1
|
Song J, Lanikova L, Kim SJ, Papadopoulos N, Meznarich J, Constantinescu SN, Parsegov B, Prchal JF, Prchal JT. Novel germline JAK2 R715T mutation causing PV-like erythrocytosis in 3 generations. Amelioration by Ropeg-Interferon. Am J Hematol 2024; 99:1220-1229. [PMID: 38629639 DOI: 10.1002/ajh.27311] [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: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 06/12/2024]
Abstract
Polycythemia vera (PV) is a clonal disorder arising from the acquired somatic mutations of the JAK2 gene, including JAK2V617F or several others in exon 12. A 38-year-old female had a stroke at age 32 and found to have elevated hemoglobin, normal leukocytes, normal platelets, and tested negative for JAK2V617F and exon 12 mutations. Next generation sequencing revealed a novel mutation: JAK2R715T in the pseudokinase domain (JH2) at 47.5%. Its presence in her nail DNA confirmed a germline origin. Her mother and her son similarly had erythrocytosis and a JAK2R715T mutation. Computer modeling indicated gain-of-function JAK2 activity. The propositus and her mother had polyclonal myelopoiesis, ruling out another somatic mutation-derived clonal hematopoiesis. Some erythroid progenitors of all three generations grew without erythropoietin, a hallmark of PV. The in vitro reporter assay confirmed increased activity of the JAK2R715T kinase. Similar to PV, the JAK2R715T native cells have increased STAT5 phosphorylation, augmented transcripts of prothrombotic and inflammatory genes, and decreased KLF2 transcripts. The propositus was not controlled by hydroxyurea, and JAK2 inhibitors were not tolerated; however, Ropeginterferon-alfa-2b (Ropeg-IFN-α) induced a remission. Ropeg-IFN-α treatment also reduced JAK2 activity in the propositus, her mother and JAK2V617F PV subjects. We report dominantly inherited erythrocytosis secondary to a novel germline JAK2R715T gain-of-function mutation with many but not all comparable molecular features to JAK2V617F PV. We also document a previously unreported inhibitory mechanism of JAK2 signaling by Ropeg-IFN-α.
Collapse
Affiliation(s)
- Jihyun Song
- Division of Hematology & Hematologic Malignancies, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Lucie Lanikova
- Department of Cell and Developmental Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Soo Jin Kim
- Division of Hematology & Hematologic Malignancies, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Nicolas Papadopoulos
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, Brussels, Belgium
| | - Jessica Meznarich
- Division of Hematology-Oncology, Department of Pediatrics, University of Utah and Primary Children's Hospital, Salt Lake City, Utah, USA
| | - Stefan N Constantinescu
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, Brussels, Belgium
- Nuffield Department of Medicine, Oxford University, Oxford, UK
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Brynn Parsegov
- Division of Hematology & Hematologic Malignancies, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | | | - Josef T Prchal
- Division of Hematology & Hematologic Malignancies, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
2
|
Benetti A, Bertozzi I, Ceolotto G, Cortella I, Regazzo D, Biagetti G, Cosi E, Randi ML. Coexistence of Multiple Gene Variants in Some Patients with Erythrocytoses. Mediterr J Hematol Infect Dis 2024; 16:e2024021. [PMID: 38468832 PMCID: PMC10927185 DOI: 10.4084/mjhid.2024.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/10/2024] [Indexed: 03/13/2024] Open
Abstract
Background Erythrocytosis is a relatively common condition; however, a large proportion of these patients (70%) remain without a clear etiologic explanation. Methods We set up a targeted NGS panel for patients with erythrocytosis, and 118 sporadic patients with idiopathic erythrocytosis were studied. Results In 40 (34%) patients, no variant was found, while in 78 (66%), we identified at least one germinal variant; 55 patients (70.5%) had 1 altered gene, 18 (23%) had 2 alterations, and 5 (6.4%) had 3. An altered HFE gene was observed in 51 cases (57.1%), EGLN1 in 18 (22.6%) and EPAS1, EPOR, JAK2, and TFR2 variants in 7.7%, 10.3%, 11.5%, and 14.1% patients, respectively. In 23 patients (19.45%), more than 1 putative variant was found in multiple genes. Conclusions Genetic variants in patients with erythrocytosis were detected in about 2/3 of our cohort. An NGS panel including more candidate genes should reduce the number of cases diagnosed as "idiopathic" erythrocytosis in which a cause cannot yet be identified. It is known that HFE variants are common in idiopathic erythrocytosis. TFR2 alterations support the existence of a relationship between genes involved in iron metabolism and impaired erythropoiesis. Some novel multiple variants were identified. Erythrocytosis appears to be often multigenic.
Collapse
Affiliation(s)
- Andrea Benetti
- First Medical Clinic, Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Irene Bertozzi
- First Medical Clinic, Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Giulio Ceolotto
- Emergency Medicine, Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Irene Cortella
- First Medical Clinic, Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Daniela Regazzo
- First Medical Clinic, Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Giacomo Biagetti
- First Medical Clinic, Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Elisabetta Cosi
- First Medical Clinic, Department of Medicine – DIMED, University of Padova, Padova, Italy
| | - Maria Luigia Randi
- First Medical Clinic, Department of Medicine – DIMED, University of Padova, Padova, Italy
| |
Collapse
|
3
|
Irene B, Andrea B, Daniela R, Francesca P, Giacomo B, Giulio C, Luigia RM. Targeted NGS analysis reveals a complex genetic background of idiopathic erythrocytosis in a large Venetian family. Genes Dis 2024; 11:561-563. [PMID: 37692494 PMCID: PMC10491904 DOI: 10.1016/j.gendis.2023.03.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/22/2023] [Accepted: 03/29/2023] [Indexed: 09/12/2023] Open
Affiliation(s)
- Bertozzi Irene
- First Medical Clinic, University of Padua Department of Medicine – DIMED, Via Giustiniani 2, Padova 35123, Italy
| | - Benetti Andrea
- First Medical Clinic, University of Padua Department of Medicine – DIMED, Via Giustiniani 2, Padova 35123, Italy
| | - Regazzo Daniela
- First Medical Clinic, University of Padua Department of Medicine – DIMED, Via Giustiniani 2, Padova 35123, Italy
| | - Polese Francesca
- Transfusion Medicine Department, Ospedale dell'Angelo - Venezia Mestre, Via Paccagnella 1, Venice 30174, Italy
| | - Biagetti Giacomo
- First Medical Clinic, University of Padua Department of Medicine – DIMED, Via Giustiniani 2, Padova 35123, Italy
| | - Ceolotto Giulio
- Emergency Medicine, University of Padua Department of Medicine – DIMED, Via Giustiniani 2, Padova 35123, Italy
| | - Randi Maria Luigia
- First Medical Clinic, University of Padua Department of Medicine – DIMED, Via Giustiniani 2, Padova 35123, Italy
| |
Collapse
|
4
|
Torres DG, Barbosa Alves EV, Araújo de Sousa M, Laranjeira WH, Paes J, Alves E, Canté D, Costa AG, Malheiro A, Abreu R, Nascimento L, Fraiji NA, Silva GA, Mourão LPDS, Tarragô AM. Molecular landscape of the JAK2 gene in chronic myeloproliferative neoplasm patients from the state of Amazonas, Brazil. Biomed Rep 2023; 19:98. [PMID: 37954635 PMCID: PMC10633817 DOI: 10.3892/br.2023.1680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/22/2023] [Indexed: 11/14/2023] Open
Abstract
JAK2V617F (dbSNP: rs77375493) is the most frequent and most-studied variant in BCR::ABL1 negative myeloproliferative neoplasms and in the JAK2 gene. The present study aimed to molecularly characterize variants in the complete coding region of the JAK2 gene in patients with BCR::ABL1 negative chronic myeloproliferative neoplasms. The study included 97 patients with BCR::ABL1 negative myeloproliferative neoplasms, including polycythemia vera (n=38), essential thrombocythemia (n=55), and myelofibrosis (n=04). Molecular evaluation was performed using conventional PCR and Sanger sequencing to detect variants in the complete coding region of the JAK2 gene. The presence of missense variants in the JAK2 gene including rs907414891, rs2230723, rs77375493 (JAK2V617F), and rs41316003 were identified. The coexistence of variants was detected in polycythemia vera and essential thrombocythemia. Thus, individuals with high JAK2V617F variant allele frequency (≥50% VAF) presented more thrombo-hemorrhagic events and manifestations of splenomegaly compared with those with low JAK2V617F variant allele frequency (<50% VAF). In conclusion, individuals with BCR::ABL1 negative neoplasms can display >1 variant in the JAK2 gene, especially rs2230722, rs2230724, and rs77375493 variants, and those with high JAK2V617F VAF show alterations in the clinical-laboratory profile compared with those with low JAK2V617F VAF.
Collapse
Affiliation(s)
- Dania G. Torres
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
- Molecular Biology Center, University of Central America, Managua 14003, Nicaragua
| | - Emanuela V. Barbosa Alves
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Miliane Araújo de Sousa
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Wanessa H. Laranjeira
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Jhemerson Paes
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Erycka Alves
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Deborah Canté
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Allyson G. Costa
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
- Post-graduate Program in Basic and Applied Immunology, Federal University of Amazonas, Manaus, Amazonas State 69067-005, Brazil
- Manaus School of Nursing, Federal University of Amazonas, Manaus, Amazonas State 69057-070, Brazil
- Amazon Genomic Health Surveillance Network Coordination, Manaus, Amazonas State 69040-010, Brazil
| | - Adriana Malheiro
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
- Post-graduate Program in Basic and Applied Immunology, Federal University of Amazonas, Manaus, Amazonas State 69067-005, Brazil
- Amazon Genomic Health Surveillance Network Coordination, Manaus, Amazonas State 69040-010, Brazil
| | - Rosângela Abreu
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Leny Nascimento
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - Nelson A. Fraiji
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
| | - George A.V. Silva
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Amazon Genomic Health Surveillance Network Coordination, Manaus, Amazonas State 69040-010, Brazil
- Leonidas and Maria Deane Institute, Oswaldo Cruz Foundation, Manaus, Amazonas State 69027-070, Brazil
| | - Lucivana P. de Souza Mourão
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Superior School of Health Sciences, Amazonas State University, Manaus, Amazonas State 69065-001, Brazil
| | - Andréa M. Tarragô
- Post-graduate Program in Sciences Applied to Hematology, University of Amazonas State, Manaus, Amazonas State 69850-001, Brazil
- Board of Teaching and Research, Hospital Foundation for Hematology and Hemotherapy of Amazonas, Manaus, Amazonas State 69050-001, Brazil
- Post-graduate Program in Basic and Applied Immunology, Federal University of Amazonas, Manaus, Amazonas State 69067-005, Brazil
- Amazon Genomic Health Surveillance Network Coordination, Manaus, Amazonas State 69040-010, Brazil
| |
Collapse
|
5
|
Dvořáček L, Marková J, Holoubek A, Grebeňová D, Kundrát D, Kuželová K, Schwarz J. A novel germline hyperactivating JAK2 mutation L604F. Ann Hematol 2023; 102:2725-2734. [PMID: 37639050 PMCID: PMC10492870 DOI: 10.1007/s00277-023-05423-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/19/2023] [Indexed: 08/29/2023]
Abstract
Somatic JAK2 mutations are the main molecular cause of the vast majority of polycythemia vera (PV) cases. According to a recent structural model, the prevalent acquired V617F mutation improves the stability of the JAK2 dimer, thereby enhancing the constitutive JAK2 kinase activity. Germline JAK2 mutations usually do not largely alter JAK2 signaling, although they may modulate the impact of V617F. We found an unusual germline JAK2 mutation L604F in homozygous form in a young PV patient, along with a low allele burden JAK2 V617F mutation, and in her apparently healthy sister. Their father with a PV-like disease had L604F in a heterozygous state, without V617F. The functional consequences of JAK2 L604Fmutation were compared with those induced by V617F in two different in vitro model systems: (i) HEK293T cells were transfected with plasmids for exogenous JAK2-GFP expression, and (ii) endogenous JAK2 modifications were introduced into HeLa cells using CRISPR/Cas9. Both mutations significantly increased JAK2 constitutive activity in transfected HEK293T cells. In the second model, JAK2 modification resulted in reduced total JAK2 protein levels. An important difference was also detected: as described previously, the effect of V617F on JAK2 kinase activity was abrogated in the absence of the aromatic residue F595. In contrast, JAK2 hyperactivation by L604F was only partially inhibited by the F595 change to alanine. We propose that the L604F mutation increases the probability of spontaneous JAK2 dimer formation, which is physiologically mediated by F595. In addition, L604F may contribute to dimer stabilization similarly to V617F.
Collapse
Affiliation(s)
- Lukáš Dvořáček
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Jana Marková
- Clinical Department, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Aleš Holoubek
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Dana Grebeňová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - David Kundrát
- Department of Genomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Kateřina Kuželová
- Department of Proteomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic.
| | - Jiří Schwarz
- Clinical Department, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| |
Collapse
|
6
|
Tun PWW, Buka RJ, Graham J, Dyer P. Heterozygous, germline JAK2 E846D substitution as the cause of familial erythrocytosis. Br J Haematol 2022; 198:923-926. [PMID: 35764421 DOI: 10.1111/bjh.18320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 05/30/2022] [Accepted: 06/08/2022] [Indexed: 01/21/2023]
Affiliation(s)
- Phyo Wint Wint Tun
- Department of Clinical Haematology, University Hospitals of North Midlands, Stoke-on-Trent, UK
| | - Richard J Buka
- Department of Clinical Haematology, University Hospitals of North Midlands, Stoke-on-Trent, UK.,Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, UK
| | - Jane Graham
- Department of Clinical Haematology, University Hospitals of North Midlands, Stoke-on-Trent, UK
| | - Peter Dyer
- Department of Clinical Haematology, University Hospitals of North Midlands, Stoke-on-Trent, UK
| |
Collapse
|
7
|
Potential limitations of diagnostic standard codes to distinguish polycythemia vera and secondary erythrocytosis. Sci Rep 2022; 12:4674. [PMID: 35304527 PMCID: PMC8933419 DOI: 10.1038/s41598-022-08606-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 02/25/2022] [Indexed: 11/18/2022] Open
Abstract
Red cell overproduction is seen in polycythemia vera (PV), a bone marrow myeloproliferative neoplasm characterized by trilinear cell proliferation (WBC, platelets), as well as in secondary erythrocytosis (SE), a group of heterogeneous disorders characterized by elevated EPO gene transcription. We aimed to verify the concordance of the International Classification of Diseases (ICD) code-based diagnosis of “polycythemia” or “erythrocytosis” with the true clinical diagnosis of these conditions. We retrospectively reviewed the electronic medical records (January 1, 2005, to December 31, 2016) of adult patients with ICD codes of polycythemia and/or erythrocytosis who had testing done for the presence of the JAK2V617F mutation. We verified the accuracy of the ICD code-based diagnoses by meticulous chart review and established whether these patients fulfilled the criteria by the evaluating physician for PV or SE and according to the World Health Organization 2016 diagnostic guidelines. The reliability of ICD coding was calculated using Cohen's kappa. We identified and chart reviewed a total of 578 patient records. Remarkably, 11% of the patients had concurrent diagnosis codes for PV and SE and were unable to be classified appropriately without individual chart review. The ICD code-based diagnostic system led to misidentification in an important fraction of cases. This represents a problem for the detection of PV or SE cases by ICD-based registries and their derived studies. Research based exclusively on ICD codes could have a potential impact on patient care and public health, and limitations must be weighed when research findings are conveyed.
Collapse
|
8
|
Stuckey R, Gómez-Casares MT. Recent Advances in the Use of Molecular Analyses to Inform the Diagnosis and Prognosis of Patients with Polycythaemia Vera. Int J Mol Sci 2021; 22:5042. [PMID: 34068690 PMCID: PMC8126083 DOI: 10.3390/ijms22095042] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 01/07/2023] Open
Abstract
Genetic studies in the past decade have improved our understanding of the molecular basis of the BCR-ABL1-negative myeloproliferative neoplasm (MPN) polycythaemia vera (PV). Such breakthroughs include the discovery of the JAK2V617F driver mutation in approximately 95% of patients with PV, as well as some very rare cases of familial hereditary MPN caused by inherited germline mutations. Patients with PV often progress to fibrosis or acute myeloid leukaemia, both associated with very poor clinical outcome. Moreover, thrombosis and major bleeding are the principal causes of morbidity and mortality. As a result of increasingly available and economical next-generation sequencing technologies, mutational studies have revealed the prognostic relevance of a few somatic mutations in terms of thrombotic risk and risk of transformation, helping to improve the risk stratification of patients with PV. Finally, knowledge of the molecular basis of PV has helped identify targets for directed therapy. The constitutive activation of the tyrosine kinase JAK2 is targeted by ruxolitinib, a JAK1/JAK2 tyrosine kinase inhibitor for PV patients who are resistant or intolerant to cytoreductive treatment with hydroxyurea. Other molecular mechanisms have also been revealed, and numerous agents are in various stages of development. Here, we will provide an update of the recent published literature on how molecular testing can improve the diagnosis and prognosis of patients with PV and present recent advances that may have prognostic value in the near future.
Collapse
Affiliation(s)
- Ruth Stuckey
- Hematology Department, Hospital Universitario de Gran Canaria Dr. Negrín, 35019 Las Palmas, Spain
| | | |
Collapse
|
9
|
Westermann J, Bullinger L. Precision medicine in myeloid malignancies. Semin Cancer Biol 2021; 84:153-169. [PMID: 33895273 DOI: 10.1016/j.semcancer.2021.03.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 03/28/2021] [Accepted: 03/29/2021] [Indexed: 12/13/2022]
Abstract
Myeloid malignancies have always been at the forefront of an improved understanding of the molecular pathogenesis of cancer. In accordance, over the last years, basic research focusing on the aberrations underlying malignant transformation of myeloid cells has provided the basis for precision medicine approaches and subsequently has led to the development of powerful therapeutic strategies. In this review article, we will recapitulate what has happened since in the 1980s the use of all-trans retinoic acid (ATRA), as a first targeted cancer therapy, has changed one of the deadliest leukemia subtypes, acute promyelocytic leukemia (APL), into one that can be cured without classical chemotherapy today. Similarly, imatinib, the first molecularly designed cancer therapy, has revolutionized the management of chronic myeloid leukemia (CML). Thus, targeted treatment approaches have become the paradigm for myeloid malignancy, but many questions still remain unanswered, especially how identical mutations can be associated with different phenotypes. This might be linked to the impact of the cell of origin, gene-gene interactions, or the tumor microenvironment including the immune system. Continuous research in the field of myeloid neoplasia has started to unravel the molecular pathways that are not only crucial for initial treatment response, but also resistance of leukemia cells under therapy. Ongoing studies focusing on leukemia cell vulnerabilities do already point to novel (targetable) "Achilles heels" that can further improve myeloid cancer therapy.
Collapse
Affiliation(s)
- Jörg Westermann
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine Berlin, Campus Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Lars Bullinger
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine Berlin, Campus Virchow Clinic, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
10
|
Favoino E, Prete M, Catacchio G, Ruscitti P, Navarini L, Giacomelli R, Perosa F. Working and safety profiles of JAK/STAT signaling inhibitors. Are these small molecules also smart? Autoimmun Rev 2021; 20:102750. [PMID: 33482338 DOI: 10.1016/j.autrev.2021.102750] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 12/14/2020] [Indexed: 12/18/2022]
Abstract
The Janus kinase (JAK)/signal transducers and activators of transcription (STAT) pathway is an important intracellular route through which many different extracellular soluble molecules, by reaching membrane receptors, can signal the nucleus. The spectrum of soluble molecules that use the JAK/STAT pathway through their corresponding receptors is quite large (almost 50 different molecules), and includes some cytokines involved in the pathogenesis of many immune-mediated diseases. Such diseases, when left untreated, present an evident hyperactivation of JAK/STAT signaling. Therefore, given the pathogenetic role of JAK/STAT, drugs known as JAK inhibitors (JAKi), that target one or more JAKs, have been developed to counteract JAK/STAT signal hyperactivation. As some hematological malignancies present an intrinsic JAK/STAT hyperactivation due to a JAK mutation, some JAKi have also been successfully used in this context. Regulatory agencies for drug administration in different countries have already approved a few JAKi in the setting of either immune-mediated diseases or hematological malignancies. Aim of this review is to describe the physiology of intracellular JAK/STAT pathway signaling and the pathological conditions associated to its dysregulation. Then, the rationale for targeting JAK in rheumatic autoimmune diseases is discussed, along with clinical data from registration studies showing the efficacy of these drugs. Finally, the excellent safety profile of JAKi is discussed in the context of the apparent poor specificity of JAK/STAT pathway signal.
Collapse
Affiliation(s)
- Elvira Favoino
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Biomedical Science and Human Oncology (DIMO), University of Bari Medical School, Italy
| | - Marcella Prete
- Internal Medicine, Department of Biomedical Science and Human Oncology (DIMO), University of Bari Medical School, Italy
| | - Giacomo Catacchio
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Biomedical Science and Human Oncology (DIMO), University of Bari Medical School, Italy
| | - Piero Ruscitti
- Rheumatology Unit, Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Luca Navarini
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome 'Campus Biomedico', Italy
| | - Roberto Giacomelli
- Rheumatology and Immunology Unit, Department of Medicine, University of Rome 'Campus Biomedico', Italy
| | - Federico Perosa
- Rheumatic and Systemic Autoimmune Diseases Unit, Department of Biomedical Science and Human Oncology (DIMO), University of Bari Medical School, Italy.
| |
Collapse
|
11
|
Bellanné-Chantelot C, Rabadan Moraes G, Schmaltz-Panneau B, Marty C, Vainchenker W, Plo I. Germline genetic factors in the pathogenesis of myeloproliferative neoplasms. Blood Rev 2020; 42:100710. [PMID: 32532454 DOI: 10.1016/j.blre.2020.100710] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 04/08/2020] [Accepted: 05/05/2020] [Indexed: 02/06/2023]
Abstract
Myeloproliferative neoplasms (MPN) are clonal hematological malignancies that lead to overproduction of mature myeloid cells. They are due to acquired mutations in genes encoding for AK2, MPL and CALR that result in the activation of the cytokine receptor/JAK2 signaling pathway. In addition, it exists germline variants that can favor the initiation of the disease or may affect its phenotype. First, they can be common risk alleles, which correspond to frequent single nucleotide variants present in control population and that contribute to the development of either sporadic or familial MPN. Second, some variants predispose to the onset of MPN with a higher penetrance and lead to familial clustering of MPN. Finally, some extremely rare genetic variants can induce MPN-like hereditary disease. We will review these different subtypes of germline genetic variants and discuss how they impact the initiation and/or development of the MPN disease.
Collapse
Affiliation(s)
- Christine Bellanné-Chantelot
- Department of Genetics, Assistance Publique-Hôpitaux de Paris (APHP), Hôpitaux Universitaires Pitié Salpêtrière-Charles Foix, Sorbonne Université, Paris, France; INSERM, UMR1287, Laboratory of Excellence GR-Ex, Villejuif, France
| | - Graciela Rabadan Moraes
- INSERM, UMR1287, Laboratory of Excellence GR-Ex, Villejuif, France; Université Paris Diderot (Paris 7), UMR1287, Gustave Roussy, Villejuif, France; Gustave Roussy, Villejuif, France
| | - Barbara Schmaltz-Panneau
- INSERM, UMR1287, Laboratory of Excellence GR-Ex, Villejuif, France; Gustave Roussy, Villejuif, France; Université Paris XI, UMR1287, Gustave Roussy, Villejuif, France
| | - Caroline Marty
- INSERM, UMR1287, Laboratory of Excellence GR-Ex, Villejuif, France; Gustave Roussy, Villejuif, France; Université Paris XI, UMR1287, Gustave Roussy, Villejuif, France
| | - William Vainchenker
- INSERM, UMR1287, Laboratory of Excellence GR-Ex, Villejuif, France; Gustave Roussy, Villejuif, France; Université Paris XI, UMR1287, Gustave Roussy, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR1287, Laboratory of Excellence GR-Ex, Villejuif, France; Gustave Roussy, Villejuif, France; Université Paris XI, UMR1287, Gustave Roussy, Villejuif, France.
| |
Collapse
|
12
|
Ilinca A, Martinez-Majander N, Samuelsson S, Piccinelli P, Truvé K, Cole J, Kittner S, Soller M, Kristoffersson U, Tatlisumak T, Puschmann A, Putaala J, Lindgren A. Whole-Exome Sequencing in 22 Young Ischemic Stroke Patients With Familial Clustering of Stroke. Stroke 2020; 51:1056-1063. [PMID: 32172663 DOI: 10.1161/strokeaha.119.027474] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Backgrounds and Purpose- Although new methods for genetic analyses are rapidly evolving, there are currently knowledge gaps in how to detect Mendelian forms of stroke. Methods- We performed whole-exome sequencing in 22 probands, under 56 years at their first ischemic stroke episode, from multi-incident stroke families. With the use of a comprehensive stroke-gene panel, we searched for variants in stroke-related genes. The probands' clinical stroke subtype was related to clinical characteristics previously associated with pathogenic variants in these genes. Relatives were genotyped in 7 families to evaluate stroke-gene variants of unknown significance. In 2 larger families with embolic stroke of unknown source, whole-exome sequencing was performed in additional members to examine the possibility of identifying new stroke genes. Results- Six of 22 probands carried pathogenic or possibly pathogenic variants in genes reported to be associated with their stroke subtype. A known pathogenic variant in NOTCH3 and a possibly pathogenic variant in ACAD9 gene were identified. A novel JAK2:c.3188G>A (p.Arg1063His) mutation was seen in a proband with embolic stroke of undetermined source and prothrombotic status. However, penetrance in the family was incomplete. COL4A2:c.3368A>G (p.Glu1123Gly) was detected in 2 probands but did not cosegregate with the disease in their families. Whole-exome sequencing in multiple members of 2 pedigrees with embolic stroke of undetermined source revealed possibly pathogenic variants in genes not previously associated with stroke, GPR142:c.148C>G (p.Leu50Val), and PTPRN2:c.2416A>G (p.Ile806Val); LRRC1 c.808A>G (p.Ile270Val), SLC7A10c.1294dupG (p.Val432fs), IKBKB: c.1070C>T (p.Ala357Val), and OXGR1 c.392G>A (p.Arg131His), respectively. Conclusions- Screening with whole-exome sequencing using a comprehensive stroke-gene panel may identify rare monogenic forms of stroke, but careful evaluation of clinical characteristics and potential pathogenicity of novel variants remain important. In our study, the majority of individuals with familial aggregation of stroke lacked any identified genetic causes.
Collapse
Affiliation(s)
- Andreea Ilinca
- From the Department of Clinical Sciences Lund, Neurology (A.I., A.P., A.L.), Lund University, Sweden.,Department of Neurology and Rehabilitation Medicine, Neurology, Skåne University Hospital, Sweden (A.L., A.P., A.L.)
| | | | - Sofie Samuelsson
- Department of Clinical Genetics and Pathology (S.S., P.P.), Lund University, Sweden
| | - Paul Piccinelli
- Department of Clinical Genetics and Pathology (S.S., P.P.), Lund University, Sweden
| | - Katarina Truvé
- Bioinformatics Core Facility, Sahlgrenska Academy at University of Gothenburg, Sweden (K.T.)
| | - John Cole
- Department of Neurology, Veterans Affairs Maryland Health Care System (J.C., S.K.), University of Maryland School of Medicine, Baltimore
| | - Steven Kittner
- Department of Neurology, Veterans Affairs Maryland Health Care System (J.C., S.K.), University of Maryland School of Medicine, Baltimore.,Department of Neurology (S.K.), University of Maryland School of Medicine, Baltimore
| | - Maria Soller
- Department of Clinical Genetics, Karolinska University Hospital, Solna, Sweden (M.S.)
| | - Ulf Kristoffersson
- Division of Clinical Genetics, Laboratory Medicine (U.K.), Lund University, Sweden
| | - Turgut Tatlisumak
- Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Sweden (T.T.)
| | - Andreas Puschmann
- From the Department of Clinical Sciences Lund, Neurology (A.I., A.P., A.L.), Lund University, Sweden.,Department of Neurology and Rehabilitation Medicine, Neurology, Skåne University Hospital, Sweden (A.L., A.P., A.L.)
| | - Jukka Putaala
- Department of Neurology, Helsinki University Hospital, Finland (N.M.-M., J.P.)
| | - Arne Lindgren
- From the Department of Clinical Sciences Lund, Neurology (A.I., A.P., A.L.), Lund University, Sweden.,Department of Neurology and Rehabilitation Medicine, Neurology, Skåne University Hospital, Sweden (A.L., A.P., A.L.)
| |
Collapse
|
13
|
Jang MA, Choi CW. Recent insights regarding the molecular basis of myeloproliferative neoplasms. Korean J Intern Med 2020; 35:1-11. [PMID: 31778606 PMCID: PMC6960053 DOI: 10.3904/kjim.2019.317] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 11/11/2019] [Indexed: 12/14/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are a heterogeneous group of clonal disorders characterized by the overproduction of mature blood cells that have an increased risk of thrombosis and progression to acute myeloid leukemia. Next-generation sequencing studies have provided key insights regarding the molecular mechanisms of MPNs. MPN driver mutations in genes associated with the JAK-STAT pathway include JAK2 V617F, JAK2 exon 12 mutations and mutations in MPL, CALR, and CSF3R. Cooperating driver genes are also frequently detected and also mutated in other myeloid neoplasms; these driver genes are involved in epigenetic methylation, messenger RNA splicing, transcription regulation, and signal transduction. In addition, other genetic factors such as germline predisposition, order of mutation acquisition, and variant allele frequency also influence disease initiation and progression. This review summarizes the current understanding of the genetic basis of MPN, and demonstrates how molecular pathophysiology can improve both our understanding of MPN heterogeneity and clinical practice.
Collapse
Affiliation(s)
- Mi-Ae Jang
- Department of Laboratory Medicine and Genetics, Soonchunhyang University Bucheon Hospital, Bucheon, Korea
| | - Chul Won Choi
- Division of Oncology and Hematology, Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
- Correspondence to Chul Won Choi, M.D. Division of Oncology and Hematology, Department of Internal Medicine, Korea University Guro Hospital, 148 Gurodong-ro, Guro-gu, Seoul 08308, Korea Tel: +82-2-2626-3058 Fax: +82-2-862-6453 E-mail:
| |
Collapse
|
14
|
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: 18] [Impact Index Per Article: 3.0] [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
|
15
|
Experimental Modeling of Myeloproliferative Neoplasms. Genes (Basel) 2019; 10:genes10100813. [PMID: 31618985 PMCID: PMC6826898 DOI: 10.3390/genes10100813] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/29/2019] [Accepted: 10/12/2019] [Indexed: 12/25/2022] Open
Abstract
Myeloproliferative neoplasms (MPN) are genetically very complex and heterogeneous diseases in which the acquisition of a somatic driver mutation triggers three main myeloid cytokine receptors, and phenotypically expresses as polycythemia vera (PV), essential thrombocytosis (ET), and primary myelofibrosis (PMF). The course of the diseases may be influenced by germline predispositions, modifying mutations, their order of acquisition and environmental factors such as aging and inflammation. Deciphering these contributory elements, their mutual interrelationships, and their contribution to MPN pathogenesis brings important insights into the diseases. Animal models (mainly mouse and zebrafish) have already significantly contributed to understanding the role of several acquired and germline mutations in MPN oncogenic signaling. Novel technologies such as induced pluripotent stem cells (iPSCs) and precise genome editing (using CRISPR/Cas9) contribute to the emerging understanding of MPN pathogenesis and clonal architecture, and form a convenient platform for evaluating drug efficacy. In this overview, the genetic landscape of MPN is briefly described, with an attempt to cover the main discoveries of the last 15 years. Mouse and zebrafish models of the driver mutations are discussed and followed by a review of recent progress in modeling MPN with patient-derived iPSCs and CRISPR/Cas9 gene editing.
Collapse
|
16
|
Hammarén HM, Virtanen AT, Raivola J, Silvennoinen O. The regulation of JAKs in cytokine signaling and its breakdown in disease. Cytokine 2019; 118:48-63. [DOI: 10.1016/j.cyto.2018.03.041] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/29/2018] [Accepted: 03/30/2018] [Indexed: 01/12/2023]
|
17
|
Cooccurring JAK2 V617F and R1063H mutations increase JAK2 signaling and neutrophilia in myeloproliferative neoplasms. Blood 2018; 132:2695-2699. [PMID: 30377194 DOI: 10.1182/blood-2018-04-843060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
18
|
Bacher U, Shumilov E, Flach J, Porret N, Joncourt R, Wiedemann G, Fiedler M, Novak U, Amstutz U, Pabst T. Challenges in the introduction of next-generation sequencing (NGS) for diagnostics of myeloid malignancies into clinical routine use. Blood Cancer J 2018; 8:113. [PMID: 30420667 PMCID: PMC6232163 DOI: 10.1038/s41408-018-0148-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/17/2018] [Accepted: 10/15/2018] [Indexed: 12/20/2022] Open
Abstract
Given the vast phenotypic and genetic heterogeneity of acute and chronic myeloid malignancies, hematologists have eagerly awaited the introduction of next-generation sequencing (NGS) into the routine diagnostic armamentarium to enable a more differentiated disease classification, risk stratification, and improved therapeutic decisions. At present, an increasing number of hematologic laboratories are in the process of integrating NGS procedures into the diagnostic algorithms of patients with acute myeloid leukemia (AML), myelodysplastic syndromes (MDS), and myeloproliferative neoplasms (MPNs). Inevitably accompanying such developments, physicians and molecular biologists are facing unexpected challenges regarding the interpretation and implementation of molecular genetic results derived from NGS in myeloid malignancies. This article summarizes typical challenges that may arise in the context of NGS-based analyses at diagnosis and during follow-up of myeloid malignancies.
Collapse
Affiliation(s)
- Ulrike Bacher
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Center for Laboratory Medicine (ZLM)/University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Evgenii Shumilov
- Department of Hematology and Medical Oncology, University Medicine Göttingen (UMG), Göttingen, Germany
| | - Johanna Flach
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Mannheim, Germany
| | - Naomi Porret
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Raphael Joncourt
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Gertrud Wiedemann
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Martin Fiedler
- Center for Laboratory Medicine (ZLM)/University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Urban Novak
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ursula Amstutz
- Center for Laboratory Medicine (ZLM)/University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Thomas Pabst
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Brown AL, Churpek JE, Malcovati L, Döhner H, Godley LA. Recognition of familial myeloid neoplasia in adults. Semin Hematol 2017. [DOI: 10.1053/j.seminhematol.2016.11.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
22
|
Diagnosis, risk stratification, and response evaluation in classical myeloproliferative neoplasms. Blood 2016; 129:680-692. [PMID: 28028026 DOI: 10.1182/blood-2016-10-695957] [Citation(s) in RCA: 162] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 12/02/2016] [Indexed: 12/14/2022] Open
Abstract
Philadelphia-negative classical myeloproliferative neoplasms (MPNs) include polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). The 2016 revision of the WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues includes new criteria for the diagnosis of these disorders. Somatic mutations in the 3 driver genes, that is, JAK2, CALR, and MPL, represent major diagnostic criteria in combination with hematologic and morphological abnormalities. PV is characterized by erythrocytosis with suppressed endogenous erythropoietin production, bone marrow panmyelosis, and JAK2 mutation. Thrombocytosis, bone marrow megakaryocytic proliferation, and presence of JAK2, CALR, or MPL mutation are the main diagnostic criteria for ET. PMF is characterized by bone marrow megakaryocytic proliferation, reticulin and/or collagen fibrosis, and presence of JAK2, CALR, or MPL mutation. Prefibrotic myelofibrosis represents an early phase of myelofibrosis, and is characterized by granulocytic/megakaryocytic proliferation and lack of reticulin fibrosis in the bone marrow. The genomic landscape of MPNs is more complex than initially thought and involves several mutant genes beyond the 3 drivers. Comutated, myeloid tumor-suppressor genes contribute to phenotypic variability, phenotypic shifts, and progression to more aggressive disorders. Patients with myeloid neoplasms are at variable risk of vascular complications, including arterial or venous thrombosis and bleeding. Current prognostic models are mainly based on clinical and hematologic parameters, but innovative models that include genetic data are being developed for both clinical and trial settings. In perspective, molecular profiling of MPNs might also allow for accurate evaluation and monitoring of response to innovative drugs that target the mutant clone.
Collapse
|