1
|
Severino P, D'Amato A, Prosperi S, Myftari V, Germanò R, Marek-Iannucci S, De Prisco A, Mariani MV, Marchiori L, Battaglia C, Tabacco L, Segato C, Mancone M, Fedele F, Vizza CD. Coronary microcirculation in myocardial ischemia: A genetic perspective. J Mol Cell Cardiol 2025; 203:67-75. [PMID: 40220989 DOI: 10.1016/j.yjmcc.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Coronary microvascular dysfunction (CMD) is a major contributor to ischemic heart disease (IHD), acting both independently and together with atherosclerosis. CMD encompasses structural and functional microcirculatory changes that result in dysregulated coronary blood flow. Structural abnormalities include microvascular remodeling, resulting in arteriolar and capillary narrowing, perivascular fibrosis and capillary rarefaction. Endothelial dysfunction and smooth muscle cell hyperactivity further impair microcirculation. Genetic factors may play a crucial role in the pathophysiology of CMD, mainly due to single nucleotide polymorphisms (SNPs) in genes that regulate coronary blood flow and microcirculation structural modifications. This manuscript aims to review the genetic determinants of CMD, with particular focus on ion channels, microRNAs (miRNAs), and proteins involved in the endothelial environment. The improving knowledge about genetic aspects of CMD opens the possibility to have new biomarkers, improving diagnosis and the development of targeted treatments in light of an even more patient-tailored approach.
Collapse
Affiliation(s)
- Paolo Severino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea D'Amato
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Silvia Prosperi
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Vincenzo Myftari
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Rosanna Germanò
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Stefanie Marek-Iannucci
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea De Prisco
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Marco Valerio Mariani
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Ludovica Marchiori
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Corinne Battaglia
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Leonardo Tabacco
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Camilla Segato
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Massimo Mancone
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | | | - Carmine Dario Vizza
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
2
|
De Moner B, Martinez-Sanchez J, Garrote M, Ramos A, Ventosa-Capell H, Moreno-Castaño A, Nomdedeu M, Ojeda A, Escolar G, Garcia-Pagan JC, Arellano-Rodrigo E, Carreras E, Alvarez-Larran A, Díaz-Ricart M. Endothelial Damage in JAK2V617F Myeloproliferative Neoplasms with Splanchnic Vein Thrombosis. Thromb Haemost 2025. [PMID: 39809451 DOI: 10.1055/a-2498-4849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
BACKGROUND JAK2V617F-mutated myeloproliferative neoplasms (MPN) exhibit abnormal proliferation of bone marrow progenitors and increased risk of thrombosis, specifically in splanchnic veins (SVT). The contribution of the endothelium to the development of the prothrombotic phenotype was explored. MATERIAL AND METHODS Plasma and serum samples from JAK2V617F MPN patients with (n=26) or without (n=7) thrombotic debut and different treatments, were obtained (n=33). Cultured endothelial cells (ECs) were exposed to serum samples from these patients and from healthy donors as controls. Changes in markers of inflammation (VCAM-1, ICAM-1), cell permeability (VE-cadherin), production of VWF, extracellular matrix (ECM) reactivity, and activation of intracellular signaling pathways related to stress, proliferation, inflammation (Akt, p44/42, IkBa), and JAK2/STAT3 pathway, were assessed by immunofluorescence, flow adhesion, SDS-PAGE and immunoblot. Additionally, circulating markers of endothelial activation and damage (VWF, sVCAM-1, sTNFRI, thrombomodulin, angiopoietin-2, a2-antiplasmin activity, PAI-1) were evaluated in Patients' plasma. RESULTS The in vitro studies showed that EC exposure to MPN thrombotic patients' sera resulted in increased VCAM-1 and ICAM-1, and reduced VE-cadherin expression (p<0.05) at the cell surface. Production and release of VWF to the ECM were higher (p<0.05), with increased platelet adhesion after perfusing whole blood, being more noticeable in response to sera from non-treated patients. Furthermore, intracellular activation of Akt, p44/42, IkBa and JAK2/STAT3 was observed. Moreover, plasma levels of VWF, TNF-R1, VCAM-1, thrombomodulin, and angiopoietin-2 were higher in JAK2V617F+ MPN patients with thrombosis. CONCLUSION The present findings suggest that circulating factors in MPNs with SVT debut induce endothelial proinflammatory and prothrombotic phenotypes, which are modulated in vitro with MPN treatment.
Collapse
Affiliation(s)
- Blanca De Moner
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Josep Carreras Leukaemia Research Institute, University of Barcelona, Hospital Clínic de Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Julia Martinez-Sanchez
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Marta Garrote
- Hematopathology Section, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, IDIBAPS, Spain
| | - Alex Ramos
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | | | - Ana Moreno-Castaño
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Meritxell Nomdedeu
- Hemostasis and Hemotherapy Department, Institut del Càncer i Malalties de la Sang (ICAMS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Asunción Ojeda
- Liver Unit, Barcelona Hepatic Haemodynamic Laboratory, Hospital Clínic de Barcelona, IDIBAPS and CIBERehd, Barcelona, Spain
| | - Gines Escolar
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| | - Joan Carles Garcia-Pagan
- Liver Unit, Barcelona Hepatic Haemodynamic Laboratory, Hospital Clínic de Barcelona, IDIBAPS and CIBERehd, Barcelona, Spain
| | - Eduardo Arellano-Rodrigo
- Hemostasis and Hemotherapy Department, Institut del Càncer i Malalties de la Sang (ICAMS), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Enric Carreras
- Barcelona Endothelium Team, Barcelona, Spain
- Fundació Carreras contra la Leucèmia, Barcelona, Spain
| | - Alberto Alvarez-Larran
- Hematology Department, Institut del Càncer i Malalties de la Sang (ICAMS), Hospital Clínic de Barcelona, IDIBAPS, Barcelona, Spain
| | - Maribel Díaz-Ricart
- Hemostasis and Erythropathology Laboratory, Hematopathology, Pathology Department, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
- Barcelona Endothelium Team, Barcelona, Spain
| |
Collapse
|
3
|
Kristiansen MH, Larsen MK, Massarenti L, Skov V, Kjær L, Enevold C, Ostrowski SR, Nielsen CH, Hasselbalch HC, Wienecke T. Thromboinflammation in ischemic cerebrovascular patients with the JAK2V617F mutation. Thromb Res 2025; 245:109236. [PMID: 39652998 DOI: 10.1016/j.thromres.2024.109236] [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: 07/09/2024] [Revised: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND The JAK2V617F mutation is a driver of Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) and is also implicated in cardiovascular diseases. Thrombosis in MPN involves JAK2V617F-associated platelet activation and endothelial dysfunction, all potentially influenced by chronic inflammation. Whether the mutation affects thromboinflammatory markers similarly in non-MPN patients remains unclear. METHOD We conducted a study involving 63 ischemic cerebrovascular patients with the JAK2V617F mutation, matched with 63 patients without the mutation. Serum samples were analyzed for 12 thromboinflammatory markers during the acute phase and at three months follow-up. RESULTS Overall, there was no significant difference in thromboinflammatory markers between cases and controls. However, subgroup analysis of patients with a JAK2V617F allele burden ≥1 % (n = 15) showed higher levels of Vascular Cell Adhesion Molecule-1 (VCAM-1) at baseline (p = 0.018), and elevated Interleukin-10 (IL-10) (p = 0.004) and Tumor Necrosis Factor α (TNF-α) (p = 0.018) at follow-up compared to controls. Regression analysis revealed an association between higher JAK2V617F allele burden and increased VCAM-1 at baseline (p < 0.001), and higher VCAM-1 (p = 0.012), IL-10 (p = 0.003), and TNF-α (p = 0.034) at follow-up. CONCLUSION In ischemic cerebrovascular patients, the JAK2V617F mutation is associated with elevated markers of endothelial dysfunction and chronic inflammation. This underscores the role of inflammation in thrombosis driven by the JAK2V617F mutation.
Collapse
Affiliation(s)
- Marie Hvelplund Kristiansen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Neurology, Zealand University Hospital, Roskilde, Denmark.
| | - Morten Kranker Larsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Laura Massarenti
- Section for Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Christian Enevold
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Claus Henrik Nielsen
- Section for Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hans Carl Hasselbalch
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Troels Wienecke
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Neurology, Zealand University Hospital, Roskilde, Denmark
| |
Collapse
|
4
|
La Rosa F, Montecucco F, Liberale L, Sessarego M, Carbone F. Venous thrombosis and obesity: from clinical needs to therapeutic challenges. Intern Emerg Med 2025; 20:47-64. [PMID: 39269539 PMCID: PMC11794390 DOI: 10.1007/s11739-024-03765-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024]
Abstract
Weight bias and stigma have limited the awareness of the systemic consequences related to obesity. As the narrative evolves, obesity is emerging as a driver and enhancer of many pathological conditions. Among these, the risk of venous thromboembolism (VTE) is a critical concern linked to obesity, ranking as the third most common cardiovascular condition. Obesity is recognized as a multifactorial risk factor for VTE, influenced by genetic, demographic, behavioral, and socio-economic conditions. Despite established links, the exact incidence of obesity related VTE in the general population remains largely unknown. The complexity of distinguishing between provoked and unprovoked VTE, coupled with gaps in obesity definition and assessment still complicates a tailored risk assessment of VTE risk. Obesity reactivity, hypercoagulability, and endothelial dysfunction are driven by the so-called 'adiposopathy'. This state of chronic inflammation and metabolic disturbance amplifies thrombin generation and alters endothelial function, promoting a pro-thrombotic environment. Additionally, the inflammation-induced clot formation-also referred to as 'immunothrombosis' further exacerbates VTE risk in people living with obesity. Furthermore, current evidence highlights significant gaps in the management of obesity related VTE, particularly concerning prophylaxis and treatment efficacy of anticoagulants in people living with obesity. This review underscores the need for tailored therapeutic approaches and well-designed clinical trials to address the unique challenges posed by obesity in VTE prevention and management. Advanced research and innovative strategies are imperative to improve outcomes and reduce the burden of VTE in people living with obesity.
Collapse
Affiliation(s)
- Federica La Rosa
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy.
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Marta Sessarego
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 Viale Benedetto XV, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa, Italy
| |
Collapse
|
5
|
Dawudi Y, Benarroch S, Helfer H, Smadja DM, Mahé I. Janus kinase inhibitor treatment for inflammatory diseases: excess or no excess risk of venous thromboembolism? Res Pract Thromb Haemost 2025; 9:102667. [PMID: 39980606 PMCID: PMC11840193 DOI: 10.1016/j.rpth.2024.102667] [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: 09/11/2024] [Revised: 12/04/2024] [Accepted: 12/24/2024] [Indexed: 02/22/2025] Open
Abstract
Janus kinase inhibitors (JAKis) have revolutionized the treatment landscape for various inflammatory and autoimmune diseases since their introduction in 2012. The expanded indications of JAKis have raised concerns about the associated risk of thrombosis, venous thromboembolic events (VTEs), and arterial thrombosis. This literature review examines studies reporting the risk of VTEs associated with JAKis in patients with inflammatory diseases. Phase I to III trials showed no increased risk of VTEs. However, these studies were not designed to detect adverse events such as VTEs. The pharmacovigilance data indicated that the frequency of VTE reports was higher than that of other adverse events. An increased risk of VTEs was also observed in the ORAL Surveillance study, a randomized, noninferiority, postmarketing phase IV safety study comparing tofacitinib with anti-tumor necrosis factor in patients with rheumatoid arthritis. However, limitations have to be acknowledged: pharmacovigilance data are declarative and subject to bias, VTE was a secondary outcome in the ORAL study, with noncomparable VTE risk factors between groups and increased thrombosis risks only at high doses of tofacitinib. Nevertheless, these data have led regulatory organizations such as the Food and Drug Administration and the European Medicines Agency to issue precautionary measures regarding the use of JAKis in inflammatory diseases. Most well-conducted real-life studies are in rheumatoid arthritis and do not confirm an excess of VTE risk associated with JAKis. Considering those conflicting results and limitations, future research should focus on specific indications and patient profiles, taking into account the complex interaction between drug treatment and underlying disease activity, to be able to draw definite conclusion about the VTE risk associated with JAKis.
Collapse
Affiliation(s)
- Yachar Dawudi
- Internal Medicine Department, Hôpital Louis-Mourier, Assistance Publique - Hôpitaux de Paris, Colombes, France
| | - Samuel Benarroch
- Internal Medicine Department, Hôpital Louis-Mourier, Assistance Publique - Hôpitaux de Paris, Colombes, France
| | - Hélène Helfer
- Internal Medicine Department, Hôpital Louis-Mourier, Assistance Publique - Hôpitaux de Paris, Colombes, France
| | - David M. Smadja
- Hematology Department, European Georges Pompidou Hospital, Assistance Publique - Hôpitaux de Paris, Paris, France
- Université Paris Cité, Paris, France
- INSERM Cardiovascular Research Center, Team « Endotheliopathy and Hemostasis Disorders », Paris, France
- Investigation Network On Venous Thrombo-Embolism (INNOVTE) - French Clinical Research Infrastructure Network, Saint-Etienne, France
| | - Isabelle Mahé
- Internal Medicine Department, Hôpital Louis-Mourier, Assistance Publique - Hôpitaux de Paris, Colombes, France
- Université Paris Cité, Paris, France
- INSERM Cardiovascular Research Center, Team « Endotheliopathy and Hemostasis Disorders », Paris, France
- Investigation Network On Venous Thrombo-Embolism (INNOVTE) - French Clinical Research Infrastructure Network, Saint-Etienne, France
| |
Collapse
|
6
|
Reda S, Chang J, Busse J, Schwarz N, McRae HL, Müller J, Strassburg CP, Oldenburg J, Pötzsch B, Jansen C, Rühl H. Assessment of Hypercoagulability in Splanchnic Vein Thrombosis by Measurement of the Hemostasis Enzymes Thrombin and Activated Protein C. Int J Mol Sci 2024; 26:292. [PMID: 39796151 PMCID: PMC11719462 DOI: 10.3390/ijms26010292] [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: 12/15/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Splanchnic vein thrombosis (SVT), which is particularly prevalent in myeloproliferative neoplasms (MPNs), has a multifactorial pathomechanism involving the anticoagulant protein C (PC) pathway. To better characterize the hypercoagulable state in SVT we assessed its key enzymes thrombin and activated PC (APC). The study population included 73 patients with SVT, thereof 36 MPN+, confirmed by bone marrow biopsy, 37 MPN-, and 30 healthy controls. Direct measurement of the active enzyme forms of thrombin and APC in the circulation was achieved by using oligonucleotide-based enzyme capture assays (OECA). Additionally, activation markers of coagulation and fibrinolysis were measured. Plasma levels of free thrombin and APC were higher in the MPN+ than in the MPN- cohort, with 0.49 vs. <0.46 pmol/L (p = 0.0057), respectively, 1.23 vs. 0.58 pmol/L (p = 0.0122), and in healthy controls (vs. <0.46 pmol/L, p = 0.0012; vs. 0.54 pmol/L, p = 0.0035). The indirect activation markers prothrombin fragment 1+2, thrombin-antithrombin complex, and D-dimer did not differ between groups. Receiver operating characteristic analysis suggested that SVT patients with MPN can be better distinguished by APC than by conventional indirect thrombin markers. A potential application of these biomarkers to guide anticoagulant therapy and to investigate the role of the PC pathway in MPN-associated hypercoagulability should be further studied.
Collapse
Affiliation(s)
- Sara Reda
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, 53127 Bonn, Germany; (S.R.); (J.B.); (N.S.); (J.O.)
- Department of Transfusion Medicine, University Hospital Cologne, 50937 Cologne, Germany
| | - Johannes Chang
- Department of Internal Medicine I, University Hospital Bonn, 53127 Bonn, Germany (C.P.S.); (C.J.)
| | - Johanna Busse
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, 53127 Bonn, Germany; (S.R.); (J.B.); (N.S.); (J.O.)
| | - Nadine Schwarz
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, 53127 Bonn, Germany; (S.R.); (J.B.); (N.S.); (J.O.)
| | - Hannah L. McRae
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, 53127 Bonn, Germany; (S.R.); (J.B.); (N.S.); (J.O.)
| | - Jens Müller
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, 53127 Bonn, Germany; (S.R.); (J.B.); (N.S.); (J.O.)
| | - Christian P. Strassburg
- Department of Internal Medicine I, University Hospital Bonn, 53127 Bonn, Germany (C.P.S.); (C.J.)
| | - Johannes Oldenburg
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, 53127 Bonn, Germany; (S.R.); (J.B.); (N.S.); (J.O.)
| | - Bernd Pötzsch
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, 53127 Bonn, Germany; (S.R.); (J.B.); (N.S.); (J.O.)
| | - Christian Jansen
- Department of Internal Medicine I, University Hospital Bonn, 53127 Bonn, Germany (C.P.S.); (C.J.)
| | - Heiko Rühl
- Institute of Experimental Hematology and Transfusion Medicine, University Hospital Bonn, 53127 Bonn, Germany; (S.R.); (J.B.); (N.S.); (J.O.)
| |
Collapse
|
7
|
Farooqi MF, Khan M, Muhammad AM, Agha A. Portal vein thrombosis in a patient on semaglutide. Qatar Med J 2024; 2024:75. [PMID: 39925824 PMCID: PMC11806721 DOI: 10.5339/qmj.2024.75] [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: 06/12/2024] [Accepted: 10/13/2024] [Indexed: 02/11/2025] Open
Abstract
Background: Obesity and type 2 diabetes mellitus (T2DM) are modern-day pandemics that have a significant impact on global healthcare. The glucagon-like peptide-1 receptor agonist (GLP1-RA) semaglutide is a novel treatment for both tbl2DM and obesity, but can be associated with an increased risk of venous thromboembolism. Case presentation: This case report describes a 59-year-old woman with tbl2DM who received semaglutide to manage glycemic levels, and also experienced the additional benefit of weight reduction. Within six months of initiating GLP1-RA, the patient experienced low back pain associated with nausea and poor oral intake. She had no known risk factors for venous thromboembolism or thrombophilia and had no history of significant illness in her family. Her physical examination revealed no significant findings. Only mild leukocytosis and neutrophilia were noted. She underwent an abdominal computed tomography scan, which revealed intrahepatic portal vein thrombosis without evidence of liver cirrhosis or abdominal malignancy. Her symptoms improved with oral anticoagulation (rivaroxaban). The result of thrombophilia examination was negative for inherited or acquired thrombophilia, except for a Janus kinase 2 mutation, which may increase the risk of thrombosis. Conclusions: The use of GLP1-RA is increasing due to the growing desire for weight loss medications. Therefore, it is important that physicians better understand the possible risks of thrombosis before initiating GLP1-RA treatment.
Collapse
Affiliation(s)
| | - Maria Khan
- Internal Medicine, Tawam Hospital, Al Ain, UAE
| | | | | |
Collapse
|
8
|
Partouche S, Goldberg I, Halperin E, Atamna B, Shacham-Abulafia A, Shapira S, Samara A, Gover-Proaktor A, Leader A, Spectre G, Raanani P, Granot G, Wolach O. Interferon-α Inhibits NET Formation in Neutrophils Derived from Patients with Myeloproliferative Neoplasms in a Neutrophil Sub-Population-Specific Manner. Int J Mol Sci 2024; 25:13473. [PMID: 39769242 PMCID: PMC11677445 DOI: 10.3390/ijms252413473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025] Open
Abstract
Neutrophils and neutrophil extracellular traps (NETs) contribute to thrombosis and hyperinflammation in myeloproliferative neoplasms (MPN). High-density neutrophils (HDNs) and low-density neutrophils (LDNs) have recently been characterized as distinct neutrophil sub-populations with distinct morphological and functional properties. We aim to study the kinetics of NET formation and inhibition with interferon-α (IFNα) in neutrophils derived from patients with MPN as compared to matched healthy controls. Ex vivo NET formation was assessed by neutrophil-elastase activity, neutrophil-associated nucleosomes, myeloperoxidase (MPO), and citrullinated histone H3 content. IFNα significantly inhibited NET formation in neutrophils derived from MPN patients. Neutrophil sub-population analysis demonstrated that HDNs drive the increase in NET formation as compared to LDNs in patients and in healthy controls and are effectively inhibited by IFNα, an effect that is lost in LDNs. In conclusion, we demonstrate that in MPN, HDNs drive excess NET formation and are more sensitive to IFNα inhibition. These observations uncover unique neutrophil sub-population biology and dynamics in MPN.
Collapse
Affiliation(s)
- Shirly Partouche
- Felsenstein Medical Research Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel; (I.G.); (P.R.)
| | - Idan Goldberg
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel; (I.G.); (P.R.)
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Internal Medicine F—Recanati, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel
| | - Erez Halperin
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel; (I.G.); (P.R.)
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Bahaa Atamna
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel; (I.G.); (P.R.)
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Adi Shacham-Abulafia
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel; (I.G.); (P.R.)
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Saar Shapira
- Felsenstein Medical Research Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel; (I.G.); (P.R.)
| | - Aladin Samara
- Felsenstein Medical Research Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel; (I.G.); (P.R.)
| | - Ayala Gover-Proaktor
- Felsenstein Medical Research Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel; (I.G.); (P.R.)
| | - Avi Leader
- Hematology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Galia Spectre
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel; (I.G.); (P.R.)
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Pia Raanani
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel; (I.G.); (P.R.)
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Galit Granot
- Felsenstein Medical Research Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel; (I.G.); (P.R.)
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ofir Wolach
- Institute of Hematology, Davidoff Cancer Center, Beilinson Hospital, Rabin Medical Center, Petah Tikva 4941492, Israel; (I.G.); (P.R.)
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
9
|
Zhang Y, Zhao Y, Liu Y, Zhang M, Zhang J. New advances in the role of JAK2 V617F mutation in myeloproliferative neoplasms. Cancer 2024; 130:4229-4240. [PMID: 39277798 DOI: 10.1002/cncr.35559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/05/2024] [Accepted: 08/26/2024] [Indexed: 09/17/2024]
Abstract
The JAK2 V617F mutation is the most common driver gene in myeloproliferative neoplasm (MPN), which means that the JAK/STAT signaling pathway is persistently activated independent of cytokines, and plays an important part in the onset and development of MPN. The JAK inhibitors, although widely used in the clinical practice, are unable to eradicate MPN. Therefore, the unavoidable long-term treatment poses a serious burden for patients with MPN. It is established that the JAK2 V617F mutation, in addition its role in the JAK/STAT pathway, can promote cell proliferation, differentiation, anti-apoptosis, DNA damage accumulation, and other key biologic processes through multiple pathways. Other than that, the JAK2 V617F mutation affects the cardiovascular system through multiple mechanisms. Although JAK inhibitors cannot eradicate MPN cells, the combined use of JAK inhibitors and other drugs may have surprising effects. This requires an in-depth understanding of the mechanism of action of the JAK2 V617F mutation. In this review, the authors explored the role of the JAK2 V617F mutation in MPN from multiple aspects, including the mechanisms of non-JAK/STAT pathways, the regulation of cellular methylation, the induction of cellular DNA damage accumulation, and effects on the cardiovascular system, with the objective of providing valuable insights into multidrug combination therapy for MPN.
Collapse
Affiliation(s)
- Yongchao Zhang
- Department of Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yue Zhao
- Department of Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yusi Liu
- Department of Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Minyu Zhang
- Department of Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jihong Zhang
- Department of Hematology Laboratory, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Chrysafi P, Barnum K, Gerhard GM, Chiasakul T, Narang A, Mcnichol M, Riva N, Semmler G, Scheiner B, Acosta S, Rautou PE, Lauw MN, Berry J, Ageno W, Zwicker JI, Patell R. Anticoagulation for splanchnic vein thrombosis in myeloproliferative neoplasms: a systematic review and meta-analysis. J Thromb Haemost 2024; 22:3479-3489. [PMID: 39127323 DOI: 10.1016/j.jtha.2024.06.029] [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: 03/26/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Optimal anticoagulation management in patients with myeloproliferative neoplasms (MPN) experiencing splanchnic vein thrombosis (SpVT) requires balancing risks of bleeding and recurrent thrombosis. OBJECTIVES We conducted a systematic review and meta-analysis to assess the incidence of bleeding and thrombosis recurrence in patients with MPN-SpVT. METHODS We included retrospective or prospective studies in English with ≥10 adult patients with MPN-SpVT. Outcomes included recurrent venous thrombosis (SpVT and non-SpVT), arterial thrombosis, and major bleeding. Pooled rates per 100 patient years with 95% CIs were calculated by DerSimonian-Laird method using random-effects model. RESULTS Out of 4624 studies screened, 9 studies with a total of 443 patients were included in the meta-analysis with median follow-up of 3.5 years. In the 364 patients with MPN-SpVT treated with anticoagulation, pooled event rate for major bleeding was 2.8 (95% CI, 1.5-5.1; I2 = 95%), for recurrent venous thrombosis was 1.4 (95% CI, 0.8-2.2; I2 = 72%), and for arterial thrombosis was 1.4 (95% CI, 0.6-3.3; I2 = 92%) per 100 patient years. Among 79 patients (n = 4 studies) who did not receive anticoagulation, pooled event rate for major bleeding was 3.2 (95% CI, 0.7-12.7; I2 = 97%), for recurrent venous thrombosis 3.5 (95% CI, 1.8-6.4; I2 = 88%), and for arterial thrombosis rate 1.6 (95% CI, 0.4-6.6; I2 = 95%) per 100 patient years. CONCLUSION Patients with MPN-SpVT treated with anticoagulation have significant risks for both major bleeding and thrombosis recurrence. Further studies are necessary to determine the optimal anticoagulation approach in patients with MPN-SpVT.
Collapse
Affiliation(s)
- Pavlina Chrysafi
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, Massachusetts, USA. https://twitter.com/PavlinaChrysafi
| | - Kevin Barnum
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Genevieve M Gerhard
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Thita Chiasakul
- Center of Excellence in Translational Hematology, Division of Hematology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Arshit Narang
- Department of Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Megan Mcnichol
- Department of Information Systems, Division of Knowledge Services, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Nicoletta Riva
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
| | - Georg Semmler
- Department of Gastroenterology & Hepatology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Scheiner
- Department of Gastroenterology & Hepatology, Medical University of Vienna, Vienna, Austria
| | - Stefan Acosta
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Pierre-Emmanuel Rautou
- Department of Immunology, Institut National de la Santé et de la Recherche Médicale (Inserm), Centre de Recherche sur l'Inflammation, Unité Mixte de Recherche (UMR) 1149, Université Paris-Cité, Paris, France; Division of Hepatology, Assistance Publique - Hôpitaux de Paris (AP-HP), Hôpital Beaujon, Service d'Hépatologie, Département Médico-Universitaire (DMU) DIGEST, Centre de Référence des Maladies Vasculaires du Foie, Filière des Maladies Hépatiques Rares (FILFOIE), European Reference Network for Rare Liver Diseases (ERN RARE-LIVER), Clichy, France
| | - Mandy N Lauw
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Jonathan Berry
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Walter Ageno
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Jeffrey I Zwicker
- Department of Medicine, Hematology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA; Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Rushad Patell
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
11
|
Qiu J, Fu Y, Tian T, Mao Y, Tian Q, Zhou L, Jin R, Zhuang L, Zhou G. Suppression of FOXC1 induces pyroptosis of the coronary artery through activation of JAK2. Atherosclerosis 2024; 396:118543. [PMID: 39182474 DOI: 10.1016/j.atherosclerosis.2024.118543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND AND AIMS Janus kinase 2 (JAK2) triggers endothelial pyroptosis and is associated with a multitude of pathological cardiovascular manifestations, including atherosclerosis. However, the associated transcriptional regulatory mechanisms remain unclear. In this study, we investigated a novel transcriptional regulator upstream of JAK2. METHODS We validated the binding and regulation of Forkhead box C1 (FOXC1) and JAK2 using chromatin immunoprecipitation and luciferase reporter assays. Immunofluorescence was used to detect protein localization in cells and tissues. Immunohistochemistry, hematoxylin-eosin (HE), Masson's trichrome, and Oil Red O staining were used to identify tissue lesions. Transcriptional functions were investigated using in vitro and in vivo coronary artery disease (CAD) atherosclerosis models. RESULTS The mRNA levels of JAK2 were considerably higher in both the cardiac tissues of mice and the peripheral blood of patients with CAD than in equivalent controls. JAK2 expression increased markedly in the coronary arteries of ApoeKO mice, whereas FOXC1 expression exhibited a decreasing trend. In vitro, FOXC1 bound to the JAK2 promoter region and inversely regulated the expression of JAK2. Mechanistic studies have revealed that the FOXC1-JAK2 pathway regulates pyroptosis and participates in the pathogenesis of human coronary artery endothelial cells (HCAECs). In vivo, the suppression of FOXC1 was confirmed to stimulate the levels of JAK2 and pyroptosis, contributing to the pathological progression of aortic and coronary artery damage. CONCLUSIONS We established the FOXC1-JAK2 regulatory pathway and verified its reverse-regulatory function in CAD pyroptosis. Our data emphasizes that FOXC1 is critical for the treatment of pyroptosis-induced injury in patients with CAD.
Collapse
Affiliation(s)
- Jiayun Qiu
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Yahong Fu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Ting Tian
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Yan Mao
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Qiang Tian
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Lanbo Zhou
- Department of Dermatology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Rui Jin
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China
| | - Lili Zhuang
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China.
| | - Guoping Zhou
- Department of Pediatrics, The First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing, 210029, China.
| |
Collapse
|
12
|
Zhang H, Kafeiti N, Masarik K, Lee S, Yang X, Zheng H, Zhan H. Decoding Endothelial MPL and JAK2V617F Mutation: Insight Into Cardiovascular Dysfunction in Myeloproliferative Neoplasms. Arterioscler Thromb Vasc Biol 2024; 44:1960-1974. [PMID: 38989576 PMCID: PMC11335084 DOI: 10.1161/atvbaha.124.321008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 06/18/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Patients with JAK2V617F-positive myeloproliferative neoplasms (MPNs) and clonal hematopoiesis of indeterminate potential face a significantly elevated risk of cardiovascular diseases. Endothelial cells carrying the JAK2V617F mutation have been detected in many patients with MPN. In this study, we investigated the molecular basis for the high incidence of cardiovascular complications in patients with MPN. METHODS We investigated the impact of endothelial JAK2V617F mutation on cardiovascular disease development using both transgenic murine models and MPN patient-derived induced pluripotent stem cell lines. RESULTS Our investigations revealed that JAK2V617F mutant endothelial cells promote cardiovascular diseases under stress, which is associated with endothelial-to-mesenchymal transition and endothelial dysfunction. Importantly, we discovered that inhibiting the endothelial TPO (thrombopoietin) receptor MPL (myeloproliferative leukemia virus oncogene) suppressed JAK2V617F-induced endothelial-to-mesenchymal transition and prevented cardiovascular dysfunction caused by mutant endothelial cells. Notably, the endothelial MPL receptor is not essential for the normal physiological regulation of blood cell counts and cardiac function. CONCLUSIONS JAK2V617F mutant endothelial cells play a critical role in the development of cardiovascular diseases in JAK2V617F-positive MPNs, and endothelial MPL could be a promising therapeutic target for preventing or ameliorating cardiovascular complications in these patients.
Collapse
Affiliation(s)
- Haotian Zhang
- Department of Medicine, Stony Brook School of Medicine, NY (H. Zhang, N.K., K.M., X.Y., H. Zhan)
- The Graduate Program in Molecular and Cellular Biology (H. Zhang), Stony Brook University, NY
| | - Nicholas Kafeiti
- Department of Medicine, Stony Brook School of Medicine, NY (H. Zhang, N.K., K.M., X.Y., H. Zhan)
| | - Kyla Masarik
- Department of Medicine, Stony Brook School of Medicine, NY (H. Zhang, N.K., K.M., X.Y., H. Zhan)
| | - Sandy Lee
- Department of Molecular and Cellular Pharmacology (S.L.), Stony Brook University, NY
| | - Xiaoxi Yang
- Department of Medicine, Stony Brook School of Medicine, NY (H. Zhang, N.K., K.M., X.Y., H. Zhan)
- Division of Rheumatology, Peking Union Medical College Hospital, Beijing, China (X.Y.)
| | - Haoyi Zheng
- Cardiac Imaging, The Heart Center, Saint Francis Hospital, Roslyn, NY (H. Zheng)
| | - Huichun Zhan
- Department of Medicine, Stony Brook School of Medicine, NY (H. Zhang, N.K., K.M., X.Y., H. Zhan)
- Medical Service, Northport VA Medical Center, NY (H. Zhan)
| |
Collapse
|
13
|
Todor SB, Ichim C, Boicean A, Mihaila RG. Cardiovascular Risk in Philadelphia-Negative Myeloproliferative Neoplasms: Mechanisms and Implications-A Narrative Review. Curr Issues Mol Biol 2024; 46:8407-8423. [PMID: 39194713 DOI: 10.3390/cimb46080496] [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: 07/08/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/29/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs), encompassing disorders like polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are characterized by clonal hematopoiesis without the Philadelphia chromosome. The JAK2 V617F mutation is prevalent in PV, ET, and PMF, while mutations in MPL and CALR also play significant roles. These conditions predispose patients to thrombotic events, with PMF exhibiting the lowest survival among MPNs. Chronic inflammation, driven by cytokine release from aberrant leukocytes and platelets, amplifies cardiovascular risk through various mechanisms, including atherosclerosis and vascular remodeling. Additionally, MPN-related complications like pulmonary hypertension and cardiac fibrosis contribute to cardiovascular morbidity and mortality. This review consolidates recent research on MPNs' cardiovascular implications, emphasizing thrombotic risk, chronic inflammation, and vascular stiffness. Understanding these associations is crucial for developing targeted therapies and improving outcomes in MPN patients.
Collapse
Affiliation(s)
- Samuel Bogdan Todor
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Cristian Ichim
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | - Adrian Boicean
- Faculty of Medicine, Lucian Blaga University of Sibiu, 550169 Sibiu, Romania
| | | |
Collapse
|
14
|
Bourrienne MC, Le Cam Duchez V, Faille D, Farkh C, Solo Nomenjanahary M, Gay J, Loyau S, Journé C, Dupont S, Ollivier V, Villeval JL, Plo I, Edmond V, Jandrot-Perrus M, Labrouche-Colomer S, Cassinat B, Verger E, Desilles JP, Ho-Tin-Noé B, Triquenot Bagan A, Mazighi M, Ajzenberg N. Exacerbation of thromboinflammation by JAK2V617F mutation worsens the prognosis of cerebral venous sinus thrombosis. Blood Adv 2024; 8:3330-3343. [PMID: 38386979 PMCID: PMC11258627 DOI: 10.1182/bloodadvances.2023011692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/22/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024] Open
Abstract
ABSTRACT Cerebral venous sinus thrombosis (CVST) is an uncommon venous thromboembolic event accounting for <1% of strokes resulting in brain parenchymal injuries. JAK2V617F mutation, the most frequent driving mutation of myeloproliferative neoplasms, has been reported to be associated with worse clinical outcomes in patients with CVST. We investigated whether hematopoietic JAK2V617F expression predisposes to specific pathophysiological processes and/or worse prognosis after CVST. Using an in vivo mouse model of CVST, we analyzed clinical, biological, and imaging outcomes in mice with hematopoietic-restricted Jak2V617F expression, compared with wild-type Jak2 mice. In parallel, we studied a human cohort of JAK2V617F-positive or -negative CVST. Early after CVST, mice with hematopoietic Jak2V617F expression had increased adhesion of platelets and neutrophils in cerebral veins located in the vicinity of CVST. On day 1, Jak2V617F mice had a worse outcome characterized by significantly more frequent and severe intracranial hemorrhages (ICHs) and higher mortality rates. Peripheral neutrophil activation was enhanced, as indicated by higher circulating platelet-neutrophil aggregates, upregulated CD11b expression, and higher myeloperoxydase plasma level. Concurrently, immunohistological and brain homogenate analysis showed higher neutrophil infiltration and increased blood-brain barrier disruption. Similarly, patients with JAK2V617F-positive CVST tended to present higher thrombotic burden and had significantly higher systemic immune-inflammation index, a systemic thromboinflammatory marker, than patients who were JAK2V617F-negative. In mice with CVST, our study corroborates that Jak2V617F mutation leads to a specific pattern including increased thrombotic burden, ICH, and mortality. The exacerbated thromboinflammatory response, observed both in mice and patients positive for JAK2V617F, could contribute to hemorrhagic complications.
Collapse
Affiliation(s)
- Marie-Charlotte Bourrienne
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
- Laboratoire d’Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Véronique Le Cam Duchez
- UNIROUEN, INSERM U1096, Normandie University, Rouen University Hospital, Hemostasis Unit and INSERM CIC-CRB 1404, Rouen, France
| | - Dorothée Faille
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
- Laboratoire d’Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Carine Farkh
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
- Laboratoire d’Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | | | - Juliette Gay
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
- Laboratoire d’Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| | - Stéphane Loyau
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
| | - Clément Journé
- Université Paris Cité, INSERM UMS34, Fédération de Recherche en Imagerie Multimodalités, Faculté de Médecine X. Bichat, Paris, France
| | - Sébastien Dupont
- Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
| | - Véronique Ollivier
- Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
| | - Jean-Luc Villeval
- INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Isabelle Plo
- INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Valérie Edmond
- INSERM U1287, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Martine Jandrot-Perrus
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
| | - Sylvie Labrouche-Colomer
- Université de Bordeaux, INSERM U1034, Biologie des maladies cardio-vasculaires & Laboratoire d’hématologie, Centre Hospitalier Universitaire de Bordeaux, Pessac, France
| | - Bruno Cassinat
- Laboratoire de Biologie Cellulaire, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Emmanuelle Verger
- Laboratoire de Biologie Cellulaire, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Jean-Philippe Desilles
- Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
- Département de Neuroradiologie interventionnelle, Hôpital Fondation Rothschild, Paris, France
| | - Benoît Ho-Tin-Noé
- Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
| | - Aude Triquenot Bagan
- Department of Neurology and INSERM CIC-CRB 1404, Rouen University Hospital, Rouen, France
| | - Mikaël Mazighi
- Université Paris Cité, Inserm, UMRS-1144, Optimisation Thérapeutique en Neuropsychopharmacologie, Paris, France
- Département de Neurologie, AP-HP, Hôpital Lariboisière, FHU NeuroVasc, Paris, France
| | - Nadine Ajzenberg
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM UMRS-1148, Laboratory for Vascular Translational Science, Paris, France
- Laboratoire d’Hématologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| |
Collapse
|
15
|
Xu Y, Carrier M, Kimpton M. Arterial Thrombosis in Patients with Cancer. Cancers (Basel) 2024; 16:2238. [PMID: 38927943 PMCID: PMC11201749 DOI: 10.3390/cancers16122238] [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: 04/25/2024] [Revised: 06/03/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Patients with cancer are at increased risk of arterial thromboembolic disease due to the presence of risk factors common to both the development of cancer and arterial thrombosis, the cancer itself, and the treatments provided to treat cancer. We review here the epidemiology and pathophysiology of arterial thromboembolic disease in cancer, along with its prevention and treatment strategies. We also propose a generalized approach for the management of arterial thromboembolic disease in this patient population.
Collapse
Affiliation(s)
| | - Marc Carrier
- Department of Medicine, The Ottawa Hospital Research Institute at University of Ottawa, Ottawa, ON K1H 8L6, Canada; (Y.X.); (M.K.)
| | | |
Collapse
|
16
|
Liu Y, Wang Y, Huang G, Wu S, Liu X, Chen S, Luo P, Liu C, Zuo X. The role of leukocytes in myeloproliferative neoplasm thromboinflammation. J Leukoc Biol 2024; 115:1020-1028. [PMID: 38527797 DOI: 10.1093/jleuko/qiae071] [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: 01/04/2024] [Revised: 03/04/2024] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
Classic myeloproliferative neoplasms lacking the Philadelphia chromosome are stem cell disorders characterized by the proliferation of myeloid cells in the bone marrow and increased counts of peripheral blood cells. The occurrence of thrombotic events is a common complication in myeloproliferative neoplasms. The heightened levels of cytokines play a substantial role in the morbidity and mortality of these patients, establishing a persistent proinflammatory condition that culminates in thrombosis. The etiology of thrombosis remains intricate and multifaceted, involving blood cells and endothelial dysfunction, the inflammatory state, and the coagulation cascade, leading to hypercoagulability. Leukocytes play a pivotal role in the thromboinflammatory process of myeloproliferative neoplasms by releasing various proinflammatory and prothrombotic factors as well as interacting with other cells, which contributes to the amplification of the clotting cascade and subsequent thrombosis. The correlation between increased leukocyte counts and thrombotic risk has been established. However, there is a need for an accurate biomarker to assess leukocyte activation. Lastly, tailored treatments to address the thrombotic risk in myeloproliferative neoplasms are needed. Therefore, this review aims to summarize the potential mechanisms of leukocyte involvement in myeloproliferative neoplasm thromboinflammation, propose potential biomarkers for leukocyte activation, and discuss promising treatment options for controlling myeloproliferative neoplasm thromboinflammation.
Collapse
Affiliation(s)
- Yu Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169, East Lake Road, Wuchang District, Wuhan 430071, China
| | - Yingying Wang
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Yixueyuan Road, Yuzhong District, Chongqing 400016, P.R. China
| | - Gang Huang
- Department of Cell Systems & Anatomy, Department of Pathology & Laboratory, Medicine UT Health San Antonio, Joe R. and Teresa Lozano Long School of Medicine, 8403 Floyd Curl Drive, San Antonio, TX 78229, United States
| | - Sanyun Wu
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169, East Lake Road, Wuchang District, Wuhan 430071, China
| | - Xiaoyan Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169, East Lake Road, Wuchang District, Wuhan 430071, China
| | - Shuo Chen
- Biomedical Sciences Graduate Program, Ohio State University, Columbus, OH 43210, United States
| | - Ping Luo
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169, East Lake Road, Wuchang District, Wuhan 430071, China
| | - Chang Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169, East Lake Road, Wuchang District, Wuhan 430071, China
| | - Xuelan Zuo
- Department of Hematology, Zhongnan Hospital of Wuhan University, 169, East Lake Road, Wuchang District, Wuhan 430071, China
| |
Collapse
|
17
|
Brett VE, Dignat George F, James C. Circulating endothelial cells in pathophysiology. Curr Opin Hematol 2024; 31:148-154. [PMID: 38362895 DOI: 10.1097/moh.0000000000000814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
PURPOSE OF REVIEW The purpose of this review is to synthesize recent insights into the roles and importance of circulating endothelial cells (CECs) as indicators of the severity, progression, and prognosis of vascular-related diseases. RECENT FINDINGS Recent studies have identified elevated counts of CECs in pathological conditions, notably inflammatory or cardiovascular diseases such as acute myocardial infarction and heart failure, underscoring their potential as sensitive indicators of disease. Furthermore, the rise in CEC levels in cancer patients, particularly with disease advancement, points to their role in cancer-associated angiogenesis and response to treatment. SUMMARY This review underscores the evolving significance of CECs as markers for evaluating the gravity and advancement of diseases with vascular injury, including cardiovascular diseases, cancer, inflammatory conditions, and thromboembolic events. These last years, efforts made to standardize flow cytometry detection of CEC and the development of highly sensitive techniques to isolate, quantify or phenotype rare cells open promising avenues for clinical application. This may yield extensive knowledge regarding the mechanisms by which endothelial cells contribute to a variety of vascular-related disorders and their clinical value as emerging biomarkers.
Collapse
Affiliation(s)
- Victor Emmanuel Brett
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034
- Laboratory of Hematology, Bordeaux University Hospital, Pessac
| | - Francoise Dignat George
- Aix Marseille Univ, INSERM, INRAE, C2VN, UFR de Pharmacie
- Hematology and Vascular Biology Department, CHU La Conception, Assistance Publique-Hôpitaux de Marseille, Marseille, France
| | - Chloe James
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034
- Laboratory of Hematology, Bordeaux University Hospital, Pessac
| |
Collapse
|
18
|
Yasuda S, Chiba M, Nishitani R, Watanabe T. Adrenal infarction with latent myelodysplastic/myeloproliferative neoplasm, unclassifiable with JAK2V617F mutation. Clin Case Rep 2024; 12:e8729. [PMID: 38601172 PMCID: PMC11004262 DOI: 10.1002/ccr3.8729] [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: 02/07/2024] [Revised: 03/13/2024] [Accepted: 03/21/2024] [Indexed: 04/12/2024] Open
Abstract
Key Clinical Message Hematopoietic neoplasms can cause adrenal infarction. In cases of thrombosis occurring at uncommon sites, it is necessary to consider evaluating for the JAK2V617F mutation, even in the absence of notable abnormalities in blood counts. Abstract Adrenal infarction, a rare ailment, has been sporadically linked to hematopoietic neoplasms. A 46-year-old male encountered left adrenal infarction, which coincided with a progressive rise in platelet counts. Subsequent diagnosis revealed myelodysplastic/myeloproliferative neoplasm-unclassifiable, featuring a JAK2V617F mutation. Simultaneously, the patient manifested multiple arteriovenous thromboses, necessitating treatment with edoxaban, aspirin, and hydroxyurea. Following thrombosis resolution, he was transferred to a transplantation center. This report delves into the thrombogenicity linked to the JAK2V617F mutation, while also examining documented instances of adrenal infarction in myeloid neoplasms. We should consider evaluating for JAK2V617F mutation even in cases of thrombosis at unusual sites, including adrenal infarction, even if there are no considerable abnormalities in blood counts.
Collapse
Affiliation(s)
| | - Momoko Chiba
- Department of HematologyTokyo Kyosai HospitalTokyoJapan
| | - Rie Nishitani
- Department of Diabetes, Endocrinology and MetabolismTokyo Kyosai HospitalTokyoJapan
| | - Takako Watanabe
- Department of Diabetes, Endocrinology and MetabolismTokyo Kyosai HospitalTokyoJapan
| |
Collapse
|
19
|
Boccatonda A, Gentilini S, Zanata E, Simion C, Serra C, Simioni P, Piscaglia F, Campello E, Ageno W. Portal Vein Thrombosis: State-of-the-Art Review. J Clin Med 2024; 13:1517. [PMID: 38592411 PMCID: PMC10932352 DOI: 10.3390/jcm13051517] [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: 02/11/2024] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 04/10/2024] Open
Abstract
Background: Portal vein thrombosis (PVT) is a rare disease with an estimated incidence of 2 to 4 cases per 100,000 inhabitants. The most common predisposing conditions for PVT are chronic liver diseases (cirrhosis), primary or secondary hepatobiliary malignancy, major infectious or inflammatory abdominal disease, or myeloproliferative disorders. Methods: PVT can be classified on the basis of the anatomical site, the degree of venous occlusion, and the timing and type of presentation. The main differential diagnosis of PVT, both acute and chronic, is malignant portal vein invasion, most frequently by hepatocarcinoma, or constriction (typically by pancreatic cancer or cholangiocarcinoma). Results: The management of PVT is based on anticoagulation and the treatment of predisposing conditions. The aim of anticoagulation in acute thrombosis is to prevent the extension of the clot and enable the recanalization of the vein to avoid the development of complications, such as intestinal infarction and portal hypertension. Conclusions: The treatment with anticoagulant therapy favors the reduction of portal hypertension, and this allows for a decrease in the risk of bleeding, especially in patients with esophageal varices. The anticoagulant treatment is generally recommended for at least three to six months. Prosecution of anticoagulation is advised until recanalization or lifelong if the patient has an underlying permanent pro-coagulant condition that cannot be corrected or if there is thrombosis extending to the mesenteric veins.
Collapse
Affiliation(s)
- Andrea Boccatonda
- Internal Medicine, Bentivoglio Hospital, Azienda Unità Sanitaria Locale (AUSL) Bologna, 40010 Bentivoglio, Italy
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
| | - Simone Gentilini
- Internal Medicine Department, IRCCS Azienda Ospedaliero-Universitaria Policlinico di Sant’Orsola, 40138 Bologna, Italy; (S.G.); (E.Z.)
| | - Elisa Zanata
- Internal Medicine Department, IRCCS Azienda Ospedaliero-Universitaria Policlinico di Sant’Orsola, 40138 Bologna, Italy; (S.G.); (E.Z.)
| | - Chiara Simion
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University Hospital of Padova, 35128 Padova, Italy (E.C.)
| | - Carla Serra
- Interventional, Diagnostic and Therapeutic Ultrasound Unit, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Paolo Simioni
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University Hospital of Padova, 35128 Padova, Italy (E.C.)
| | - Fabio Piscaglia
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Elena Campello
- General Medicine and Thrombotic and Hemorrhagic Diseases Unit, Department of Medicine, University Hospital of Padova, 35128 Padova, Italy (E.C.)
| | - Walter Ageno
- Research Center on Thromboembolic Diseases and Antithrombotic Therapies, Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy
| |
Collapse
|
20
|
Andreescu M, Andreescu B. A Review About the Assessment of the Bleeding and Thrombosis Risk for Patients With Myeloproliferative Neoplasms Scheduled for Surgery. Cureus 2024; 16:e56008. [PMID: 38606222 PMCID: PMC11007487 DOI: 10.7759/cureus.56008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 04/13/2024] Open
Abstract
Myeloproliferative neoplasms (MPNs) present a unique challenge in surgical management due to their inherent predisposition to both bleeding and thrombosis. MPNs are a heterogenous group of acquired clonal conditions. The three classic MPNs are essential thrombocythemia (ET), myelofibrosis (PMF), and polycythemia vera (PV). All subtypes of MPN are associated with both thrombotic and bleeding complications. There are four risk categories for thrombosis in MPN patients: age, thrombosis history, and JAK-2 mutation. They are further classified as very low, low, intermediate, and high risk. The genetic landscape of MPN is fascinating and complex like all myeloid disorders. Bleeding risk can be assessed through leukocytosis, thrombocytosis, acquired von Willebrand syndrome (AVWS), and a previous history of bleeding in a patient. Risk assessment and perioperative management are important aspects of improving the quality of life and preventing complications in surgeries. Preoperative management includes a risk assessment of venous thromboembolism, use of appropriate pharmacological treatment, platelet count control, and correction and cardiovascular risk factors. This review summarizes the assessment of bleeding and thrombosis risk for patients with MPNs scheduled for surgery. Furthermore, this review discusses various tools that can be used to identify MPN patients at risk of thrombosis prior to surgery.
Collapse
Affiliation(s)
- Mihaela Andreescu
- Faculty of Medicine, Titu Maiorescu University, Bucharest, ROU
- Hematology, Colentina Clinical Hospital, Bucharest, ROU
| | | |
Collapse
|
21
|
Tirandi A, Schiavetta E, Maioli E, Montecucco F, Liberale L. Inflammation as a cause of acute myocardial infarction in patients with myeloproliferative neoplasm. World J Cardiol 2024; 16:58-63. [PMID: 38456066 PMCID: PMC10915890 DOI: 10.4330/wjc.v16.i2.58] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/01/2024] [Accepted: 01/18/2024] [Indexed: 02/21/2024] Open
Abstract
Myeloproliferative neoplasms (MPN) are a group of diseases characterized by the clonal proliferation of hematopoietic progenitor or stem cells. They are clinically classifiable into four main diseases: chronic myeloid leukemia, essential thrombocythemia, polycythemia vera, and primary myelofibrosis. These pathologies are closely related to cardio- and cerebrovascular diseases due to the increased risk of arterial thrombosis, the most common underlying cause of acute myocardial infarction. Recent evidence shows that the classical Virchow triad (hypercoagulability, blood stasis, endothelial injury) might offer an explanation for such association. Indeed, patients with MPN might have a higher number and more reactive circulating platelets and leukocytes, a tendency toward blood stasis because of a high number of circulating red blood cells, endothelial injury or overactivation as a consequence of sustained inflammation caused by the neoplastic clonal cell. These abnormal cancer cells, especially when associated with the JAK2V617F mutation, tend to proliferate and secrete several inflammatory cytokines. This sustains a pro-inflammatory state throughout the body. The direct consequence is the induction of a pro-thrombotic state that acts as a determinant in favoring both venous and arterial thrombus formation. Clinically, MPN patients need to be carefully evaluated to be treated not only with cytoreductive treatments but also with cardiovascular protective strategies.
Collapse
Affiliation(s)
- Amedeo Tirandi
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
| | - Elisa Schiavetta
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
| | - Elia Maioli
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa 16132, Italy.
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa - Italian Cardiovascular Network, Genoa 16132, Italy
| |
Collapse
|
22
|
Tsumita T, Takeda R, Maishi N, Hida Y, Sasaki M, Orba Y, Sato A, Toba S, Ito W, Teshirogi T, Sakurai Y, Iba T, Naito H, Ando H, Watanabe H, Mizuno A, Nakanishi T, Matsuda A, Zixiao R, Lee J, Iimura T, Sawa H, Hida K. Viral uptake and pathophysiology of the lung endothelial cells in age-associated severe SARS-CoV-2 infection models. Aging Cell 2024; 23:e14050. [PMID: 38098255 PMCID: PMC10861199 DOI: 10.1111/acel.14050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/10/2023] [Accepted: 11/13/2023] [Indexed: 12/20/2023] Open
Abstract
Thrombosis is the major cause of death in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, and the pathology of vascular endothelial cells (ECs) has received much attention. Although there is evidence of the infection of ECs in human autopsy tissues, their detailed pathophysiology remains unclear due to the lack of animal model to study it. We used a mouse-adapted SARS-CoV-2 virus strain in young and mid-aged mice. Only mid-aged mice developed fatal pneumonia with thrombosis. Pulmonary ECs were isolated from these infected mice and RNA-Seq was performed. The pulmonary EC transcriptome revealed that significantly higher levels of viral genes were detected in ECs from mid-aged mice with upregulation of viral response genes such as DDX58 and IRF7. In addition, the thrombogenesis-related genes encoding PLAT, PF4, F3 PAI-1, and P-selectin were upregulated. In addition, the inflammation-related molecules such as CXCL2 and CXCL10 were upregulated in the mid-aged ECs upon viral infection. Our mouse model demonstrated that SARS-CoV-2 virus entry into aged vascular ECs upregulated thrombogenesis and inflammation-related genes and led to fatal pneumonia with thrombosis. Current results of EC transcriptome showed that EC uptake virus and become thrombogenic by activating neutrophils and platelets in the aged mice, suggesting age-associated EC response as a novel finding in human severe COVID-19.
Collapse
Affiliation(s)
- Takuya Tsumita
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Ryo Takeda
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
- Department of Oral Diagnosis and Medicine, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Nako Maishi
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Yasuhiro Hida
- Department of Advanced Robotic and Endoscopic SurgeryFujita Health UniversityToyoakeJapan
| | - Michihito Sasaki
- Division of Molecular Pathobiology, International Institute for Zoonosis ControlHokkaido UniversitySapporoJapan
| | - Yasuko Orba
- Division of Molecular Pathobiology, International Institute for Zoonosis ControlHokkaido UniversitySapporoJapan
- International Collaboration Unit, International Institute for Zoonosis ControlHokkaido UniversitySapporoJapan
| | - Akihiko Sato
- Division of Molecular Pathobiology, International Institute for Zoonosis ControlHokkaido UniversitySapporoJapan
- Drug Discovery and Disease Research LaboratoryShionogi and Co., Ltd.OsakaJapan
| | - Shinsuke Toba
- Division of Molecular Pathobiology, International Institute for Zoonosis ControlHokkaido UniversitySapporoJapan
- Drug Discovery and Disease Research LaboratoryShionogi and Co., Ltd.OsakaJapan
| | - Wataru Ito
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
- Department of Oral and Maxillofacial Surgery, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Takahito Teshirogi
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
- Department of Dental Anesthesiology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Yuya Sakurai
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
- Department of Dental Anesthesiology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Tomohiro Iba
- Department of Vascular Physiology, Graduate School of Medical SciencesKanazawa UniversityKanazawaJapan
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Hisamichi Naito
- Department of Vascular Physiology, Graduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Hitoshi Ando
- Department of Cellular and Molecular Function Analysis, Graduate School of Medical SciencesKanazawa UniversityKanazawaJapan
| | - Haruhisa Watanabe
- Department of Pharmacology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Amane Mizuno
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Toshiki Nakanishi
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Aya Matsuda
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Ren Zixiao
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
- Department of Oral and Maxillofacial Surgery, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Ji‐Won Lee
- Department of Pharmacology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Tadahiro Iimura
- Department of Pharmacology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| | - Hirofumi Sawa
- Division of Molecular Pathobiology, International Institute for Zoonosis ControlHokkaido UniversitySapporoJapan
- International Collaboration Unit, International Institute for Zoonosis ControlHokkaido UniversitySapporoJapan
- One Health Research CenterHokkaido UniversitySapporoJapan
- Institute for Vaccine Research and DevelopmentHokkaido UniversitySapporoJapan
| | - Kyoko Hida
- Department of Vascular Biology and Molecular Pathology, Faculty and Graduate School of Dental MedicineHokkaido UniversitySapporoJapan
| |
Collapse
|
23
|
Pasquer H, Daltro de Oliveira R, Vasseur L, Soret-Dulphy J, Maslah N, Zhao LP, Marcault C, Cazaux M, Gauthier N, Verger E, Parquet N, Vainchenker W, Raffoux E, Ugo V, Luque Paz D, Roy L, Lambert WC, Ianotto JC, Lippert E, Giraudier S, Cassinat B, Kiladjian JJ, Benajiba L. Distinct clinico-molecular arterial and venous thrombosis scores for myeloproliferative neoplasms risk stratification. Leukemia 2024; 38:326-339. [PMID: 38148396 DOI: 10.1038/s41375-023-02114-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 12/28/2023]
Abstract
Current recommended risk scores to predict thrombotic events associated with myeloproliferative neoplasms (MPN) do not discriminate between arterial and venous thrombosis despite their different physiopathology. To define novel stratification systems, we delineated a comprehensive landscape of MPN associated thrombosis across a large long-term follow-up MPN cohort. Prior arterial thrombosis, age >60 years, cardiovascular risk factors and presence of TET2 or DNMT3A mutations were independently associated with arterial thrombosis in multivariable analysis. ARTS, an ARterial Thrombosis Score, based on these four factors, defined low- (0.37% patients-year) and high-risk (1.19% patients-year) patients. ARTS performance was superior to the two-tiered conventional risk stratification in our training cohort, across all MPN subtypes, as well as in two external validation cohorts. Prior venous thrombosis and presence of a JAK2V617F mutation with a variant allelic frequency ≥50% were independently associated with venous thrombosis. The discrimination potential of VETS, a VEnous Thrombosis Score based on these two factors, was poor, similar to the two-tiered conventional risk stratification. Our study pinpoints arterial and venous thrombosis clinico-molecular differences and proposes an arterial risk score for more accurate patients' stratification. Further improvement of venous risk scores, accounting for additional factors and considering venous thrombosis as a heterogeneous entity is warranted.
Collapse
Affiliation(s)
- Hélène Pasquer
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
- INSERM UMR 944, Institut de Recherche Saint-Louis, Paris, France
| | - Rafael Daltro de Oliveira
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Loic Vasseur
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Juliette Soret-Dulphy
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Nabih Maslah
- Université Paris Cité, APHP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Lin-Pierre Zhao
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Clémence Marcault
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Marine Cazaux
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Nicolas Gauthier
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Emmanuelle Verger
- Université Paris Cité, APHP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Nathalie Parquet
- Université Paris Cité, APHP, Hôpital Saint-Louis, Département d'hématologie et d'Immunologie, Paris, France
| | - William Vainchenker
- APHP, Hôpital Saint-Louis, Département d'hématologie et d'Immunologie, Paris, France
| | - Emmanuel Raffoux
- Université Paris Cité, APHP, Hôpital Saint-Louis, Département d'hématologie et d'Immunologie, Paris, France
| | - Valérie Ugo
- Univ Angers, Nantes Université, CHU Angers, Inserm, CNRS, CRCI2NA, Angers, France
| | - Damien Luque Paz
- Univ Angers, Nantes Université, CHU Angers, Inserm, CNRS, CRCI2NA, Angers, France
| | - Lydia Roy
- Université Paris Est Créteil, APHP, Hôpital Henri Mondor, Service d'hématologie, Créteil, France
| | - Wayne-Corentin Lambert
- Université de Bretagne Occidentale, CHU de Brest, Service d'Hématologie Biologique, Brest, France
| | - Jean-Christophe Ianotto
- Université de Bretagne Occidentale, CHU de Brest, Service d'Hématologie et d'Hémostase Clinique, Brest, France
| | - Eric Lippert
- Université de Bretagne Occidentale, CHU de Brest, Service d'Hématologie Biologique, Brest, France
| | - Stéphane Giraudier
- Université Paris Cité, APHP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Bruno Cassinat
- Université Paris Cité, APHP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Jean-Jacques Kiladjian
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Lina Benajiba
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France.
- INSERM UMR 944, Institut de Recherche Saint-Louis, Paris, France.
| |
Collapse
|
24
|
Grenier JMP, El Nemer W, De Grandis M. Red Blood Cell Contribution to Thrombosis in Polycythemia Vera and Essential Thrombocythemia. Int J Mol Sci 2024; 25:1417. [PMID: 38338695 PMCID: PMC10855956 DOI: 10.3390/ijms25031417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Polycythemia vera (PV) and essential thrombocythemia (ET) are myeloproliferative neoplasms (MPN) characterized by clonal erythrocytosis and thrombocytosis, respectively. The main goal of therapy in PV and ET is to prevent thrombohemorrhagic complications. Despite a debated notion that red blood cells (RBCs) play a passive and minor role in thrombosis, there has been increasing evidence over the past decades that RBCs may play a biological and clinical role in PV and ET pathophysiology. This review summarizes the main mechanisms that suggest the involvement of PV and ET RBCs in thrombosis, including quantitative and qualitative RBC abnormalities reported in these pathologies. Among these abnormalities, we discuss increased RBC counts and hematocrit, that modulate blood rheology by increasing viscosity, as well as qualitative changes, such as deformability, aggregation, expression of adhesion proteins and phosphatidylserine and release of extracellular microvesicles. While the direct relationship between a high red cell count and thrombosis is well-known, the intrinsic defects of RBCs from PV and ET patients are new contributors that need to be investigated in depth in order to elucidate their role and pave the way for new therapeutical strategies.
Collapse
Affiliation(s)
- Julien M. P. Grenier
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France
- Laboratoire d’Excellence GR-Ex, 75015 Paris, France
| | - Wassim El Nemer
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France
- Laboratoire d’Excellence GR-Ex, 75015 Paris, France
| | - Maria De Grandis
- Etablissement Français du Sang PACA-Corse, Aix Marseille University, CNRS, ADES UMR 7268, 13005 Marseille, France
- Laboratoire d’Excellence GR-Ex, 75015 Paris, France
| |
Collapse
|
25
|
Guy A, Garcia G, Gourdou-Latyszenok V, Wolff-Trombini L, Josserand L, Kimmerlin Q, Favre S, Kilani B, Marty C, Boulaftali Y, Labrouche-Colomer S, Mansier O, James C. Platelets and neutrophils cooperate to induce increased neutrophil extracellular trap formation in JAK2V617F myeloproliferative neoplasms. J Thromb Haemost 2024; 22:172-187. [PMID: 37678548 DOI: 10.1016/j.jtha.2023.08.028] [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: 02/28/2023] [Revised: 08/24/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Neutrophils participate in the pathogenesis of thrombosis through the formation of neutrophil extracellular traps (NETs). Thrombosis is the main cause of morbidity and mortality in patients with myeloproliferative neoplasms (MPNs). Recent studies have shown an increase in NET formation (NETosis) both in patients with JAK2V617F neutrophils and in mouse models, and reported the participation of NETosis in the pathophysiology of thrombosis in mice. OBJECTIVES This study investigated whether JAK2V617F neutrophils are sufficient to promote thrombosis or whether their cooperation with other blood cell types is necessary. METHODS NETosis was studied in PF4iCre;Jak2V617F/WT mice expressing JAK2V617F in all hematopoietic lineages, as occurs in MPNs, and in MRP8Cre;Jak2V617F/WT mice in which JAK2V617F is expressed only in leukocytes. RESULTS In PF4iCre;Jak2V617F/WT mice, an increase in NETosis and spontaneous lung thrombosis abrogated by DNAse administration were observed. The absence of spontaneous NETosis or lung thrombosis in MRP8Cre;Jak2V617F/WT mice suggested that mutated neutrophils alone are not sufficient to induce thrombosis. Ex vivo experiments demonstrated that JAK2V617F-mutated platelets trigger NETosis by JAK2V617F-mutated neutrophils. Aspirin treatment in PF4iCre;Jak2V617F/WT mice reduced NETosis and reduced lung thrombosis. In cytoreductive-therapy-free patients with MPN treated with aspirin, plasma NET marker concentrations were lower than that in patients with MPN not treated with aspirin. CONCLUSION Our study demonstrates that JAK2V617F neutrophils alone are not sufficient to promote thrombosis; rather, platelets cooperate with neutrophils to promote NETosis in vivo. A new role for aspirin in thrombosis prevention in MPNs was also identified.
Collapse
Affiliation(s)
- Alexandre Guy
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France; Laboratory of Hematology, Bordeaux University Hospital, Pessac, France. https://twitter.com/Alexandreguy6
| | - Geoffrey Garcia
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France. https://twitter.com/GeofGarciaVirginie
| | - Virginie Gourdou-Latyszenok
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France. https://twitter.com/GourdouV
| | - Laura Wolff-Trombini
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France. https://twitter.com/TrombiniWolff
| | - Lara Josserand
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France
| | - Quentin Kimmerlin
- Department of Biomedicine, Experimental Hematology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Simon Favre
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France
| | - Badr Kilani
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France
| | - Caroline Marty
- Institut national de la santé et de la recherche médicale, UMR1287, University of Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Yacine Boulaftali
- Paris Diderot University, Institut national de la santé et de la recherche médicale, Unité Mixte de Recherche_S1148, Laboratory for Vascular Translational Science, Paris, France
| | - Sylvie Labrouche-Colomer
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France; Laboratory of Hematology, Bordeaux University Hospital, Pessac, France
| | - Olivier Mansier
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France; Laboratory of Hematology, Bordeaux University Hospital, Pessac, France
| | - Chloé James
- University of Bordeaux, Institut national de la santé et de la recherche médicale, Biologie des maladies cardio-vasculaires, U1034, Pessac, France; Laboratory of Hematology, Bordeaux University Hospital, Pessac, France.
| |
Collapse
|
26
|
Pescia C, Lopez G, Cattaneo D, Bucelli C, Gianelli U, Iurlo A. The molecular landscape of myeloproliferative neoplasms associated with splanchnic vein thrombosis: Current perspective. Leuk Res 2024; 136:107420. [PMID: 38016412 DOI: 10.1016/j.leukres.2023.107420] [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: 06/26/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/30/2023]
Abstract
BCR::ABL1-negative myeloproliferative neoplasms (MPNs) are classically represented by polycythemia vera, essential thrombocythemia, and primary myelofibrosis. BCR::ABL1-negative MPNs are significantly associated with morbidity and mortality related to an increased risk of thrombo-hemorrhagic events. They show a consistent association with splanchnic vein thrombosis (SVT), either represented by the portal, mesenteric or splenic vein thrombosis, or Budd-Chiari Syndrome. SVT is also a frequent presenting manifestation of MPN. MPNs associated with SVT show a predilection for younger women, high association with JAK2V617F mutation, low JAK2V617F variant allele frequency (generally <10 %), and low rates of CALR, MPL, or JAK2 exon 12 mutations. Next-Generation Sequencing techniques have contributed to deepening our knowledge of the molecular landscape of such cases, with potential diagnostic and prognostic implications. In this narrative review, we analyze the current perspective on the molecular background of MPN associated with SVT, pointing as well future directions in this field.
Collapse
Affiliation(s)
- Carlo Pescia
- Unit of Anatomic Pathology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Gianluca Lopez
- Unit of Anatomic Pathology, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Biomedical, Surgical and Dental Sciences, University of Milan, Italy
| | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Cristina Bucelli
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Umberto Gianelli
- Department of Health Sciences, University of Milan, Milan, Italy; Unit of Anatomic Pathology, ASST Santi Paolo e Carlo, Milan, Italy
| | - Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
27
|
Beleva EA. Splanchnic Vein Thrombosis in Myelofibrosis-An Underappreciated Hallmark of Disease Phenotype. Int J Mol Sci 2023; 24:15717. [PMID: 37958701 PMCID: PMC10649007 DOI: 10.3390/ijms242115717] [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: 09/30/2023] [Revised: 10/21/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Splanchnic vein thrombosis (SVT) encompasses thrombosis in the vessels of the splanchnic basin and has a relatively rare occurrence with a reported frequency in the general population of 1-2%. An episode of seemingly unprovoked SVT almost always triggers a diagnostic work-up for a Philadelphia chromosome-negative myeloproliferative neoplasm (MPN), since atypical site thrombosis is a hallmark of MPN-associated thrombophilia. Primary myelofibrosis (PMF) is a rare MPN with an estimated incidence between 0.1 and 1/100,000 per year. Although prothrombotic tendency in PMF is not envisioned as a subject of specific therapeutic management, unlike other MPNs, such as polycythemia vera (PV) and essential thrombocythemia (ET), thrombotic risk and SVT prevalence in PMF may be comparably high. Additionally, unlike PV and ET, SVT development in PMF may depend more on procoagulant mechanisms involving endothelium than on blood cell activation. Emerging results from registry data also suggest that PMF patients with SVT may exhibit lower risk and better prognosis, thus highlighting the need for better thrombotic risk stratification and identifying a subset of patients with potential benefit from antithrombotic prophylaxis. This review highlights specific epidemiological, pathogenetic, and clinical features pertinent to SVT in myelofibrosis.
Collapse
Affiliation(s)
- Elina A. Beleva
- Clinic of Hematology, Military Medical Academy, 1606 Sofia, Bulgaria;
- QSAR and Molecular Modelling, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| |
Collapse
|
28
|
Montani D, Thoré P, Mignard X, Jaïs X, Boucly A, Jevnikar M, Seferian A, Jutant EM, Cottin V, Fadel E, Simonneau G, Savale L, Sitbon O, Humbert M. Clinical Phenotype and Outcomes of Pulmonary Hypertension Associated with Myeloproliferative Neoplasms: A Population-based Study. Am J Respir Crit Care Med 2023; 208:600-612. [PMID: 37311222 DOI: 10.1164/rccm.202210-1941oc] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 06/13/2023] [Indexed: 06/15/2023] Open
Abstract
Rationale: Precapillary pulmonary hypertension (PH) is a rare and largely unrecognized complication of myeloproliferative neoplasms (MPNs), including polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (MF). Objectives: To describe characteristics and outcomes of MPN-associated PH. Methods: We report clinical, functional, and hemodynamic characteristics, classification, and outcomes of patients with PV, ET, or primary MF in the French PH registry. Measurements and Main Results: Ninety patients with MPN (42 PV, 35 ET, 13 primary MF) presented with precapillary PH with severe hemodynamic impairment, with a median mean pulmonary arterial pressure and pulmonary vascular resistance of 42 mm Hg and 6.7 Wood units, respectively, and impaired clinical conditions, with 71% in New York Heart Association functional classes III/IV and having a median 6-minute-walk distance of 310 m. Half of the patients were diagnosed with chronic thromboembolic PH (CTEPH); the other half were considered to have group 5 PH. MF was preferentially associated with group 5 PH, whereas PV and ET were generally related to CTEPH. Proximal lesions were diagnosed in half of the patients with CTEPH. Thromboendarterectomy was performed in 18 selected patients with high risk of complications (5 early deaths). Overall survival at 1, 3, and 5 years was 67%, 50%, and 34% in group 5 PH and 81%, 66%, and 42% in CTEPH, respectively. Conclusions: PH is a life-threatening condition potentially occurring in MPN. There are multiple mechanisms, with equal diagnoses of CTEPH and group 5 PH. Physicians should be aware that PH strongly affects the burden of patients with MPN, especially in group 5 PH, with unknown pathophysiological mechanisms.
Collapse
Affiliation(s)
- David Montani
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies and
| | - Pierre Thoré
- Assistance Publique - Hôpitaux de Paris, Department of Respiratory Medicine, Histiocytosis National Referral Center, Hôpital Saint-Louis, Paris, France
- School of Medicine, Université Paris Cité, Paris, France
| | - Xavier Mignard
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies and
| | - Xavier Jaïs
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies and
| | - Athénaïs Boucly
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies and
| | - Mitja Jevnikar
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies and
| | - Andrei Seferian
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies and
| | - Etienne-Marie Jutant
- Department of Pneumology, University Hospital of Poitiers, Institut National de la Santé et de la Recherche Médicale Centre D'investigation Clinique 1402, University of Poitiers, Poitiers, France; and
| | - Vincent Cottin
- Centre Hospitalier Universitaire de Lyon Hospices Civils de Lyon, Service de Pneumologie, Centre de Référence des Maladies Pulmonaires Rares, Groupement Hospitalier Est, Hôpital Louis Pradel, Bron, France
| | - Elie Fadel
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies and
- Department of Thoracic Surgery, Hôpital Marie Lannelongue, Le Plessis-Robinson, France
| | - Gérald Simonneau
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies and
| | - Laurent Savale
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies and
| | - Olivier Sitbon
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies and
| | - Marc Humbert
- School of Medicine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique - Hôpitaux de Paris, Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Hôpital Bicêtre, Le Kremlin-Bicêtre, France
- Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 999 Pulmonary Hypertension: Pathophysiology and Novel Therapies and
| |
Collapse
|
29
|
Giuli L, Pallozzi M, Venturini G, Gasbarrini A, Ponziani FR, Santopaolo F. Molecular Mechanisms Underlying Vascular Liver Diseases: Focus on Thrombosis. Int J Mol Sci 2023; 24:12754. [PMID: 37628933 PMCID: PMC10454315 DOI: 10.3390/ijms241612754] [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: 07/15/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
Vascular liver disorders (VLDs) comprise a wide spectrum of clinical-pathological entities that primarily affect the hepatic vascular system of both cirrhotic and non-cirrhotic patients. VLDs more frequently involve the portal and the hepatic veins, as well as liver sinusoids, resulting in an imbalance of liver homeostasis with serious consequences, such as the development of portal hypertension and liver fibrosis. Surprisingly, many VLDs are characterized by a prothrombotic phenotype. The molecular mechanisms that cause thrombosis in VLD are only partially explained by the alteration in the Virchow's triad (hypercoagulability, blood stasis, and endothelial damage) and nowadays their pathogenesis is incompletely described and understood. Studies about this topic have been hampered by the low incidence of VLDs in the general population and by the absence of suitable animal models. Recently, the role of coagulation imbalance in liver disease has been postulated as one of the main mechanisms linked to fibrogenesis, so a novel interest in vascular alterations of the liver has been renewed. This review provides a detailed analysis of the current knowledge of molecular mechanisms of VLD. We also focus on the promising role of anticoagulation as a strategy to prevent liver complications and to improve the outcome of these patients.
Collapse
Affiliation(s)
- Lucia Giuli
- Hepatology Unit, CEMAD Centro Malattie Dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (M.P.); (G.V.); (F.R.P.); (F.S.)
| | - Maria Pallozzi
- Hepatology Unit, CEMAD Centro Malattie Dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (M.P.); (G.V.); (F.R.P.); (F.S.)
| | - Giulia Venturini
- Hepatology Unit, CEMAD Centro Malattie Dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (M.P.); (G.V.); (F.R.P.); (F.S.)
| | - Antonio Gasbarrini
- Hepatology Unit, CEMAD Centro Malattie Dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (M.P.); (G.V.); (F.R.P.); (F.S.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Hepatology Unit, CEMAD Centro Malattie Dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (M.P.); (G.V.); (F.R.P.); (F.S.)
| | - Francesco Santopaolo
- Hepatology Unit, CEMAD Centro Malattie Dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy; (L.G.); (M.P.); (G.V.); (F.R.P.); (F.S.)
| |
Collapse
|
30
|
Guijarro-Hernández A, Vizmanos JL. Transcriptomic comparison of bone marrow CD34 + cells and peripheral blood neutrophils from ET patients with JAK2 or CALR mutations. BMC Genom Data 2023; 24:40. [PMID: 37550636 PMCID: PMC10408115 DOI: 10.1186/s12863-023-01142-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 07/25/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Essential thrombocythemia (ET) is one of the most common types of Ph-negative myeloproliferative neoplasms, an infrequent group of blood cancers that arise from a CD34 + hematopoietic stem cell (HSC) in the bone marrow (BM) primarily due to driver mutations in JAK2, CALR or MPL. These aberrations result in an overproduction of mature myeloid cells in peripheral blood (PB). To date, no targeted therapies have been approved for ET patients, so the study of the molecular mechanisms behind the disease and the identification of new therapeutic targets may be of interest. For this reason, in this study, we have compared the transcriptomic profile of undifferentiated CD34 + cells and mature myeloid cells from ET patients (CALR and JAK2-mutated) and healthy donors deposited in publicly available databases. The study of the similarities and differences between these samples might help to better understand the molecular mechanisms behind the disease according to the degree of maturation of the malignant clone and the type of mutation and ultimately help identify new therapeutic targets for these patients. RESULTS The results show that most of the altered hallmarks in neutrophils were also found in CD34 + cells. However, only a few genes showed a similar aberrant expression pattern in both types of cells. We have identified a signature of six genes common to patients with CALR and JAK2 mutations (BPI, CRISP3, LTF, MMP8, and PTGS1 upregulated, and PBXIP1 downregulated), a different signature of seven genes for patients with CALR mutations (BMP6, CEACAM8, ITK, LCN2, and PRG2 upregulated, and MAN1A1 and MME downregulated) and a signature of 13 genes for patients with JAK2 mutations (ARG1, CAST, CD177, CLEC5A, DAPP1, EPS15, IL18RAP, OLFM4, OLR1, RIOK3, SELP, and THBS1 upregulated, and IGHM downregulated). CONCLUSIONS Our results highlight transcriptomic similarities and differences in ET patients according to the degree of maturation of the malignant clone and the type of mutation. The genes and processes altered in both CD34 + cells and mature neutrophils may reveal altered sustained processes that could be studied as future therapeutic targets for ET patients.
Collapse
Affiliation(s)
- Ana Guijarro-Hernández
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - José Luis Vizmanos
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain.
| |
Collapse
|
31
|
Beckman JD, DaSilva A, Aronovich E, Nguyen A, Nguyen J, Hargis G, Reynolds D, Vercellotti GM, Betts B, Wood DK. JAK-STAT inhibition reduces endothelial prothrombotic activation and leukocyte-endothelial proadhesive interactions. J Thromb Haemost 2023; 21:1366-1380. [PMID: 36738826 PMCID: PMC10246778 DOI: 10.1016/j.jtha.2023.01.027] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
BACKGROUND Vascular activation is characterized by increased proinflammatory, pro thrombotic, and proadhesive signaling. Several chronic and acute conditions, including Bcr-abl-negative myeloproliferative neoplasms (MPNs), graft-vs-host disease, and COVID-19 have been noted to have increased activation of the janus kinase (JAK)-signal transducer and downstream activator of transcription (STAT) pathways. Two notable inhibitors of the JAK-STAT pathway are ruxolitinib (JAK1/2 inhibitor) and fedratinib (JAK2 inhibitor), which are currently used to treat MPN patients. However, in some conditions, it has been noted that JAK inhibitors can increase the risk of thromboembolic complications. OBJECTIVES We sought to define the anti-inflammatory and antithrombotic effects of JAK-STAT inhibitors in vascular endothelial cells. METHODS We assessed endothelial activation in the presence or absence of ruxolitinib or fedratinib by using immunoblots, immunofluorescence, qRT-PCR, and function coagulation assays. Finally, we used endothelialized microfluidics perfused with blood from normal and JAK2V617F+ individuals to evaluate whether ruxolitinib and fedratinib changed cell adhesion. RESULTS We found that both ruxolitinib and fedratinib reduced endothelial cell phospho-STAT1 and STAT3 signaling and attenuated nuclear phospho-NK-κB and phospho-c-Jun localization. JAK-STAT inhibition also limited secretion of proadhesive and procoagulant P-selectin and von Willebrand factor and proinflammatory IL-6. Likewise, we found that JAK-STAT inhibition reduced endothelial tissue factor and urokinase plasminogen activator expression and activity. CONCLUSIONS By using endothelialized microfluidics perfused with whole blood samples, we demonstrated that endothelial treatment with JAK-STAT inhibitors prevented rolling of both healthy control and JAK2V617F MPN leukocytes. Together, these findings demonstrate that JAK-STAT inhibitors reduce the upregulation of critical prothrombotic pathways and prevent increased leukocyte-endothelial adhesion.
Collapse
Affiliation(s)
- Joan D Beckman
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA.
| | - Angelica DaSilva
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Elena Aronovich
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Aithanh Nguyen
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Julia Nguyen
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Geneva Hargis
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - David Reynolds
- Department of Biomedical Engineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Gregory M Vercellotti
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - Brian Betts
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, Minnesota, USA
| | - David K Wood
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
32
|
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
|
33
|
Iizuka K, Morishita S, Nishizaki Y, Iizuka Y, Iriyama N, Ochiai T, Yanagisawa N, Yasuda H, Ando J, Gotoh A, Takei M, Hatta Y, Nakamura H, Nakayama T, Komatsu N. Von Willebrand factor activity levels are influenced by driver mutation status in polycythemia vera and essential thrombocythemia patients with well-controlled platelet counts. Eur J Haematol Suppl 2022; 109:779-786. [PMID: 36130908 DOI: 10.1111/ejh.13866] [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: 05/04/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 01/19/2023]
Abstract
Von Willebrand factor ristocetin cofactor (VWF activity) and platelet count (PLT) are negatively correlated in patients with polycythemia vera (PV) and essential thrombocythemia (ET). However, VWF activity does not always normalize upon controlling PLT in those patients. To address this issue, we investigated the correlation between VWF activity and PLT in PV and ET patients. The negative correlation between VWF activity and PLT was stronger in calreticulin mutation-positive (CALR+) ET than in Janus kinase 2 mutation-positive (JAK2+) PV or ET groups. When PLT were maintained at a certain level (<600 × 109 /L), low VWF activity (<50%) was more frequently observed in JAK2+ PV patients than in JAK2+ ET (p=0.013) or CALR+ ET (p=0.013) groups, and in PV and ET patients with ≥50% JAK2+ allele burden than in those with allele burden <50% (p=0.015). High VWF activity (>150%) was more frequent in the JAK2+ ET group than in the CALR+ ET group (p=0.005), and often associated with vasomotor symptoms (p=0.002). This study suggests that some patients with JAK2+ PV or ET have VWF activity outside the standard range even with well-controlled PLT, and that the measurement of VWF activity is useful for assessing the risk of thrombosis and hemorrhage.
Collapse
Affiliation(s)
- Kazuhide Iizuka
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan.,Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan.,Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan.,Internal Medicine, Atami Tokoro Memorial Hospital, Shizuoka, Japan
| | - Soji Morishita
- Laboratory for the development of therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuji Nishizaki
- Medical Technology Innovation Center, Juntendo University, Tokyo, Japan
| | - Yoshikazu Iizuka
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Noriyoshi Iriyama
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Tomonori Ochiai
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Hajime Yasuda
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | - Jun Ando
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan.,Department of Cell Therapy and Transfusion Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akihiko Gotoh
- Department of Hematology, Tokyo Medical University, Tokyo, Japan
| | - Masami Takei
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Yoshihiro Hatta
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Hideki Nakamura
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Tomohiro Nakayama
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan.,Laboratory for the development of therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Pharmaessentia Japan KK, Tokyo, Japan
| |
Collapse
|
34
|
Malara A, Gruppi C, Massa M, Tira ME, Rosti V, Balduini A, Barosi G. Elevated plasma EDA fibronectin in primary myelofibrosis is determined by high allele burden of JAK2V617F mutation and strongly predicts splenomegaly progression. Front Oncol 2022; 12:987643. [PMID: 36212480 PMCID: PMC9532599 DOI: 10.3389/fonc.2022.987643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/31/2022] [Indexed: 11/18/2022] Open
Abstract
In primary myelofibrosis, extra-domain A fibronectin (EDA-FN), the result of alternative splicing of FN gene, sustains megakaryocyte proliferation and confers a pro-inflammatory phenotype to bone marrow cell niches. In this work we assessed the levels of circulating EDA-FN in plasma samples of 122 patients with primary myelofibrosis. Patients with a homozygous JAK2V617F genotype displayed the higher level of plasma EDA-FN. Increased EDA-FN levels were associated with anemia, elevated high-sensitivity C-reactive protein, bone marrow fibrosis and splanchnic vein thrombosis at diagnosis. While no correlation was observed with CD34+ hematopoietic stem cell mobilization, elevated blood level of EDA-FN at diagnosis was a predictor of large splenomegaly (over 10 cm from the left costal margin) outcome. Thus, EDA-FN expression in primary myelofibrosis may represent the first marker of disease progression, and a novel target to treat splenomegaly.
Collapse
Affiliation(s)
- Alessandro Malara
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- *Correspondence: Alessandro Malara, ; Alessandra Balduini,
| | - Cristian Gruppi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Margherita Massa
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnostics, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Maria Enrica Tira
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Vittorio Rosti
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnostics, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
- *Correspondence: Alessandro Malara, ; Alessandra Balduini,
| | - Giovanni Barosi
- Center for the Study of Myelofibrosis, Laboratory of Biochemistry, Biotechnology and Advanced Diagnostics, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| |
Collapse
|
35
|
Prediction of thrombosis in post-polycythemia vera and post-essential thrombocythemia myelofibrosis: a study on 1258 patients. Leukemia 2022; 36:2453-2460. [PMID: 36042316 DOI: 10.1038/s41375-022-01673-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 11/08/2022]
Abstract
Patients with Philadelphia-negative myeloproliferative neoplasms are at high risk of thrombotic events (TEs). Predisposing factors have been identified in essential thrombocythemia (ET), polycythemia vera (PV) and primary myelofibrosis (primary MF, PMF), while yet not recognized in post PV/ET-MF (known as secondary MF, SMF). Within the 1258 SMF of the MYSEC (MYelofibrosis SECondary to PV and ET) dataset, 135 (10.7%) developed a TE at a median follow-up of 3.5 years (range, 1-21.4), with an incidence of 2.3% patients per year. Venous events accounted for two-thirds of the total. Cox multivariable analysis, supported by Fine-Gray models with death as competitive risk, showed that being on cytoreductive therapy at time of SMF evolution is associated with an absolute risk reduction of thrombosis equal to 3.3% within 3 years. Considering individually cytoreductive therapies, univariate regression model found that both conventional cytoreduction, mainly hydroxyurea, (HR 0.41, 95% CI: 0.26-0.65, p = 0.0001) and JAK inhibitors, mostly ruxolitinib, (HR 0.50, 95% CI: 0.24-1.02, p = 0.05) were associated with fewer thrombosis. Our study informs treating physicians of a non-low incidence of TEs in post PV/ET-MF and of the potential protective role of cytoreductive therapy in terms of thrombotic events.
Collapse
|
36
|
Splanchnic vein thrombosis associated with myeloproliferative neoplasms. Thromb Res 2022; 218:8-16. [PMID: 35963121 DOI: 10.1016/j.thromres.2022.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/30/2022] [Accepted: 08/03/2022] [Indexed: 11/22/2022]
|
37
|
Bhuria V, Baldauf CK, Schraven B, Fischer T. Thromboinflammation in Myeloproliferative Neoplasms (MPN)-A Puzzle Still to Be Solved. Int J Mol Sci 2022; 23:ijms23063206. [PMID: 35328626 PMCID: PMC8954909 DOI: 10.3390/ijms23063206] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 02/04/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs), a group of malignant hematological disorders, occur as a consequence of somatic mutations in the hematopoietic stem cell compartment and show excessive accumulation of mature myeloid cells in the blood. A major cause of morbidity and mortality in these patients is the marked prothrombotic state leading to venous and arterial thrombosis, including myocardial infarction (MI), deep vein thrombosis (DVT), and strokes. Additionally, many MPN patients suffer from inflammation-mediated constitutional symptoms, such as fever, night sweats, fatigue, and cachexia. The chronic inflammatory syndrome in MPNs is associated with the up-regulation of various inflammatory cytokines in patients and is involved in the formation of the so-called MPN thromboinflammation. JAK2-V617F, the most prevalent mutation in MPNs, has been shown to activate a number of integrins on mature myeloid cells, including granulocytes and erythrocytes, which increase adhesion and drive venous thrombosis in murine knock-in/out models. This review aims to shed light on the current understanding of thromboinflammation, involvement of neutrophils in the prothrombotic state, plausible molecular mechanisms triggering the process of thrombosis, and potential novel therapeutic targets for developing effective strategies to reduce the MPN disease burden.
Collapse
Affiliation(s)
- Vikas Bhuria
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention—ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Conny K. Baldauf
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention—ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Correspondence: ; Tel.: +49-391-67-15338; Fax: +49-391-67-15852
| | - Thomas Fischer
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany; (V.B.); (C.K.B.); (T.F.)
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention—ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
38
|
Bhuria V, Baldauf CK, Schraven B, Fischer T. Thromboinflammation in Myeloproliferative Neoplasms (MPN)-A Puzzle Still to Be Solved. Int J Mol Sci 2022. [PMID: 35328626 DOI: 10.3390/ijms23063206.pmid:35328626;pmcid:pmc8954909] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs), a group of malignant hematological disorders, occur as a consequence of somatic mutations in the hematopoietic stem cell compartment and show excessive accumulation of mature myeloid cells in the blood. A major cause of morbidity and mortality in these patients is the marked prothrombotic state leading to venous and arterial thrombosis, including myocardial infarction (MI), deep vein thrombosis (DVT), and strokes. Additionally, many MPN patients suffer from inflammation-mediated constitutional symptoms, such as fever, night sweats, fatigue, and cachexia. The chronic inflammatory syndrome in MPNs is associated with the up-regulation of various inflammatory cytokines in patients and is involved in the formation of the so-called MPN thromboinflammation. JAK2-V617F, the most prevalent mutation in MPNs, has been shown to activate a number of integrins on mature myeloid cells, including granulocytes and erythrocytes, which increase adhesion and drive venous thrombosis in murine knock-in/out models. This review aims to shed light on the current understanding of thromboinflammation, involvement of neutrophils in the prothrombotic state, plausible molecular mechanisms triggering the process of thrombosis, and potential novel therapeutic targets for developing effective strategies to reduce the MPN disease burden.
Collapse
Affiliation(s)
- Vikas Bhuria
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention-ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Conny K Baldauf
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Burkhart Schraven
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention-ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Thomas Fischer
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Health-Campus Immunology, Infectiology, and Inflammation, Medical Center, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Center for Health and Medical Prevention-ChaMP, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| |
Collapse
|
39
|
Shapira Cohen T, Chodick G, Steinberg DM, Grossman E, Shohat M, Salomon O. JAK2V617F Is a Risk Factor for TIA/Stroke in Young Patients. Thromb Haemost 2022; 122:1333-1340. [PMID: 35288888 DOI: 10.1055/s-0042-1743470] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The objective of this study was to assess the risk of arterial thrombosis in patients who harbor the JAK2V617F allele burden ≥1% detected during workup for myeloproliferative neoplasms (MPNs). We conducted a large cross-sectional analysis consisted of 5,220 patients who were tested for JAK2V617F and 1,047,258 people matched in age from health care insurance provider, taking into account age, sex, hypertension, diabetes, atrial fibrillation. Compared with noncarriers, mutation carriers were older, less likely to be current or past smokers and had lower body mass index. There was no significant difference between the groups regarding myocardial infarction and peripheral vascular disease. However, JAK2V617F ≥1% at age 34 to 54 years was associated with eightfold more likely to have transient ischemic attack (TIA)/stroke history unrelated to hypertension, diabetes, or atrial fibrillation. Association of JAK2V617F with TIA/stroke was also observed in the older age group, albeit a weaker association and not statistically significant. Prevalence of TIA/stroke was higher in patients with JAK2V617F negative, with odds ratio of 3.93 when compared with the general population after confounder adjustments. Further research is warranted to verify the relation between allele burden of JAK2V617F mutation and TIA/stroke and the role of JAK2V617F per se as a risk factor for arterial thrombosis in the absence of overt MPN. Also, consideration should be paid to the screened group with JAK2V617F negative due to the high incidence of TIA/stroke among them in comparison to the general population.
Collapse
Affiliation(s)
| | - Gabriel Chodick
- Maccabitech, Maccabi Institute for Research and Innovation, Maccabi Healthcare Services, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - David M Steinberg
- Department of Statistics and Operations Research, Faculty of Exact Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ehud Grossman
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Division of Internal Medicine, Sheba Medical Center, Tel Hashomer, Israel
| | - Mordechai Shohat
- Institute of Medical Genetics, Maccabi HMO, Rehovot, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Cancer Research Center, Wohl Institute of Translational Medicine, Sheba Medical Center, Tel Hashomer, Israel
| | - Ophira Salomon
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Thrombosis Unit, Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
40
|
Laktib N, Mahtat EM, Lahlafi Z, Mouine N, Asfalou I, Aghoutane N, Chaib A, Lakhal Z, Doghmi K, Benyass A. Essential thrombocythemia and aortic dissection,causal or incidental association? JOURNAL DE MEDECINE VASCULAIRE 2022; 47:39-42. [PMID: 35393092 DOI: 10.1016/j.jdmv.2022.01.136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 01/12/2022] [Indexed: 06/14/2023]
Abstract
Only few cases of vascular dissection and essential thrombocythemia association have been reported. To the best of our knowledge, we reported the second case of aortic dissection and essential thrombocythemia association in a 60-year-old man with positive JAK2V617F mutation who had no history of hypertension or connective tissue disorders. Through this case, we discussed the eventual existence of a causal relationship between the two conditions. We also suggested the use of hydroxyurea as a prevention treatment of thrombosis in myeloproliferative neoplasms.
Collapse
Affiliation(s)
- Nabil Laktib
- Department of Cardiology Intensive Care Unit, Cardiology Center of Mohammed V Military Training Hospital, Rabat, Morocco.
| | - El Mehdi Mahtat
- Department of Hematology, Mohammed V Military Training Hospital, Rabat, Morocco
| | - Zakaria Lahlafi
- Catheterization laboratory, Cardiology Center of Mohammed V Military Training Hospital, Rabat, Morocco
| | - Najat Mouine
- Department of Clinical Cardiology, Cardiology Center of Mohammed V Military Training Hospital, Rabat, Morocco
| | - Iliyasse Asfalou
- Department of non-Invasive Cardiac Explorations, Cardiology Center of Mohammed V Military Training Hospital, Rabat, Morocco
| | - Nabil Aghoutane
- Department of Vascular Surgery, Mohammed V Military Training Hospital, Rabat, Morocco
| | - Ali Chaib
- Department of Rhythmology, Cardiology Center of Mohammed V Military Training Hospital, Rabat, Morocco
| | - Zouhair Lakhal
- Department of Cardiology Intensive Care Unit, Cardiology Center of Mohammed V Military Training Hospital, Rabat, Morocco
| | - Kamal Doghmi
- Department of Hematology, Mohammed V Military Training Hospital, Rabat, Morocco
| | - Aatif Benyass
- Cardiology Center of Mohammed V Military Training Hospital, Rabat, Morocco
| |
Collapse
|
41
|
Bader MS, Meyer SC. JAK2 in Myeloproliferative Neoplasms: Still a Protagonist. Pharmaceuticals (Basel) 2022; 15:ph15020160. [PMID: 35215273 PMCID: PMC8874480 DOI: 10.3390/ph15020160] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
The discovery of the activating V617F mutation in Janus kinase 2 (JAK2) has been decisive for the understanding of myeloproliferative neoplasms (MPN). Activated JAK2 signaling by JAK2, CALR, and MPL mutations has become a focus for the development of targeted therapies for patients with MPN. JAK2 inhibitors now represent a standard of clinical care for certain forms of MPN and offer important benefits for MPN patients. However, several key aspects remain unsolved regarding the targeted therapy of MPN with JAK2 inhibitors, such as reducing the MPN clone and how to avoid or overcome a loss of response. Here, we summarize the current knowledge on the structure and signaling of JAK2 as central elements of MPN pathogenesis and feature benefits and limitations of therapeutic JAK2 targeting in MPN.
Collapse
Affiliation(s)
| | - Sara Christina Meyer
- Division of Hematology, University Hospital Basel, CH-4031 Basel, Switzerland;
- Department of Biomedicine, University Hospital Basel and University of Basel, CH-4031 Basel, Switzerland
- Correspondence:
| |
Collapse
|
42
|
Feng Y, Zhang Y, Shi J. Thrombosis and hemorrhage in myeloproliferative neoplasms: The platelet perspective. Platelets 2022; 33:955-963. [PMID: 35081860 DOI: 10.1080/09537104.2021.2019210] [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: 10/19/2022]
Abstract
Classical myeloproliferative neoplasm (MPN), also known as BCR-ABL-negative MPN, is a clonal disease characterized by abnormal expansion of hematopoietic stem cells. It has been demonstrated that MPN patients are more susceptible to thrombotic events compared to the general population. Therefore, researchers have been exploring the treatment for MPN thrombosis. However, antithrombotic therapies have brought another concern for the clinical management of MPN because they may cause bleeding events. When thrombosis and bleeding, two seemingly contradictory complications, occur in MPN patients at the same time, they will lead to more serious consequences. Therefore, it is a major challenge to achieving the best antithrombotic effect and minimizing bleeding events simultaneously. To date, there has yet been a perfect strategy to meet this challenge and therefore a new treatment method needs to be established. In this article, we describe the mechanism of thrombosis and bleeding events in MPN from the perspective of platelets for the first time. Based on the double-sided role of platelets in MPN, optimal antithrombotic treatment strategies that can simultaneously control thrombosis and bleeding at the same time may be formulated by adjusting the administration time and dosage of antiplatelet drugs. We argue that more attention should be paid to the critical role of platelets in MPN thrombosis and MPN bleeding in the future, so as to better manage adverse vascular events in MPN.
Collapse
Affiliation(s)
- Yiming Feng
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Yue Zhang
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Jialan Shi
- Department of Hematology, The First Affiliated Hospital, Harbin Medical University, Harbin, China.,Departments of Medical Oncology and Research, Dana-Farber Cancer Institute, Va Boston Healthcare System, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
43
|
Papageorgiou L, Elalamy I, Vandreden P, Gerotziafas GT. Thrombotic and Hemorrhagic Issues Associated with Myeloproliferative Neoplasms. Clin Appl Thromb Hemost 2022; 28:10760296221097969. [PMID: 35733370 PMCID: PMC9234921 DOI: 10.1177/10760296221097969] [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] [Indexed: 12/04/2022] Open
Abstract
Thrombotic and hemorrhagic complications are related to a significant rate of
morbidity and mortality in patients with myeloproliferative neoplasms (MPNs),
they are therefore called “thrombohemorrhagic” syndromes. Several clinical
factors, such as age and presence of cardiovascular comorbidities are
responsible for thrombotic complications. High blood counts, platelet
alterations, presence of JAK2 mutation and possibly of other CHIP mutations such
as TET2, DNMT3A, and ASXL1, procoagulant microparticles, NETs formation,
endothelial activation and neo-angiogenesis are some of the parameters
accounting for hypercoagulability in patients with myeloproliferative neoplasms.
Bleeding complications emerge as a result of platelet exhaustion. They can be
also linked to a functional deficiency of von Willebrand factor, when platelet
counts rise above 1000G/L. The mainstay of management consists on preventing
hemostatic complications, by antiplatelet and/or anticoagulant treatment and
myelosuppressive agents in high-risk patients.Circumstances related to a high
thrombohemorrhagic risk, such as pregnancy and the perioperative period, prompt
for specific management with regards to anticoagulation and myelosuppression
treatment type. In order to apply a patient-specific treatment strategy, there
is a need for a risk score assessment tool encompassing clinical parameters and
hemostasis biomarkers.
Collapse
Affiliation(s)
- Loula Papageorgiou
- Hrombosis Center, 432215Service d'Hématologie Biologique Hôpital Tenon, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Faculté de Médecine Sorbonne Université, Paris, France.,Faculty of Medicine, Research Group "Cancer, Haemostasis and Angiogenesis", INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Sorbonne University, Paris, France
| | - Ismail Elalamy
- Hrombosis Center, 432215Service d'Hématologie Biologique Hôpital Tenon, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Faculté de Médecine Sorbonne Université, Paris, France.,Faculty of Medicine, Research Group "Cancer, Haemostasis and Angiogenesis", INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Sorbonne University, Paris, France.,The First I.M. Sechenov Moscow State Medical University, Moscow, Russia
| | - Patrick Vandreden
- Faculty of Medicine, Research Group "Cancer, Haemostasis and Angiogenesis", INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Sorbonne University, Paris, France.,Clinical Research Department, Diagnostica Stago, Gennevilliers, France
| | - Grigoris T Gerotziafas
- Hrombosis Center, 432215Service d'Hématologie Biologique Hôpital Tenon, Hôpitaux Universitaires de l'Est Parisien, Assistance Publique Hôpitaux de Paris, Faculté de Médecine Sorbonne Université, Paris, France.,Faculty of Medicine, Research Group "Cancer, Haemostasis and Angiogenesis", INSERM U938, Centre de Recherche Saint-Antoine, Institut Universitaire de Cancérologie, Sorbonne University, Paris, France
| |
Collapse
|
44
|
Karki NR, Saunders K, Kutlar A. A critical evaluation of crizanlizumab for the treatment of sickle cell disease. Expert Rev Hematol 2021; 15:5-13. [PMID: 34942078 DOI: 10.1080/17474086.2022.2023007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION P-selectin is a key adhesion molecule in the pathogenesis of sickle cell disease, including acute painful event(s). Many of the mediators activated in prototypical pain crisis are also involved in other complications seen in this population. Crizanlizumab is a monoclonal antibody approved in the US in 2019 for patients of all genotypes of sickle cell disease. By blocking P-selectin, it effectively prevents acute painful event(s) and has a manageable safety profile. AREAS COVERED In this review, we provide an overview of the (i) biology of P-selectin in sickle cell disease, (ii) various agents inhibiting P-selectin, (iii) pharmacology of crizanlizumab, (iv) preclinical and clinical data on crizanlizumab, and (v) its potential for other indications, ongoing studies, regulatory status, and cost issues. Further, we describe its position among other approved agents in sickle cell disease and project future directions as well. EXPERT OPINION Crizanlizumab holds great promise in modulating the natural history of sickle cell disease and may have pleotropic effects. Studies are ongoing to define its role in preventing other sickle cell-related complications, non-sickle cell inflammatory states, and thrombotic disorders.
Collapse
Affiliation(s)
- Nabin Raj Karki
- Division of Hematology/Oncology, Augusta University, Augusta, GA, USA
| | | | - Abdullah Kutlar
- Division of Hematology/Oncology, Augusta University, Augusta, GA, USA
| |
Collapse
|
45
|
Progression of Myeloproliferative Neoplasms (MPN): Diagnostic and Therapeutic Perspectives. Cells 2021; 10:cells10123551. [PMID: 34944059 PMCID: PMC8700229 DOI: 10.3390/cells10123551] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 12/21/2022] Open
Abstract
Classical BCR-ABL-negative myeloproliferative neoplasms (MPN) are a heterogeneous group of hematologic malignancies, including essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF), as well as post-PV-MF and post-ET-MF. Progression to more symptomatic disease, such as overt MF or acute leukemia, represents one of the major causes of morbidity and mortality. There are clinically evident but also subclinical types of MPN progression. Clinically evident progression includes evolution from ET to PV, ET to post-ET-MF, PV to post-PV-MF, or pre-PMF to overt PMF, and transformation of any of these subtypes to myelodysplastic neoplasms or acute leukemia. Thrombosis, major hemorrhage, severe infections, or increasing symptom burden (e.g., pruritus, night sweats) may herald progression. Subclinical types of progression may include increases in the extent of bone marrow fibrosis, increases of driver gene mutational allele burden, and clonal evolution. The underlying causes of MPN progression are diverse and can be attributed to genetic alterations and chronic inflammation. Particularly, bystander mutations in genes encoding epigenetic regulators or splicing factors were associated with progression. Finally, comorbidities such as systemic inflammation, cardiovascular diseases, and organ fibrosis may augment the risk of progression. The aim of this review was to discuss types and mechanisms of MPN progression and how their knowledge might improve risk stratification and therapeutic intervention. In view of these aspects, we discuss the potential benefits of early diagnosis using molecular and functional imaging and exploitable therapeutic strategies that may prevent progression, but also highlight current challenges and methodological pitfalls.
Collapse
|
46
|
MPN and thrombosis was hard enough . . . now there's COVID-19 thrombosis too. Hematology 2021; 2021:710-717. [DOI: 10.1182/hematology.2021000315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Both myeloproliferative neoplasms (MPNs) and coronavirus disease 2019 (COVID-19) are characterized by an intrinsic thrombotic risk. Little is known about the incidence and the outcome of thrombotic events in patients with MPN infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but common mechanisms of coagulation activation, typical of both disorders, suggest that these patients can be at particularly high risk. To define the best thromboprophylaxis and treatment regimens in both MPN and COVID-19, individual- and disease-specific thrombotic risk factors, bleeding risk, and concomitant specific treatments need to be considered. In this case-based review, an individualized approach is presented in a case of SARS-CoV-2 infection occurring in a man with polycythemia vera (PV). A primary anticoagulant thromboprophylaxis strategy and adjustment of his PV treatment were implemented. However, during the hospital stay, he experienced pulmonary embolism and therapeutic anticoagulation had to be set. Then his condition improved, and discharge was planned. Postdischarge decisions had to be made about the type and duration of venous thromboembolism treatment as well as the management of PV-specific drugs. The steps of our decisions and recommendations are presented.
Collapse
|
47
|
Mancini SJC, Balabanian K, Corre I, Gavard J, Lazennec G, Le Bousse-Kerdilès MC, Louache F, Maguer-Satta V, Mazure NM, Mechta-Grigoriou F, Peyron JF, Trichet V, Herault O. Deciphering Tumor Niches: Lessons From Solid and Hematological Malignancies. Front Immunol 2021; 12:766275. [PMID: 34858421 PMCID: PMC8631445 DOI: 10.3389/fimmu.2021.766275] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Knowledge about the hematopoietic niche has evolved considerably in recent years, in particular through in vitro analyzes, mouse models and the use of xenografts. Its complexity in the human bone marrow, in particular in a context of hematological malignancy, is more difficult to decipher by these strategies and could benefit from the knowledge acquired on the niches of solid tumors. Indeed, some common features can be suspected, since the bone marrow is a frequent site of solid tumor metastases. Recent research on solid tumors has provided very interesting information on the interactions between tumoral cells and their microenvironment, composed notably of mesenchymal, endothelial and immune cells. This review thus focuses on recent discoveries on tumor niches that could help in understanding hematopoietic niches, with special attention to 4 particular points: i) the heterogeneity of carcinoma/cancer-associated fibroblasts (CAFs) and mesenchymal stem/stromal cells (MSCs), ii) niche cytokines and chemokines, iii) the energy/oxidative metabolism and communication, especially mitochondrial transfer, and iv) the vascular niche through angiogenesis and endothelial plasticity. This review highlights actors and/or pathways of the microenvironment broadly involved in cancer processes. This opens avenues for innovative therapeutic opportunities targeting not only cancer stem cells but also their regulatory tumor niche(s), in order to improve current antitumor therapies.
Collapse
Affiliation(s)
- Stéphane J C Mancini
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM UMR1236, Rennes 1 University, Etablissement Français du Sang Bretagne, Rennes, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France
| | - Karl Balabanian
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Saint-Louis Research Institute, University of Paris, EMiLy, INSERM U1160, Paris, France.,The Organization for Partnerships in Leukemia (OPALE) Carnot Institute, The Organization for Partnerships in Leukemia, Paris, France
| | - Isabelle Corre
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France.,Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), Signaling in Oncogenesis Angiogenesis and Permeability (SOAP), INSERM UMR1232, Centre National de la Recherche scientifique (CNRS) ERL600, Université de Nantes, Nantes, France
| | - Julie Gavard
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France.,Center for Research in Cancerology and Immunology Nantes-Angers (CRCINA), Signaling in Oncogenesis Angiogenesis and Permeability (SOAP), INSERM UMR1232, Centre National de la Recherche scientifique (CNRS) ERL600, Université de Nantes, Nantes, France.,Integrated Center for Oncology, St. Herblain, France
| | - Gwendal Lazennec
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Centre National de la Recherche scientifique (CNRS) UMR9005, SYS2DIAG-ALCEDIAG, Montpellier, France
| | - Marie-Caroline Le Bousse-Kerdilès
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM UMRS-MD1197, Paris-Saclay University, Paul-Brousse Hospital, Villejuif, France
| | - Fawzia Louache
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM UMRS-MD1197, Paris-Saclay University, Paul-Brousse Hospital, Villejuif, France
| | - Véronique Maguer-Satta
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancer Research Center of Lyon (CRCL), CNRS UMR5286, INSERM U1052, Lyon 1 university, Lean Bérard Center, Lyon, France
| | - Nathalie M Mazure
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM U1065, C3M, University of Côte d'Azur (UCA), Nice, France
| | - Fatima Mechta-Grigoriou
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Stress and Cancer Laboratory, Institut Curie, INSERM U830, Paris Sciences et Lettres (PSL) Research University, Team Babelized Ligue Nationale Contre le Cancer (LNCC), Paris, France
| | - Jean-François Peyron
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,INSERM U1065, C3M, University of Côte d'Azur (UCA), Nice, France
| | - Valérie Trichet
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France.,INSERM UMR1238 Phy-Os, Université de Nantes, Nantes, France
| | - Olivier Herault
- Centre National de la Recherche scientifique (CNRS) GDR3697, Micronit "Microenvironment of Tumor Niches", Tours, France.,Cancéropole Grand-Ouest, NET network "Niches and Epigenetics of Tumors", Nantes, France.,The Organization for Partnerships in Leukemia (OPALE) Carnot Institute, The Organization for Partnerships in Leukemia, Paris, France.,Centre National de la Recherche scientifique (CNRS) ERL7001 LNOx, EA7501, Tours University, Tours, France.,Department of Biological Hematology, Tours University Hospital, Tours, France
| |
Collapse
|
48
|
Thrombosis in myeloproliferative neoplasms: A clinical and pathophysiological perspective. THROMBOSIS UPDATE 2021. [DOI: 10.1016/j.tru.2021.100081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
49
|
Comparative Mutational Profiling of Hematopoietic Progenitor Cells and Circulating Endothelial Cells (CECs) in Patients with Primary Myelofibrosis. Cells 2021; 10:cells10102764. [PMID: 34685741 PMCID: PMC8534986 DOI: 10.3390/cells10102764] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 01/08/2023] Open
Abstract
A role of endothelial cells (ECs) in Primary Myelofibrosis (PMF) was supposed since JAK2 mutation was found in endothelial precursor cells (EPCs) and in ECs captured by laser microdissection. By Cell Search method, the circulating endothelial cells (CECs) from 14 PMF patients and 5 healthy controls have been isolated and compared by NGS with CD34+Hematopoietic stem and progenitors cells (HSPCs) for panel of 54 myeloid-associated mutations. PMF patients had higher levels of CECs. No mutation was found in HSPCs and CECs from controls, while CECs from PMF patients presented several somatic mutations. 72% of evaluable patients shared at least one mutation between HSPCs and CECs. 2 patients shared the JAK2 mutation, together with ABL1, IDH1, TET2 and ASXL1, KMT2A, respectively. 6 out of 8 shared only NON MPN-driver mutations: TET2 and NOTCH1 in one case; individual paired mutations in TP53, KIT, SRSF2, NOTCH1 and WT1, in the other cases. In conclusion, 70% of PMF patients shared at least one mutation between HSPCs and CECs. These latter harbored several myeloid-associated mutations, besides JAK2V617F mutation. Our results support a primary involvement of EC in PMF and provide a new methodological approach for further studies exploring the role of the “neoplastic” vascular niche.
Collapse
|
50
|
Sharda AV, Bogue T, Barr A, Mendez LM, Flaumenhaft R, Zwicker JI. Circulating Protein Disulfide Isomerase Is Associated with Increased Risk of Thrombosis in JAK2-Mutated Myeloproliferative Neoplasms. Clin Cancer Res 2021; 27:5708-5717. [PMID: 34400417 DOI: 10.1158/1078-0432.ccr-21-1140] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/07/2021] [Accepted: 08/12/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Thromboembolic events (TE) are the most common complications of myeloproliferative neoplasms (MPN). Clinical parameters, including patient age and mutation status, are used to risk-stratify patients with MPN, but a true biomarker of TE risk is lacking. Protein disulfide isomerase (PDI), an endoplasmic reticulum protein vital for protein folding, also possesses essential extracellular functions, including regulation of thrombus formation. Pharmacologic PDI inhibition prevents thrombus formation, but whether pathologic increases in PDI increase TE risk remains unknown. EXPERIMENTAL DESIGN We evaluated the association of plasma PDI levels and risk of TE in a cohort of patients with MPN with established diagnosis of polycythemia vera (PV) or essential thrombocythemia (ET), compared with healthy controls. Plasma PDI was measured at enrollment and subjects followed prospectively for development of TE. RESULTS A subset of patients, primarily those with JAK2-mutated MPN, had significantly elevated plasma PDI levels as compared with controls. Plasma PDI was functionally active. There was no association between PDI levels and clinical parameters typically used to risk-stratify patients with MPN. The risk of TE was 8-fold greater in those with PDI levels above 2.5 ng/mL. Circulating endothelial cells from JAK2-mutated MPN patients, but not platelets, demonstrated augmented PDI release, suggesting endothelial activation as a source of increased plasma PDI in MPN. CONCLUSIONS The observed association between plasma PDI levels and increased risk of TE in patients with JAK2-mutated MPN has both prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Anish V Sharda
- Division of Hematology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts.,Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical School and Harvard Medical School, Boston, Massachusetts
| | - Thomas Bogue
- Division of Hematology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Alexandra Barr
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical School and Harvard Medical School, Boston, Massachusetts
| | - Lourdes M Mendez
- Division of Hematology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Robert Flaumenhaft
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical School and Harvard Medical School, Boston, Massachusetts
| | - Jeffrey I Zwicker
- Division of Hematology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts. .,Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical School and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|