be difficult in some cases, to do gene sequence analysis to identify other clonal marker gene mutations than the driver mutations, as only very few were found. METHODS IGF-1R quantification by flow cytometry in mononuclear cells (MNC) from peripheral blood was performed in 33 patients with ET (untreated or off treatment with hydroxyurea), 28 patients with RT, and 16 normal volunteer controls. RESULTS We found IGF-1R levels were significantly elevated in ET patients compared to RT patients or controls. A cutoff value of 253 was chosen from the logistic regression to predict each patient's group, a value ≥253 meant that a patient belonged to the ET group (sensitivity 96.4% and specificity 68.6%). CONCLUSION We suggest that adding quantification of IGF-1R in blood MNC by flow cytometry is useful in differentiating ET from RT.
Collapse
Affiliation(s)
- Jen C Wang
- Division of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Guanfang Shi
- Division of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Ching Wong
- Division of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Vladimir Gotlieb
- Division of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Preethi Ramachandran
- Division of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY, USA
| | - Hui Chen
- Division of Hematology/Oncology, Brookdale University Hospital Medical Center, Brooklyn, NY, USA
| |
Collapse
|
495
|
Vu HA, Thao TT, Dong CV, Vuong NL, Chuong HQ, Van PNT, Nghia H, Binh NT, Dung PC, Xinh PT. Clinical and Hematological Relevance of JAK2V617F, CALR, and MPL Mutations in Vietnamese Patients with Essential Thrombocythemia. Asian Pac J Cancer Prev 2019; 20:2775-2780. [PMID: 31554376 PMCID: PMC6976857 DOI: 10.31557/apjcp.2019.20.9.2775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Indexed: 12/22/2022] Open
Abstract
Background: The picture of Vietnamese patients with essential thrombocythemia (ET) remains mostly undetermined. Our study intended to determine the frequency of JAK2V617F, CALR exon 9, and MPL exon 10 mutations as well as to analyze clinical characteristics associated with different mutational status in Vietnamese ET patients. Methods: We explored mutations of JAK2V617F, MPL, and CALR from 395 patients using allele specific oligonucleotide – polymerase chain reaction and Sanger sequencing techniques; then, the clinical and hematological features were compared according to mutation patterns. Results: We found that JAK2V617F, CALR exon 9, and MPL exon 10 mutations were present in 56.2%, 27.6%, and 1% of the 395 patients with ET, respectively. Twelve different types of CALR mutation were detected in 109 patients, with the CALR type 1 mutation (c.1099_1150del; L367fs*46) was the most common, followed by CALR type 2 mutation (c.1154_1155insTTGTC; K385fs*47). The JAK2V617F-positive patients had older age, higher white blood cell counts and higher hemoglobin levels but lower platelet counts than patients with CALR mutations or patients negative for triple tests. There was no significant difference regarding sex ratio, white blood cell counts, platelet counts and hemoglobin levels among CALR mutation subtypes. Conclusion: we reported high frequency of JAK2V617F, CALR, and MPL mutations in Vietnamese patients with ET and underscored the importance of combined genetic tests for diagnosis and classification of ET into different subtypes.
Collapse
Affiliation(s)
- Hoang Anh Vu
- Center for Molecular Biomedicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Tran Thi Thao
- Department of Hematology, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam.
| | - Cao Van Dong
- Ho Chi Minh City Blood Transfusion and Hematology Hospital, Ho Chi Minh City, Vietnam
| | - Nguyen Lam Vuong
- Department of Medical Statistics and Informatics, Faculty of Public Health, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Ho Quoc Chuong
- Center for Molecular Biomedicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Phan Nguyen Thanh Van
- Ho Chi Minh City Blood Transfusion and Hematology Hospital, Ho Chi Minh City, Vietnam
| | - Huynh Nghia
- Department of Hematology, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam. ,Ho Chi Minh City Blood Transfusion and Hematology Hospital, Ho Chi Minh City, Vietnam
| | - Nguyen Tan Binh
- Ho Chi Minh City Blood Transfusion and Hematology Hospital, Ho Chi Minh City, Vietnam
| | - Phu Chi Dung
- Ho Chi Minh City Blood Transfusion and Hematology Hospital, Ho Chi Minh City, Vietnam
| | - Phan Thi Xinh
- Department of Hematology, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam. ,Ho Chi Minh City Blood Transfusion and Hematology Hospital, Ho Chi Minh City, Vietnam
| |
Collapse
|
496
|
Knock-in of murine Calr del52 induces essential thrombocythemia with slow-rising dominance in mice and reveals key role of Calr exon 9 in cardiac development. Leukemia 2019; 34:510-521. [PMID: 31471561 DOI: 10.1038/s41375-019-0538-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 05/23/2019] [Accepted: 05/28/2019] [Indexed: 12/13/2022]
Abstract
Frameshifting mutations (-1/+2) of the calreticulin (CALR) gene are responsible for the development of essential thrombocythemia (ET) and primary myelofibrosis (PMF). The mutant CALR proteins activate the thrombopoietin receptor (TpoR) inducing cytokine-independent megakaryocyte progenitor proliferation. Here, we generated via CRISPR/Cas9 technology two knock-in mouse models that are heterozygous for a type-I murine Calr mutation. These mice exhibit an ET phenotype with elevated circulating platelets compared with wild-type controls, consistent with our previous results showing that murine CALR mutants activate TpoR. We also show that the mutant CALR proteins can be detected in plasma. The phenotype of Calr del52 is transplantable, and the Calr mutated hematopoietic cells have a slow-rising advantage over wild-type hematopoiesis. Importantly, a homozygous state of a type-1 Calr mutation is lethal at a late embryonic development stage, showing narrowed ventricular myocardium walls, similar to the murine Calr knockout phenotype, pointing to the C terminus of CALR as crucial for heart development.
Collapse
|
497
|
Calreticulin Mutation by Immunohistochemistry: Can It Replace PCR? Appl Immunohistochem Mol Morphol 2019; 28:621-626. [DOI: 10.1097/pai.0000000000000804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
498
|
Mutant calreticulin interacts with MPL in the secretion pathway for activation on the cell surface. Leukemia 2019; 34:499-509. [PMID: 31462733 DOI: 10.1038/s41375-019-0564-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 06/30/2019] [Accepted: 07/15/2019] [Indexed: 01/18/2023]
Abstract
Studies have shown that mutant calreticulin (CALR) constitutively activates the thrombopoietin (TPO) receptor MPL and thus plays a causal role in the development of myeloproliferative neoplasms (MPNs). To further elucidate the molecular mechanism by which mutant CALR promotes MPN development, we studied the subcellular localization of mutant CALR and its importance for the oncogenic properties of mutant CALR. Here, mutant CALR accumulated in the Golgi apparatus, and its entrance into the secretion pathway and capacity to interact with N-glycan were required for its oncogenic capacity via the constitutive activation of MPL. Mutant CALR-dependent MPL activation was resistant to blockade of intracellular protein trafficking, suggesting that MPL is activated before reaching the cell surface. However, removal of MPL from the cell surface with trypsin shut down downstream activation, implying that the surface localization of MPL is required for mutant CALR-dependent activation. Furthermore, we found that mutant CALR and MPL interact on the cell surface. Based on these findings, we propose a model in which mutant CALR induces MPL activation on the cell surface to promote MPN development.
Collapse
|
499
|
Imetelstat, a telomerase inhibitor, is capable of depleting myelofibrosis stem and progenitor cells. Blood Adv 2019; 2:2378-2388. [PMID: 30242099 DOI: 10.1182/bloodadvances.2018022012] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 08/18/2018] [Indexed: 01/26/2023] Open
Abstract
Clinical trials of imetelstat therapy have indicated that this telomerase inhibitor might have disease-modifying effects in a subset of patients with myelofibrosis (MF). The mechanism by which imetelstat induces such clinical responses has not been clearly elucidated. Using in vitro hematopoietic progenitor cell (HPC) assays and in vivo hematopoietic stem cell (HSC) assays, we examined the effects of imetelstat on primary normal and MF HSCs/HPCs. Treatment of CD34+ cells with imetelstat reduced the numbers of MF but not cord blood HPCs (colony-forming unit-granulocyte/macrophage, burst-forming unit-erythroid, and colony-forming unit-granulocyte/erythroid/macrophage/megakaryocyte) as well as MF but not normal CD34+ALDH+ cells irrespective of the patient's mutational status. Moreover, imetelstat treatment resulted in depletion of mutated HPCs from JAK2V617F+ MF patients. Furthermore, treatment of immunodeficient mice that had been previously transplanted with MF splenic CD34+ cells with imetelstat at a dose of 15 mg/kg, 3 times per week for 4 weeks had a limited effect on the degree of chimerism achieved by normal severe combined immunodeficiency repopulating cells but resulted in a significant reduction in the degree of human MF cell chimerism as well as the proportion of mutated donor cells. These effects were sustained for at least 3 months after drug treatment was discontinued. These actions of imetelstat on MF HSCs/HPCs were associated with inhibition of telomerase activity and the induction of apoptosis. Our findings indicate that the effects of imetelstat therapy observed in MF patients are likely attributable to the greater sensitivity of imetelstat against MF as compared with normal HSCs/HPCs as well as the intensity of the imetelstat dose schedule.
Collapse
|
500
|
Iurlo A, Elli EM, Palandri F, Cattaneo D, Bossi A, Cortinovis I, Bucelli C, Orofino N, Brioschi F, Auteri G, Bianchi P, Fabris S, Isimbaldi G, Sabattini E, Baldini L, Gianelli U. Integrating clinical, morphological, and molecular data to assess prognosis in patients with primary myelofibrosis at diagnosis: A practical approach. Hematol Oncol 2019; 37:424-433. [PMID: 31359447 DOI: 10.1002/hon.2658] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 12/29/2022]
Abstract
Currently available prognostic scoring systems in primary myelofibrosis (PMF) do not integrate clinical, histological, and molecular data, or they also required information on "other" mutations that are available in the clinical practice only in a very limited number of laboratories. In the present multicenter study, including 401 PMF patients, an integrated International Prognostic Scoring System (I-IPSS) was developed by combining IPSS, grade of bone marrow fibrosis (GBMF), and driver mutations molecular status (MS) to define PMF prognosis at diagnosis. Four prognostic categories were identified: I-IPSS-low risk (113 patients), I-IPSS-intermediate-1 risk (56 patients), I-IPSS-intermediate-2 risk (154 patients), and I-IPSS-high risk (78 patients). Median overall survival was 26.7 years in I-IPSS-intermediate-1, 10.8 in I-IPSS-intermediate-2, and 6.4 in I-IPSS-high-risk patients (log-rank test <0.0001); instead, it was not reached in the I-IPSS-low-risk cohort because of the extremely low number of registered deaths. The addition of GBMF and MS to IPSS improved the efficacy for predicting the risk of death. Indeed, the sensitivity of I-IPSS was significantly higher (P < .05) than that of IPSS, considering both total deaths and 5- and 10-year mortality. This comprehensive approach allows clinicians to evaluate mutual interactions between IPSS, GBMF, and MS and identify high-risk patients with poor prognosis who may benefit from aggressive treatments. More importantly, this integrated score can be easily applicable worldwide as it only required information that represent the good clinical practice in the management of PMF patients.
Collapse
Affiliation(s)
- Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Elena Maria Elli
- Hematology Division, San Gerardo Hospital, ASST Monza, Monza, Italy
| | - Francesca Palandri
- Institute of Hematology, "L. and A. Seràgnoli", S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Anna Bossi
- Laboratory G. A. Maccacaro, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Ivan Cortinovis
- Laboratory G. A. Maccacaro, Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Cristina Bucelli
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Nicola Orofino
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Filippo Brioschi
- Hematology Division, San Gerardo Hospital, ASST Monza, Monza, Italy
| | - Giuseppe Auteri
- Institute of Hematology, "L. and A. Seràgnoli", S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Paola Bianchi
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Sonia Fabris
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | | | - Elena Sabattini
- Institute of Hematology, "L. and A. Seràgnoli", S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Luca Baldini
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Umberto Gianelli
- Division of Pathology, Department of Pathophysiology and Transplantation, University of Milan, and Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|