1
|
Hormoz S, Sankaran VG, Mullally A. Evolution of myeloproliferative neoplasms from normal blood stem cells. Haematologica 2025; 110:840-849. [PMID: 39633553 PMCID: PMC11959262 DOI: 10.3324/haematol.2023.283951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
Over the course of the last decade, genomic studies in the context of normal human hematopoiesis have provided new insights into the early pathogenesis of myeloproliferative neoplasms (MPN). A preclinical phase of MPN, termed clonal hematopoiesis was identified and subsequent lineage tracing studies revealed a multi-decade long time interval from acquisition of an MPN phenotypic driver mutation in a hematopoietic stem cell to the development of overt MPN. Multiple germline variants associated with MPN risk have been identified through genome-wide association studies and in some cases functional interrogation of the impact of the variant has uncovered new insights into hematopoietic stem cell biology and MPN development. Increasingly sophisticated methods to study clonal contributions to human hematopoiesis and measure hematopoietic stem cell fitness have helped to discern the biology underlying the tremendous clinical heterogeneity observed in MPN. Despite these advances, significant knowledge gaps remain, particularly with respect to germline genetic contributors to both MPN pathogenesis and phenotypic diversity, as well as limitations in the ability to prospectively quantify rates of clonal expansion in individual MPN patients. Ultimately, we envisage a personalized approach to MPN care in the future, in which an individualized genetic assessment can predict MPN trajectory and this information will be used to inform and guide therapy. MPN is particularly amenable to precision medicine strategies and our increased understanding of the evolution of MPN from normal blood stem cells provides a unique opportunity for early therapeutic intervention approaches and potentially MPN prevention strategies.
Collapse
Affiliation(s)
- Sahand Hormoz
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA.
| | - Vijay G Sankaran
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA
| | - Ann Mullally
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Hematology Division, VA Palo Alto Health Care System, Palo Alto, CA.
| |
Collapse
|
2
|
Elbadry MI, Abdelkreem E, Tawfeek A, Seo GH, Aziz SP. Genetic variants in NHEJ1 and related DNA repair disorders: insights into phenotypic heterogeneity and links to hypoplastic myelodysplastic syndromes and familial hematological malignancies susceptibility. Ann Hematol 2025; 104:1633-1653. [PMID: 40047910 DOI: 10.1007/s00277-025-06257-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/11/2025] [Indexed: 04/26/2025]
Abstract
This study investigates the burden, phenotypes, progression, and outcomes of familial hematological malignancies (FHM) through clinical evaluation, gene panel testing, and whole exome sequencing, highlighting the significance of identifying genetic causes for personalized treatment. Over six years, 357 patients initially diagnosed with bone marrow failure (BMF) were evaluated, with 152 patients lacking identifiable causes undergoing further analysis. Among these, 53 (34.9%) exhibited features of inherited BMF syndromes, and 13 (24.5%) developed FHM. In a separate cohort of 27 patients with inherited immunodeficiency disorders, 8 (29.6%) developed FHM associated with NHEJ1 or LYST variants, underscoring the familial clustering of hematologic disorders. Notably, 6 of 7 patients from the same family (family-1) with homozygous NHEJ1 variants progressed to secondary myelodysplastic syndrome (sMDS), acute myeloid leukemia (AML), or lymphoma. Among 780 patients diagnosed with hematological malignancies during the study period, 45 (5.8%) were confirmed to have FHM, with 33 patients enrolled for detailed analysis. Of these, 16 (48.5%) had DNA-repair deficiencies (DNA-RD), including eight with Fanconi anemia, six with NHEJ1 variants, and two with BRCA2 mutations. The remaining 17 patients presented conditions such as familial myeloproliferative neoplasms, dyskeratosis congenita (DC) [TERT, DKC1 variants], and Chediak-Higashi syndrome. Two siblings (family-3) with a rare TERT variant and a unique DC phenotype developed sMDS after prolonged BMF. Patients with DNA-RD were younger and exhibited higher rates of growth failure, recurrent infections, and endocrinopathies. These cases frequently progressed to sMDS or AML. A comparative analysis of 319 individuals with DNA double-strand break repair deficiencies revealed a 45% frequency of hematological malignancies. Lymphoma was most common in Nijmegen breakage syndrome (79.4%) while MDS/AML was prevalent in Cernunnos deficiency (66.6%). The findings emphasize the importance of early diagnosis, genetic testing, and personalized management, including timely transplantation, to improve outcomes in FHM. This research underscores the need for clinical awareness and surveillance to facilitate timely interventions and mitigate disease progression.
Collapse
Affiliation(s)
- Mahmoud I Elbadry
- Department of Internal Medicine, Division of Haematology, Faculty of Medicine, Sohag University, Sohag, 82524, Egypt.
| | - Elsayed Abdelkreem
- Department of Paediatrics, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Ahmed Tawfeek
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Sohag University, Sohag, 82524, Egypt
| | - Go Hun Seo
- Medical Genetics Division, 3billion Inc, Seoul, South Korea
| | - Shereen Philip Aziz
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Sohag University, Sohag, 82524, Egypt
| |
Collapse
|
3
|
Hermouet S. Mutations, inflammation and phenotype of myeloproliferative neoplasms. Front Oncol 2023; 13:1196817. [PMID: 37284191 PMCID: PMC10239955 DOI: 10.3389/fonc.2023.1196817] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/09/2023] [Indexed: 06/08/2023] Open
Abstract
Knowledge on the myeloproliferative neoplasms (MPNs) - polycythemia vera (PV), essential thrombocythemia (ET), primary myelofibrosis (PMF) - has accumulated since the discovery of the JAK/STAT-activating mutations associated with MPNs: JAK2V617F, observed in PV, ET and PMF; and the MPL and CALR mutations, found in ET and PMF. The intriguing lack of disease specificity of these mutations, and of the chronic inflammation associated with MPNs, triggered a quest for finding what precisely determines that MPN patients develop a PV, ET or PMF phenoptype. The mechanisms of action of MPN-driving mutations, and concomitant mutations (ASXL1, DNMT3A, TET2, others), have been extensively studied, as well as the role played by these mutations in inflammation, and several pathogenic models have been proposed. In parallel, different types of drugs have been tested in MPNs (JAK inhibitors, interferons, hydroxyurea, anagrelide, azacytidine, combinations of those), some acting on both JAK2 and inflammation. Yet MPNs remain incurable diseases. This review aims to present current, detailed knowledge on the pathogenic mechanisms specifically associated with PV, ET or PMF that may pave the way for the development of novel, curative therapies.
Collapse
Affiliation(s)
- Sylvie Hermouet
- Nantes Université, INSERM, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, Nantes, France
- Laboratoire d'Hématologie, CHU Nantes, Nantes, France
| |
Collapse
|
4
|
Luque Paz D, Kralovics R, Skoda RC. Genetic basis and molecular profiling in myeloproliferative neoplasms. Blood 2023; 141:1909-1921. [PMID: 36347013 PMCID: PMC10646774 DOI: 10.1182/blood.2022017578] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/03/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
BCR::ABL1-negative myeloproliferative neoplasms (MPNs) are clonal diseases originating from a single hematopoietic stem cell that cause excessive production of mature blood cells. The 3 subtypes, that is, polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are diagnosed according to the World Health Organization (WHO) and international consensus classification (ICC) criteria. Acquired gain-of-function mutations in 1 of 3 disease driver genes (JAK2, CALR, and MPL) are the causative events that can alone initiate and promote MPN disease without requiring additional cooperating mutations. JAK2-p.V617F is present in >95% of PV patients, and also in about half of the patients with ET or PMF. ET and PMF are also caused by mutations in CALR or MPL. In ∼10% of MPN patients, those referred to as being "triple negative," none of the known driver gene mutations can be detected. The common theme between the 3 driver gene mutations and triple-negative MPN is that the Janus kinase-signal transducer and activator of transcription (JAK/STAT) signaling pathway is constitutively activated. We review the recent advances in our understanding of the early events after the acquisition of a driver gene mutation. The limiting factor that determines the frequency at which MPN disease develops with a long latency is not the acquisition of driver gene mutations, but rather the expansion of the clone. Factors that control the conversion from clonal hematopoiesis to MPN disease include inherited predisposition, presence of additional mutations, and inflammation. The full extent of knowledge of the mutational landscape in individual MPN patients is now increasingly being used to predict outcome and chose the optimal therapy.
Collapse
Affiliation(s)
- Damien Luque Paz
- Univ Angers, Nantes Université, CHU Angers, Inserm, CNRS, CRCI2NA, Angers, France
| | - Robert Kralovics
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Radek C. Skoda
- Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel, Basel, Switzerland
| |
Collapse
|
5
|
Torres DG, Paes J, da Costa AG, Malheiro A, Silva GV, Mourão LPDS, Tarragô AM. JAK2 Variant Signaling: Genetic, Hematologic and Immune Implication in Chronic Myeloproliferative Neoplasms. Biomolecules 2022; 12:291. [PMID: 35204792 PMCID: PMC8961666 DOI: 10.3390/biom12020291] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 02/04/2023] Open
Abstract
The JAK2V617F variant constitutes a genetic alteration of higher frequency in BCR/ABL1 negative chronic myeloproliferative neoplasms, which is caused by a substitution of a G ˃ T at position 1849 and results in the substitution of valine with phenylalanine at codon 617 of the polypeptide chain. Clinical, morphological and molecular genetic features define the diagnosis criteria of polycythemia vera, essential thrombocythemia and primary myelofibrosis. Currently, JAK2V617F is associated with clonal hematopoiesis, genomic instability, dysregulations in hemostasis and immune response. JAK2V617F clones induce an inflammatory immune response and lead to a process of immunothrombosis. Recent research has shown great interest in trying to understand the mechanisms associated with JAK2V617F signaling and activation of cellular and molecular responses that progressively contribute to the development of inflammatory and vascular conditions in association with chronic myeloproliferative neoplasms. Thus, the aim of this review is to describe the main genetic, hematological and immunological findings that are linked to JAK2 variant signaling in chronic myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Dania G. Torres
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
| | - Jhemerson Paes
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
| | - Allyson G. da Costa
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus 69067-005, AM, Brazil
| | - Adriana Malheiro
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
- Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus 69067-005, AM, Brazil
| | - George V. Silva
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
- Fundação Oswaldo Cruz–Instituto Leônidas e Maria Deane (Fiocruz), Manaus 69027-070, AM, Brazil
- Fundação Centro de Controle de Oncologia do Amazonas (FCECON), Manaus 69040-010, AM, Brazil
| | - Lucivana P. de Souza Mourão
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
| | - Andréa M. Tarragô
- Programa de Pós-Graduação em Ciências Aplicadas à Hematologia, Universidade do Estado do Amazonas (UEA), Manaus 69850-000, AM, Brazil; (D.G.T.); (J.P.); (A.G.d.C.); (A.M.); (G.V.S.)
- Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (FHEMOAM), Manaus 69050-001, AM, Brazil
| |
Collapse
|
6
|
Germline ATG2B/GSKIP-containing 14q32 duplication predisposes to early clonal hematopoiesis leading to myeloid neoplasms. Leukemia 2022; 36:126-137. [PMID: 34172895 DOI: 10.1038/s41375-021-01319-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/26/2021] [Accepted: 06/02/2021] [Indexed: 02/06/2023]
Abstract
The germline predisposition associated with the autosomal dominant inheritance of the 14q32 duplication implicating ATG2B/GSKIP genes is characterized by a wide clinical spectrum of myeloid neoplasms. We analyzed 12 asymptomatic carriers and 52 patients aged 18-74 years from six families, by targeted sequencing of 41 genes commonly mutated in myeloid malignancies. We found that 75% of healthy carriers displayed early clonal hematopoiesis mainly driven by TET2 mutations. Molecular landscapes of patients revealed two distinct routes of clonal expansion and leukemogenesis. The first route is characterized by the clonal dominance of myeloproliferative neoplasms (MPN)-driver events associated with TET2 mutations in half of cases and mutations affecting splicing and/or the RAS pathway in one-third of cases, leading to the early development of MPN, mostly essential thrombocythemia, with a high risk of transformation (50% after 10 years). The second route is distinguished by the absence of MPN-driver mutations and leads to AML without prior MPN. These patients mostly harbored a genomic landscape specific to acute myeloid leukemia secondary to myelodysplastic syndrome. An unexpected result was the total absence of DNMT3A mutations in this cohort. Our results suggest that the germline duplication constitutively mimics hematopoiesis aging by favoring TET2 clonal hematopoiesis.
Collapse
|
7
|
Bohiltea RE, Niculescu-Mizil E, Mihai BM, Furtunescu F, Ducu I, Munteanu O, Georgescu TA, Grigoriu C. Polycythemia vera in pregnancy represents a challenge for a multidisciplinary collaboration: A case report and literature review. Exp Ther Med 2021; 23:19. [PMID: 34815771 DOI: 10.3892/etm.2021.10941] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/24/2021] [Indexed: 01/10/2023] Open
Abstract
Polycythemia vera (PV) is a rare chronic myeloproliferative neoplasm which represents an additional thrombotic factor in pregnancy. PV may be difficult to diagnose, particularly as its incidence is extremely uncommon among young women. The main diagnostic method involves a bone marrow biopsy, and high hemoglobin and platelet counts are usually indicative of the condition, after excluding other more frequent pathologies. PV is associated with a high risk of thrombosis, particularly in pregnancy, and requires anti-platelet treatment. At present, only a limited number of PV cases in pregnancy have been reported in the literature, at least to the best of our knowledge, with the largest case series being a retrospective study that included 25 pregnancies in 15 women. The present study describes the case of a patient diagnosed with JAK2-positive PV and also discusses this rare condition with particular focus on the following: i) The management of PV in pregnancy along with the additional pathologies in this specific case; and ii) the particularities of the pregnancy course. By identifying women suffering from PV superimposed by other possible procoagulant factors and applying the latest standard in healthcare, fetal and maternal prognosis may be significantly improved.
Collapse
Affiliation(s)
- Roxana Elena Bohiltea
- Department of Obstetrics and Gynecology, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, 050098 Bucharest, Romania.,Department of Obstetrics and Gynecology, Life Memorial Hospital, 012244 Bucharest, Romania
| | | | - Bianca Margareta Mihai
- Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, 050098 Bucharest, Romania
| | - Florentina Furtunescu
- Department of Public Health and Management, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050463 Bucharest, Romania
| | - Ionita Ducu
- Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, 050098 Bucharest, Romania
| | - Octavian Munteanu
- Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, 050098 Bucharest, Romania.,Department of Anatomy, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Tiberiu Augustin Georgescu
- Department of Pathology, Faculty of Medicine, 'Carol Davila' University of Medicine and Pharmacy, 050463 Bucharest, Romania
| | - Corina Grigoriu
- Department of Obstetrics and Gynecology, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Obstetrics and Gynecology, University Emergency Hospital Bucharest, 050098 Bucharest, Romania
| |
Collapse
|
8
|
Patel AB, Franzini A, Leroy E, Kim SJ, Pomicter AD, Genet L, Xiao M, Yan D, Ahmann JM, Agarwal AM, Clair P, Addada J, Lambert J, Salmon M, Gleich GJ, Cross NCP, Constantinescu SN, O'Hare T, Prchal JT, Deininger MW. JAK2 ex13InDel drives oncogenic transformation and is associated with chronic eosinophilic leukemia and polycythemia vera. Blood 2019; 134:2388-2398. [PMID: 31697804 PMCID: PMC6933291 DOI: 10.1182/blood.2019001385] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/01/2019] [Indexed: 02/06/2023] Open
Abstract
The V617F mutation in the JH2 domain of Janus kinase 2 (JAK2) is an oncogenic driver in several myeloproliferative neoplasms (MPNs), including essential thrombocythemia, myelofibrosis, and polycythemia vera (PV). Other mutations in JAK2 have been identified in MPNs, most notably exon 12 mutations in PV. Here, we describe a novel recurrent mutation characterized by a common 4-amino-acid deletion and variable 1-amino-acid insertion (Leu583-Ala586DelInsSer/Gln/Pro) within the JH2 domain of JAK2. All 4 affected patients had eosinophilia, and both patients with Leu583-Ala586DelInsSer fulfilled diagnostic criteria of both PV and chronic eosinophilic leukemia (CEL). Computational and functional studies revealed that Leu583-Ala586DelInsSer (herein referred to as JAK2ex13InDel) deregulates JAK2 through a mechanism similar to JAK2V617F, activates signal transducer and activator of transcription 5 and extracellular signal-regulated kinase, and transforms parental Ba/F3 cells to growth factor independence. In contrast to JAK2V617F, JAK2ex13InDel does not require an exogenous homodimeric type 1 cytokine receptor to transform Ba/F3 cells and is capable of activating β common chain family cytokine receptor (interleukin-3 receptor [IL-3R], IL-5R, and granulocyte-macrophage colony stimulating factor receptor) signaling in the absence of ligand, with the maximum effect observed for IL-5R, consistent with the clinical phenotype of eosinophilia. Recognizing this new PV/CEL-overlap MPN has significant clinical implications, as both PV and CEL patients are at high risk for thrombosis, and concomitant cytoreduction of red cells, neutrophils, and eosinophils may be required for prevention of thromboembolic events. Targeted next-generation sequencing for genes recurrently mutated in myeloid malignancies in patients with unexplained eosinophilia may reveal additional cases of Leu583-Ala586DelInsSer/Gln/Pro, allowing for complete characterization of this unique MPN.
Collapse
Affiliation(s)
- Ami B Patel
- Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, UT
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Anca Franzini
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Emilie Leroy
- Ludwig Cancer Research Brussels and de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels, Belgium
| | - Soo Jin Kim
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | | | - Lidvine Genet
- Ludwig Cancer Research Brussels and de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels, Belgium
| | - Michael Xiao
- Department of Biochemistry, The University of Utah School of Medicine, Salt Lake City, UT
| | - Dongqing Yan
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Jonathan M Ahmann
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Archana M Agarwal
- Division of Clinical Pathology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT
| | - Phillip Clair
- Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, UT
| | - Juanah Addada
- Department of Haematology, Royal Derby Hospital, Derby, United Kingdom
| | - Jonathan Lambert
- Department of Clinical Haematology, University College London Hospitals, London, United Kingdom
| | - Matthew Salmon
- Wessex Regional Genetics Laboratory, Salisbury NHS Foundation Trust, Salisbury, United Kingdom
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Gerald J Gleich
- Department of Dermatology and
- Department of Medicine, The University of Utah, Salt Lake City, UT; and
| | - Nicholas C P Cross
- Wessex Regional Genetics Laboratory, Salisbury NHS Foundation Trust, Salisbury, United Kingdom
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Stefan N Constantinescu
- Ludwig Cancer Research Brussels and de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- WELBIO (Walloon Excellence in Life Sciences and Biotechnology), Brussels, Belgium
| | - Thomas O'Hare
- Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, UT
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| | - Josef T Prchal
- Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, UT
- Veteran Administration Medical Center, Salt Lake City, UT
| | - Michael W Deininger
- Division of Hematology and Hematologic Malignancies, The University of Utah, Salt Lake City, UT
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, UT
| |
Collapse
|
9
|
Sud A, Chattopadhyay S, Thomsen H, Sundquist K, Sundquist J, Houlston RS, Hemminki K. Analysis of 153 115 patients with hematological malignancies refines the spectrum of familial risk. Blood 2019; 134:960-969. [PMID: 31395603 PMCID: PMC6789511 DOI: 10.1182/blood.2019001362] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 06/26/2019] [Indexed: 02/08/2023] Open
Abstract
Estimating familial cancer risks is clinically important in being able to discriminate between individuals in the population at differing risk for malignancy. To gain insight into the familial risk for the different hematological malignancies and their possible inter-relationship, we analyzed data on more than 16 million individuals from the Swedish Family-Cancer Database. After identifying 153 115 patients diagnosed with a primary hematological malignancy, we quantified familial relative risks (FRRs) by calculating standardized incident ratios (SIRs) in 391 131 of their first-degree relatives. The majority of hematological malignancies showed increased FRRs for the same tumor type, with the highest FRRs being observed for mixed cellularity Hodgkin lymphoma (SIR, 16.7), lymphoplasmacytic lymphoma (SIR, 15.8), and mantle cell lymphoma (SIR, 13.3). There was evidence for pleiotropic relationships; notably, chronic lymphocytic leukemia was associated with an elevated familial risk for other B-cell tumors and myeloproliferative neoplasms. Collectively, these data provide evidence for shared etiological factors for many hematological malignancies and provide information for identifying individuals at increased risk, as well as informing future gene discovery initiatives.
Collapse
Affiliation(s)
- Amit Sud
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre, Heidelberg, Germany
| | - Subhayan Chattopadhyay
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre, Heidelberg, Germany
- Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | - Hauke Thomsen
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre, Heidelberg, Germany
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
- Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Community-based Healthcare Research and Education, Department of Functional Pathology, School of Medicine, Shimane University, Matsue, Japan; and
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
- Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY
- Center for Community-based Healthcare Research and Education, Department of Functional Pathology, School of Medicine, Shimane University, Matsue, Japan; and
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, United Kingdom
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre, Heidelberg, Germany
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| |
Collapse
|
10
|
Crysandt M, Brings K, Beier F, Thiede C, Brümmendorf TH, Jost E. Germ line predisposition to myeloid malignancies appearing in adulthood. Expert Rev Hematol 2018; 11:625-636. [PMID: 29958021 DOI: 10.1080/17474086.2018.1494566] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Germ line predisposition to myeloid neoplasms has been incorporated in the WHO 2016 classification of myeloid neoplasms and acute leukemia. The new category of disease is named hereditary myeloid disorder (HMD). Although most myeloid neoplasms are sporadic, germ line mutations and familial predisposition can contribute to development of chronic myeloid diseases and acute myeloid leukemia. This finding and upcoming frequent use of genome wide detection of molecular aberrations will lead to a higher detection rate of a genetic predisposition and influence treatment decisions. Hereditary predisposition is responsible for 5-10% of myeloid malignancies. Management of affected patients begins by the awareness of treating physicians of the problem and a precise work up of the patient and family members. Areas covered: This review focuses on current knowledge about germ line predisposition for myeloid neoplasms including diagnostic, prognostic, and therapeutic aspects in adult patients. Essential information for clinical routine is provided. Expert commentary: Compared to a patient without predisposition, adaptation of treatment strategy for patients with an HMD is often necessary, especially to avoid higher risk of relapse or higher toxicity during chemotherapy or transplantation. Mistakes in choice of a related donor can be omitted. Relatives at risk of developing a HMD need specific surveillance.
Collapse
Affiliation(s)
- Martina Crysandt
- a Medical Faculty, Dept. of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation , University Hospital RWTH Aachen , Aachen , Germany
| | - Kira Brings
- a Medical Faculty, Dept. of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation , University Hospital RWTH Aachen , Aachen , Germany
| | - Fabian Beier
- a Medical Faculty, Dept. of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation , University Hospital RWTH Aachen , Aachen , Germany
| | - Christian Thiede
- b Medizinische Klinik und Poliklinik I , Universitätsklinikum Carl Gustav Carus der TU Dresden , Dresden , Germany
| | - Tim H Brümmendorf
- a Medical Faculty, Dept. of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation , University Hospital RWTH Aachen , Aachen , Germany
| | - Edgar Jost
- a Medical Faculty, Dept. of Hematology, Oncology, Hemostaseology and Stem Cell Transplantation , University Hospital RWTH Aachen , Aachen , Germany
| |
Collapse
|
11
|
Abstract
Chronic myeloproliferative neoplasms (MPN) characteristically arise from a somatic mutation in the pluripotent hematopoietic stem cell, and most common recurring mutations are in the JAK2, CALR, and cMPL genes. However, these mutations are not founder mutations, but mainly drive the disease phenotype and a pre-existing germline predisposition has been long speculated, but has not been clearly defined to date. Genome-wide association studies in family clusters of MPN have identified a number of genetic variants that are associated with increased germline risk for developing clonal MPN. The strongest association discovered so far is the presence of JAK2 46/1 haplotype, and subsequently, many studies have found additional variants in other genes, most notably in TERT gene. However, these still account for a small fraction of familial MPN, and more in-depth studies including whole genome sequencing are needed to gain better insight into familial genetic predisposition of clonal MPNs.
Collapse
|
12
|
Hirvonen EAM, Pitkänen E, Hemminki K, Aaltonen LA, Kilpivaara O. Whole-exome sequencing identifies novel candidate predisposition genes for familial polycythemia vera. Hum Genomics 2017; 11:6. [PMID: 28427458 PMCID: PMC5397753 DOI: 10.1186/s40246-017-0102-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/09/2017] [Indexed: 12/12/2022] Open
Abstract
Background Polycythemia vera (PV), characterized by massive production of erythrocytes, is one of the myeloproliferative neoplasms. Most patients carry a somatic gain-of-function mutation in JAK2, c.1849G > T (p.Val617Phe), leading to constitutive activation of JAK-STAT signaling pathway. Familial clustering is also observed occasionally, but high-penetrance predisposition genes to PV have remained unidentified. Results We studied the predisposition to PV by exome sequencing (three cases) in a Finnish PV family with four patients. The 12 shared variants (maximum allowed minor allele frequency <0.001 in Finnish population in ExAC database) predicted damaging in silico and absent in an additional control set of over 500 Finns were further validated by Sanger sequencing in a fourth affected family member. Three novel predisposition candidate variants were identified: c.1254C > G (p.Phe418Leu) in ZXDC, c.1931C > G (p.Pro644Arg) in ATN1, and c.701G > A (p.Arg234Gln) in LRRC3. We also observed a rare, predicted benign germline variant c.2912C > G (p.Ala971Gly) in BCORL1 in all four patients. Somatic mutations in BCORL1 have been reported in myeloid malignancies. We further screened the variants in eight PV patients in six other Finnish families, but no other carriers were found. Conclusions Exome sequencing provides a powerful tool for the identification of novel variants, and understanding the familial predisposition of diseases. This is the first report on Finnish familial PV cases, and we identified three novel candidate variants that may predispose to the disease. Electronic supplementary material The online version of this article (doi:10.1186/s40246-017-0102-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elina A M Hirvonen
- Genome-Scale Biology Research Program, Research Programs Unit and Department of Medical and Clinical Genetics, Medicum, University of Helsinki, P.O. Box 63, 00014, Helsinki, Finland
| | - Esa Pitkänen
- Genome-Scale Biology Research Program, Research Programs Unit and Department of Medical and Clinical Genetics, Medicum, University of Helsinki, P.O. Box 63, 00014, Helsinki, Finland
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lauri A Aaltonen
- Genome-Scale Biology Research Program, Research Programs Unit and Department of Medical and Clinical Genetics, Medicum, University of Helsinki, P.O. Box 63, 00014, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, SE-17177, Stockholm, Sweden
| | - Outi Kilpivaara
- Genome-Scale Biology Research Program, Research Programs Unit and Department of Medical and Clinical Genetics, Medicum, University of Helsinki, P.O. Box 63, 00014, Helsinki, Finland.
| |
Collapse
|
13
|
|
14
|
Braunstein EM, Moliterno AR. Back to biology: new insights on inheritance in myeloproliferative disorders. Curr Hematol Malig Rep 2015; 9:311-8. [PMID: 25195195 DOI: 10.1007/s11899-014-0232-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The myeloproliferative disorders (MPDs) are a group of hematologic diseases with significant overlap in both clinical phenotype and genetic etiology. While most often caused by acquired somatic mutations in hematopoietic stem cells, the presence of familial clustering in MPD cases suggests that inheritance is an important factor in the etiology of this disease. Though far less common than sporadic disease, inherited MPDs can be clinically indistinguishable from sporadic disease. Recently, germline mutations in Janus kinase 2 (JAK2) and MPL, two genes frequently mutated in sporadic MPD, have been shown to cause inherited thrombocytosis. Study of the function of these mutant proteins has led to a new understanding of the biological mechanisms that produce myeloproliferative disease. In this review, we summarize the data regarding inherited mutations that cause or predispose to MPDs, with a focus on the biological effects of mutant proteins. We propose that defining inherited MPDs in this manner has the potential to simplify diagnosis in a group of disorders that can be difficult to differentiate clinically.
Collapse
Affiliation(s)
- Evan M Braunstein
- Division of Hematology, Department of Medicine, School of Medicine, Johns Hopkins University, 720 Rutland Ave., Ross Research Building Room 1025, Baltimore, MD, 21205, USA,
| | | |
Collapse
|
15
|
El Nemer W, De Grandis M, Brusson M. Abnormal adhesion of red blood cells in polycythemia vera: a prothrombotic effect? Thromb Res 2015; 133 Suppl 2:S107-11. [PMID: 24862129 DOI: 10.1016/s0049-3848(14)50018-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Polycythemia vera (PV) is a myeloproliferative neoplasm (MPN) characterised by the V617F activating mutation in the tyrosine kinase JAK2. PV patients exhibit increased haemoglobin levels and red cell mass because of uncontrolled proliferation of the erythroid lineage. Thrombosis and transformation to acute leukaemia are the major causes of morbidity and mortality in this disease. Increased thrombotic risk in PV patients is multifactorial and complex; it is associated with high levels of haemoglobin, impaired rheology and increased viscosity resulting from erythrocytosis. An additional parameter that might contribute to this risk was recently brought to light by work from our group showing abnormal activation of adhesion proteins in PV RBCs. In this review we provide an overview of these recent findings and discuss how the pro-adhesive features of JAK2V617F-positive red blood cells might initiate and contribute to the circulatory complications described in PV.
Collapse
Affiliation(s)
- Wassim El Nemer
- INSERM U1134, F-75739 Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMR_S 1134, F-75739 Paris, France; Institut National de la Transfusion Sanguine F-75739 Paris, France Laboratoire d'Excellence GR-Ex France.
| | - Maria De Grandis
- INSERM U1134, F-75739 Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMR_S 1134, F-75739 Paris, France; Institut National de la Transfusion Sanguine F-75739 Paris, France Laboratoire d'Excellence GR-Ex France
| | - Mégane Brusson
- INSERM U1134, F-75739 Paris, France; Univ Paris Diderot, Sorbonne Paris Cité, UMR_S 1134, F-75739 Paris, France; Institut National de la Transfusion Sanguine F-75739 Paris, France Laboratoire d'Excellence GR-Ex France
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Knowledge related to gastrointestinal stromal tumor (GIST) in the setting of nonhereditary and hereditary multiple tumor syndromes continues to expand. This review describes associations between sporadic GIST and second malignancies, as well as new contributions to our knowledge about hereditary GIST multiple tumor syndromes. RECENT FINDINGS Sporadic GIST patients have increased risk of developing synchronous/metachronous cancers, including nonhematologic and hematologic malignancies. Data suggest these associations are nonrandom, more prevalent in men and increase with age. New adrenal tumors have also been associated with nonhereditary Carney's triad. Meanwhile, understanding of the molecular basis of heritable GIST syndromes has improved. Several new familial GIST kindreds have been reported, including those with germline KIT and PDGFRα mutations. Knowledge about succinate dehydrogenase (SDH) deficiency and mutations in hereditary GIST syndromes has expanded. It is now known that neurofibromatosis-1-associated GISTs are SDHB-positive, whereas Carney-Stratakis syndrome-associated GISTs are SDHB-deficient with underlying germline mutations in SDH subunits A-D. SUMMARY Recognition and early diagnosis of GIST syndromes allows for improved comprehensive medical care. With additional understanding of the molecular pathogenesis of GIST multiple tumor syndromes, we can refine our screening programs and management of these patients and their families.
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW This review discusses the unexpected role of red blood cell (RBC) adhesiveness in the pathophysiology of two red cell diseases, hereditary spherocytosis and polycythemia vera, and two 'nonerythroid' disorders, central retinal vein occlusion and Gaucher disease. These pathologies share common clinical manifestations, that is vaso-occlusion and/or thrombotic events. RECENT FINDINGS Recently, the direct involvement of RBC adhesion to the vascular endothelium has been demonstrated in the occurrence of vaso-occlusive events, in particular in sickle cell disease (SCD). Several erythroid adhesion molecules and their ligands have been identified that belong to different molecular classes (integrins, Ig-like molecules, lipids...) and are activated by a variety of signaling pathways. Among these, the laminin receptor, Lutheran/basal cell adhesion molecule, which is activated by phosphorylation, appears to play a central role in several pathologies. SUMMARY RBC adhesiveness might be involved in complications such as the vaso-occlusive crisis in SCD, thrombosis in polycythemia vera, splenic sequestration in hereditary spherocytosis, occlusions in central retinal vein occlusion and bone infarcts in Gaucher disease. Characterization of this pathological process at the cellular and molecular levels should prove useful to develop new therapeutic approaches based on the blockade of RBC abnormal interactions with vascular endothelium and/or circulating blood cells.
Collapse
|
18
|
Ye Z, Liu CF, Lanikova L, Dowey SN, He C, Huang X, Brodsky RA, Spivak JL, Prchal JT, Cheng L. Differential sensitivity to JAK inhibitory drugs by isogenic human erythroblasts and hematopoietic progenitors generated from patient-specific induced pluripotent stem cells. Stem Cells 2014; 32:269-78. [PMID: 24105986 DOI: 10.1002/stem.1545] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/25/2013] [Accepted: 08/02/2013] [Indexed: 01/31/2023]
Abstract
Disease-specific induced pluripotent stem cells (iPSCs) provide an unprecedented opportunity to establish novel disease models and accelerate drug development using distinct tissue target cells generated from isogenic iPSC lines with and without disease-causing mutations. To realize the potential of iPSCs in modeling acquired diseases which are usually heterogeneous, we have generated multiple iPSC lines including two lines that are JAK2-wild-type and four lines homozygous for JAK2-V617F somatic mutation from a single polycythemia vera (PV) patient blood. In vitro differentiation of the same patient-derived iPSC lines have demonstrated the differential contributions of their parental hematopoietic clones to the abnormal erythropoiesis including the formation of endogenous erythroid colonies. This iPSC approach thus may provide unique and valuable insights into the genetic events responsible for disease development. To examine the potential of iPSCs in drug testing, we generated isogenic hematopoietic progenitors and erythroblasts from the same iPSC lines derived from PV patients and normal donors. Their response to three clinical JAK inhibitors, INCB018424 (Ruxolitinib), TG101348 (SAR302503), and the more recent CYT387 was evaluated. All three drugs similarly inhibited erythropoiesis from normal and PV iPSC lines containing the wild-type JAK2 genotype, as well as those containing a homozygous or heterozygous JAK2-V617F activating mutation that showed increased erythropoiesis without a JAK inhibitor. However, the JAK inhibitors had less inhibitory effect on the self-renewal of CD34+ hematopoietic progenitors. The iPSC-mediated disease modeling thus underlies the ineffectiveness of the current JAK inhibitors and provides a modeling system to develop better targeted therapies for the JAK2 mutated hematopoiesis.
Collapse
Affiliation(s)
- Zhaohui Ye
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Stem Cell Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Prick J, de Haan G, Green AR, Kent DG. Clonal heterogeneity as a driver of disease variability in the evolution of myeloproliferative neoplasms. Exp Hematol 2014; 42:841-51. [PMID: 25201757 DOI: 10.1016/j.exphem.2014.07.268] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 01/01/2023]
Abstract
Myeloproliferative neoplasms (MPNs) are clonal hematological diseases in which cells of the myelo-erythroid lineage are overproduced and patients are predisposed to leukemic transformation. Hematopoietic stem cells are the suspected disease-initiating cells, and these cells must acquire a clonal advantage relative to nonmutant hematopoietic stem cells to perpetuate disease. In 2005, several groups identified a single gain-of-function point mutation in JAK2 that associated with the majority of MPNs, and subsequent studies have led to a comprehensive understanding of the mutational landscape in MPNs. However, confusion still exists as to how a single genetic aberration can be associated with multiple distinct disease entities. Many explanations have been proposed, including JAK2V617F homozygosity, individual patient heterogeneity, and the differential regulation of downstream JAK2 signaling pathways. Several groups have made knock-in mouse models expressing JAK2V617F and have observed divergent phenotypes, each recapitulating some aspects of disease. Intriguingly, most of these models do not observe a strong hematopoietic stem cell self-renewal advantage compared with wild-type littermate controls, raising the question of how a clonal advantage is established in patients with MPNs. This review summarizes the current molecular understanding of MPNs and the diversity of disease phenotypes and proposes that the increased proliferation induced by JAK2V617F applies a selection pressure on the mutant clone that results in highly diverse clonal evolution in individuals.
Collapse
Affiliation(s)
- Janine Prick
- Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gerald de Haan
- Laboratory of Ageing Biology and Stem Cells, European Research Institute for the Biology of Ageing, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Anthony R Green
- Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - David G Kent
- Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, University of Cambridge, Cambridge, United Kingdom; Department of Haematology, Addenbrooke's Hospital, Cambridge, United Kingdom.
| |
Collapse
|
20
|
From Janus kinase 2 to calreticulin: the clinically relevant genomic landscape of myeloproliferative neoplasms. Blood 2014; 123:3714-9. [PMID: 24786775 DOI: 10.1182/blood-2014-03-530865] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Our understanding of the genetic basis of myeloproliferative neoplasms began in 2005, when the JAK2 (V617F) mutation was identified in polycythemia vera, essential thrombocythemia, and primary myelofibrosis. JAK2 exon 12 and MPL exon 10 mutations were then detected in subsets of patients, and subclonal driver mutations in other genes were found to be associated with disease progression. Recently, somatic mutations in the gene CALR, encoding calreticulin, have been found in most patients with essential thrombocythemia or primary myelofibrosis with nonmutated JAK2 and MPL. The JAK-STAT pathway appears to be activated in all myeloproliferative neoplasms, regardless of founding driver mutations. These latter, however, have different effects on clinical course and outcomes. Thus, evaluation of JAK2, MPL, and CALR mutation status is important not only for diagnosis but also for prognostication. These genetic data should now also be considered in designing clinical trials.
Collapse
|
21
|
Relationship between the 46/1 haplotype of the JAK2 gene and the JAK2 mutational status and allele burden, the initial findings, and the survival of patients with myelofibrosis. Ann Hematol 2013; 93:797-802. [DOI: 10.1007/s00277-013-1989-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 12/02/2013] [Indexed: 10/25/2022]
|
22
|
Abstract
Abstract
The discovery of mutations activating JAK-STAT signaling in the majority of patients with myeloproliferative neoplasms (MPNs) led to identification of tyrosine kinase activation as a predominant mechanism driving MPN pathogenesis. Despite this, the existence of additional genetic events that modify the MPN phenotype, predate JAK2 mutations, and/or contribute to leukemic transformation of MPNs has been suggested. Recently, mutations in several epigenetic modifiers have been described in patients with MPNs, including mutations in ASXL1, DNMT3A, EZH2, IDH1, IDH2, and TET2. Moreover, the mutant JAK2 itself has been shown recently to affect histone posttranslational modifications directly. Here we review the biological and clinical implications of epigenetic alterations in the pathogenesis of MPNs.
Collapse
|
23
|
Abstract
Myeloproliferative neoplasms (MPNs) are haematological disorders characterized by an overproduction of mature myeloid cells with a tendency to transform to acute myeloid leukaemia. Clonal proliferation of myeloid progenitor cells is driven by somatically acquired mutations, most notably JAK2 V617F, but there are important features relating to pathogenesis and phenotypic diversity that cannot be explained by acquired mutations alone. In this review we consider what is currently known about the role that inherited factors play in the development and biology of both sporadic and familial forms of MPN. Although most MPN cases appear to be sporadic, familial predisposition has been recognized for many years in a subset of cases and epidemiological studies have indicated the presence of common susceptibility alleles. Currently the JAK2 46/1 haplotype (also referred to as 'GGCC') is the strongest known predisposition factor for sporadic MPNs carrying a JAK2 V617F mutation, explaining a large proportion of the heritability of this disorder. Less is known about what genetic variants predispose to MPNs that lack JAK2 V617F, but there have been recent reports of interesting associations in biologically plausible candidates, and more loci are set to emerge with the application of systematic genome-wide association methodologies. Several highly penetrant predisposition variants that affect erythropoietin signalling, thrombopoietin signalling or oxygen sensing have been characterized in families with nonclonal hereditary erythrocytosis or thrombocytosis, but much less is known about familial predisposition to true clonal MPN. The heterogeneous pattern of inheritance and presumed genetic heterogeneity in these families makes analysis difficult, but whole exome or genome sequencing should provide novel insights into these elusive disorders.
Collapse
Affiliation(s)
- Amy V Jones
- Wessex Regional Genetics Laboratory, Salisbury, UK, Faculty of Medicine, University of Southampton, Southampton, UK
| | | |
Collapse
|
24
|
Abstract
The association between somatic JAK2 mutation and myeloproliferative neoplasms (MPNs) is now well established. However, because JAK2 mutations are associated with heterogeneous clinical phenotypes and often occur as secondary genetic events, some aspects of JAK2 mutation biology remain to be understood. We recently described a germline JAK2V617I mutation in a family with hereditary thrombocytosis and herein characterize the hematopoietic and signaling impact of JAK2V617I. Through targeted sequencing of MPN-associated mutations, exome sequencing, and clonality analysis, we demonstrate that JAK2V617I is likely to be the sole driver mutation in JAK2V617I-positive individuals with thrombocytosis. Phenotypic hematopoietic stem cells (HSCs) were increased in the blood and bone marrow of JAK2V617I-positive individuals and were sustained at higher levels than controls after xenotransplantation. In signaling and transcriptional assays, JAK2V617I demonstrated more activity than wild-type JAK2 but substantially less than JAK2V617F. After cytokine stimulation, JAK2V617I resulted in markedly increased downstream signaling compared with wild-type JAK2 and comparable with JAK2V617F. These findings demonstrate that JAK2V617I induces sufficient cytokine hyperresponsiveness in the absence of other molecular events to induce a homogeneous MPN-like phenotype. We also provide evidence that the JAK2V617I mutation may expand the HSC pool, providing insights into both JAK2 mutation biology and MPN disease pathogenesis.
Collapse
|
25
|
Milosevic JD, Kralovics R. Genetic and epigenetic alterations of myeloproliferative disorders. Int J Hematol 2012; 97:183-97. [PMID: 23233154 DOI: 10.1007/s12185-012-1235-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 11/27/2012] [Accepted: 11/27/2012] [Indexed: 01/07/2023]
Abstract
The classical BCR-ABL negative myeloproliferative neoplasms (MPN) polycythemia vera, essential thrombocythemia, and primary myelofibrosis are clonal hematopoietic disorders characterized by excessive production of terminally differentiated myeloid cells. In MPN patients, the disease can progress to secondary myelofibrosis or acute myeloid leukemia. Clonal hematopoiesis, disease phenotype, and progression are caused by somatically acquired genetic lesions of genes involved in cytokine signaling, RNA splicing, as well as epigenetic regulation. This review provides an overview of point mutations and cytogenetic lesions associated with MPN and addresses the role of these somatic lesions in MPN disease progression.
Collapse
Affiliation(s)
- Jelena D Milosevic
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT25.3, 1090 Vienna, Austria
| | | |
Collapse
|
26
|
|
27
|
Abstract
It is thought that myeloproliferative neoplasms (MPNs) are driven by somatic mutations, although hereditary factors also play a prominent role in the pathogenesis of the disease. Hereditary thrombocytosis and erythrocytosis are not malignant disorders but are clinically similar to MPNs. Several mutations have been found that explain a proportion of hereditary thrombocytosis and hereditary erythrocytosis. Germline variants can influence the risk of leukemic transformation in MPNs and the course of the disease through interaction with acquired chromosomal aberrations. Overall, it has been shown that germline factors play an important part in MPN pathogenesis.
Collapse
|
28
|
Abdel-Wahab O, Tefferi A, Levine RL. Role of TET2 and ASXL1 mutations in the pathogenesis of myeloproliferative neoplasms. Hematol Oncol Clin North Am 2012; 26:1053-64. [PMID: 23009937 DOI: 10.1016/j.hoc.2012.07.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Since the discovery of activating mutations in JAK2 in patients with myeloproliferative neoplasms (MPNs) in 2005, gene discovery efforts have identified additional disease alleles, which can predate or occur subsequent to acquisition of JAK2/MPL mutations. In 2009, somatic copy number loss and mutations in the genes TET2 and ASXL1 were identified in MPN patients. Genetic analysis of MPN patient cohorts with adequate sample size and clear clinical annotation are needed to understand the importance of these mutations on MPN phenotype, risk of transformation to leukemia, response to therapy, and influence on overall survival.
Collapse
Affiliation(s)
- Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | | | | |
Collapse
|
29
|
Malak S, Labopin M, Saint-Martin C, Bellanne-Chantelot C, Najman A. Long term follow up of 93 families with myeloproliferative neoplasms: life expectancy and implications of JAK2V617F in the occurrence of complications. Blood Cells Mol Dis 2012; 49:170-6. [PMID: 22818858 DOI: 10.1016/j.bcmd.2012.06.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 06/06/2012] [Indexed: 02/02/2023]
Abstract
The long-term evolution of familial myeloproliferative neoplasms was studied in 93 families with 227 subjects including 97 with polycythemia vera (PV), 105 essential thrombocythemia (ET), 14 primary myelofibrosis (PMF) and 11 chronic myeloid leukemia (CML). In PV patients, with 12years of median follow-up, overall survival was 83% at 10years and 37% at 20years. A high JAK2(V617F) allele burden was correlated with the transformation to myelofibrosis (p<0.0001), but not with the transformation to acute leukemia. Among the 105 ET, with 8years of median follow-up, overall survival was 83% at 10years and 57% at 20years. Progression to acute leukemia and progression to myelofibrosis were 10% and 13%. There was a trend toward a more frequent evolution to myelofibrosis when the JAK2(V617F) mutated allele burden was >50% (p=0.09), but not to AML. Hematologic transformation of the MPN was responsible for 69% of the deaths, cerebral stroke for 7% and 4% died of myocardial infarction. Eleven JAK2(V617F) mutated patients developed 13 deep splanchnic thromboses in PV and ET. Finally whereas patients with familial PV and ET have a comparable prognosis to non-familial MPN, the JAK2(V617F) mutation was associated with a more frequent occurrence of thrombosis in the entire population.
Collapse
Affiliation(s)
- Sandra Malak
- Department of Hematology, Assistance Publique-Hopitaux de Paris (AP-HP) Saint-Antoine, Universite Pierre et Marie Curie, Paris, France
| | | | | | | | | | | |
Collapse
|
30
|
Abstract
Clonality can be established by a lack of mosaicism in a female because of random inactivation of either the maternal or paternal X chromosome early in embryogenesis. The methylation status of CpG sites close to the trinucleotide repeats in exon 1 of the human androgen receptor (AR) X chromosome gene assay (HUMARA) has been used to determine clonality. This HUMARA at times indicated clonal hematopoiesis in healthy elderly women, thus precluding its applicability. We used a clonality assay based on quantitative expression of polymorphic X chromosome genes (qTCA) and found no evidence of clonal hematopoiesis in healthy nonanemic elderly persons. We found instances of discordance between HUMARA results and those obtained by pyrosequencing and qTCA methods, as well as by directly quantifying AR gene expression. To determine the basis of this discrepancy we examined the methylation pattern of the AR locus subject to HUMARA. Notably, we found the extent of DNA methylation to be highly variable at the AR gene in granulocytes of persons with discordant results and also in erythroid burst-forming unit colonies but not in those with clonal hematopoiesis. These data provide the molecular basis of incomplete correlation with the pattern of DNA methylation of this X chromosome AR gene locus.
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Since the discovery of the JAK2V617F mutation in 2005, an increasing number of somatic and germline genetic events responsible for myeloproliferative neoplasm (MPN) pathogenesis have been uncovered. The purpose of this review is to outline the most recent discoveries of the genetic alterations found in patients with MPNs. RECENT FINDINGS In addition to the JAK2V617F mutation, additional mutations in the JAK–STAT pathway have been discovered including a series of mutations in exon 12 of JAK2, the thrombopoietic receptor gene MPL, and in the gene encoding the JAK–STAT inhibitory adaptor protein LNK. Additionally, mutations in genes which appear to affect the epigenome of MPN patients have been discovered including mutations in TET2, IDH1/ 2, EZH2, and ASXL1. Lastly, some insights into the genetic events which contribute to transformation of a chronic MPN phenotype to acute myeloid leukemia have been elucidated, including deletion of the transcription factor Ikaros. SUMMARY The spectrum of genetic abnormalities found in the classic MPNs has increased over the last 6 years and somatic mutations in JAK2, MPL, LNK, TET2, EZH2, ASXL1, and IDH1/2 have all been described. Despite this, the initiating genetic events responsible for the development of MPNs is still not totally understood.
Collapse
|
32
|
Agrawal M, Garg RJ, Cortes J, Kantarjian H, Verstovsek S, Quintas-Cardama A. Experimental therapeutics for patients with myeloproliferative neoplasias. Cancer 2011; 117:662-76. [PMID: 20922795 DOI: 10.1002/cncr.25672] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2010] [Revised: 06/28/2010] [Accepted: 08/05/2010] [Indexed: 02/03/2023]
Abstract
Philadelphia chromosome (Ph)-negative myeloproliferative neoplasms (MPNs) are characterized by stem cell-derived, unrestrained clonal myeloproliferation. The World Health Organization classification system, proposed in 2008, identifies 7 distinct categories of Ph-negative MPNs including essential thrombocythemia (ET); polycythemia vera (PV); primary myelofibrosis (PMF); mastocytosis; chronic eosinophilic leukemia; chronic neutrophilic leukemia; and MPN, unclassifiable. For many years, the treatment of ET, PV, and PMF, the most frequently diagnosed Ph-negative MPNs, has been largely supportive. In recent years, that paradigm has been challenged because of the discovery of a recurrent point mutation in the Janus kinase 2 (JAK2) gene (JAK2(V617F)). This mutation can be detected in the vast majority of patients with PV and approximately half of patients with ET or PMF and serves as both a diagnostic marker as well as representing a putative molecular target for drug development. Several putative targeted agents with significant in vitro JAK2 inhibitory activity and various degrees of JAK2 specificity are currently undergoing clinical evaluation. Furthermore, other investigational non-tyrosine kinase inhibitor approaches such as immunomodulatory agents and pegylated interferon- have also shown promising results in MPNs.
Collapse
Affiliation(s)
- Meetu Agrawal
- Department of Leukemia, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA.
| | | | | | | | | | | |
Collapse
|
33
|
Swierczek SI, Yoon D, Bellanné-Chantelot C, Kim SJ, Saint-Martin C, Delhommeau F, Najman A, Prchal JT. Extent of hematopoietic involvement by TET2 mutations in JAK2V⁶¹⁷F polycythemia vera. Haematologica 2011; 96:775-8. [PMID: 21273266 DOI: 10.3324/haematol.2010.029678] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
TET2 mutations are found in polycythemia vera and it was initially reported that there is a greater TET2 mutational burden than JAK2(V617F) in polycythemia vera stem cells and that TET2 mutations precede JAK2(V617F). We quantified the proportion of TET2, JAK2(V617F) mutations and X-chromosome allelic usage in polycythemia vera cells, BFU-Es and in vitro expanded erythroid progenitors and found clonal reticulocytes, granulocytes, platelets and CD34(+) cells. We found that TET2 mutations may also follow rather than precede JAK2(V617F) as recently reported by others. Only a fraction of clonal early hematopoietic precursors and largely polyclonal T cells carry the TET2 mutation. We showed that in vitro the concomitant presence of JAK2(V617F) and TET2 mutations favors clonal polycythemia vera erythroid progenitors in contrast with non-TET2 mutated progenitors. We conclude that loss-of-function TET2 mutations are not the polycythemia vera initiating events and that the acquisition of TET2 somatic mutations may increase the aggressivity of the polycythemia vera clone.
Collapse
|
34
|
Abstract
The process of megakaryopoiesis and platelet production is complex, with the potential for regulation at multiple stages. Megakaryocytes are derived from the hematopoietic stem cell through successive lineage commitment steps, and they undergo a unique maturation process that includes polyploidization, development of an extensive internal demarcation membrane system, and finally formation of pro-platelet processes. Platelets are shed from these processes into vascular sinusoids within the bone marrow. Megakaryocyte differentiation is regulated both positively and negatively by transcription factors and cytokine signaling. Thrombopoietin (TPO) is the most important hematopoietic cytokine for platelet production. Clinically, acquired and inherited mutations affecting megakaryocytic transcription factors and thrombopoietin signaling have been identified in disorders of thrombocytopenia and thrombocytosis.
Collapse
Affiliation(s)
- Amy E Geddis
- Pediatric Hematology-Oncology, University of California, San Diego, La Jolla, CA 92023, USA.
| |
Collapse
|
35
|
Olcaydu D, Rumi E, Harutyunyan A, Passamonti F, Pietra D, Pascutto C, Berg T, Jäger R, Hammond E, Cazzola M, Kralovics R. The role of the JAK2 GGCC haplotype and the TET2 gene in familial myeloproliferative neoplasms. Haematologica 2010; 96:367-74. [PMID: 21173100 DOI: 10.3324/haematol.2010.034488] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Myeloproliferative neoplasms constitute a group of diverse chronic myeloid malignancies that share pathogenic features such as acquired mutations in the JAK2, TET2, CBL and MPL genes. There are recent reports that a JAK2 gene haplotype (GGCC or 46/1) confers susceptibility to JAK2 mutation-positive myeloproliferative neoplasms. The aim of this study was to examine the role of the JAK2 GGCC haplotype and germline mutations of TET2, CBL and MPL in familial myeloproliferative neoplasms. DESIGN AND METHODS We investigated patients with familial (n=88) or sporadic (n=684) myeloproliferative neoplasms, and a control population (n=203) from the same demographic area in Italy. Association analysis was performed using tagged single nucleotide polymorphisms (rs10974944 and rs12343867) of the JAK2 haplotype. Sequence analysis of TET2, CBL and MPL was conducted in the 88 patients with familial myeloproliferative neoplasms. RESULTS Association analysis revealed no difference in haplotype frequency between familial and sporadic cases of myeloproliferative neoplasms (P=0.6529). No germline mutations in TET2, CBL or MPL that segregate with the disease phenotype were identified. As we observed variability in somatic mutations in the affected members of a pedigree with myeloproliferative neoplasms, we postulated that somatic mutagenesis is increased in familial myeloproliferative neoplasms. Accordingly, we compared the incidence of malignant disorders between sporadic and familial patients. Although the overall incidence of malignant disorders did not differ significantly between cases of familial and sporadic myeloproliferative neoplasms, malignancies were more frequent in patients with familial disease aged between 50 to 70 years (P=0.0198) than in patients in the same age range with sporadic myeloproliferative neoplasms. CONCLUSIONS We conclude that the JAK2 GGCC haplotype and germline mutations of TET2, CBL or MPL do not explain familial clustering of myeloproliferative neoplasms. As we observed an increased frequency of malignant disorders in patients with familial myeloproliferative neoplasms, we hypothesize that the germline genetic lesions that underlie familial clustering of myeloproliferative neoplasms predispose to somatic mutagenesis that is not restricted to myeloid hematopoietic cells but cause an increase in overall carcinogenesis.
Collapse
Affiliation(s)
- Damla Olcaydu
- Center for Molecular Medicine, Austrian Academy of Sciences, Lazarettgasse 14, AKH BT25.3, 1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Ye Z, Cheng L. Potential of human induced pluripotent stem cells derived from blood and other postnatal cell types. Regen Med 2010; 5:521-30. [PMID: 20632856 DOI: 10.2217/rme.10.38] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Human induced pluripotent stem (iPS) cells have been generated from various cell types including blood cells, and offer certain advantages as a starting population for reprogramming postnatal somatic cells. Unlike adult stem cells, iPS cells can proliferate limitlessly in culture while retaining their potential to differentiate into any cell type, including hematopoietic lineages. Derivation of patient-specific iPS cells, in combination with improved hematopoietic differentiation protocols, provides an alternative to generate histocompatible stem cells for bone marrow transplantation. In addition, the ability to reprogram blood cells and redifferentiate iPS cells back to hematopoietic lineages provides opportunities to establish novel models for acquired and inherited blood diseases. This article will summarize recent progress in human iPS cells derived from blood cells and hematopoietic differentiation from iPS cells. Advantages of blood as a source for reprogramming and applications in regenerative medicine will be discussed.
Collapse
Affiliation(s)
- Zhaohui Ye
- Division of Hematology & Stem Cell Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Broadway Research Building, Room 747, 733 N. Broadway, Baltimore, MD 21205, USA
| | | |
Collapse
|
37
|
Ayad MW, Nafea D. Acquired mutation of the tyrosine kinase JAK2V617F in Egyptian patients with myeloid disorders. Genet Test Mol Biomarkers 2010; 15:17-21. [PMID: 21034166 DOI: 10.1089/gtmb.2010.0093] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Janus Kinase 2 (JAK2) is a member of a family of four Janus Kinases, 2, and 3 and tyrosine kinase 2. Mutated JAK2 (V617F) has the ability to activate downstream signal transducer and activator of transcription (STAT)-mediated transcription in the absence of the ligand erythropoietin. The autoinhibitory activity of JAK2 is disrupted by the presence of the V617F mutation. Somatic mutation in JAK2 (V617F) gene has been reported in myeloid disorders. This study reports the prevalence of JAK2V617F using amplification refractory mutation system (ARMS)-polymerase chain reaction in 246 Egyptian patients with different myeloid disorders and studied the relationship between the JAK2V617F mutation and parameters in peripheral blood. The mutation was detected among 88 patients (35.8%) with different myeloid disorders. JAK2V617F was found among 81.4% of polycythemia vera (PV), 50% of essential thrombocythemia, 46.1% of primary myelofibrosis (PMF), 33.3% of philadelphia (Ph)-negative chronic myeloid leukemia, 33.3% of myelodysplastic syndrome (MDS)/myeloproliferative neoplasm (MPN), and 50% of refractory anemia with ringed sideroblasts associated with marked thrombocytosis (RARS-T) patients. Hemoglobin and white blood cells were significantly higher in the mutated group of MPN including PV, essential thrombocythemia, and PMF, whereas platelet counts were higher among the mutated PV, PMF, RARS-T, and MDS/MPN group. The identification of JAK2V617F mutations has raised the prospect of developing specific JAK2V617F inhibitors to treat mutated patients.
Collapse
Affiliation(s)
- Mona Wagdy Ayad
- Clinical Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | | |
Collapse
|
38
|
Churpek JE, Onel K. Heritability of hematologic malignancies: from pedigrees to genomics. Hematol Oncol Clin North Am 2010; 24:939-72. [PMID: 20816581 DOI: 10.1016/j.hoc.2010.06.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Many hematologic malignancies have an underlying heritable component. Although not as well characterized as the acquired genetic abnormalities that define important prognostic and therapeutic subgroups of myeloid and lymphoid neoplasms, investigations are beginning to unravel the role of germline genetic variation in the predisposition to hematologic malignancies. Information gained from the study of striking family pedigrees, epidemiologic data, and candidate genes are now being combined with unbiased genome-wide investigations to outline the network of genetic abnormalities that contribute to hematologic malignancy risk. This article reviews the current understanding of the heritability of hematologic malignancies in the genomics era.
Collapse
Affiliation(s)
- Jane E Churpek
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
39
|
Abstract
The 46/1 JAK2 haplotype predisposes to V617F-positive myeloproliferative neoplasms, but the underlying mechanism is obscure. We analyzed essential thrombocythemia patients entered into the PT-1 studies and, as expected, found that 46/1 was overrepresented in V617F-positive cases (n = 404) versus controls (n = 1492, P = 3.9 x 10(-11)). The 46/1 haplotype was also overrepresented in cases without V617F (n = 347, P = .009), with an excess seen for both MPL exon 10 mutated and V617F, MPL exon 10 nonmutated cases. Analysis of further MPL-positive, V617F-negative cases confirmed an excess of 46/1 (n = 176, P = .002), but no association between MPL mutations and MPL haplotype was seen. An excess of 46/1 was also seen in JAK2 exon 12 mutated cases (n = 69, P = .002), and these mutations preferentially arose on the 46/1 chromosome (P = .029). No association between 46/1 and clinical or laboratory features was seen in the PT-1 cohort either with or without V617F. The excess of 46/1 in JAK2 exon 12 cases is compatible with both the "hypermutability" and "fertile ground" hypotheses, but the excess in MPL-mutated cases argues against the former. No difference in sequence, splicing, or expression of JAK2 was found on 46/1 compared with other haplotypes, suggesting that any functional difference of JAK2 on 46/1, if it exists, must be relatively subtle.
Collapse
|
40
|
Seaman V, Dearwent SM, Gable D, Lewis B, Metcalf S, Orloff K, Tierney B, Zhu J, Logue J, Marchetto D, Ostroff S, Hoffman R, Xu M, Carey D, Erlich P, Gerhard G, Roda P, Iannuzzo J, Lewis R, Mellow J, Mulvihill L, Myles Z, Wu M, Frank A, Gross-Davis CA, Klotz J, Lynch A, Weissfeld J, Weinberg R, Cole H. A multidisciplinary investigation of a polycythemia vera cancer cluster of unknown origin. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2010; 7:1139-52. [PMID: 20617023 PMCID: PMC2872321 DOI: 10.3390/ijerph7031139] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Revised: 03/13/2010] [Accepted: 03/16/2010] [Indexed: 11/16/2022]
Abstract
Cancer cluster investigations rarely receive significant public health resource allocations due to numerous inherent challenges and the limited success of past efforts. In 2008, a cluster of polycythemia vera, a rare blood cancer with unknown etiology, was identified in northeast Pennsylvania. A multidisciplinary group of federal and state agencies, academic institutions, and local healthcare providers subsequently developed a multifaceted research portfolio designed to better understand the cause of the cluster. This research agenda represents a unique and important opportunity to demonstrate that cancer cluster investigations can produce desirable public health and scientific outcomes when necessary resources are available.
Collapse
Affiliation(s)
- Vincent Seaman
- Agency for Toxic Substances and Disease Registry, 4770 Buford Highway NE, Atlanta, GA, 30341, USA; E-Mails:
(S.D.);
(D.G.);
(B.L.);
(S.M.);
(K.O.);
(B.T.);
(J.Z.)
- Author to whom correspondence should be addressed; E-Mail:
; Tel.: +1-770-488-3682; Fax: +1-770-488-1537
| | - Steve M Dearwent
- Agency for Toxic Substances and Disease Registry, 4770 Buford Highway NE, Atlanta, GA, 30341, USA; E-Mails:
(S.D.);
(D.G.);
(B.L.);
(S.M.);
(K.O.);
(B.T.);
(J.Z.)
| | - Debra Gable
- Agency for Toxic Substances and Disease Registry, 4770 Buford Highway NE, Atlanta, GA, 30341, USA; E-Mails:
(S.D.);
(D.G.);
(B.L.);
(S.M.);
(K.O.);
(B.T.);
(J.Z.)
| | - Brian Lewis
- Agency for Toxic Substances and Disease Registry, 4770 Buford Highway NE, Atlanta, GA, 30341, USA; E-Mails:
(S.D.);
(D.G.);
(B.L.);
(S.M.);
(K.O.);
(B.T.);
(J.Z.)
| | - Susan Metcalf
- Agency for Toxic Substances and Disease Registry, 4770 Buford Highway NE, Atlanta, GA, 30341, USA; E-Mails:
(S.D.);
(D.G.);
(B.L.);
(S.M.);
(K.O.);
(B.T.);
(J.Z.)
| | - Ken Orloff
- Agency for Toxic Substances and Disease Registry, 4770 Buford Highway NE, Atlanta, GA, 30341, USA; E-Mails:
(S.D.);
(D.G.);
(B.L.);
(S.M.);
(K.O.);
(B.T.);
(J.Z.)
| | - Bruce Tierney
- Agency for Toxic Substances and Disease Registry, 4770 Buford Highway NE, Atlanta, GA, 30341, USA; E-Mails:
(S.D.);
(D.G.);
(B.L.);
(S.M.);
(K.O.);
(B.T.);
(J.Z.)
| | - Jane Zhu
- Agency for Toxic Substances and Disease Registry, 4770 Buford Highway NE, Atlanta, GA, 30341, USA; E-Mails:
(S.D.);
(D.G.);
(B.L.);
(S.M.);
(K.O.);
(B.T.);
(J.Z.)
| | - James Logue
- Pennsylvania Department of Health, 7 & Forster Streets, Harrisburg, PA 17120, USA; E-Mails:
(J.L.);
(D.M.);
(S.O.)
| | - David Marchetto
- Pennsylvania Department of Health, 7 & Forster Streets, Harrisburg, PA 17120, USA; E-Mails:
(J.L.);
(D.M.);
(S.O.)
| | - Stephen Ostroff
- Pennsylvania Department of Health, 7 & Forster Streets, Harrisburg, PA 17120, USA; E-Mails:
(J.L.);
(D.M.);
(S.O.)
| | - Ronald Hoffman
- Mt. Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029-6574, USA; E-Mails:
(R.H.);
(M.X.)
| | - Mingjiang Xu
- Mt. Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029-6574, USA; E-Mails:
(R.H.);
(M.X.)
| | - David Carey
- Geisinger Health System/Clinic, 100 N. Academy Ave, Danville, PA 17822, USA; E-Mails:
(D.C.);
(P.E.);
(G.G.);
(P.R.)
| | - Porat Erlich
- Geisinger Health System/Clinic, 100 N. Academy Ave, Danville, PA 17822, USA; E-Mails:
(D.C.);
(P.E.);
(G.G.);
(P.R.)
| | - Glenn Gerhard
- Geisinger Health System/Clinic, 100 N. Academy Ave, Danville, PA 17822, USA; E-Mails:
(D.C.);
(P.E.);
(G.G.);
(P.R.)
| | - Paul Roda
- Geisinger Health System/Clinic, 100 N. Academy Ave, Danville, PA 17822, USA; E-Mails:
(D.C.);
(P.E.);
(G.G.);
(P.R.)
| | - Joseph Iannuzzo
- Pennsylvania Department of Environmental Protection, 2 Public Square, Wilkes-Barre, PA 18711, USA; E-Mails:
(J.I.);
(R.L.);
(J.M.)
| | - Robert Lewis
- Pennsylvania Department of Environmental Protection, 2 Public Square, Wilkes-Barre, PA 18711, USA; E-Mails:
(J.I.);
(R.L.);
(J.M.)
| | - John Mellow
- Pennsylvania Department of Environmental Protection, 2 Public Square, Wilkes-Barre, PA 18711, USA; E-Mails:
(J.I.);
(R.L.);
(J.M.)
| | - Linda Mulvihill
- Centers for Disease Control and Prevention, National Program of Cancer Registries, 1600 Clifton, Rd NE, Atlanta, GA 30333, USA; E-Mails:
(L.M.);
(Z.M.);
(M.W.)
| | - Zachary Myles
- Centers for Disease Control and Prevention, National Program of Cancer Registries, 1600 Clifton, Rd NE, Atlanta, GA 30333, USA; E-Mails:
(L.M.);
(Z.M.);
(M.W.)
| | - Manxia Wu
- Centers for Disease Control and Prevention, National Program of Cancer Registries, 1600 Clifton, Rd NE, Atlanta, GA 30333, USA; E-Mails:
(L.M.);
(Z.M.);
(M.W.)
| | - Arthur Frank
- Drexel University School of Public Health, 1505 Race Street, Bellet Building 13th Floor, Philadelphia, PA, 19102, USA; E-Mails:
(A.F.);
(C.A.G);
(J.K.)
| | - Carol Ann Gross-Davis
- Drexel University School of Public Health, 1505 Race Street, Bellet Building 13th Floor, Philadelphia, PA, 19102, USA; E-Mails:
(A.F.);
(C.A.G);
(J.K.)
| | - Judith Klotz
- Drexel University School of Public Health, 1505 Race Street, Bellet Building 13th Floor, Philadelphia, PA, 19102, USA; E-Mails:
(A.F.);
(C.A.G);
(J.K.)
| | - Adam Lynch
- Drexel University School of Public Health, 1505 Race Street, Bellet Building 13th Floor, Philadelphia, PA, 19102, USA; E-Mails:
(A.F.);
(C.A.G);
(J.K.)
| | - Joel Weissfeld
- UPMC Cancer Pavilion, 3rd Floor, 5150 Centre Avenue, Pittsburgh, PA 15232, USA; E-Mail:
| | - Rona Weinberg
- New York Blood Center, 310 East 67th Street, 2-47B, New York, NY 10065, USA; E-Mail:
| | - Henry Cole
- Henry S. Cole & Associates, 7611 S. Osborne Rd, Ste 201, Upper Marlboro, MD 20772, USA; E-Mail:
| |
Collapse
|
41
|
Abstract
Single nucleotide polymorphism arrays (SNP-A) have recently been widely applied as a powerful karyotyping tool in numerous translational cancer studies. SNP-A complements traditional metaphase cytogenetics with the unique ability to delineate a previously hidden chromosomal defect, copy neutral loss of heterozygosity (CN-LOH). Emerging data demonstrate that selected hematologic malignancies exhibit abundant CN-LOH, often in the setting of a normal metaphase karyotype and no previously identified clonal marker. In this review, we explore emerging biologic and clinical features of CN-LOH relevant to hematologic malignancies. In myeloid malignancies, CN-LOH has been associated with the duplication of oncogenic mutations with concomitant loss of the normal allele. Examples include JAK2, MPL, c-KIT, and FLT3. More recent investigations have focused on evaluation of candidate genes contained in common CN-LOH and deletion regions and have led to the discovery of tumor suppressor genes, including c-CBL and family members, as well as TET2. Investigations into the underlying mechanisms generating CN-LOH have great promise for elucidating general cancer mechanisms. We anticipate that further detailed characterization of CN-LOH lesions will probably facilitate our discovery of a more complete set of pathogenic molecular lesions, disease and prognosis markers, and better understanding of the initiation and progression of hematologic malignancies.
Collapse
|
42
|
Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm. Blood 2009; 115:2891-900. [PMID: 20008300 DOI: 10.1182/blood-2009-08-236596] [Citation(s) in RCA: 231] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Acute myeloid leukemia (AML) may follow a JAK2-positive myeloproliferative neoplasm (MPN), although the mechanisms of disease evolution, often involving loss of mutant JAK2, remain obscure. We studied 16 patients with JAK2-mutant (7 of 16) or JAK2 wild-type (9 of 16) AML after a JAK2-mutant MPN. Primary myelofibrosis or myelofibrotic transformation preceded all 7 JAK2-mutant but only 1 of 9 JAK2 wild-type AMLs (P = .001), implying that JAK2-mutant AML is preceded by mutation(s) that give rise to a "myelofibrosis" phenotype. Loss of the JAK2 mutation by mitotic recombination, gene conversion, or deletion was excluded in all wild-type AMLs. A search for additional mutations identified alterations of RUNX1, WT1, TP53, CBL, NRAS, and TET2, without significant differences between JAK2-mutant and wild-type leukemias. In 4 patients, mutations in TP53, CBL, or TET2 were present in JAK2 wild-type leukemic blasts but absent from the JAK2-mutant MPN. By contrast in a chronic-phase patient, clones harboring mutations in JAK2 or MPL represented the progeny of a shared TET2-mutant ancestral clone. These results indicate that different pathogenetic mechanisms underlie transformation to JAK2 wild-type and JAK2-mutant AML, show that TET2 mutations may be present in a clone distinct from that harboring a JAK2 mutation, and emphasize the clonal heterogeneity of the MPNs.
Collapse
|
43
|
Caocci G, Atzeni S, Vacca A, Orrù N, Ledda A, La Nasa G. Familial occurrence of chronic myeloid leukemia. Leuk Lymphoma 2009; 50:854-6. [PMID: 19452324 DOI: 10.1080/10428190902842634] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
44
|
Usenko T, Eskinazi D, Correa PN, Amato D, Ben-David Y, Axelrad AA. Overexpression of SOCS-2 and SOCS-3 genes reverses erythroid overgrowth and IGF-I hypersensitivity of primary polycythemia vera (PV) cells. Leuk Lymphoma 2009; 48:134-46. [PMID: 17325857 DOI: 10.1080/10428190601043138] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Polycythemia vera (PV), an acquired, chronic, clonal disorder arising in a multipotential hematopoietic progenitor cell, is characterized by hyperplasia of three major myeloid lineages, with a pronounced increase in cells of the erythroid lineage. Erythroid progenitor cells in PV are strikingly hypersensitive to insulin-like growth factor-I (IGF-I); this effect is specific and is mediated through the IGF-I receptor. To investigate the possibility that in PV the increase in number of erythroid progenitors and their hypersensitivity to IGF-I result from a defect in negative regulation of cytokine activity, we examined the expression of members of the SOCS gene family. Circulating mononuclear cells, grown in serum-free methylcellulose medium in the presence of IGF-I, produced BFU-E-derived colonies whose cells revealed a reduction of SOCS-2 and SOCS-3 expression in PV only. Overexpression of these genes in transfected PV cells reduced their erythroid overgrowth and IGF-I hypersensitivity. We hypothesize that a defect in expression of SOCS-2 and SOCS-3 genes may be crucial for the IGF-I hypersensitivity and progressive increase in erythroid cell population size characteristic of PV.
Collapse
Affiliation(s)
- Tatiana Usenko
- Department of Molecular and Cellular Biology, Sunnybrook and Women's College Health Sciences Centre, Toronto, Canada
| | | | | | | | | | | |
Collapse
|
45
|
Analysis of the ten-eleven translocation 2 (TET2) gene in familial myeloproliferative neoplasms. Blood 2009; 114:1628-32. [PMID: 19564637 DOI: 10.1182/blood-2009-01-197525] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The JAK2(V617F) mutation does not elucidate the phenotypic variability observed in myeloproliferative neoplasm (MPN) families. A putative tumor suppressor gene, TET2, was recently implicated in MPN and myelodysplastic syndromes through the identification of acquired mutations affecting hematopoietic stem cells. The present study analyzed the TET2 gene in 61 MPN cases from 42 families. Fifteen distinct mutations were identified in 12 (20%) JAK2(V617F)-positive or -negative patients. In a patient with 2 TET2 mutations, the analysis of 5 blood samples at different phases of her disease showed the sequential occurrence of JAK2(V617F) and TET2 mutations concomitantly to the disease evolution. Analysis of familial segregation confirmed that TET2 mutations were not inherited but somatically acquired. TET2 mutations were mainly observed (10 of 12) in patients with primary myelofibrosis or patients with polycythemia vera or essential thrombocythemia who secondarily evolved toward myelofibrosis or acute myeloid leukemia.
Collapse
|
46
|
Vannucchi AM, Masala G, Antonioli E, Chiara Susini M, Guglielmelli P, Pieri L, Maggi L, Caini S, Palli D, Bogani C, Ponziani V, Pancrazzi A, Annunziato F, Bosi A. Increased Risk of Lymphoid Neoplasms in Patients with Philadelphia Chromosome–Negative Myeloproliferative Neoplasms. Cancer Epidemiol Biomarkers Prev 2009; 18:2068-73. [DOI: 10.1158/1055-9965.epi-09-0353] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
47
|
Percy MJ, Rumi E. Genetic origins and clinical phenotype of familial and acquired erythrocytosis and thrombocytosis. Am J Hematol 2009; 84:46-54. [PMID: 19006225 DOI: 10.1002/ajh.21313] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Familial and acquired erythrocytosis and thrombocytosis are characterized by myeloid lineage hyperproliferation, which is either single or multi-lineage in origin. The single lineage disorders exhibit Mendelian inheritance with polyclonal hematopoiesis and often arise from a single genetic defect. In contrast, the multi-lineage disorders exhibit complex patterns of inheritance with multi-genetic origins and clonal hematopoiesis. They have the potential to acquire JAK2 somatic mutations, but this is not the primary event. Identification of the disease-causing genes will enable better classification of familial and acquired erythrocytosis and thrombocytosis. Furthermore, it will provide an insight into the mechanisms regulating myeloid cell proliferation.
Collapse
Affiliation(s)
- Melanie J Percy
- Department of Haematology, Belfast City Hospital, Belfast, Northern Ireland, UK.
| | | |
Collapse
|
48
|
Abstract
Major progress in understanding the pathogenesis in patients with thrombocytosis has been made by identifying mutations in the key regulators of thrombopoietin: the thrombopoietin receptor MPL and JAK2. Together, these mutations can be found in 50% to 60% of patients with essential thrombocythemia or primary myelofibrosis and in 10% to 20% of hereditary thrombocytosis. A decrease in expression of the Mpl protein can cause thrombocytosis even in the absence of mutations in the coding sequence, due to a shift in the balance between stimulation of signaling in megakaryopoiesis and removal of thrombopoietin by receptor mediated internalization in platelets. When present in a heterozygous state the JAK2-V617F mutation preferentially stimulates megakaryopoiesis and in most cases manifests as essential thrombocythemia (ET), whereas homozygous JAK2-V617F reduces megakaryopoiesis in favor of increased erythropoiesis, resulting in polycythemia vera and/or myelofibrosis. In 30% to 40% of patients with ET or primary myelofibrosis (PMF) and in 80% to 90% of pedigrees with hereditary thrombocytosis the disease-causing gene remains unknown. Ongoing genetic and genomic screens have identified genes that, when mutated, can cause thrombocytosis in mouse models. A more complete picture of the pathways that regulate megakaryopoisis and platelet production will be important for finding new ways of controlling platelet production in patients with thrombocytosis.
Collapse
Affiliation(s)
- Radek C Skoda
- Experimental Hematology, Department of Biomedicine, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
49
|
BCR-ABL1-positive CML and BCR-ABL1-negative chronic myeloproliferative disorders: some common and contrasting features. Leukemia 2008; 22:1975-89. [PMID: 19002192 DOI: 10.1038/leu.2008.231] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
50
|
|