1
|
Szuber N, Guglielmelli P, Gangat N. Topics of Interest in Women With Myeloproliferative Neoplasms. Am J Hematol 2025. [PMID: 40084464 DOI: 10.1002/ajh.27665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/16/2025]
Abstract
OVERVIEW Sex and gender have emerged as central modifiers of disease biology, phenotype, and clinical outcomes in myeloproliferative neoplasms (MPNs). This review will uniquely highlight issues affecting women with MPN and articulate their relevant determinants. EPIDEMIOLOGY AND DIAGNOSIS A higher overall prevalence of MPN has been established in women. The incidence of essential thrombocythemia (ET) predominates, while, conversely, polycythemia vera (PV) and myelofibrosis (MF) are seen in lower frequencies as compared to men. Diagnostic criteria are dictated by sex-driven physiological variances in hemoglobin and hematocrit levels in PV, mandating separate diagnostic thresholds, respectively: > 16.0 g/dL and > 48% in women vs. > 16.5 and > 49% in men. GENETIC FRAMEWORK AND PHENOTYPE Women with MPN harbor fewer acquired somatic mutations and a lower frequency of high-risk mutations than their male counterparts; lower JAK2V617F driver variant allele frequency and attenuated allele burden kinetics have also been reported. Women with MPN are younger at diagnosis than men and, contingent on subtype, display more indolent disease features. Importantly, validated symptom burden assessments consistently disclose higher scores in women vs. men. THROMBOSIS AND OUTCOMES Women with MPN have a unique thrombotic diathesis with respect to men, more frequently involving the splanchnic venous system in those ultimately diagnosed with PV. Outcomes data depict female sex as a variable associated with more favorable clinical trajectories, including lower rates of MF/leukemic transformation and secondary cancers, as well as improved overall survival rates vis-à-vis men. LIFE-CYCLE WINDOWS, PREGNANCY, AND POSTPARTUM Potential challenges at each significant life stage will be addressed: puberty, preconception and fertility, and perimenopause; these include issues surrounding oral contraceptives and hormone use. Prospective studies suggest overall favorable maternal and fetal outcomes with pregnancy in women with MPN. Full details on risks and reported outcomes will be discussed, as well as a risk-adapted approach to management informed by obstetric and thrombosis history. Recommendations include aspirin 81 mg daily in all patients and cytoreduction with interferon-α in those with antecedent thrombosis, as well as in low-risk cases with higher-risk features (e.g., poorly controlled hematocrit and recurrent fetal loss). Antepartum anticoagulation with low molecular weight heparin (LMWH) is recommended in cases with previous venous thromboembolism. CONCLUSIONS AND FUTURE DIRECTIONS This review highlights female sex and gender as critical drivers of MPN incidence, presentation, and natural history. It further outlines the impact and management of MPN as related to unique female reproductive phases. A sex-informed lens will be required in order to recalibrate current prognostic tools, a requisite to refining patient counselling and clinical decision-making in line with precision medicine. Moreover, while several mechanisms underpinning sex-defined discrepancies have been defined, these mandate further prospective study. Finally, sex and gender-based differences must be weighted in clinical trials with systematized procedures to correct participation imbalances in favor of sex and gender equity.
Collapse
Affiliation(s)
- Natasha Szuber
- Division of Hematology, Department of Internal Medicine, Université de Montréal, Montréal, Quebec, Canada
| | - Paola Guglielmelli
- Department of Experimental and Clinical Medicine, CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| | - Naseema Gangat
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
2
|
Chiasakul T, Baker RI. Management of Bleeding, Thrombotic and Pregnancy-Related Complications in Women with Myeloproliferative Neoplasms: A Case-Based Review Focusing on Sex-Specific Challenges. J Clin Med 2025; 14:1537. [PMID: 40095471 PMCID: PMC11900594 DOI: 10.3390/jcm14051537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Myeloproliferative neoplasms (MPNs) are a heterogeneous group of clonal hematopoietic disorders that pose unique challenges in women, particularly regarding thrombosis, bleeding, fertility, and pregnancy. Women with MPN exhibit distinct thrombotic and sometimes contradictory bleeding profiles, including a higher prevalence of unusual thrombosis such as cerebral and splanchnic vein thrombosis and increased risk of hemorrhage from anti-thrombotic medication, acquired von Willebrand syndrome and platelet dysfunction. Estrogen-containing contraceptives should generally be avoided due to thrombotic risk. Around 10-20% of newly diagnosed MPN cases are women of childbearing age and the number is increasing annually. MPN patients when compared to controls have a lower rate of live birth rate of 71% vs. 80% with a hazard ratio of 0.78 (95% CI: 0.68-0.90), and increased preterm birth (14% vs. 4%), low birth weight (<2500 g, 10% vs. 4%), and increased cesarean section rate (32% vs. 17%). Management of MPN-related pregnancy requires specific considerations regarding the prevention of thrombosis, bleeding, and pregnancy-related complications. Management strategies during pregnancy include low-dose aspirin and consideration of low-molecular-weight heparin and interferon. Despite these challenges, most women with MPN can achieve successful pregnancies with optimized care. In this case-based review, we present two cases that illustrate key aspects of managing MPN in women, summarize the current literature, and propose a diagnostic and management framework tailored to these complexities.
Collapse
Affiliation(s)
- Thita Chiasakul
- Center of Excellence in Translational Hematology, Division of Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok 10330, Thailand;
| | - Ross I. Baker
- Western Australia Centre for Thrombosis and Haemostasis, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia
- Clinical Research Unit, Perth Blood Institute, Perth, WA 6005, Australia
| |
Collapse
|
3
|
Spivak JL. Myeloproliferative Neoplasms: Challenging Dogma. J Clin Med 2024; 13:6957. [PMID: 39598101 PMCID: PMC11595126 DOI: 10.3390/jcm13226957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024] Open
Abstract
Myeloproliferative neoplasms, polycythemia vera, essential thrombocytosis, and primary myelofibrosis are a unique group of clonal hematopoietic stem cell neoplasms that share somatic, gain-in-function driver mutations in JAK2, CALR, and MPL. As a consequence, these disorders exhibit similar phenotypic features, the most common of which are the ceaseless production of normal erythrocytes, myeloid cells, platelets alone or in combination, extramedullary hematopoiesis, myelofibrosis, and a potential for leukemic transformation. In the case of polycythemia vera and essential thrombocytosis, however, prolonged survival is possible. With an incidence value in the range of 0.5-2.0/100,000, myeloproliferative neoplasms are rare disorders, but they are not new disorders, and after a century of scrutiny, their clinical features and natural histories are well-defined, though their individual management continues to be controversial. With respect to polycythemia vera, there has been a long-standing dispute between those who believe that the suppression of red blood cell production by chemotherapy is superior to phlebotomy to prevent thrombosis, and those who do not. With respect to essential thrombocytosis, there is a similar dispute about the role of platelets in veinous thrombosis, and the role of chemotherapy in preventing thrombosis by suppressing platelet production. Linked to these disputes is another: whether therapy with hydroxyurea promotes acute leukemia in disorders with a substantial possibility of longevity. The 21st century revealed new insights into myeloproliferative neoplasms with the discovery of their three somatic, gain-of-function driver mutations. Almost immediately, this triggered changes in the diagnostic criteria for myeloproliferative neoplasms and their therapy. Most of these changes, however, conflicted with prior well-validated, phenotypically driven diagnostic criteria and the management of these disorders. The aim of this review is to examine these conflicts and demonstrate how genomic discoveries in myeloproliferative neoplasms can be used to effectively complement the known phenotypic features of these disorders for their diagnosis and management.
Collapse
Affiliation(s)
- Jerry L Spivak
- Hematology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
4
|
Tiu A, McKinnell Z, Liu S, Gill P, Antonio M, Shancer Z, Srinivasa N, Diao G, Subrahmanyam R, Kessler CM, Jain M. Risk of myeloproliferative neoplasms among U.S. Veterans from Korean, Vietnam, and Persian Gulf War eras. Am J Hematol 2024; 99:1969-1978. [PMID: 39023278 DOI: 10.1002/ajh.27438] [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: 05/23/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/20/2024]
Abstract
The Promise to Address Comprehensive Toxics (PACT) Act expanded U.S. Veterans' health care and benefits for conditions linked to service-connected exposures (e.g., Burn Pits, Agent Orange). However, myeloproliferative neoplasms (MPN) are not recognized as presumptive conditions for Veterans exposed to these toxic substances. This study evaluated the development of MPN among U.S. Veterans from the Korean, Vietnam, and Persian Gulf War eras. This retrospective cohort study included 65 425 Korean War era Veterans; 211 927 Vietnam War era Veterans; and 214 007 Persian Gulf War era Veterans from January 1, 2006, to January 26, 2023. Veterans with MPN, thrombosis, bleeding, and cardiovascular risk factors were identified through ICD-9 and -10 codes. Veterans from the Persian Gulf War era had the highest risk of developing MPN compared with Veterans from the Korean and Vietnam War eras, hazard ratio (HR) 4.92, 95% confidence interval (CI) 4.20-5.75 and HR 2.49, 95% CI 2.20-2.82, both p < .0001, respectively. Vietnam War era Veterans also had a higher risk of MPN development compared with Korean War era Veterans, HR 1.97, 95% CI 1.77-2.21, p < .0001. Persian Gulf War era Veterans were diagnosed with MPN at an earlier age, had higher risks of thrombosis and bleeding, and had lower survival rates compared with Korean War and Vietnam War era Veterans. This study reinforces evidence that environmental and occupational hazards increase the risk of clonal myeloid disorders and related complications, impacting overall survival with MPN. Limitations include the inability to confirm clonality and fully verify deployment and exposure status.
Collapse
Affiliation(s)
- Andrew Tiu
- Division of Hematology-Oncology, Lombardi Comprehensive Cancer Center, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Zoe McKinnell
- Division of Hematology-Oncology, The George Washington University, Washington, DC, USA
| | - Shanshan Liu
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | - Puneet Gill
- Institute for Clinical Research, Washington, DC, USA
| | | | - Zoe Shancer
- The George Washington University School of Medicine, Washington, DC, USA
| | - Nandan Srinivasa
- The George Washington University School of Medicine, Washington, DC, USA
| | - Guoqing Diao
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC, USA
| | | | - Craig M Kessler
- Division of Hematology-Oncology, Lombardi Comprehensive Cancer Center, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Maneesh Jain
- The George Washington University School of Medicine, Washington, DC, USA
- Washington DC VA Medical Center, Washington, DC, USA
| |
Collapse
|
5
|
Yan J, Hammami MB, Wei JX, Shah N, Goldfinger M, Mantzaris I, Kornblum N, Gritsman K, Sica A, Cooper D, Feldman E, Konopleva M, Pradhan K, Thakur R, Vegivinti C, Qasim A, Verma A, Goel S. Socio-demographic determinants of myelofibrosis outcomes in an underserved center and the SEER national database. Ann Hematol 2024; 103:3543-3551. [PMID: 39046510 PMCID: PMC11358356 DOI: 10.1007/s00277-024-05894-7] [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: 06/29/2024] [Accepted: 07/11/2024] [Indexed: 07/25/2024]
Abstract
The influence of demographic characteristics and social determinants on cancer outcomes is widely recognized in various malignancies but remains understudied in myelofibrosis (MF). This study aims to investigate social and demographic variables associated with MF survival. We retrospectively reviewed data of biopsy-proven MF patients from the Surveillance, Epidemiology and End Results (SEER) database (2000-2021) and Montefiore Medical Center (2000-2023), an underserved inner-city hospital. The SEER cohort included 5,403 MF patients and was predominantly Non-Hispanic (NH) White (82%) with a median age of 69 years. The age-adjusted incidence rate of MF was 0.32 cases per 100,000 person-years, increasing annually by 1.3% from 2000 to 2021. Two- and five- year overall survival rates were 69% and 42%, respectively. Worse cause-specific survival was associated with older age, male sex, and diagnosis before 2011 (year of Ruxolitinib approval). NH-Black ethnicity, unmarried status and lower median income were independent predictors of worse overall survival. The single-center analysis included 84 cases, with a median age of 66 years. NH-White patients comprised 37% of the sample, followed by NH-Black (28.5%). Two- and five- year overall survival rates were 90% and 61%, respectively, with NH-Black patients exhibiting the lowest median survival, although the difference was not statistically significant. Age was a significant predictor of worse survival in this cohort. NH-Black and Hispanic patients lived in areas with higher socioeconomic and demographic stress compared to NH-White patients. Overall, this study highlights the association of social and demographic factors with MF survival and emphasizes the need for equitable healthcare and further exploration of social-demographic factors affecting MF survival.
Collapse
Affiliation(s)
- John Yan
- Department of Internal Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - M Bakri Hammami
- Department of Medicine, Jacobi Medical Center, Bronx, NY, USA
| | - John X Wei
- Department of Internal Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Nishi Shah
- Department of Hematology/Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Mendel Goldfinger
- Department of Hematology/Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Ioannis Mantzaris
- Department of Hematology/Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Noah Kornblum
- Department of Hematology/Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Kira Gritsman
- Department of Hematology/Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Alejandro Sica
- Department of Hematology/Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Dennis Cooper
- Department of Hematology/Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Eric Feldman
- Department of Hematology/Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Marina Konopleva
- Department of Hematology/Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Kith Pradhan
- Department of Hematology/Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Rahul Thakur
- Department of Medicine, Jacobi Medical Center, Bronx, NY, USA
| | | | - Asma Qasim
- Department of Medicine, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - Amit Verma
- Department of Hematology/Oncology, Montefiore Medical Center, Bronx, NY, USA
| | - Swati Goel
- Department of Hematology/Oncology, Montefiore Medical Center, Bronx, NY, USA.
- Department of Oncology, Montefiore Einstein Comprehensive Cancer Center, 3411 Wayne Avenue, Bronx, NY, 10467-2509, USA.
| |
Collapse
|
6
|
Reeves B. Blood cell JAKtivation aggravates cerebral venous thrombosis. Blood Adv 2024; 8:3327-3329. [PMID: 38916898 PMCID: PMC11258618 DOI: 10.1182/bloodadvances.2024012977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/26/2024] Open
Affiliation(s)
- Brandi Reeves
- Department of Medicine, Division of Hematology, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
7
|
Vachhani P, Mascarenhas J, Bose P, Hobbs G, Yacoub A, Palmer JM, Gerds AT, Masarova L, Kuykendall AT, Rampal RK, Mesa R, Verstovsek S. Interferons in the treatment of myeloproliferative neoplasms. Ther Adv Hematol 2024; 15:20406207241229588. [PMID: 38380373 PMCID: PMC10878223 DOI: 10.1177/20406207241229588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/14/2023] [Indexed: 02/22/2024] Open
Abstract
Interferons are cytokines with immunomodulatory properties and disease-modifying effects that have been used to treat myeloproliferative neoplasms (MPNs) for more than 35 years. The initial use of interferons was limited due to difficulties with administration and a significant toxicity profile. Many of these shortcomings were addressed by covalently binding polyethylene glycol to the interferon structure, which increases the stability, prolongs activity, and reduces immunogenicity of the molecule. In the current therapeutic landscape, pegylated interferons are recommended for use in the treatment of polycythemia vera, essential thrombocythemia, and primary myelofibrosis. We review recent efficacy, molecular response, and safety data for the two available pegylated interferons, peginterferon alfa-2a (Pegasys) and ropeginterferon alfa-2b-njft (BESREMi). The practical management of interferon-based therapies is discussed, along with our opinions on whether to and how to switch from hydroxyurea to one of these therapies. Key topics and questions related to use of interferons, such as their safety and tolerability, the significance of variant allele frequency, advantages of early treatment, and what the future of interferon therapy may look like, will be examined. Pegylated interferons represent an important therapeutic option for patients with MPNs; however, more research is still required to further refine interferon therapy.
Collapse
Affiliation(s)
- Pankit Vachhani
- Hematology Oncology at The Kirklin Clinic of UAB Hospital, North Pavilion, Room 2540C, 1720 2 Ave S, Birmingham, AL 35294-3300, USA
| | - John Mascarenhas
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Prithviraj Bose
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriela Hobbs
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Abdulraheem Yacoub
- Division of Hematologic Malignancies and Cellular Therapeutics, The University of Kansas Cancer Center, Westwood, KS, USA
| | | | - Aaron T. Gerds
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA
| | - Lucia Masarova
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew T. Kuykendall
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Raajit K. Rampal
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ruben Mesa
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - Srdan Verstovsek
- Division of Cancer Medicine, Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
8
|
Bewersdorf JP, How J, Masarova L, Bose P, Pemmaraju N, Mascarenhas J, Rampal RK. Moving toward disease modification in polycythemia vera. Blood 2023; 142:1859-1870. [PMID: 37729609 DOI: 10.1182/blood.2023021503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/01/2023] [Accepted: 09/15/2023] [Indexed: 09/22/2023] Open
Abstract
Polycythemia vera (PV) belongs to the BCR-ABL1-negative myeloproliferative neoplasms and is characterized by activating mutations in JAK2 and clinically presents with erythrocytosis, variable degrees of systemic and vasomotor symptoms, and an increased risk of both thromboembolic events and progression to myelofibrosis and acute myeloid leukemia (AML). Treatment selection is based on a patient's age and a history of thrombosis in patients with low-risk PV treated with therapeutic phlebotomy and aspirin alone, whereas cytoreductive therapy with either hydroxyurea or interferon alfa (IFN-α) is added for high-risk disease. However, other disease features such as significant disease-related symptoms and splenomegaly, concurrent thrombocytosis and leukocytosis, or intolerance of phlebotomy can constitute an indication for cytoreductive therapy in patients with otherwise low-risk disease. Additionally, recent studies demonstrating the safety and efficacy (ie, reduction in phlebotomy requirements and molecular responses) of ropegylated IFN-α2b support its use for patients with low-risk PV. Additionally, emerging data suggest that early treatment is associated with higher rates of molecular responses, which might eventually enable time-limited therapy. Nonetheless, longer follow-up is needed to assess whether molecular responses associate with clinically meaningful outcome measures such as thrombosis and progression to myelofibrosis or AML. In this article, we provide an overview of the current and evolving treatment landscape of PV and outline our vision for a patient-centered, phlebotomy-free, treatment approach using time-limited, disease-modifying treatment modalities early in the disease course, which could ultimately affect the natural history of the disease.
Collapse
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Joan How
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Lucia Masarova
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Prithviraj Bose
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naveen Pemmaraju
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - John Mascarenhas
- Division of Hematology/Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Raajit K Rampal
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
9
|
Orbell LY, Abutheraa N, Duncombe AS, McMullin MF, Mesa R, McShane CM, James G, Anderson LA. The JAK2V617F mutation and the role of therapeutic agents in alleviating myeloproliferative neoplasm symptom burden. EJHAEM 2023; 4:1071-1080. [PMID: 38024634 PMCID: PMC10660120 DOI: 10.1002/jha2.805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 12/01/2023]
Abstract
Alleviating symptom burden in patients with myeloproliferative neoplasms (MPNs) is imperative to achieving optimal management. Research remains to elucidate the relationship between the JAK2V617F (Janus kinase 2) mutation present in many MPN patients, and the symptomatology they experience. This retrospective study analysed data collected from MPN patients included in the Myeloproliferative Neoplasms: An In-depth Case-Control (MOSAICC) pilot study. The MPN Symptom Assessment Form was administered, and median symptom scores were compared between JAK2V617F-positive and JAK2V617F-negative groups. Multivariate logistic regression analysis adjusted for confounding variables. Overall, 106 MPN patients participated: 65.1% were JAK2V617F positive, 30.2% were JAK2V617F negative and 4.7% had an unknown status. Multivariate analysis revealed a low symptom burden for early satiety (p < 0.01), dizziness (p < 0.05), cough (p < 0.05) and bone pain (p < 0.01) in those receiving venesection alone. Interferon alpha was significantly associated (p < 0.05) with severe burden for 16 of the 27 symptoms. JAK2V617F-positive females experienced a greater symptom burden than JAK2V617F-positive males. There was no discernible relationship between the JAK2V617F mutation and symptom burden in MPN patients, unlike the therapeutic agents investigated. Larger studies are required to validate these results and identify mechanisms of symptom development and control in MPN patients.
Collapse
Affiliation(s)
- Lai Yee Orbell
- School of Medicine, Medical Sciences, and NutritionUniversity of AberdeenAberdeenScotlandUK
| | - Nouf Abutheraa
- School of Medicine, Medical Sciences, and NutritionUniversity of AberdeenAberdeenScotlandUK
- Aberdeen Centre for Health Data ScienceUniversity of AberdeenAberdeenScotlandUK
| | - Andrew S Duncombe
- University Hospital Southampton NHS Foundation Trust, SouthamptonEnglandUK
| | - Mary Frances McMullin
- School of Medicine, Dentistry and Biomedical SciencesQueen's University BelfastBelfastNorthern IrelandUK
| | - Ruben Mesa
- Mays Cancer Center at UT Health San AntonioSan AntonioTexasUSA
| | - Charlene M McShane
- School of Medicine, Dentistry and Biomedical SciencesQueen's University BelfastBelfastNorthern IrelandUK
- Centre for Public HealthQueen's University BelfastBelfastNorthern IrelandUK
| | - Glen James
- Centre for Public HealthQueen's University BelfastBelfastNorthern IrelandUK
| | - Lesley A Anderson
- Aberdeen Centre for Health Data ScienceUniversity of AberdeenAberdeenScotlandUK
| |
Collapse
|
10
|
Moliterno AR, Kaizer H, Reeves BN. JAK2 V617F allele burden in polycythemia vera: burden of proof. Blood 2023; 141:1934-1942. [PMID: 36745865 PMCID: PMC10163319 DOI: 10.1182/blood.2022017697] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/09/2023] [Accepted: 01/23/2023] [Indexed: 02/08/2023] Open
Abstract
Polycythemia vera (PV) is a hematopoietic stem cell neoplasm defined by activating somatic mutations in the JAK2 gene and characterized clinically by overproduction of red blood cells, platelets, and neutrophils; a significant burden of disease-specific symptoms; high rates of vascular events; and evolution to a myelofibrosis phase or acute leukemia. The JAK2V617F variant allele frequency (VAF) is a key determinant of outcomes in PV, including thrombosis and myelofibrotic progression. Here, we critically review the dynamic role of JAK2V617F mutation burden in the pathogenesis and natural history of PV, the suitability of JAK2V617F VAF as a diagnostic and prognostic biomarker, and the utility of JAK2V617F VAF reduction in PV treatment.
Collapse
Affiliation(s)
- Alison R. Moliterno
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Hannah Kaizer
- Division of Hematology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Brandi N. Reeves
- Division of Hematology, Department of Medicine, Blood Research Center, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
11
|
Chia YC, Siti Asmaa MJ, Ramli M, Woon PY, Johan MF, Hassan R, Islam MA. Molecular Genetics of Thrombotic Myeloproliferative Neoplasms: Implications in Precision Oncology. Diagnostics (Basel) 2023; 13:163. [PMID: 36611455 PMCID: PMC9818412 DOI: 10.3390/diagnostics13010163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/24/2022] [Accepted: 11/28/2022] [Indexed: 01/06/2023] Open
Abstract
Classical BCR-ABL-negative myeloproliferative neoplasms (MPN) include polycythaemia vera, essential thrombocythaemia, and primary myelofibrosis. Unlike monogenic disorders, a more complicated series of genetic mutations are believed to be responsible for MPN with various degrees of thromboembolic and bleeding complications. Thrombosis is one of the early manifestations in patients with MPN. To date, the driver genes responsible for MPN include JAK2, CALR, MPL, TET2, ASXL1, and MTHFR. Affords have been done to elucidate these mutations and the incidence of thromboembolic events. Several lines of evidence indicate that mutations in JAK2, MPL, TET2 and ASXL1 gene and polymorphisms in several clotting factors (GPIa, GPIIa, and GPIIIa) are associated with the occurrence and prevalence of thrombosis in MPN patients. Some polymorphisms within XRCC1, FBG, F2, F5, F7, F12, MMP9, HPA5, MTHFR, SDF-1, FAS, FASL, TERT, ACE, and TLR4 genes may also play a role in MPN manifestation. This review aims to provide an insightful overview on the genetic perspective of thrombotic complications in patients with MPN.
Collapse
Affiliation(s)
- Yuh Cai Chia
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Mat Jusoh Siti Asmaa
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Marini Ramli
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Peng Yeong Woon
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien 97004, Taiwan
| | - Muhammad Farid Johan
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Rosline Hassan
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Md Asiful Islam
- Department Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
12
|
Splanchnic Vein Thrombosis in Myeloproliferative Neoplasms: Treatment Considerations and Unmet Needs. Cancers (Basel) 2022; 15:cancers15010011. [PMID: 36612008 PMCID: PMC9817858 DOI: 10.3390/cancers15010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Patients who develop splanchnic vein thrombosis (SVT) in the setting of a myeloproliferative neoplasm (MPN) are at risk for complications including portal hypertension, bleeding, thrombosis, and death. Prompt multidisciplinary treatment is thus necessary to prevent long-term sequelae. However, optimal management strategies are not well established due to a paucity of data. In this review, we very briefly discuss the epidemiology, pathophysiology, and prognosis of MPN-SVT and then more comprehensively explore treatment considerations of MPN-SVT, including anticoagulation, endovascular/surgical intervention, and cytoreductive therapy. We will also highlight current gaps in our knowledge of MPN-SVT and conclude by suggesting future directions to optimize the treatment of MPN-SVT and improve outcomes.
Collapse
|
13
|
Osho P, Ojo M, Osho E. Profile of polycythaemia vera in South Western Nigeria. NIGERIAN JOURNAL OF MEDICINE 2022. [DOI: 10.4103/njm.njm_56_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
14
|
Spivak JL. Advances in polycythemia vera and lessons for acute leukemia. Best Pract Res Clin Haematol 2021; 34:101330. [PMID: 34865702 DOI: 10.1016/j.beha.2021.101330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The myeloproliferative neoplasms (MPN), polycythemia vera (PV), essential thrombocytosis and primary myelofibrosis, are an unusual group of myeloid neoplasms, which arise in a pluripotent hematopoietic stem cell (HSC) due to gain of function driver mutations in the JAK2, CALR and MPL genes that constitutively activate JAK2, the cognate tyrosine kinase of the type 1 hematopoietic growth factor (HGF) receptors. PV is the ultimate phenotypic expression of constitutive JAK2 activation since it alone of the three MPN is characterized by overproduction of normal red cells, white cells and platelets. Paradoxically, however, although PV is a panmyelopathy involving myeloid, erythroid and megakaryocytic progenitor cells, pluripotent HSC only express a single type of HGF receptor, the thrombopoietin receptor, MPL. In this review, the basis for how a pluripotent HSC with one type of HGF can give rise to three separate types of myeloid cells will be explained and it will be demonstrated that PV is actually a hormone-sensitive disorder, characterized by elevated thrombopoietin levels. Finally, it will be shown that the most common form of acute leukemia in PV is due to the inappropriate use of chemotherapy, including hydroxyurea, which facilitates expansion of DNA-damaged, mutated HSC at the expense of their normal counterparts.
Collapse
Affiliation(s)
- Jerry L Spivak
- Division of Hematology, Johns Hopkins University School of Medicine, Traylor 924, 720 Rutland Avenue, Baltimore, MD, 20037, USA.
| |
Collapse
|
15
|
Abstract
Myeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell (HSC) disorders with overproduction of mature myeloid blood cells, including essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF). In 2005, several groups identified a single gain-of-function point mutation JAK2V617F in the majority of MPN patients. The JAK2V617F mutation confers cytokine independent proliferation to hematopoietic progenitor cells by constitutively activating canonical and non-canonical downstream pathways. In this chapter, we focus on (1) the regulation of JAK2, (2) the molecular mechanisms used by JAK2V617F to induce MPNs, (3) the factors that are involved in the phenotypic diversity in MPNs, and (4) the effects of JAK2V617F on hematopoietic stem cells (HSCs). The discovery of the JAK2V617F mutation led to a comprehensive understanding of MPN; however, the question still remains about how one mutation can give rise to three distinct disease entities. Various mechanisms have been proposed, including JAK2V617F allele burden, differential STAT signaling, and host genetic modifiers. In vivo modeling of JAK2V617F has dramatically enhanced the understanding of the pathophysiology of the disease and provided the pre-clinical platform. Interestingly, most of these models do not show an increased hematopoietic stem cell self-renewal and function compared to wildtype controls, raising the question of whether JAK2V617F alone is sufficient to give a clonal advantage in MPN patients. In addition, the advent of modern sequencing technologies has led to a broader understanding of the mutational landscape and detailed JAK2V617F clonal architecture in MPN patients.
Collapse
|
16
|
Castiglione M, Zhang H, Kaushansky K, Zhan H. Cell competition between wild-type and JAK2V617F mutant cells in a murine model of a myeloproliferative neoplasm. Exp Hematol 2021; 100:52-62. [PMID: 34153382 PMCID: PMC9911310 DOI: 10.1016/j.exphem.2021.06.003] [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: 05/27/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 10/21/2022]
Abstract
The myeloproliferative neoplasms (MPNs) are characterized by overproduction of mature blood cells and increased risk of transformation to frank leukemia. The acquired kinase mutation JAK2V617F plays a central role in a majority of patients with these diseases. As MPNs are clonal stem cell disorders (i.e. arise from a single stem cell which eventually expands), the hematopoietic stem/progenitor cell (HSPC) compartment in MPNs is heterogeneous with the presence of both JAK2 wild-type and JAK2V617F mutant cells. Mechanisms responsible for the mutant stem cell expansion in MPNs are not fully understood. Utilizing in vitro co-culture assays and in vivo competitive transplantation assays, we show that the presence of wild-type cells alters both the gene expression profile and cellular function of JAK2V617F mutant HSPCs and inhibits the expansion of co-existing JAK2V617F mutant cells in a normal microenvironment. In contrast, we found that a microenvironment bearing the mutant kinase promotes JAK2V617F mutant HSPC expansion over wild-type cells due in part to altered CXCL12/CXCR4 signaling. Further understanding of the molecular mechanisms controlling the competitive interactions between normal and JAK2V617F mutant cells, and how these mechanisms break down during MPN disease progression hold great potential for advances in treating patients with these diseases.
Collapse
Affiliation(s)
| | - Haotian Zhang
- Graduate Program in Molecular & Cellular Biology, Stony Brook University, Stony Brook, NY
| | - Kenneth Kaushansky
- Office of the Sr. Vice President, Health Sciences, Stony Brook School of Medicine, Stony Brook, NY
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY; Medical Service, Northport VA Medical Center, Northport, NY.
| |
Collapse
|
17
|
Clinical insights into the origins of thrombosis in myeloproliferative neoplasms. Blood 2021; 137:1145-1153. [PMID: 33237986 DOI: 10.1182/blood.2020008043] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs), polycythemia vera, essential thrombocythemia, and primary myelofibrosis, are hematopoietic stem cell disorders that are defined by activating mutations in signal transduction pathways and are characterized clinically by the overproduction of platelets, red blood cells, and neutrophils, significant burden of disease-specific symptoms, and high rates of vascular events. The focus of this review is to critically reevaluate the clinical burden of thrombosis in MPNs, to review the clinical associations among clonal hematopoiesis, JAK2V617F burden, inflammation, and thrombosis, and to provide insights into novel primary and secondary thrombosis-prevention strategies.
Collapse
|
18
|
Greenfield G, McMullin MF, Mills K. Molecular pathogenesis of the myeloproliferative neoplasms. J Hematol Oncol 2021; 14:103. [PMID: 34193229 PMCID: PMC8246678 DOI: 10.1186/s13045-021-01116-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
The Philadelphia negative myeloproliferative neoplasms (MPN) compromise a heterogeneous group of clonal myeloid stem cell disorders comprising polycythaemia vera, essential thrombocythaemia and primary myelofibrosis. Despite distinct clinical entities, these disorders are linked by morphological similarities and propensity to thrombotic complications and leukaemic transformation. Current therapeutic options are limited in disease-modifying activity with a focus on the prevention of thrombus formation. Constitutive activation of the JAK/STAT signalling pathway is a hallmark of pathogenesis across the disease spectrum with driving mutations in JAK2, CALR and MPL identified in the majority of patients. Co-occurring somatic mutations in genes associated with epigenetic regulation, transcriptional control and splicing of RNA are variably but recurrently identified across the MPN disease spectrum, whilst epigenetic contributors to disease are increasingly recognised. The prognostic implications of one MPN diagnosis may significantly limit life expectancy, whilst another may have limited impact depending on the disease phenotype, genotype and other external factors. The genetic and clinical similarities and differences in these disorders have provided a unique opportunity to understand the relative contributions to MPN, myeloid and cancer biology generally from specific genetic and epigenetic changes. This review provides a comprehensive overview of the molecular pathophysiology of MPN exploring the role of driver mutations, co-occurring mutations, dysregulation of intrinsic cell signalling, epigenetic regulation and genetic predisposing factors highlighting important areas for future consideration.
Collapse
Affiliation(s)
- Graeme Greenfield
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK.
| | | | - Ken Mills
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| |
Collapse
|
19
|
Karantanos T, Jain T, Moliterno AR, Jones RJ, DeZern AE. Sex-Related Differences in Chronic Myeloid Neoplasms: From the Clinical Observation to the Underlying Biology. Int J Mol Sci 2021; 22:2595. [PMID: 33807519 PMCID: PMC7961949 DOI: 10.3390/ijms22052595] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/01/2021] [Accepted: 03/02/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic myeloid neoplasms are clonal diseases with variable clinical course and outcomes and despite the introduction of novel therapies, patients with high-risk disease continue to have overall poor outcomes. Different groups have highlighted that men have overall worse survival and higher incidence of transformation to acute leukemia compared to women across neoplasms such as myelodysplastic syndrome (MDS), myeloproliferative neoplasms (MPN), MDS/MPN overlap neoplasms, and CML. More recent studies evaluating the genomic profile of patients with these neoplasms demonstrated a male predominance for mutations in high-risk genes including ASXL1, U2AF1, SRSF2 and ZRSR2. The understanding of the underlying biology is limited but a number of hypotheses have been developed and are currently being investigated. This review summarizes the current knowledge about sex-related differences in the clinical outcomes and genomic profile of patients with chronic myeloid neoplasms and discusses the hypothesized biologic mechanisms as an attempt to explain these observations.
Collapse
MESH Headings
- Animals
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Mutation/genetics
- Myelodysplastic Syndromes/genetics
- Myelodysplastic Syndromes/pathology
- Myeloproliferative Disorders/genetics
- Myeloproliferative Disorders/pathology
- Sex Characteristics
Collapse
Affiliation(s)
- Theodoros Karantanos
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA; (T.J.); (R.J.J.); (A.E.D.)
| | - Tania Jain
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA; (T.J.); (R.J.J.); (A.E.D.)
| | - Alison R. Moliterno
- Division of Adult Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD 21231, USA;
| | - Richard J. Jones
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA; (T.J.); (R.J.J.); (A.E.D.)
| | - Amy E. DeZern
- Division of Hematological Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA; (T.J.); (R.J.J.); (A.E.D.)
| |
Collapse
|
20
|
Shah SZ, Raza N, Nasir MI, Zaidi SMH. Frequency of Zygosity in Jak-2 Positive Patients with Polycythemia Vera-Pakistan's Perspective. Asian Pac J Cancer Prev 2021; 22:559-564. [PMID: 33639674 PMCID: PMC8190342 DOI: 10.31557/apjcp.2021.22.2.559] [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: 02/24/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Estimation of JAK2V617F mutational load in Polycythemia Vera (PV) helps to determine the severity of the disease phenotype, the risk of thrombotic events, progression to post-PV myelofibrosis and survival. Amplification Refractory Mutation Screening (ARMS) PCR or Allele Specific (AS) PCR is a simple easy method with a reasonable sensitivity for screening of zygosity.The purpose of this study was to see the frequency of disease burden and phenotypic characteristics in Pakistani patients diagnosed with JAK2V617F mutation positive PV. MATERIALS AND METHODS A cross-sectional study using non probability consecutive sampling was conducted at Hematology Department, Liaquat National Hospital Karachi from October 2018 to July 2019.Adult newly diagnosed JAK2V617F positive PV patients of either gender were included. Patients' demographics, clinical characteristics and baseline CBC were noted. JAK2V617F zygosity was qualitatively analyzed by ARMS-PCR technique. Age and gender were stratified to see to see the result of qualitative and quantitative effect modifiers on these patients using Chi Square and fisher exact test as appropriate while mean comparison was done by independent t-test and one way ANOVAtest. P value of ≤0.05 was considered as significant. RESULTS Fifty one patients were included in the study with an average age of 59.60±14.29years.90.2% of patients had hypertension.All patients tested positive for heterozygous state. Significant association of gender was found with smoking (p=0.001) while age was significantly linked with hypertension (p-0.033). CONCLUSION JAK2V617F positive PV patients are mainly heterozygous males showing significant association with smoking and hypertension. ARMS-PCR is a robust technique to determine zygosity which can be used for screening purposes.<br />.
Collapse
Affiliation(s)
- Syed Zubair Shah
- Department of Hematology, Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Naila Raza
- Department of Hematology, Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Muhammad Israr Nasir
- Department of Molecular Pathology, Liaquat National Hospital and Medical College, Karachi, Pakistan
| | | |
Collapse
|
21
|
Zhan H, Kaushansky K. The Hematopoietic Microenvironment in Myeloproliferative Neoplasms: The Interplay Between Nature (Stem Cells) and Nurture (the Niche). ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1273:135-145. [PMID: 33119879 DOI: 10.1007/978-3-030-49270-0_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Hematopoietic stem cells (HSCs) rely on instructive cues from the marrow microenvironment for their maintenance and function. Accumulating evidence indicates that the survival and proliferation of hematopoietic neoplasms are dependent not only on cell-intrinsic, genetic mutations, and other molecular alterations present within neoplastic stem cells, but also on the ability of the surrounding microenvironmental cells to nurture and promote the malignancy. It is anticipated that a better understanding of the molecular and cellular events responsible for these microenvironmental features of neoplastic hematopoiesis will lead to improved treatment for patients. This review will focus on the myeloproliferative neoplasms (MPNs), polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), in which an acquired signaling kinase mutation (JAK2V617F) plays a central, pathogenetic role in 50-100% of patients with these disorders. Evidence is presented that the development of an MPN requires both an abnormal, mutation-bearing (i.e., neoplastic) HSC and an abnormal, mutation-bearing microenvironment.
Collapse
Affiliation(s)
- Huichun Zhan
- Division of Hematology-Oncology, Department of Medicine, Stony Brook School of Medicine, Stony Brook, NY, USA. .,Northport VA Medical Center, Northport, NY, USA.
| | | |
Collapse
|
22
|
Chia YC, Ramli M, Woon PY, Johan MF, Hassan R, Islam MA. WITHDRAWN: Molecular genetics of thrombotic myeloproliferative neoplasms: Implications in precision oncology. Genes Dis 2021. [DOI: 10.1016/j.gendis.2021.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
23
|
Palandri F, Mora B, Gangat N, Catani L. Is there a gender effect in polycythemia vera? Ann Hematol 2021; 100:11-25. [PMID: 33006021 PMCID: PMC7782364 DOI: 10.1007/s00277-020-04287-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022]
Abstract
In recent times, there has been a growing interest in understanding the impact of gender on disease biology and clinical outcomes in Philadelphia-negative chronic myeloproliferative neoplasms. Among those, polycythemia vera (PV) is characterized by increased thrombotic risk, systemic symptoms, and overall reduced survival. Here, we aim to summarize data on whether and to what extent female sex can affect PV biology and outcome. To this end, we will discuss the latest acquisitions in terms of pathogenesis, diagnosis, epidemiology, clinical presentation and symptoms burden, thrombotic risk and related treatment strategies, and prognosis in female patients affected by PV.
Collapse
Affiliation(s)
- Francesca Palandri
- Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy
| | - Barbara Mora
- Hematology, ASST Sette Laghi, Ospedale di Circolo, Varese, Italy
| | | | - Lucia Catani
- Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, Bologna, Italy
| |
Collapse
|
24
|
Moliterno AR, Kaizer H. Applied genomics in MPN presentation. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2020; 2020:434-439. [PMID: 33275725 PMCID: PMC7727573 DOI: 10.1182/hematology.2020000128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Polycythemia vera, essential thrombocytosis (ET), and primary myelofibrosis (PMF) are grouped together as myeloproliferative neoplasms (MPNs) because of shared clinical, pathologic, and molecular features. The 2005 discovery of the driver mutation JAK2V617F, found in more than 70% of individuals with MPNs and 98% of those with PV, has transformed the diagnosis and management of MPNs. Although PV is the most common phenotype associated with JAK2V617F, roughly 60% of individuals with ET or PMF also have the mutation, and JAK2V617F is now recognized as a common lesion in clonal hematopoiesis (CH). JAK2V617F+ CH and MPN are indolent disorders that evolve over time, with transitions to different disease phases, transformation to bone marrow failure or leukemia, and high thrombosis rates. Genomic assessment has taken center stage as an important tool to define disease phenotype, disease burden, prognosis, and even thrombosis risk of MPNs. Genomics has also unveiled the causes and factors that modify the risk of acquiring and expanding CH and MPNs and points to new pathways for targeted therapies to treat and ultimately prevent them. Genomic assessment of patients with MPNs, like other cancers, enables the clinician to capitalize on large population data sets to inform the individual patient of risk, identify treatment, and improve outcomes.
Collapse
Affiliation(s)
| | - Hannah Kaizer
- Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
25
|
Macauda A, Giaccherini M, Sainz J, Gemignani F, Sgherza N, Sánchez-Maldonado JM, Gora-Tybor J, Martinez-Lopez J, Carreño-Tarragona G, Jerez A, Spadano R, Gołos A, Jurado M, Hernández-Mohedo F, Mazur G, Tavano F, Butrym A, Várkonyi J, Canzian F, Campa D. Do myeloproliferative neoplasms and multiple myeloma share the same genetic susceptibility loci? Int J Cancer 2020; 148:1616-1624. [PMID: 33038278 DOI: 10.1002/ijc.33337] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/04/2020] [Accepted: 08/27/2020] [Indexed: 01/22/2023]
Abstract
Myeloproliferative neoplasms (MPNs) are a group of diseases that cause myeloid hematopoietic cells to overproliferate. Epidemiological and familial studies suggest that genetic factors contribute to the risk of developing MPN, but the genetic susceptibility of MPN is still not well known. Indeed, only few loci are known to have a clear role in the predisposition to this disease. Some studies reported a diagnosis of MPNs and multiple myeloma (MM) in the same patients, but the biological causes are still unclear. We tested the hypothesis that the two diseases share at least partly the same genetic risk loci. In the context of a European multicenter study with 460 cases and 880 controls, we analyzed the effect of the known MM risk loci, individually and in a polygenic risk score (PRS). The most significant result was obtained among patients with chronic myeloid leukemia (CML) for PS0RS1C1-rs2285803, which showed to be associated with an increased risk (OR = 3.28, 95% CI 1.79-6.02, P = .00012, P = .00276 when taking into account multiple testing). Additionally, the PRS showed an association with MPN risk when comparing the last with the first quartile of the PRS (OR = 2.39, 95% CI 1.64-3.48, P = 5.98 × 10-6 ). In conclusion, our results suggest a potential common genetic background between MPN and MM, which needs to be further investigated.
Collapse
Affiliation(s)
- Angelica Macauda
- Department of Biology, University of Pisa, Pisa, Italy
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matteo Giaccherini
- Department of Biology, University of Pisa, Pisa, Italy
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Juan Sainz
- Genomic Oncology Area, GENYO. Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain
- Monoclonal Gammopathies Unit, University Hospital Virgen de las Nieves, Granada, Spain
- Pharmacogenetics Unit, Instituto de Investigación Biosanitaria de Granada (Ibs. Granada), Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
- Department of Medicine, University of Granada, Granada, Spain
| | | | - Nicola Sgherza
- Division of Hematology, Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - José Manuel Sánchez-Maldonado
- Pharmacogenetics Unit, Instituto de Investigación Biosanitaria de Granada (Ibs. Granada), Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
- Department of Medicine, University of Granada, Granada, Spain
| | | | | | | | - Andrés Jerez
- Hematology and Medical Oncology Department, Hospital Morales Meseguer, IMIB, Murcia, Spain
| | - Raffaele Spadano
- Division of Hematology, Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Aleksandra Gołos
- Department of Clinical Oncology and Chemotherapy, Magodent Hospital, Warsaw, Poland
| | - Manuel Jurado
- Genomic Oncology Area, GENYO. Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain
- Pharmacogenetics Unit, Instituto de Investigación Biosanitaria de Granada (Ibs. Granada), Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
- Department of Medicine, University of Granada, Granada, Spain
| | - Francisca Hernández-Mohedo
- Genomic Oncology Area, GENYO. Centre for Genomics and Oncological Research: Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, Granada, Spain
- Pharmacogenetics Unit, Instituto de Investigación Biosanitaria de Granada (Ibs. Granada), Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
- Department of Medicine, University of Granada, Granada, Spain
| | - Grzegorz Mazur
- Department of Internal Medicine, Occupational Diseases, Hypertension and Clinical Oncology, Wroclaw Medical University, Wroclaw, Poland
| | - Francesca Tavano
- Division of Gastroenterology and Research Laboratory, Fondazione IRCCS Casa Sollievo della Sofferenza, Foggia, Italy
| | - Aleksandra Butrym
- Department of Cancer Prevention and Therapy, Wroclaw Medical University, Wroclaw, Poland
| | - Judit Várkonyi
- Third Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Federico Canzian
- Genomic Epidemiology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| |
Collapse
|
26
|
Karantanos T, Chaturvedi S, Braunstein EM, Spivak J, Resar L, Karanika S, Williams DM, Rogers O, Gocke CD, Moliterno AR. Sex determines the presentation and outcomes in MPN and is related to sex-specific differences in the mutational burden. Blood Adv 2020; 4:2567-2576. [PMID: 32542392 PMCID: PMC7322953 DOI: 10.1182/bloodadvances.2019001407] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
The factors underlying the variable presentation and clinical course of myeloproliferative neoplasms (MPNs) remain unclear. The aim of this study was to evaluate the independent effect of sex on MPN presentation and outcomes. A total of 815 patients with essential thrombocytosis, polycythemia vera, or primary myelofibrosis were evaluated between 2005 and 2019, and the association of sex with presenting phenotype, JAK2 V617F burden, progression, and survival was examined. Men presented more often with primary myelofibrosis vs essential thrombocytosis (relative risk, 3.2; P < .001) and polycythemia vera (relative risk, 2.1; P < .001), had higher rates of transformation to secondary myelofibrosis (hazard ratio [HR], 1.55; P = .013) and acute myeloid leukemia (HR, 3.67; P < .001), and worse survival (HR, 1.63; P = .001) independent of age, phenotype at diagnosis, and MPN-specific mutation. Men had higher JAK2 V617F allele burdens in their CD34+ cells (P = .001), acquired more somatic mutations (P = .012) apart from the MPN-specific mutations, and had an increased frequency of 1 (odds ratio, 2.35; P = .017) and 2 (odds ratio, 20.20; P = .011) high-risk mutations independent of age, phenotype, and driver mutation. Male sex is an independent predictor of poor outcomes in MPNs. This seems to be due to an increased risk of non-MPN-specific somatic mutations, particularly high-risk mutations, rather than MPN-specific mutation allele frequency. Conversely, disease progression in female subjects is more dependent on JAK2 mutation allele burden than on acquisition of other somatic mutations. Sex should be considered in prognostic models and when evaluating therapeutic strategies in MPNs.
Collapse
Affiliation(s)
- Theodoros Karantanos
- Department of Oncology, Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center
| | | | | | - Jerry Spivak
- Division of Adult Hematology, Department of Medicine
| | - Linda Resar
- Department of Oncology, Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center
- Division of Adult Hematology, Department of Medicine
- Department of Pathology, and
| | - Styliani Karanika
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | | | | | |
Collapse
|
27
|
Sacco M, Ranalli P, Lancellotti S, Petrucci G, Dragani A, Rocca B, De Cristofaro R. Increased von Willebrand factor levels in polycythemia vera and phenotypic differences with essential thrombocythemia. Res Pract Thromb Haemost 2020; 4:413-421. [PMID: 32211575 PMCID: PMC7086469 DOI: 10.1002/rth2.12315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/13/2020] [Accepted: 01/17/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Acquired von Willebrand factor (VWF) deficiency was described in Philadelphia-negative myeloproliferative neoplasms, especially in essential thrombocythemia (ET). VWF phenotype in contemporary patients with polycythemia vera (PV) remains less explored. OBJECTIVES To characterize the VWF phenotype in PV and to compare VWF phenotype in PV with matched healthy subjects and ET patients. PATIENTS/METHODS We studied 48 PV patients, treated according to current recommendations (hematocrit ≤ 45%, on low-dose aspirin prophylaxis); 48 healthy and 41 subjects with ET, all sex, age, and blood group matched. We measured VWF antigen, activity, multimeric pattern, ADAMTS-13, and factor VIII (FVIII) antigen. RESULTS In patients with PV, VWF antigen and activity were significantly higher than in healthy subjects (antigen: 119[96-137] vs 93[79-107] IU/dL; activity: 114[95-128] vs 90[79-107] IU/dL, respectively, medians and interquartile, P < 0.01), with normal multimeric distribution. ADAMTS-13 levels were similar between patients with PV and healthy subjects. FVIII levels were higher in PV than in healthy subjects (141[119-169] versus 98[88-123] IU/dL, respectively, P < 0.01). By multivariable analysis, JAK2-p.V617F allelic burden, erythrocyte count, and male sex significantly predicted VWF antigen and activity levels. As compared to patients with ET, patients with PV showed similar VWF antigen levels but approximately 40% higher activity (79[49-104] vs 112[93-125] IU/dL, respectively, P < 0.01). CONCLUSIONS Patients with PV show increased VWF and FVIII levels, predicted by JAK2-p.V617F burden and erythrocyte count. At variance with ET, acquired VWF defect was not observed in PV. High VWF/FVIII levels may sustain the thrombotic diathesis of PV and may be investigated as biomarkers for risk stratification.
Collapse
Affiliation(s)
- Monica Sacco
- Servizio Malattie Emorragiche e TromboticheFondazione Policlinico Universitario “A. Gemelli” IRCCSRomaItaly
| | - Paola Ranalli
- Dipartimento di Medicina Interna e Chirurgia TraslazionaleFacoltà di Medicina e Chirurgia “A. Gemelli”Università Cattolica del Sacro CuoreRomaItaly
| | | | - Giovanna Petrucci
- Istituto di FarmacologiaFacoltà di Medicina e Chirurgia “A. Gemelli”Università Cattolica del S. Cuore and Fondazione Policlinico Universitario “A. Gemelli” IRCCSRomeItaly
| | - Alfredo Dragani
- Dipartimento di Medicina Interna e Chirurgia TraslazionaleFacoltà di Medicina e Chirurgia “A. Gemelli”Università Cattolica del Sacro CuoreRomaItaly
| | - Bianca Rocca
- Istituto di FarmacologiaFacoltà di Medicina e Chirurgia “A. Gemelli”Università Cattolica del S. Cuore and Fondazione Policlinico Universitario “A. Gemelli” IRCCSRomeItaly
| | - Raimondo De Cristofaro
- Servizio Malattie Emorragiche e TromboticheFondazione Policlinico Universitario “A. Gemelli” IRCCSRomaItaly
- Dipartimento di EmatologiaOspedale S. SpiritoPescaraItaly
| |
Collapse
|
28
|
Cuthbert D, Stein BL. Polycythemia Vera-Associated Complications: Pathogenesis, Clinical Manifestations, And Effects On Outcomes. J Blood Med 2019; 10:359-371. [PMID: 31695542 PMCID: PMC6805785 DOI: 10.2147/jbm.s189922] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/18/2019] [Indexed: 12/13/2022] Open
Abstract
Polycythemia vera is a Philadelphia-negative chronic myeloproliferative neoplasm, characterized by erythrocytosis, which is unique, compared to essential thrombocytosis and primary myelofibrosis. Though longevity can usually be expected, vascular morbidity is associated with this condition, as well as a propensity to evolve into myelofibrosis (post-PV MF) and acute myeloid leukemia. In addition, patients can have a pronounced symptom burden. Herein, contributors to the symptomatic burden, as well as the thrombotic and transformative tendencies are reviewed. From a symptom perspective, some are explained by cytokine release, others by microvascular complications, whereas certain symptoms can herald disease evolution. Thrombosis has multifactorial contributors, including but not limited to gender, and inflammatory stress; investigators have recently hypothesized that microparticles and Neutrophil Extracellular Trap Formations may add to thrombotic burden. Finally, we examine the progression to post-PV MF as well as leukemic transformation, highlighting well-established risk factors including age and leukocytosis, certain treatments, and the presence of “non-driver” mutations.
Collapse
Affiliation(s)
- Danielle Cuthbert
- McGaw Medical Center of Northwestern University, Department of Internal Medicine, Chicago, IL 60611, USA
| | - Brady Lee Stein
- Northwestern University Feinberg School of Medicine, Division of Hematology/Oncology, Department of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
29
|
Chapeau EA, Mandon E, Gill J, Romanet V, Ebel N, Powajbo V, Andraos-Rey R, Qian Z, Kininis M, Zumstein-Mecker S, Ito M, Hynes NE, Tiedt R, Hofmann F, Eshkind L, Bockamp E, Kinzel B, Mueller M, Murakami M, Baffert F, Radimerski T. A conditional inducible JAK2V617F transgenic mouse model reveals myeloproliferative disease that is reversible upon switching off transgene expression. PLoS One 2019; 14:e0221635. [PMID: 31600213 PMCID: PMC6786561 DOI: 10.1371/journal.pone.0221635] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 08/12/2019] [Indexed: 11/19/2022] Open
Abstract
Aberrant activation of the JAK/STAT pathway is thought to be the critical event in the pathogenesis of the chronic myeloproliferative neoplasms, polycythemia vera, essential thrombocythemia and primary myelofibrosis. The most frequent genetic alteration in these pathologies is the activating JAK2V617F mutation, and expression of the mutant gene in mouse models was shown to cause a phenotype resembling the human diseases. Given the body of genetic evidence, it has come as a sobering finding that JAK inhibitor therapy only modestly suppresses the JAK2V617F allele burden, despite showing clear benefits in terms of reducing splenomegaly and constitutional symptoms in patients. To gain a better understanding if JAK2V617F is required for maintenance of myeloproliferative disease once it has evolved, we generated a conditional inducible transgenic JAK2V617F mouse model using the SCL-tTA-2S tet-off system. Our model corroborates that expression of JAK2V617F in hematopoietic stem and progenitor cells recapitulates key hallmarks of human myeloproliferative neoplasms, and exhibits gender differences in disease manifestation. The disease was found to be transplantable, and importantly, reversible when transgenic JAK2V617F expression was switched off. Our results indicate that mutant JAK2V617F-specific inhibitors should result in profound disease modification by disabling the myeloproliferative clone bearing mutant JAK2.
Collapse
Affiliation(s)
- Emilie A. Chapeau
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
- * E-mail:
| | - Emeline Mandon
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Jason Gill
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Vincent Romanet
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Nicolas Ebel
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Violetta Powajbo
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Rita Andraos-Rey
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Zhiyan Qian
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Miltos Kininis
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | | | - Moriko Ito
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Nancy E. Hynes
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Ralph Tiedt
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Francesco Hofmann
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Leonid Eshkind
- Institute for Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Ernesto Bockamp
- Institute for Translational Immunology and Research Center for Immunotherapy, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Bernd Kinzel
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Matthias Mueller
- Chemical Biology and Therapeutics, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Masato Murakami
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Fabienne Baffert
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Thomas Radimerski
- Disease Area Oncology, Novartis Institutes for BioMedical Research, Basel, Switzerland
| |
Collapse
|
30
|
Farge D, Le Maignan C, Doucet L, Frere C. Women, thrombosis, and cancer. Thromb Res 2019; 181 Suppl 1:S47-S53. [DOI: 10.1016/s0049-3848(19)30367-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/12/2019] [Accepted: 02/15/2019] [Indexed: 12/12/2022]
|
31
|
Abstract
Since its discovery, polycythemia vera (PV) has challenged clinicians responsible for its diagnosis and management and scientists investigating its pathogenesis. As a clonal hematopoietic stem cell (HSC) disorder, PV is a neoplasm but its driver mutations result in overproduction of morphologically and functionally normal blood cells. PV arises in an HSC but it can present initially as isolated erythrocytosis, leukocytosis, thrombocytosis, or any combination of these together with splenomegaly or myelofibrosis, and it can take years for a true panmyelopathy to appear. PV shares the same JAK2 mutation as essential thrombocytosis and primary myelofibrosis, but erythrocytosis only occurs in PV. However, unlike secondary causes of erythrocytosis, in PV, the plasma volume is frequently expanded, masking the erythrocytosis and making diagnosis difficult if this essential fact is ignored. PV is not a monolithic disorder: female patients deregulate fewer genes and clinically behave differently than their male counterparts, while some PV patients are genetically predisposed to an aggressive clinical course. Nevertheless, based on what we have learned over the past century, most PV patients can lead long and productive lives. In this review, using clinical examples, I describe how I diagnose and manage PV in an evidence-based manner without relying on chemotherapy.
Collapse
|
32
|
Farge D, Le Maignan C, Doucet L, Frere C. WITHDRAWN: Women, thrombosis, and cancer. Thromb Res 2019. [DOI: 10.1016/j.thromres.2019.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
33
|
Grunwald MR, Stein BL, Boccia RV, Oh ST, Paranagama D, Parasuraman S, Colucci P, Mesa R. Clinical and Disease Characteristics From REVEAL at Time of Enrollment (Baseline): Prospective Observational Study of Patients With Polycythemia Vera in the United States. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2018; 18:788-795.e2. [PMID: 30245100 PMCID: PMC8148982 DOI: 10.1016/j.clml.2018.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 08/17/2018] [Accepted: 08/21/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Polycythemia vera (PV) has a prevalence of 44 to 57 per 100,000 people in the United States. Prospective data concerning the demographics, clinical characteristics, and treatment patterns of patients with PV in the United States are lacking. PATIENTS AND METHODS The ongoing, prospective, observational REVEAL study evaluates demographics, disease burden, clinical management, patient-reported outcomes, and health care resource utilization of adult patients with PV in the United States. This report summarizes the demographics and clinical characteristics of patients at enrollment (baseline). RESULTS Patients (n = 2510) were a median age of 67.0 years, 54.2% were male, and 89.1% were white. The median time from PV diagnosis to study enrollment was 4.0 (range, 0-56.3) years. Most patients (89.7%) were diagnosed after an abnormal blood test. Less than half (49.2%) underwent JAK2 mutation analysis, of whom 95.8% were JAK2 V617F mutation positive; < 1% were positive for JAK2 exon 12 mutations. At enrollment, 47.7% of patients had elevated hematocrit (> 45%), 35.8% had elevated platelets (> 400 × 109/L), and 37.0% had elevated leukocytes (> 10 × 109/L). Most patients (94.5%) were receiving active PV treatment, predominantly therapeutic phlebotomy alone (33.6%), hydroxyurea monotherapy (29.0%), or hydroxyurea plus phlebotomy (23.7%). Thrombotic events occurred in 11.9% of patients before PV diagnosis (venous, 6.7%; arterial, 5.7%), and 8.3% between diagnosis and enrollment. Hypertension (70.6%) was the most common previous medical condition. CONCLUSION REVEAL enrollment data inform our understanding of the baseline demographics, diagnostic approach, disease characteristics, and treatment patterns of patients with PV in the United States. Longitudinal real-world data collected in this study will complement information collected during randomized controlled clinical trials.
Collapse
Affiliation(s)
| | - Brady L Stein
- Northwestern University Feinberg School of Medicine, Department of Hematology and Oncology, Chicago, IL
| | | | - Stephen T Oh
- Washington University School of Medicine, Divisions of Hematology and Oncology, St Louis, MO
| | | | | | | | - Ruben Mesa
- San Antonio Cancer Center (an NCI Designated Cancer Center), San Antonio, TX
| |
Collapse
|
34
|
Barraco D, Mora B, Guglielmelli P, Rumi E, Maffioli M, Rambaldi A, Caramella M, Komrokji R, Gotlib J, Kiladjian JJ, Cervantes F, Devos T, Palandri F, De Stefano V, Ruggeri M, Silver RT, Benevolo G, Albano F, Merli M, Pietra D, Barbui T, Rotunno G, Cazzola M, Giorgino T, Vannucchi AM, Passamonti F. Gender effect on phenotype and genotype in patients with post-polycythemia vera and post-essential thrombocythemia myelofibrosis: results from the MYSEC project. Blood Cancer J 2018; 8:89. [PMID: 30291232 PMCID: PMC6173714 DOI: 10.1038/s41408-018-0128-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/03/2018] [Accepted: 08/28/2018] [Indexed: 12/25/2022] Open
Affiliation(s)
- Daniela Barraco
- Hematology, Department of Medicine and Surgery, University of Insubria & Ospedale di Circolo, ASST Sette Laghi, Varese, Italy
| | - Barbara Mora
- Hematology, Department of Medicine and Surgery, University of Insubria & Ospedale di Circolo, ASST Sette Laghi, Varese, Italy
| | - Paola Guglielmelli
- CRIMM-Centro Ricerca e Innovazione delle Malattie Mieloproliferative, Department of Experimental and Clinical Medicine, Azienda ospedaliera-Universitaria Careggi, University of Florence, Florence, Italy
| | - Elisa Rumi
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Università di Pavia, Pavia, Italy
| | - Margherita Maffioli
- Hematology, Department of Medicine and Surgery, University of Insubria & Ospedale di Circolo, ASST Sette Laghi, Varese, Italy
| | - Alessandro Rambaldi
- Department of Oncology-Hematology, University of Milan and BMT Unit, ASST Papa Giovanni XXIII, Bergamo, Italy
| | | | | | | | | | | | - Timothy Devos
- Department of Hematology, University Hospitals Leuven and Laboratory of Experimental Transplantation, Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
| | | | | | | | | | - Giulia Benevolo
- SC Hematology, A.O. Città della Salute e della Scienza, Turin, Italy
| | | | - Michele Merli
- Hematology, Department of Medicine and Surgery, University of Insubria & Ospedale di Circolo, ASST Sette Laghi, Varese, Italy
| | - Daniela Pietra
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Università di Pavia, Pavia, Italy
| | - Tiziano Barbui
- Research Foundation, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Giada Rotunno
- CRIMM-Centro Ricerca e Innovazione delle Malattie Mieloproliferative, Department of Experimental and Clinical Medicine, Azienda ospedaliera-Universitaria Careggi, University of Florence, Florence, Italy
| | - Mario Cazzola
- Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Università di Pavia, Pavia, Italy
| | - Toni Giorgino
- Biophysics Institute, National Research Council of Italy, c/o Department of Biosciences, University of Milan, via Celoria 26, I-20133, Milan, Italy
| | - Alessandro Maria Vannucchi
- CRIMM-Centro Ricerca e Innovazione delle Malattie Mieloproliferative, Department of Experimental and Clinical Medicine, Azienda ospedaliera-Universitaria Careggi, University of Florence, Florence, Italy
| | - Francesco Passamonti
- Hematology, Department of Medicine and Surgery, University of Insubria & Ospedale di Circolo, ASST Sette Laghi, Varese, Italy.
| |
Collapse
|
35
|
Horvat I, Boban A, Zadro R, Antolic MR, Serventi-Seiwerth R, Roncevic P, Radman I, Sertic D, Vodanovic M, Pulanic D, Basic-Kinda S, Durakovic N, Zupancic-Salek S, Vrhovac R, Aurer I, Nemet D, Labar B. Influence of Blood Count, Cardiovascular Risks, Inherited Thrombophilia, and JAK2 V617F Burden Allele on Type of Thrombosis in Patients With Philadelphia Chromosome Negative Myeloproliferative Neoplasms. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2018; 19:53-63. [PMID: 30301673 DOI: 10.1016/j.clml.2018.08.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/31/2018] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Thrombosis is the most common complication in Philadelphia chromosome negative (Ph-) myeloproliferative neoplasms patients. PATIENTS AND METHODS In a cohort of 258 Ph- myeloproliferative neoplasm patients, the difference between patients with and without thrombosis was analyzed according to genetic thrombophilia factors, JAK2 V617F status and burden allele, blood count, cardiovascular risk factors and age. Patients were also divided in polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) subgroups as well as by the type of thrombosis. RESULTS Analysis of cardiovascular risk factors regarding arterial thrombosis showed that PV patients with thrombosis had higher incidence of diabetes (P = .030), ET patients more often had hypertension (P = .003) and hyperlipidemia (P = .005), while PMF patients had hyperlipidemia (P = .046) and at least one cardiovascular risk factor (P = .044). Moreover, leukocytes > 18 × 109/L and V617F burden allele > 25.7% were statistically significantly different in PV patients (P = .019 and borderline significant at P = .055, respectively), while in ET patients leukocytes > 9.2 × 109/L (P < .001) and age at diagnosis of > 55 years were statistically significantly different (P = .002). PMF patients with V617F burden allele ≤ 34.8% were more prone to thrombosis (P = .032). When comparing patients with and without venous thrombosis, cutoff value of V617F burden allele > 90.4% was significant for PV patients with thrombosis (P = .036), as was > 56.7% for PMF patients with thrombosis (P = .046). Platelets ≤ 536 × 109/L and age at diagnosis > 54 years showed statistically significant difference for ET patients with thrombosis (P = .015 and P = .041, respectively). CONCLUSION On the basis of our results, a new scoring system for thrombosis risk in PV could be made, while PMF prognostic model may be expanded for better recognition of potential thrombotic risk factors.
Collapse
Affiliation(s)
- Ivana Horvat
- Department of Laboratory Diagnostics, University Hospital Centre Zagreb, Zagreb, Croatia.
| | - Ana Boban
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Renata Zadro
- Department of Laboratory Diagnostics, University Hospital Centre Zagreb, Zagreb, Croatia; Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | | | | | - Pavle Roncevic
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Ivo Radman
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Dubravka Sertic
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Marijo Vodanovic
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Drazen Pulanic
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Sandra Basic-Kinda
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Nadira Durakovic
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Silva Zupancic-Salek
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia; School of Medicine, University of Osijek, Osijek, Croatia
| | - Radovan Vrhovac
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Igor Aurer
- Department of Hematology, University Hospital Centre Zagreb, Zagreb, Croatia; School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Damir Nemet
- School of Medicine, University of Zagreb, Zagreb, Croatia; International University Libertas, Zagreb, Croatia
| | - Boris Labar
- School of Medicine, University of Zagreb, Zagreb, Croatia; Center for Medical Experts, Zagreb, Croatia
| |
Collapse
|
36
|
Mesa R, Boccia RV, Grunwald MR, Oh ST, Colucci P, Paranagama D, Parasuraman S, Stein BL. Patient-Reported Outcomes Data From REVEAL at the Time of Enrollment (Baseline): A Prospective Observational Study of Patients With Polycythemia Vera in the United States. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2018; 18:590-596. [PMID: 30122202 PMCID: PMC8148983 DOI: 10.1016/j.clml.2018.05.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/21/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND Patients with polycythemia vera (PV) often experience symptoms that adversely affect their quality of life (QoL). The ongoing, prospective, observational REVEAL (Prospective Observational Study of Patients With Polycythemia Vera in US Clinical Practices) study was designed to collect contemporary data regarding burden of disease, clinical management, patient-reported outcomes (PROs), and health care resource utilization from adult patients with PV in the United States. PATIENTS AND METHODS Data on PROs were collected at enrollment using the Myeloproliferative Neoplasm Symptom Assessment Form Total Symptom Score (MPN-SAF TSS; range, 0-100); the European Organization for Research and Treatment of Cancer-Core Quality of Life Questionnaire, version 3.0 (EORTC QLQ-C30; range, 0-100); and the Work Productivity and Activity Impairment Questionnaire-Specific Health Problem (WPAI-SHP; range, 0%-100%). RESULTS Among 2309 patients, mean (SD) disease duration was 5.8 (6.1) years and Charlson Comorbidity Index was 3.4 (0.8); 54.0% (1247/2309) were male. Mean (SD) MPN-SAF TSS was 18.8 (15.5). The most common symptoms were fatigue (80.1% [1844/2302]), early satiety (60.9% [1402/2302]), and inactivity (57.6% [1324/2302]). The most common severe symptoms were fatigue (16.8% [387/2302]), itching (13.4% [308/2302]), and inactivity (11.8% [271/2302]). The mean (SD) EORTC QLQ-C30 global health status/QoL score was 73.1 (23.2): mean functional subscale scores ranged from 80.5 (23.9) for cognitive functioning to 85.7 (24.6) for social functioning. The mean WPAI-SHP activity impairment score was 19.7% (n = 2300). Employed patients had mean WPAI-SHP scores for absenteeism, presenteeism, and overall work impairment of 3.2% (n = 810), 12.1% (n = 807), and 13.4% (n = 802), respectively. CONCLUSION These data confirm that many patients with PV experience symptoms, QoL impairments, and work productivity impairments that negatively affect their lives. Longitudinal data from REVEAL will be important for evaluating how PROs change over time in these patients.
Collapse
Affiliation(s)
- Ruben Mesa
- Mays Cancer Center, UT Health San Antonio Cancer Center - An NCI Designated Cancer Center, San Antonio, TX.
| | | | | | - Stephen T Oh
- Divisions of Hematology & Oncology, Washington University School of Medicine, St Louis, MO
| | | | | | | | - Brady L Stein
- Department of Medicine (Hematology and Oncology), Northwestern University Feinberg School of Medicine, Chicago, IL
| |
Collapse
|
37
|
|
38
|
Patterson-Fortin J, Moliterno AR. Molecular Pathogenesis of Myeloproliferative Neoplasms: Influence of Age and Gender. Curr Hematol Malig Rep 2018; 12:424-431. [PMID: 28948454 DOI: 10.1007/s11899-017-0411-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The myeloproliferative neoplasms polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) display distinct clinical and pathologic features but are characterized by mutations in JAK2, MPL, and CALR leading to activation of the JAK-STAT pathway. This review addresses the pathogenesis and mechanisms of these mutant alleles and the unique interactions of both of age and gender.
Collapse
Affiliation(s)
- Jeffrey Patterson-Fortin
- Department of Medicine, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Nelson-Harvey 808, Baltimore, MD, 21287, USA
| | - Alison R Moliterno
- Division of Hematology, Department of Medicine, The Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building, #1025, Baltimore, MD, 21205, USA.
| |
Collapse
|
39
|
How J, Trinkaus KM, Oh ST. Distinct clinical, laboratory and molecular features of myeloproliferative neoplasm patients with splanchnic vein thrombosis. Br J Haematol 2017; 183:310-313. [PMID: 29048104 DOI: 10.1111/bjh.14958] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Joan How
- Division of Hematology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kathryn M Trinkaus
- Biostatistics Shared Resource, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Stephen T Oh
- Division of Hematology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
40
|
Zhan H, Lin CHS, Segal Y, Kaushansky K. The JAK2V617F-bearing vascular niche promotes clonal expansion in myeloproliferative neoplasms. Leukemia 2017; 32:462-469. [PMID: 28744010 PMCID: PMC5783797 DOI: 10.1038/leu.2017.233] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 06/15/2017] [Accepted: 07/10/2017] [Indexed: 12/17/2022]
Abstract
The acquired kinase mutation JAK2V617F plays a central role in myeloproliferative neoplasms (MPNs). However, the mechanisms responsible for the malignant hematopoietic stem/progenitor cell (HSPC) expansion seen in patients with MPNs are not fully understood, limiting the effectiveness of current treatment. Endothelial cells (ECs) are an essential component of the hematopoietic niche, and they have been shown to express the JAK2V617F mutation in patients with MPNs. We show that the JAK2V617F-bearing vascular niche promotes the expansion of the JAK2V617F HSPCs in preference to JAK2WT HSPCs, potentially contributing to poor donor cell engraftment and disease relapse following stem cell transplantation. The expression of Chemokine (C-X-C motif) ligand 12 (CXCL12) and stem cell factor (SCF) were upregulated in JAK2V617F-bearing ECs compared to wild-type ECs, potentially accounting for this observation. We further identify that the thrombopoietin (TPO)/MPL signaling pathway is critical for the altered vascular niche function. A better understanding of how the vascular niche contributes to HSPC expansion and MPN development is essential for the design of more effective therapeutic strategies for patients with MPNs.
Collapse
Affiliation(s)
- H Zhan
- Northport VA Medical Center, Northport, NY, USA.,Department of Medicine, Stony Brook Medicine, Stony Brook, NY, USA
| | - C H S Lin
- Department of Medicine, Stony Brook Medicine, Stony Brook, NY, USA
| | - Y Segal
- Northport VA Medical Center, Northport, NY, USA
| | - K Kaushansky
- Department of Medicine, Stony Brook Medicine, Stony Brook, NY, USA.,Office of the Sr. Vice President, Health Sciences, Stony Brook Medicine, Stony Brook, NY, USA
| |
Collapse
|
41
|
Nangalia J, Grinfeld J, Green AR. Pathogenesis of Myeloproliferative Disorders. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 11:101-26. [PMID: 27193452 DOI: 10.1146/annurev-pathol-012615-044454] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Myeloproliferative neoplasms (MPNs) are a set of chronic hematopoietic neoplasms with overlapping clinical and molecular features. Recent years have witnessed considerable advances in our understanding of their pathogenetic basis. Due to their protracted clinical course, the evolution to advanced hematological malignancies, and the accessibility of neoplastic tissue, the study of MPNs has provided a window into the earliest stages of tumorigenesis. With the discovery of mutations in CALR, the majority of MPN patients now bear an identifiable marker of clonal disease; however, the mechanism by which mutated CALR perturbs megakaryopoiesis is currently unresolved. We are beginning to understand better the role of JAK2(V617F) homozygosity, the function of comutations in epigenetic regulators and spliceosome components, and how these mutations cooperate with JAK2(V617F) to modulate MPN phenotype.
Collapse
Affiliation(s)
- Jyoti Nangalia
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge CB2 0XY, United Kingdom; .,Department of Haematology, Addenbrooke's Hospital, Cambridge CB2 2QR, United Kingdom
| | - Jacob Grinfeld
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge CB2 0XY, United Kingdom; .,Department of Haematology, Addenbrooke's Hospital, Cambridge CB2 2QR, United Kingdom
| | - Anthony R Green
- Department of Haematology, Cambridge Institute for Medical Research and Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge CB2 0XY, United Kingdom; .,Department of Haematology, Addenbrooke's Hospital, Cambridge CB2 2QR, United Kingdom
| |
Collapse
|
42
|
Affiliation(s)
- Jerry L Spivak
- From the Hematology Division, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore
| |
Collapse
|
43
|
Grinfeld J, Godfrey AL. After 10 years of JAK2V617F: Disease biology and current management strategies in polycythaemia vera. Blood Rev 2017; 31:101-118. [DOI: 10.1016/j.blre.2016.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 11/08/2016] [Accepted: 11/14/2016] [Indexed: 12/12/2022]
|
44
|
How J, Zhou A, Oh ST. Splanchnic vein thrombosis in myeloproliferative neoplasms: pathophysiology and molecular mechanisms of disease. Ther Adv Hematol 2017; 8:107-118. [PMID: 28246554 PMCID: PMC5305004 DOI: 10.1177/2040620716680333] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are the most common underlying prothrombotic disorder found in patients with splanchnic vein thrombosis (SVT). Clinical risk factors for MPN-associated SVTs include younger age, female sex, concomitant hypercoagulable disorders, and the JAK2 V617F mutation. These risk factors are distinct from those associated with arterial or deep venous thrombosis (DVT) in MPN patients, suggesting disparate disease mechanisms. The pathophysiology of SVT is thought to derive from local interactions between activated blood cells and the unique splanchnic endothelial environment. Other mutations commonly found in MPNs, including CALR and MPL, are rare in MPN-associated SVT. The purpose of this article is to review the clinical and molecular risk factors for MPN-associated SVT, with particular focus on the possible mechanisms of SVT formation in MPN patients.
Collapse
Affiliation(s)
- Joan How
- Division of Hematology, Washington University School of Medicine, St Louis, MO, USA
| | - Amy Zhou
- Division of Hematology, Washington University School of Medicine, St Louis, MO, USA
| | - Stephen T. Oh
- Division of Hematology, Washington University School of Medicine, 660 South Euclid Avenue, Campus Box 8125, St Louis, MO 63110, USA
| |
Collapse
|
45
|
Abstract
Myeloproliferative neoplasms (MPNs) are a group of related clonal hematologic disorders characterized by excess accumulation of one or more myeloid cell lineages and a tendency to transform to acute myeloid leukemia. Deregulated JAK2 signaling has emerged as the central phenotypic driver of BCR -ABL1-negative MPNs and a unifying therapeutic target. In addition, MPNs show unexpected layers of genetic complexity, with multiple abnormalities associated with disease progression, interactions between inherited factors and phenotype driver mutations, and effects related to the order in which mutations are acquired. Although morphology and clinical laboratory analysis continue to play an important role in defining these conditions, genomic analysis is providing a platform for better disease definition, more accurate diagnosis, direction of therapy, and refined prognostication. There is an emerging consensus with regard to many prognostic factors, but there is a clear need to synthesize genomic findings into robust, clinically actionable and widely accepted scoring systems as well as the need to standardize the laboratory methodologies that are used.
Collapse
Affiliation(s)
- Katerina Zoi
- Katerina Zoi, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Nicholas C.P. Cross, Salisbury District Hospital, Salisbury; and University of Southampton, Southampton, United Kingdom
| | - Nicholas C P Cross
- Katerina Zoi, Biomedical Research Foundation of the Academy of Athens, Athens, Greece; Nicholas C.P. Cross, Salisbury District Hospital, Salisbury; and University of Southampton, Southampton, United Kingdom
| |
Collapse
|
46
|
Geyer HL, Kosiorek H, Dueck AC, Scherber R, Slot S, Zweegman S, Te Boekhorst PA, Senyak Z, Schouten HC, Sackmann F, Fuentes AK, Hernández-Maraver D, Pahl HL, Griesshammer M, Stegelmann F, Döhner K, Lehmann T, Bonatz K, Reiter A, Boyer F, Etienne G, Ianotto JC, Ranta D, Roy L, Cahn JY, Harrison CN, Radia D, Muxi P, Maldonado N, Besses C, Cervantes F, Johansson PL, Barbui T, Barosi G, Vannucchi AM, Paoli C, Passamonti F, Andreasson B, Ferrari ML, Rambaldi A, Samuelsson J, Cannon K, Birgegard G, Xiao Z, Xu Z, Zhang Y, Sun X, Xu J, Kiladjian JJ, Zhang P, Gale RP, Mesa RA. Associations between gender, disease features and symptom burden in patients with myeloproliferative neoplasms: an analysis by the MPN QOL International Working Group. Haematologica 2016; 102:85-93. [PMID: 27540137 DOI: 10.3324/haematol.2016.149559] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 08/12/2016] [Indexed: 12/15/2022] Open
Abstract
The myeloproliferative neoplasms, including polycythemia vera, essential thrombocythemia and myelofibrosis, are distinguished by their debilitating symptom profiles, life-threatening complications and profound impact on quality of life. The role gender plays in the symptomatology of myeloproliferative neoplasms remains under-investigated. In this study we evaluated how gender relates to patients' characteristics, disease complications and overall symptom expression. A total of 2,006 patients (polycythemia vera=711, essential thrombocythemia=830, myelofibrosis=460, unknown=5) were prospectively evaluated, with patients completing the Myeloproliferative Neoplasm-Symptom Assessment Form and Brief Fatigue Inventory Patient Reported Outcome tools. Information on the individual patients' characteristics, disease complications and laboratory data was collected. Consistent with known literature, most female patients were more likely to have essential thrombocythemia (48.6% versus 33.0%; P<0.001) and most male patients were more likely to have polycythemia vera (41.8% versus 30.3%; P<0.001). The rate of thrombocytopenia was higher among males than females (13.9% versus 8.2%; P<0.001) and males also had greater red-blood cell transfusion requirements (7.3% versus 4.9%; P=0.02) with shorter mean disease duration (6.4 versus 7.2 years, P=0.03). Despite there being no statistical differences in risk scores, receipt of most therapies or prior complications (hemorrhage, thrombosis), females had more severe and more frequent symptoms for most individual symptoms, along with overall total symptom score (22.8 versus 20.3; P<0.001). Females had particularly high scores for abdominal-related symptoms (abdominal pain/discomfort) and microvascular symptoms (headache, fatigue, insomnia, concentration difficulties, dizziness; all P<0.01). Despite complaining of more severe symptom burden, females had similar quality of life scores to those of males. The results of this study suggest that gender contributes to the heterogeneity of myeloproliferative neoplasms by influencing phenotypic profiles and symptom expression.
Collapse
Affiliation(s)
- Holly L Geyer
- Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Heidi Kosiorek
- Section of Biostatistics, Mayo Clinic, Scottsdale, AZ, USA
| | - Amylou C Dueck
- Section of Biostatistics, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Stefanie Slot
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | - Sonja Zweegman
- Department of Hematology, VU University Medical Center, Amsterdam, the Netherlands
| | | | | | | | | | | | | | - Heike L Pahl
- Department of Molecular Hematology, University Hospital Freiburg, Germany
| | | | - Frank Stegelmann
- Department of Internal Medicine III, University Hospital of Ulm, Germany
| | - Konstanze Döhner
- Department of Internal Medicine III, University Hospital of Ulm, Germany
| | - Thomas Lehmann
- Hematology Department, University Hospital, Basel, Switzerland
| | - Karin Bonatz
- Medizinische Klinik, Universitätsmedizin, Mannheim, Germany
| | - Andreas Reiter
- Medizinische Klinik, Universitätsmedizin, Mannheim, Germany
| | | | | | | | - Dana Ranta
- Hospitalier Universitaire, Nancy, France
| | - Lydia Roy
- Centre Hospitalier Universitaire, Poitiers, France
| | | | - Claire N Harrison
- Department of Haematology, Guy's and St. Thomas NHS Foundation Trust, London, UK
| | - Deepti Radia
- Department of Haematology, Guy's and St. Thomas NHS Foundation Trust, London, UK
| | - Pablo Muxi
- Unidadde Hematología, Hospital Británico, Montevideo, Uruguay
| | - Norman Maldonado
- University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Carlos Besses
- Hematology Department, Hospital del Mar, Barcelona, Spain
| | - Francisco Cervantes
- Hematology Department, Hospital Clínic, IDIBAPS, University of Barcelona, Spain
| | | | - Tiziano Barbui
- Research Foundation (FROM), Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Giovanni Barosi
- Laboratory of Clinical Epidemiology, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Alessandro M Vannucchi
- Center for Research and Innovation of Myeloproliferative Neoplasms, University of Florence, Italy
| | - Chiara Paoli
- Center for Research and Innovation of Myeloproliferative Neoplasms, University of Florence, Italy
| | - Francesco Passamonti
- Ematologia, Dipartimento di Medicina Clinica e Sperimentale, University of Insubria, Varese, Italy
| | | | | | | | - Jan Samuelsson
- Department of Internal Medicine, Stockholm South Hospital, Sweden
| | - Keith Cannon
- Department of Internal Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Zhijian Xiao
- MDS and MPN Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Zefeng Xu
- MDS and MPN Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yue Zhang
- MDS and MPN Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiujuan Sun
- MDS and MPN Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Junqing Xu
- MDS and MPN Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | | | - Peihong Zhang
- Department of Pathology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | | | - Ruben A Mesa
- Department of Hematology and Oncology, Mayo Clinic, Scottsdale, AZ, USA
| |
Collapse
|
47
|
Ferdowsi S, Ghaffari SH, Amirizadeh N, Azarkeivan A, Atarodi K, Faranoush M, Toogeh G, Shirkoohi R, Vaezi M, Maghsoodlu M, Alimoghaddam K, Ghavamzadeh A, Teimori Naghadeh H. JAK2V617F Allele Burden Measurement in Peripheral Blood of Iranian Patients with Myeloproliferative Neoplasms and Effect of Hydroxyurea on JAK2V617F Allele Burden. Int J Hematol Oncol Stem Cell Res 2016; 10:70-8. [PMID: 27252806 PMCID: PMC4888151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2022] Open
Abstract
BACKGROUND Myeloproliferative neoplasms (MPNs) are clonal malignant diseases that represent a group of conditions including polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF). The aim of this study was to evaluate possible correlations between JAK2V617F allele burden and clinicohematologic characteristics in Iranian patients with MPNs. We also aimed at determining the correlation between JAK2V617F allele burden and use of cyto reductive treatment (hydroxyurea). MATERIALS AND METHODS We performed ARMS-PCR for all MPNs samples and subsequently performed real-time quantitative polymerase chain reaction (qRT-PCR) for JAK2V617F allele burden measurement using DNA from peripheral blood leukocytes. RESULTS Two distinct groups of patients were examined at a single time point: group A (n=40; 20 PV, 20 ET) was examined at the time of diagnosis; group B (n=85; 40 PV, 30 ET and 15 PMF) while under treatment with hydroxyurea (HU). The median allele burden of the JAK2 V617F was 72% for PV and 49% for ET patients at the time of diagnosis (p=0.01). For patients with HU treatment, we determined the median JAK2V617F allele burden to be 43%, 40%, and 46.5 % in PV, ET and PMF patients; respectively. HU-treated PV patients had a significant lower %JAK2V617F than PV patients at the time of diagnosis (43% vs. 72%, p=0.005). In ET group, the relationship between the JAK2 V617F allele burden and leukocyte count was significant (p=0.02 and p=0.01 in untreated and treated patients, respectively). CONCLUSIONS Our results showed that patients with PV have a higher JAK2V617F allele burden. Moreover, our study demonstrated that the JAK2V617F allele burden correlates with clinical features in ET group. We also showed hydroxyurea can affect the JAK2V617F allele burden in PV patients.
Collapse
Affiliation(s)
- Shirin Ferdowsi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Seyed H. Ghaffari
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Naser Amirizadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Azita Azarkeivan
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Kamran Atarodi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mohammad Faranoush
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Gholamreza Toogeh
- Hematology-Oncology and BMT Research Center, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Shirkoohi
- Molecular Genetics, Cancer Research Center, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Vaezi
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahtab Maghsoodlu
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Kamran Alimoghaddam
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ardeshir Ghavamzadeh
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hosein Teimori Naghadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| |
Collapse
|
48
|
Pemmaraju N, Moliterno AR. From Philadelphia-Negative to JAK2-Positive: Effect of Genetic Discovery on Risk Stratification and Management. Am Soc Clin Oncol Educ Book 2016:139-45. [PMID: 25993152 DOI: 10.14694/edbook_am.2015.35.139] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The 2005 discovery of the JAK2 mutation redefined the diagnosis and natural history of myeloproliferative neoplasms (MPNs). Most importantly, this improvement in the pathobiologic conceptualization has focused our evolution of this field from being defined as what it is not (e.g., Philadelphia [Ph]-negative) to what it is (e.g., JAK2-positive, CALR-positive) in the majority of MPN cases. In the ensuing 10 years, the field has experienced a paradigm shift in terms of understanding of the biologic basis of the development of MPNs, an explosion of knowledge of the genetics of MPNs, and has translated disease knowledge into effective targeted therapies. With greater uniformity and agreement on the diagnosis and differences among the individual MPNs, augmented by improved cytogenetic and molecular classification, attention has turned now to addressing the need for uniformity in risk stratification of patients in the clinic for both disease complications and disease transformation. This article will highlight the developments in the field with regard to risk stratification and prognostication in MPNs with focus on the clinical aspects of the patient who presents with either essential thrombocytosis (ET), polycythemia vera (PV), or myelofibrosis (MF).
Collapse
Affiliation(s)
- Naveen Pemmaraju
- From the Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD; the Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Alison R Moliterno
- From the Department of Medicine, The Johns Hopkins Hospital, Baltimore, MD; the Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
49
|
Zhan H, Ciano K, Dong K, Zucker S. Targeting glutamine metabolism in myeloproliferative neoplasms. Blood Cells Mol Dis 2015; 55:241-7. [PMID: 26227854 DOI: 10.1016/j.bcmd.2015.07.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 07/13/2015] [Accepted: 07/13/2015] [Indexed: 12/29/2022]
Abstract
JAK2(V617F) mutation can be detected in the majority of myeloproliferative neoplasm (MPN) patients. The JAK2 inhibitor Ruxolitinib is the first FDA-approved treatment for MPNs. However, its use is limited by various dose related toxicities. Here, we studied the metabolic state and glutamine metabolism of BaF3-hEPOR-JAK2V617F and BaF3-hEPOR-JAK2WT cells. We found that the JAK2(V617F)-mutant cells were associated with increased oxygen consumption rate and extracellular acidification rate than the JAK2(WT) cells and there was an increased glutamine metabolism in JAK2(V617F)-mutant cells compared to wild-type cells. Glutaminase (GLS), the key enzyme in glutamine metabolism, was upregulated in the JAK2(V617F)-mutant BaF3 cells compared to the JAK2(WT) BaF3 cells. In MPN patient peripheral blood CD34+ cells, GLS expression was increased in JAK2(V617F)-mutant progenitor cells compared to JAK2 wild-type progenitor cells from the same patients and GLS levels were increased at the time of disease progression compared to at earlier time points. Moreover, GLS inhibitor increased the growth inhibitory effect of Ruxolitinib in both JAK2(V617F)-mutant cell lines and peripheral blood CD34+ cells from MPN patients. Therefore, GLS inhibitor should be further explored to enhance the therapeutic effectiveness of JAK2 inhibitor and allow the administration of lower doses of the drug to avoid its toxicity.
Collapse
Affiliation(s)
- Huichun Zhan
- Northport VA Medical Center, Northport, NY, USA; Department of Medicine, Stony Brook University, Stony Brook, NY, USA.
| | | | - Katherine Dong
- Rice University and Baylor College of Medicine, Houston, TX, USA
| | - Stanley Zucker
- Northport VA Medical Center, Northport, NY, USA; Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
50
|
Stein BL, Platanias LC. Calreticulin gene mutations in the myeloproliferative neoplasms: Dameshek's other “myelostimulatory” factor. Leuk Lymphoma 2015; 56:1573-4. [DOI: 10.3109/10428194.2014.981178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|