1
|
Uslu-Biçak İ, Nalçaci M, Sözer S. Targeting PAR1 activation in JAK2V617F-driven philadelphia-negative myeloproliferative neoplasms: Unraveling its role in thrombosis and disease progression. Neoplasia 2025; 63:101153. [PMID: 40088673 PMCID: PMC11951995 DOI: 10.1016/j.neo.2025.101153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Philadelphia chromosome-negative myeloproliferative neoplasms (Ph-MPNs) are clonal disorders marked by high morbidity and mortality, driven by uncontrolled myeloid proliferation from hematopoietic stem/progenitor cells (HSCs) and associated with a significant risk of thrombosis. This study explored the relationship between JAK2V617F and protease-activated receptor 1 (PAR1) by examining PAR1 expression and activation across various hematopoietic stem/progenitor cell (HSPC) subgroups, assessing their contribution to the hypercoagulable state in Ph-MPNs. We investigated the effects of thrombin, a PAR1 antagonist (vorapaxar), and a JAK2 inhibitor (ruxolitinib) on Ph-MPN cells. Mononuclear cells (MNCs) were isolated from Ph-MPN patients (n = 18), cord blood (CB) samples (n = 5) and healthy volunteers (n = 11). Specific subpopulations were sorted and analyzed for PAR1 expression and JAK2V617F status using qRT-PCR. PAR1 expression changes, along with other PAR pathway-related genes, were assessed post-treatment. Our results revealed that most PAR1+ cells (∼95 %) co-expressed CD34+, with a smaller JAK2V617F+ PAR1+ population lacking CD34. PAR1 expression was significantly higher in Ph-MPN MNCs compared to CB (p = 0.0005), particularly in EMP, HSC/EPC, and EPC subsets. Thrombin treatment reduced surface PAR1 expression, while PAR1 antagonist treatment further decrease the expression level. Combined PAR1 antagonist and ruxolitinib treatment significantly downregulated PAR1 expression (p < 0.0001), and several PAR-pathway-related genes were notably downregulated after treatment. This study highlights that elevated PAR1 expression in primitive hematopoietic subpopulations is linked to disease progression and thrombosis in Ph-MPNs, suggesting PAR1 as a potential therapeutic target. Combining PAR1 antagonists with JAK2 inhibitors shows promise in reducing PAR1 expression and mitigating thrombotic events in Ph-MPN patients.
Collapse
Affiliation(s)
- İldeniz Uslu-Biçak
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Türkiye; Institute of Health Sciences, Istanbul University, Istanbul, Türkiye
| | - Meliha Nalçaci
- Department of Internal Medicine, Division of Hematology, Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| | - Selçuk Sözer
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Türkiye.
| |
Collapse
|
2
|
Cvetković M, Arsenović I, Smiljanić M, Sobas M, Bogdanović A, Leković D. Neutrophil-to-lymphocyte and platelet-to-lymphocyte ratio as novel prognostic biomarkers in BCR-ABL negative myeloproliferative neoplasms. Ann Hematol 2024; 103:4545-4556. [PMID: 39331155 DOI: 10.1007/s00277-024-06023-0] [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: 08/22/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024]
Abstract
Higher neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) have been associated with increased risk of thrombosis, cardiovascular mortality, but their role in myeloproliferative neoplasms (MPN) remains unclear. We analyzed NLR and PLR as prognostic markers for thrombosis and overall survival (OS) in the study that included 461 consecutive MPN patients who were diagnosed from 2018 to 2022 at University center. Twenty age-matched patients without hematological disorder were used as controls. NLR and PLR were significantly increased in whole MPN group compared to controls. NLR was highest in PV > PMF > ET (p < 0.001) while PLR was highest in ET > PMF > PV (p < 0.001). Thrombosis occurrence during follow-up correlated with NLR, NLR ≥ 4.5, presence of ≥ 2 CV factors and previous thrombosis. Arterial thrombosis was associated with previous thrombosis, NLR and NLR ≥ 4.5. Similarly in venous thrombosis previous thrombosis was risk factor, together with NLR, NLR ≥ 4.5, PLR, but also secondary malignancy and female gender. In multivariate Cox model, most important factors for thrombosis development during follow-up were previous thrombosis, NLR ≥ 4.5 and PLR ≥ 500; for arterial thrombosis, NLR ≥ 4.5 and previous thrombosis; for venous thrombosis PLR ≥ 500 and previous thrombosis. Patients with pre-PMF had significantly higher NLR than ET patients. In multivariate Cox regression model, most important factors associated with survival were NLR ≥ 4.5 and PLR ≥ 500. This study highlights strong prognostic correlation of NLR ≥ 4.5 and PLR ≥ 500 with development of thrombosis and OS in MPN. Besides previous thrombosis, most important factor associated with development of arterial thrombosis is NLR ≥ 4.5 and for venous PLR ≥ 500. Our results revealed that NLR ≥ 4.5 could be used as additional marker to distinguish ET from prePMF.
Collapse
Affiliation(s)
- Mirjana Cvetković
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovića, Belgrade, 11000, Serbia.
| | - Isidora Arsenović
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovića, Belgrade, 11000, Serbia
| | - Mihailo Smiljanić
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovića, Belgrade, 11000, Serbia
| | - Marta Sobas
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Andrija Bogdanović
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovića, Belgrade, 11000, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Danijela Leković
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovića, Belgrade, 11000, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
3
|
Wang Z, Zou J. Potential biomarkers and immune characteristics for polycythemia vera-related atherosclerosis using bulk RNA and single-cell RNA datasets: a combined comprehensive bioinformatics and machine learning analysis. Front Cardiovasc Med 2024; 11:1426278. [PMID: 39188323 PMCID: PMC11345232 DOI: 10.3389/fcvm.2024.1426278] [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: 05/20/2024] [Accepted: 07/25/2024] [Indexed: 08/28/2024] Open
Abstract
Background Polycythemia vera (PV) is a myeloproliferative disease characterized by significantly higher hemoglobin levels and positivity for JAK2 mutation. Thrombosis is the main risk event of this disease. Atherosclerosis (AS) can markedly increase the risk of arterial thrombosis in patients with PV. The objectives of our study were to identify potential biomarkers for PV-related AS and to explore the molecular biological association between PV and AS. Methods We extracted microarray datasets from the Gene Expression Omnibus (GEO) dataset for PV and AS. Common differentially expressed genes (CGs) were identified by differential expression analysis. Functional enrichment and protein-protein interaction (PPI) networks were constructed from the CG by random forest models using LASSO regression to identify pathogenic genes and their underlying processes in PV-related AS. The expression of potential biomarkers was validated using an external dataset. A diagnostic nomogram was constructed based on potential biomarkers to predict PV-related AS, and its diagnostic performance was assessed using ROC, calibration, and decision curve analyses. Subsequently, we used single-cell gene set enrichment analysis (GSEA) to analyze the immune signaling pathways associated with potential biomarkers. We also performed immune infiltration analysis of AS with "CIBERSORT" and calculated Pearson's correlation coefficients for potential biomarkers and infiltrating immune cells. Finally, we observed the expression of potential biomarkers in immune cells based on the single-cell RNA dataset. Results Fifty-two CGs were identified based on the intersection between up-regulated and down-regulated genes in PV and AS. Most biological processes associated with CGs were cytokines and factors associated with chemotaxis of immune cells. The PPI analysis identified ten hub genes, and of these, CCR1 and MMP9 were selected as potential biomarkers with which to construct a diagnostic model using machine learning methods and external dataset validation. These biomarkers could regulate Toll-like signaling, NOD-like signaling, and chemokine signaling pathways associated with AS. Finally, we determined that these potential biomarkers had a strong correlation with macrophage M0 infiltration. Further, the potential biomarkers were highly expressed in macrophages from patients with AS. Conclusion We identified two CGs (CCR1 and MMP9) as potential biomarkers for PV-related AS and established a diagnostic model based on them. These results may provide insight for future experimental studies for the diagnosis and treatment of PV-related AS.
Collapse
Affiliation(s)
- Ziqing Wang
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jixuan Zou
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
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
|
5
|
Brambilla M, Becchetti A, Rovati GE, Cosentino N, Conti M, Canzano P, Giesen PL, Loffreda A, Bonomi A, Cattaneo M, De Candia E, Podda GM, Trabattoni D, Werba PJ, Campodonico J, Pinna C, Marenzi G, Tremoli E, Camera M. Cell Surface Platelet Tissue Factor Expression: Regulation by P2Y 12 and Link to Residual Platelet Reactivity. Arterioscler Thromb Vasc Biol 2023; 43:2042-2057. [PMID: 37589138 PMCID: PMC10521789 DOI: 10.1161/atvbaha.123.319099] [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: 04/21/2023] [Accepted: 07/26/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND ADP-induced platelet activation leads to cell surface expression of several proteins, including TF (tissue factor). The role of ADP receptors in platelet TF modulation is still unknown. We aimed to assess the (1) involvement of P2Y1 and P2Y12 receptors in ADP-induced TF exposure; (2) modulation of TFpos-platelets in anti-P2Y12-treated patients with coronary artery disease. Based on the obtained results, we revisited the intracellular localization of TF in platelets. METHODS The effects of P2Y1 or P2Y12 antagonists on ADP-induced TF expression and activity were analyzed in vitro by flow cytometry and thrombin generation assay in blood from healthy subjects, P2Y12-/-, and patients with gray platelet syndrome. Ex vivo, P2Y12 inhibition of TF expression by clopidogrel/prasugrel/ticagrelor, assessed by VASP (vasodilator-stimulated phosphoprotein) platelet reactivity index, was investigated in coronary artery disease (n=238). Inhibition of open canalicular system externalization and electron microscopy (TEM) were used for TF localization. RESULTS In blood from healthy subjects, stimulated in vitro by ADP, the percentage of TFpos-platelets (17.3±5.5%) was significantly reduced in a concentration-dependent manner by P2Y12 inhibition only (-81.7±9.5% with 100 nM AR-C69931MX). In coronary artery disease, inhibition of P2Y12 is paralleled by reduction of ADP-induced platelet TF expression (VASP platelet reactivity index: 17.9±11%, 20.9±11.3%, 40.3±13%; TFpos-platelets: 10.5±4.8%, 9.8±5.9%, 13.6±6.3%, in prasugrel/ticagrelor/clopidogrel-treated patients, respectively). Despite this, 15% of clopidogrel good responders had a level of TFpos-platelets similar to the poor-responder group. Indeed, a stronger P2Y12 inhibition (130-fold) is required to inhibit TF than VASP. Thus, a VASP platelet reactivity index <20% (as in prasugrel/ticagrelor-treated patients) identifies patients with TFpos-platelets <20% (92% sensitivity). Finally, colchicine impaired in vitro ADP-induced TF expression but not α-granule release, suggesting that TF is open canalicular system stored as confirmed by TEM and platelet analysis of patients with gray platelet syndrome. CONCLUSIONS Data show that TF expression is regulated by P2Y12 and not P2Y1; P2Y12 antagonists downregulate the percentage of TFpos-platelets. In clopidogrel good-responder patients, assessment of TFpos-platelets highlights those with residual platelet reactivity. TF is stored in open canalicular system, and its membrane exposure upon activation is prevented by colchicine.
Collapse
Affiliation(s)
- Marta Brambilla
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Alessia Becchetti
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Gian Enrico Rovati
- Department of Pharmaceutical Sciences (G.E.R., C.P., M. Camera), Università degli Studi di Milano, Italy
| | - Nicola Cosentino
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Maria Conti
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Paola Canzano
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | | | - Alessia Loffreda
- Experimental Imaging Center, San Raffaele Scientific Institute, Milan, Italy (A.L.)
| | - Alice Bonomi
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Marco Cattaneo
- Unità di Medicina II, ASST Santi Paolo e Carlo, Department of Scienze della Salute (M. Cattaneo, G.M.P.), Università degli Studi di Milano, Italy
| | - Erica De Candia
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy (E.D.C.)
| | - Gian Marco Podda
- Unità di Medicina II, ASST Santi Paolo e Carlo, Department of Scienze della Salute (M. Cattaneo, G.M.P.), Università degli Studi di Milano, Italy
| | - Daniela Trabattoni
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Pablo Josè Werba
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Jeness Campodonico
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | - Christian Pinna
- Department of Pharmaceutical Sciences (G.E.R., C.P., M. Camera), Università degli Studi di Milano, Italy
| | - Giancarlo Marenzi
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
| | | | - Marina Camera
- Centro Cardiologico Monzino IRCCS, Milan, Italy (M.B., A. Becchetti, N.C., M. Conti, P.C., A. Bonomi, D.T., P.J.W., J.C., G.M., M. Camera)
- Department of Pharmaceutical Sciences (G.E.R., C.P., M. Camera), Università degli Studi di Milano, Italy
| |
Collapse
|
6
|
Bekendam RH, Ravid K. Mechanisms of platelet activation in cancer-associated thrombosis: a focus on myeloproliferative neoplasms. Front Cell Dev Biol 2023; 11:1207395. [PMID: 37457287 PMCID: PMC10342211 DOI: 10.3389/fcell.2023.1207395] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/22/2023] [Indexed: 07/18/2023] Open
Abstract
Platelets are anucleate blood cells that play key roles in thrombosis and hemostasis. Platelets are also effector cells in malignancy and are known to home into the microenvironment of cancers. As such, these cells provide central links between the hemostatic system, inflammation and cancer progression. Activation of platelets by cancers has been postulated to contribute to metastasis and progression of local tumor invasion. Similarly, cancer-activated platelets can increase the risk of development of both arterial and venous thrombosis; a major contributor to cancer-associated morbidity. Platelet granules secretion within the tumor environment or the plasma provide a rich source of potential biomarkers for prediction of thrombotic risk or tumor progression. In the case of myeloproliferative neoplasms (MPNs), which are characterized by clonal expansion of myeloid precursors and abnormal function and number of erythrocytes, leukocytes and platelets, patients suffer from thrombotic and hemorrhagic complications. The mechanisms driving this are likely multifactorial but remain poorly understood. Several mouse models developed to recapitulate MPN phenotype with one of the driving mutations, in JAK2 (JAK2V617F) or in calreticulin (CALR) or myeloproliferative leukemia virus oncogene receptor (MPL), have been studied for their thrombotic phenotype. Variability and discrepancies were identified within different disease models of MPN, emphasizing the complexity of increased risk of clotting and bleeding in these pathologies. Here, we review recent literature on the role of platelets in cancer-associated arterial and venous thrombosis and use MPN as case study to illustrate recent advances in experimental models of thrombosis in a malignant phenotype. We address major mechanisms of tumor-platelet communication leading to thrombosis and focus on the role of altered platelets in promoting thrombosis in MPN experimental models and patients with MPN. Recent identification of platelet-derived biomarkers of MPN-associated thrombosis is also reviewed, with potential therapeutic implications.
Collapse
Affiliation(s)
- Roelof H. Bekendam
- Division of Hematology and Hematologic Malignancies, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Katya Ravid
- Department of Medicine and Biochemistry, Whitaker Cardiovascular Institute, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| |
Collapse
|
7
|
Abstract
The Philadelphia-negative myeloproliferative neoplasms (MPNs)-essential thrombocythemia (ET), polycythemia vera (PV) and myelofibrosis (MF), are characterized by a propensity for thrombotic events and variable risks for transformation to MF (for ET and PV) and acute leukemia. Leukocytosis, which serves a minor criterion for the diagnosis of MF, is present in a significant portion of patients with MPNs. The relation and impact of leukocytosis on disease course and outcomes of patients with MPNs has been studied in multiple, large retrospective and prospective studies. Despite this, the association of leukocytosis and thrombosis, fibrosis and leukemic transformation remains unclear. This article details the published investigations regarding the impact of leukocytosis in MPNs and explores the changing role of leukocytosis in disease prognostication as increasing emphasis is placed on molecular and genetic studies.
Collapse
Affiliation(s)
- Alexander Coltoff
- Department of Hematology/Oncology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
8
|
Stuckey R, Ianotto JC, Santoro M, Czyż A, Perez Encinas MM, Gómez-Casares MT, Noya Pereira MS, de Nałęcz AK, Gołos A, Lewandowski K, Szukalski Ł, González-Martín JM, Wróbel T, Sobas MA. Validation of thrombotic risk factors in 1381 patients with essential thrombocythaemia: A multicentre retrospective real-life study. Br J Haematol 2022; 199:86-94. [PMID: 35906782 DOI: 10.1111/bjh.18387] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/07/2022] [Accepted: 07/17/2022] [Indexed: 11/29/2022]
Abstract
Thrombosis and haemorrhage are frequent in patients with essential thrombocythaemia (ET). The 2016 revised International Prognostic Score for Thrombosis in Essential Thrombocythaemia-thrombosis (r-IPSET-t) score stratifies patients into very-low- (VLR), low- (LR), intermediate- (IR) and high-risk (HR) groups. We validated the r-IPSET-t in the biggest population of patients with ET (n = 1381) to date and found it to be a better fit than the earlier IPSET-t score. With an average follow-up of 87.7 months, there were 0.578 thrombotic events/person-year and 0.286 bleeding events/person-year after diagnosis. The 10-year thrombosis-free survival was 88% and 99% for the r-IPSET-t LR and VLR groups (p < 0.001). Cytoreduction was a thrombotic risk factor in younger patients (aged <60 years, hazard ratio 9.49, p = 0.026; aged ≥60 years, hazard ratio 1.04, p = 0.93). In multivariable Cox regression analysis, anti-aggregation after diagnosis was protective for thrombosis (hazard ratio 0.31, p = 0.005) but a risk factor for major bleeding (hazard ratio 10.56, p = 0.021). Of the IPSET-t HR and LR groups, 132/780 and 249/301 were re-classified as LR and VLR respectively (p < 0.001). The European LeukemiaNET (ELN) does not recommend aspirin for VLR patients but in this real-life analysis 83.1% of VLR patients received it. Our results validate the r-IPSET-t score as more predictive for thrombosis than the ELN-recommended IPSET-t and raise concerns about unnecessary cytoreductive and anti-aggregative therapy.
Collapse
Affiliation(s)
- Ruth Stuckey
- Hematology Department, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas, Spain
| | - Jean-Christophe Ianotto
- Service d'Hématologie Clinique, Institut de Cancéro-Hématologie, Centre Hospitalier Régional et Universitaire de Brest, Brest, France
| | - Marco Santoro
- Hematology Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Anna Czyż
- Department of Hematology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Manuel M Perez Encinas
- Hematology Department, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | | | | | | | - Aleksandra Gołos
- Department of Hematology, Institute of Hematology, Warsaw, Poland
| | - Krzysztof Lewandowski
- Department of Hematology and Bone Marrow Transplantation, Poznan University of Medical Sciences, Poznan, Poland
| | - Łukasz Szukalski
- Department of Haematology CM UMK in Bydgoszcz, Nicolaus Copernicus University in Toruń, Toruń, Poland
| | | | - Tomasz Wróbel
- Department of Hematology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Marta Anna Sobas
- Department of Hematology and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
9
|
Ng TL, Tsui DCC, Wang S, Usari T, Patil T, Wilner K, Camidge DR. Association of anticoagulant use with clinical outcomes from crizotinib in ALK- and ROS1-rearranged advanced non-small cell lung cancers: A retrospective analysis of PROFILE 1001. Cancer Med 2022; 11:4422-4429. [PMID: 35510711 PMCID: PMC9741966 DOI: 10.1002/cam4.4789] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 04/11/2022] [Accepted: 04/17/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND ROS1- and ALK-rearranged advanced NSCLCs are associated with increased thromboembolic risk. We hypothesized that a prothrombotic phenotype offers an evolutionary advantage to subsets of these cancers. The impact of this phenotype could alter outcomes from targeted therapy. METHODS In a retrospective analysis of ROS1- and ALK-rearranged NSCLCs treated with crizotinib in a phase 1 trial, we compared progression-free survival (PFS) and objective response rate (ORR) based on the history of anticoagulation use (a possible surrogate of thromboembolism) at baseline (within 90 days before study enrollment) or within 90 days of study treatment. RESULTS Twelve out of 53 (22.6%) ROS1- and 39 out of 153 (25.5%) ALK-rearranged NSCLCs received anticoagulation before or during the trial. Most ROS1 and ALK patients on anticoagulation received low-molecular-weight heparin (75% and 64.1%, respectively). In the ROS1-rearranged group, the median PFS (95% CI) values were 5.1 (4.4-14.4) and 29.0 (16.5-48.8) months, and the ORR values were 41.7% (95% CI: 15.2 to 72.3) and 80.5% (95% CI: 65.1 to 91.2) among those with and without anticoagulation treatment, respectively. In the ALK-rearranged group, the median PFS (95% CI) was 7.1 (5.4-7.7) and 12.0 (9.4-18.3) months, and the ORR was 41% (95% CI: 25.6 to 57.9) and 74.3% (95% CI: 65.3 to 82.1) among those with and without anticoagulation, respectively. CONCLUSIONS Anticoagulation (as a potential surrogate of a prothrombotic subset) in ROS1- and ALK-rearranged NSCLCs may be associated with a lower PFS and ORR to crizotinib. CLINICALTRIAL gov: NCT00585195.
Collapse
Affiliation(s)
- Terry L. Ng
- Division of Medical Oncology, Department of MedicineUniversity of OttawaOttawaCanada
| | - David C. C. Tsui
- Division of Medical Oncology, Department of MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
| | | | | | - Tejas Patil
- Division of Medical Oncology, Department of MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
| | | | - David R. Camidge
- Division of Medical Oncology, Department of MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
| |
Collapse
|
10
|
Tawil N, Rak J. Blood coagulation and cancer genes. Best Pract Res Clin Haematol 2022; 35:101349. [DOI: 10.1016/j.beha.2022.101349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022]
|
11
|
Mantha S, Rak J. Cancer genetic alterations and risk of venous thromboembolism. Thromb Res 2022; 213 Suppl 1:S29-S34. [DOI: 10.1016/j.thromres.2021.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/02/2021] [Accepted: 12/09/2021] [Indexed: 10/18/2022]
|
12
|
Manoharan A, Gemmell R, Cavanaugh L, Shadood N. Thrombosis in Myeloproliferative Neoplasms: A Single Center Experience of Using Whole Blood Platelet Aggregation Studies for Risk Assessment and Thromboprophylaxis. Clin Appl Thromb Hemost 2022; 28:10760296221117482. [PMID: 35898172 PMCID: PMC9340402 DOI: 10.1177/10760296221117482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Thromboembolic complications are the most common causes of morbidity and
mortality in patients with Philadelphia chromosome-negative myeloproliferative
neoplasms (MPN); and prevention of these complications remains a significant
clinical challenge. Effective thromboprophylaxis in MPN patients generally
requires use of anti-platelet therapy, commonly aspirin; however, there are no
standardized or universally accepted guidelines regarding the dose of aspirin.
This study evaluates the usefulness of whole blood platelet aggregation (WBPA)
studies to identify patients at risk for thrombosis and to achieve safe and
effective long term thromboprophylaxis. One hundred and thirty-two consecutive
patients were enrolled into this study. WBPA studies were performed at diagnosis
in 125 patients to identify those with platelet hyperactivity (deemed to be at
risk for thrombosis) and repeated 4 weeks after commencement of anti-platelet
therapy to ascertain the efficacy. In patients with incomplete drug effect,
treatment was revised and the study repeated until optimum effect was achieved.
Results of the WBPA studies and anti-platelet therapy requirements were
correlated with the underlying driver mutations and various international
prognostic score of thrombosis for essential thrombocythemia (IPSET- Thrombosis)
sub-groups. WBPA studies showed varying degrees of platelet hyper-activity in
115 patients. Based on these results, the patients were commenced on
anti-platelet therapy comprising aspirin (dose ranging from 100mg twice or
thrice weekly to 400mg daily) and clopidogrel (75mg daily) alone or in
combination with aspirin or odorless garlic. None of the patients developed
thrombosis during the follow up period ranging from 1-23 years (median 8yrs),
while on the prescribed, individualized anti-platelet therapy. No significant
differences were noted in terms of aspirin dose requirements between the JAK-2
positive and CALR or MPL positive patients, and, among the four IPSET-Thrombosis
sub-groups. Patients with normal (9) or hypo (1) – activity were not given any
anti-platelet therapy at diagnosis.
Collapse
Affiliation(s)
- Arumugam Manoharan
- Southern Sydney Haematology, St George Private Hosipital, Sydney, Australia.,Faculty of Health, Medicine & Applied Sciences, 8691University of Wollongong, Wollongong, New South Wales, Australia
| | - Rosalie Gemmell
- Department of Haematology, ST. George Hospital, Sydney, Australia
| | - Lauren Cavanaugh
- Department of Haematology, ST. George Hospital, Sydney, Australia
| | - Noor Shadood
- Department of Haematology, ST. George Hospital, Sydney, Australia
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Lucchesi A, Napolitano R, Bochicchio MT, Giordano G, Napolitano M. Platelets Contribution to Thrombin Generation in Philadelphia-Negative Myeloproliferative Neoplasms: The "Circulating Wound" Model. Int J Mol Sci 2021; 22:ijms222111343. [PMID: 34768772 PMCID: PMC8583863 DOI: 10.3390/ijms222111343] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
Current cytoreductive and antithrombotic strategies in MPNs are mostly based on cell counts and on patient's demographic and clinical history. Despite the numerous studies conducted on platelet function and on the role of plasma factors, an accurate and reliable method to dynamically quantify the hypercoagulability states of these conditions is not yet part of clinical practice. Starting from our experience, and after having sifted through the literature, we propose an in-depth narrative report on the contribution of the clonal platelets of MPNs-rich in tissue factor (TF)-in promoting a perpetual procoagulant mechanism. The whole process results in an unbalanced generation of thrombin and is self-maintained by Protease Activated Receptors (PARs). We chose to define this model as a "circulating wound", as it indisputably links the coagulation, inflammation, and fibrotic progression of the disease, in analogy with what happens in some solid tumours. The platelet contribution to thrombin generation results in triggering a vicious circle supported by the PARs/TGF-beta axis. PAR antagonists could therefore be a good option for target therapy, both to contain the risk of vascular events and to slow the progression of the disease towards end-stage forms. Both the new and old strategies, however, will require tools capable of measuring procoagulant or prohaemorrhagic states in a more extensive and dynamic way to favour a less empirical management of MPNs and their potential clinical complications.
Collapse
MESH Headings
- Animals
- Biological Assay
- Blood Platelets/metabolism
- Humans
- Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/drug therapy
- Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/metabolism
- Models, Biological
- Receptors, Fibrinogen/metabolism
- Thrombin/antagonists & inhibitors
- Thrombin/biosynthesis
- Thrombophilia/physiopathology
Collapse
Affiliation(s)
- Alessandro Lucchesi
- Hematology Unit, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Roberta Napolitano
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
- Correspondence:
| | - Maria Teresa Bochicchio
- Biosciences Laboratory, IRCCS Istituto Scientifico Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Giulio Giordano
- Internal Medicine Division, Hematology Service, Regional Hospital “A. Cardarelli”, 86100 Campobasso, Italy;
| | - Mariasanta Napolitano
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties and Infectious Disease Unit, University Hospital “P. Giaccone”, 90127 Palermo, Italy;
| |
Collapse
|
16
|
Baldini C, Moriconi FR, Galimberti S, Libby P, De Caterina R. The JAK-STAT pathway: an emerging target for cardiovascular disease in rheumatoid arthritis and myeloproliferative neoplasms. Eur Heart J 2021; 42:4389-4400. [PMID: 34343257 DOI: 10.1093/eurheartj/ehab447] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 05/21/2021] [Accepted: 07/31/2021] [Indexed: 01/07/2023] Open
Abstract
Inflammation contributes centrally to cardiovascular diseases, and anti-inflammatory treatments can reduce cardiovascular events. The JAK-STAT pathway is an emerging target in inflammation, mainly in rheumatoid arthritis (RA) and chronic myeloproliferative neoplasms (MPNs), disorders that heighten cardiovascular risk. The aim of this study was to review the international literature on the relationship between dysregulation of the JAK-STAT pathway in RA/MPNs and cardiovascular risk and on the potential cardiovascular effects of JAK-STAT inhibitors. The JAK-STAT pathway sustains inflammatory and thrombotic events in autoimmune disorders such as RA and MPNs. Here, an imbalance exists between pro- and anti-inflammatory cytokines [increased levels of interleukin (IL)-6, IL-1-β, tumour necrosis factor-α, decreased levels of IL-10] and the over-expression of some prothrombotic proteins, such as protein kinase Cε, on the surface of activated platelets. This pathway also operates in atherosclerotic cardiovascular disease. JAK-STAT inhibitors may reduce cardiovascular events and related deaths in such conditions, but the potential of these agents requires more studies, especially with regard to cardiovascular safety, and particularly for potential prothrombotic effects. JAK-STAT inhibitors merit consideration to curb heightened cardiovascular risk in patients with RA and MPNs, with rigorous assessment of the potential benefits and risks.
Collapse
Affiliation(s)
- Chiara Baldini
- Division of Rheumatology, University of Pisa and Pisa University Hospital, Via Paradisa, 2, Pisa 56124, Italy
| | - Francesca Romana Moriconi
- Division of Rheumatology, University of Pisa and Pisa University Hospital, Via Paradisa, 2, Pisa 56124, Italy.,Division of Cardiology, University of Pisa and Pisa University Hospital, Via Paradisa, 2, Pisa 56124, Italy
| | - Sara Galimberti
- Division of Hematology, University of Pisa and Pisa University Hospital, Via Paradisa, 2, Pisa 56124, Italy
| | - Peter Libby
- Cardiovascular Division, Brigham and Women's Hospital-Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - Raffaele De Caterina
- Division of Cardiology, University of Pisa and Pisa University Hospital, Via Paradisa, 2, Pisa 56124, Italy
| |
Collapse
|
17
|
Lambert J, Saliba J, Calderon C, Sii-Felice K, Salma M, Edmond V, Alvarez JC, Delord M, Marty C, Plo I, Kiladjian JJ, Soler E, Vainchenker W, Villeval JL, Rousselot P, Prost S. PPARγ agonists promote the resolution of myelofibrosis in preclinical models. J Clin Invest 2021; 131:136713. [PMID: 33914703 DOI: 10.1172/jci136713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/22/2021] [Indexed: 12/14/2022] Open
Abstract
Myelofibrosis (MF) is a non-BCR-ABL myeloproliferative neoplasm associated with poor outcomes. Current treatment has little effect on the natural history of the disease. MF results from complex interactions between (a) the malignant clone, (b) an inflammatory context, and (c) remodeling of the bone marrow (BM) microenvironment. Each of these points is a potential target of PPARγ activation. Here, we demonstrated the therapeutic potential of PPARγ agonists in resolving MF in 3 mouse models. We showed that PPARγ agonists reduce myeloproliferation, modulate inflammation, and protect the BM stroma in vitro and ex vivo. Activation of PPARγ constitutes a relevant therapeutic target in MF, and our data support the possibility of using PPARγ agonists in clinical practice.
Collapse
Affiliation(s)
- Juliette Lambert
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Department of Hematology and Oncology, Centre Hospitalier de Versailles, Le Chesnay, France.,Opale Carnot Institute, Paris, France
| | - Joseph Saliba
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Carolina Calderon
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Opale Carnot Institute, Paris, France
| | - Karine Sii-Felice
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Mohammad Salma
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Université de Paris, Laboratory of Excellence GR-Ex, Paris, France
| | - Valérie Edmond
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Jean-Claude Alvarez
- Département de Pharmacologie-Toxicologie, Hôpitaux Universitaires Paris Ile-de-France Ouest, AP-HP, Hôpital Raymond-Poincaré, FHU Sepsis, Garches, France.,MasSpecLab, Plateforme de spectrométrie de masse, INSERM U-1173, Université Paris-Saclay (Versailles Saint-Quentin-en-Yvelines), UFR des sciences de la santé, Montigny-le-Bretonneux, France
| | - Marc Delord
- Recherche Clinique, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Caroline Marty
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Isabelle Plo
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Jean-Jacques Kiladjian
- Opale Carnot Institute, Paris, France.,Université de Paris, AP-HP, Hôpital Saint-Louis, Centre d'Investigations Cliniques CIC 1427, INSERM, Paris, France
| | - Eric Soler
- Institut de Génétique Moléculaire de Montpellier, University of Montpellier, CNRS, Montpellier, France.,Université de Paris, Laboratory of Excellence GR-Ex, Paris, France
| | | | - Jean-Luc Villeval
- INSERM, UMR1287, Université Paris-Saclay, Gustave Roussy, Villejuif, France
| | - Philippe Rousselot
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Department of Hematology and Oncology, Centre Hospitalier de Versailles, Le Chesnay, France.,Opale Carnot Institute, Paris, France.,Université Paris-Saclay (Versailles Saint-Quentin-en-Yvelines), UFR des sciences de la santé, Montigny-le-Bretonneux, France
| | - Stéphane Prost
- Division of Innovative Therapies, CEA/DRF/François Jacob Biology Institute, UMR1184 IMVA-HB/IDMIT, Université Paris-Saclay, Fontenay-aux-Roses, France.,Opale Carnot Institute, Paris, France
| |
Collapse
|
18
|
Fisher DAC, Fowles JS, Zhou A, Oh ST. Inflammatory Pathophysiology as a Contributor to Myeloproliferative Neoplasms. Front Immunol 2021; 12:683401. [PMID: 34140953 PMCID: PMC8204249 DOI: 10.3389/fimmu.2021.683401] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Myeloid neoplasms, including acute myeloid leukemia (AML), myeloproliferative neoplasms (MPNs), and myelodysplastic syndromes (MDS), feature clonal dominance and remodeling of the bone marrow niche in a manner that promotes malignant over non-malignant hematopoiesis. This take-over of hematopoiesis by the malignant clone is hypothesized to include hyperactivation of inflammatory signaling and overproduction of inflammatory cytokines. In the Ph-negative MPNs, inflammatory cytokines are considered to be responsible for a highly deleterious pathophysiologic process: the phenotypic transformation of polycythemia vera (PV) or essential thrombocythemia (ET) to secondary myelofibrosis (MF), and the equivalent emergence of primary myelofibrosis (PMF). Bone marrow fibrosis itself is thought to be mediated heavily by the cytokine TGF-β, and possibly other cytokines produced as a result of hyperactivated JAK2 kinase in the malignant clone. MF also features extramedullary hematopoiesis and progression to bone marrow failure, both of which may be mediated in part by responses to cytokines. In MF, elevated levels of individual cytokines in plasma are adverse prognostic indicators: elevated IL-8/CXCL8, in particular, predicts risk of transformation of MF to secondary AML (sAML). Tumor necrosis factor (TNF, also known as TNFα), may underlie malignant clonal dominance, based on results from mouse models. Human PV and ET, as well as MF, harbor overproduction of multiple cytokines, above what is observed in normal aging, which can lead to cellular signaling abnormalities separate from those directly mediated by hyperactivated JAK2 or MPL kinases. Evidence that NFκB pathway signaling is frequently hyperactivated in a pan-hematopoietic pattern in MPNs, including in cells outside the malignant clone, emphasizes that MPNs are pan-hematopoietic diseases, which remodel the bone marrow milieu to favor persistence of the malignancy. Clinical evidence that JAK2 inhibition by ruxolitinib in MF neither reliably reduces malignant clonal burden nor eliminates cytokine elevations, suggests targeting cytokine mediated signaling as a therapeutic strategy, which is being pursued in new clinical trials. Greater knowledge of inflammatory pathophysiology in MPNs can therefore contribute to the development of more effective therapy.
Collapse
Affiliation(s)
- Daniel Arthur Corpuz Fisher
- Divisions of Hematology & Oncology, School of Medicine, Washington University in St. Louis, Saint Louis, MO, United States
| | - Jared Scott Fowles
- Divisions of Hematology & Oncology, School of Medicine, Washington University in St. Louis, Saint Louis, MO, United States
| | - Amy Zhou
- Divisions of Hematology & Oncology, School of Medicine, Washington University in St. Louis, Saint Louis, MO, United States
| | - Stephen Tracy Oh
- Divisions of Hematology & Oncology, School of Medicine, Washington University in St. Louis, Saint Louis, MO, United States
| |
Collapse
|
19
|
The pathobiology of thrombosis, microvascular disease, and hemorrhage in the myeloproliferative neoplasms. Blood 2021; 137:2152-2160. [PMID: 33649757 DOI: 10.1182/blood.2020008109] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 01/11/2021] [Indexed: 12/19/2022] Open
Abstract
Thrombotic, vascular, and bleeding complications are the most common causes of morbidity and mortality in the Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs). In these disorders, circulating red cells, leukocytes, and platelets, as well as some vascular endothelial cells, each have abnormalities that are cell-intrinsic to the MPN driver mutations they harbor (eg, JAK2 V617F). When these cells are activated in the MPNs, their interactions with each other create a highly proadhesive and prothrombotic milieu in the circulation that predisposes patients with MPN to venous, arterial, and microvascular thrombosis and occlusive disease. Bleeding problems in the MPNs are caused by the MPN blood cell-initiated development of acquired von Willebrand disease. The inflammatory state created by MPN stem cells in their microenvironment extends systemically to amplify the clinical thrombotic tendency and, at the same time, preferentially promote further MPN stem cell clonal expansion, thereby generating a vicious cycle that favors a prothrombotic state in these diseases.
Collapse
|
20
|
Reeves BN, Beckman JD. Novel Pathophysiological Mechanisms of Thrombosis in Myeloproliferative Neoplasms. Curr Hematol Malig Rep 2021; 16:304-313. [PMID: 33876389 DOI: 10.1007/s11899-021-00630-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW Thrombosis remains a leading cause of morbidity and mortality in BCR/ABL negative myeloproliferative neoplasms (MPN). Circulating blood cells are both increased in quantity and qualitatively abnormal in MPN, resulting in an increased thrombotic risk. Herein, we review recently elucidated mechanisms of MPN thrombosis and discuss implications of drugs currently under investigation for MPN. RECENT FINDINGS Recent studies highlight that in JAK2V617F granulocytes and platelets, thrombo-inflammatory genes are upregulated. Furthermore, in JAK2V617F granulocytes, protein expression of integrin CD11b, tissue factor, and leukocyte alkaline phosphatase are all increased. Overall, myeloid cells, namely neutrophils, may contribute in several ways, such as through increased adhesion via β1 integrin binding to VCAM1, increased infiltration, and enhanced inducibility to extrude neutrophil extracellular traps. Non-myeloid inflammatory cells may also contribute via secretion of cytokines. With regard to red blood cells, number, rigidity, adhesion, and generation of microvesicles may lead to increased vascular resistance as well as increased cell-cell interactions that promote rolling and adhesion. Platelets may also contribute in a similar fashion. Lastly, the vasculature is also increasingly appreciated, as several studies have demonstrated increased endothelial expression of pro-coagulant and pro-adhesive proteins, such as von Willebrand factor or P-selectin in JAK2V617F endothelial cells. With the advent of molecular diagnostics, MPN therapeutics are advancing beyond cytoreduction. Our increased understanding of pro-inflammatory and thrombotic pathophysiology in MPN provides a rational basis for evaluation of in-development MPN therapeutics to reduce thrombosis.
Collapse
Affiliation(s)
- Brandi N Reeves
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
- Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joan D Beckman
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
- Department of Medicine, Division of Hematology, Oncology and Transplantation, University of Minnesota, 420 Delaware St. SE, MMC 480, Minneapolis, MN, 55455, USA.
| |
Collapse
|
21
|
Guy A, Poisson J, James C. Pathogenesis of cardiovascular events in BCR-ABL1-negative myeloproliferative neoplasms. Leukemia 2021; 35:935-955. [PMID: 33658660 DOI: 10.1038/s41375-021-01170-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/11/2021] [Accepted: 01/28/2021] [Indexed: 02/07/2023]
Abstract
Thrombosis, both in arterial and venous territories, is the major complication of myeloproliferative neoplasms and is responsible for a high rate of morbidity and mortality. The currently accepted risk factors are an age over 60 years and a history of thrombosis. However, many complex mechanisms contribute to this increased prothrombotic risk, with involvement of all blood cell types, plasmatic factors, and endothelial cells. Besides, some cardiovascular events may originate from arterial vasospasm that could contribute to thrombotic complications. In this review, we discuss recent results obtained in mouse models in the light of data obtained from clinical studies. We emphasize on actors of thrombosis that are currently not targeted with current therapeutics but could be promising targets, i.e, neutrophil extracellular traps and vascular reactivity.
Collapse
Affiliation(s)
- Alexandre Guy
- UMR1034, Inserm, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France.,Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Johanne Poisson
- Inserm, Centre de recherche sur l'inflammation, University of Paris, Paris, France.,Geriatrics Department, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Chloe James
- UMR1034, Inserm, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France. .,Laboratoire d'Hématologie, CHU de Bordeaux, Pessac, France.
| |
Collapse
|
22
|
Low-Risk Essential Thrombocythemia: A Comprehensive Review. Hemasphere 2021; 5:e521. [PMID: 33880431 PMCID: PMC8051994 DOI: 10.1097/hs9.0000000000000521] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/13/2020] [Indexed: 12/25/2022] Open
Abstract
Essential thrombocythemia (ET) is a chronic myeloproliferative neoplasm characterized by a persistently elevated platelet count in the absence of a secondary cause. The clinical consequences of uncontrolled thrombocytosis can include both thrombosis and hemorrhage. Patients with features conferring a “high risk” of vascular events benefit from reduction of the platelet count through cytoreductive therapy. The management of patients who lack such high-risk features has until recently been less well defined, but it is now apparent that many require minimal or even no intervention. In this review, we discuss the diagnostic pathway for younger patients with unexplained thrombocytosis, including screening molecular investigations, the role of bone marrow biopsy, and investigations in those patients negative for the classic myeloproliferative neoplasm driver mutations (JAK2, CALR, MPL). We discuss conventional and novel risk stratification methods in essential thrombocythemia and how these can be best applied in clinical practice, particularly in the era of more comprehensive genomic testing. The treatment approach for “low risk” patients is discussed including antiplatelets and the options for cytoreductive therapy, if indicated, together with areas of clinical need for future study.
Collapse
|
23
|
Polokhov DM, Ershov NM, Ignatova AA, Ponomarenko EA, Gaskova MV, Zharkov PA, Fedorova DV, Poletaev AV, Seregina EA, Novichkova GA, Smetanina NS, Panteleev MA. Platelet function and blood coagulation system status in childhood essential thrombocythemia. Platelets 2020; 31:1001-1011. [PMID: 31856623 DOI: 10.1080/09537104.2019.1704710] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Childhood essential thrombocythemia (ET) is a rare chronic myeloproliferative disorder. The quality of life of ET patients may decrease as a result of ischemic and hemorrhagic complications of unclear origin. Our goal was to characterize the hemostatic system in children with ET. We genotyped and investigated blood samples from 20 children with ET in a prospective case series study using platelet aggregation, functional flow cytometry (FC) assay and standard clotting assays. Three children had a JAK2V617F mutation, 4 had mutations in CALR and 13 were triple-negative. Myelofibrosis in stage 1-2 was detected in 3 children. Three patients had bleeding episodes and seven had ischemic events. Aggregation in response to collagen, adenosine diphosphate, and ristomycin was decreased in all patients. In FC, significant changes in the whole patient group compared to the healthy children control group were decrease in the resting forward scatter and PAC1 binding (activated GPIIb/IIIa) level. For the activated platelets, dense granules release (by mepacrine), PAC1, and GPIIb/IIIa levels were significantly decreased. GPIb/V/IX, P-selectin, and phosphatidylserine levels manifested only moderate differences. Forward and side scatter changes in response to stimulation (representing shape change) and dense granules release were significantly lower in the 3 patients with bleeding than in the 17 patients without hemorrhage. Activated partial thromboplastin time was slightly prolonged, prothrombin index was slightly shortened and thrombin time was normal, while fibrinogen was mildly decreased in the ET patients. It could be concluded that the observed platelet function defects could be related to bleeding in ET, and be potentially used as a marker.
Collapse
Affiliation(s)
- Dmitrii M Polokhov
- Oncology and Immunology, Federal Research and Clinical Centre of Pediatric Hematology , Moscow, Russian Federation, Russia
| | - Nikolay M Ershov
- Oncology and Immunology, Federal Research and Clinical Centre of Pediatric Hematology , Moscow, Russian Federation, Russia
| | - Anastasia A Ignatova
- Oncology and Immunology, Federal Research and Clinical Centre of Pediatric Hematology , Moscow, Russian Federation, Russia
| | - Evgeniya A Ponomarenko
- Oncology and Immunology, Federal Research and Clinical Centre of Pediatric Hematology , Moscow, Russian Federation, Russia
| | - Marina V Gaskova
- Oncology and Immunology, Federal Research and Clinical Centre of Pediatric Hematology , Moscow, Russian Federation, Russia
| | - Pavel A Zharkov
- Oncology and Immunology, Federal Research and Clinical Centre of Pediatric Hematology , Moscow, Russian Federation, Russia
| | - Daria V Fedorova
- Oncology and Immunology, Federal Research and Clinical Centre of Pediatric Hematology , Moscow, Russian Federation, Russia
| | - Alexandr V Poletaev
- Oncology and Immunology, Federal Research and Clinical Centre of Pediatric Hematology , Moscow, Russian Federation, Russia
| | - Elena A Seregina
- Oncology and Immunology, Federal Research and Clinical Centre of Pediatric Hematology , Moscow, Russian Federation, Russia
| | - Galina A Novichkova
- Oncology and Immunology, Federal Research and Clinical Centre of Pediatric Hematology , Moscow, Russian Federation, Russia
| | - Nataliya S Smetanina
- Oncology and Immunology, Federal Research and Clinical Centre of Pediatric Hematology , Moscow, Russian Federation, Russia
| | - Mikhail A Panteleev
- Oncology and Immunology, Federal Research and Clinical Centre of Pediatric Hematology , Moscow, Russian Federation, Russia.,Center for Theoretical Problems of Physicochemical Pharmacology , Moscow, Russia.,Faculty of Physics, Moscow State University , Moscow, Russia.,Faculty of Biological and Medical Physics, Moscow Institute of Physics and Technology , Dolgoprudny, Russia
| |
Collapse
|
24
|
Iurlo A, Cattaneo D, Bucelli C, Baldini L. New Perspectives on Polycythemia Vera: From Diagnosis to Therapy. Int J Mol Sci 2020; 21:ijms21165805. [PMID: 32823537 PMCID: PMC7461104 DOI: 10.3390/ijms21165805] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
Polycythemia vera (PV) is mainly characterized by elevated blood cell counts, thrombotic as well as hemorrhagic predisposition, a variety of symptoms, and cumulative risks of fibrotic progression and/or leukemic evolution over time. Major changes to its diagnostic criteria were made in the 2016 revision of the World Health Organization (WHO) classification, with both hemoglobin and hematocrit diagnostic thresholds lowered to 16.5 g/dL and 49% for men, and 16 g/dL and 48% for women, respectively. The main reason leading to these changes was represented by the recognition of a new entity, namely the so-called “masked PV”, as individuals suffering from this condition have a worse outcome, possibly owing to missed or delayed diagnoses and lower intensity of treatment. Thrombotic risk stratification is of crucial importance to evaluate patients’ prognosis at diagnosis. Currently, patients are stratified into a low-risk group, in the case of younger age (<60 years) and no previous thromboses, and a high-risk group, in the case of patients older than 60 years and/or with a previous thrombotic complication. Furthermore, even though they have not yet been formally included in a scoring system, generic cardiovascular risk factors, particularly hypertension, smoking, and leukocytosis, contribute to the thrombotic overall risk. In the absence of agents proven to modify its natural history and prevent progression, PV management has primarily been focused on minimizing the thrombotic risk, representing the main cause of morbidity and mortality. When cytoreduction is necessary, conventional therapies include hydroxyurea as a first-line treatment and ruxolitinib and interferon in resistant/intolerant cases. Each therapy, however, is burdened by specific drawbacks, underlying the need for improved strategies. Currently, the therapeutic landscape for PV is still expanding, and includes several molecules that are under investigation, like long-acting pegylated interferon alpha-2b, histone deacetylase inhibitors, and murine double minute 2 (MDM2) inhibitors.
Collapse
Affiliation(s)
- Alessandra Iurlo
- Hematology Division, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (D.C.); (C.B.); (L.B.)
- Correspondence: ; Tel.: +39-02-5503-3463; Fax: +39-02-5503-4105
| | - Daniele Cattaneo
- Hematology Division, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (D.C.); (C.B.); (L.B.)
| | - Cristina Bucelli
- Hematology Division, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (D.C.); (C.B.); (L.B.)
| | - Luca Baldini
- Hematology Division, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (D.C.); (C.B.); (L.B.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
25
|
Essential thrombocythemia: a hemostatic view of thrombogenic risk factors and prognosis. Mol Biol Rep 2020; 47:4767-4778. [PMID: 32472297 DOI: 10.1007/s11033-020-05536-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 05/15/2020] [Indexed: 01/03/2023]
Abstract
Essential thrombocythemia (ET) is a classical myeloproliferative neoplasm that is susceptible to hypercoagulable state due to impaired hemostatic system, so that thrombotic complications are the leading cause of mortality in ET patients. The content used in this article has been obtained by the PubMed database and Google Scholar search engine from English-language articles (2000-2019) using the following keywords: "Essential thrombocythemia," "Thrombosis," "Risk factors" and "Hemostasis. In this neoplasm, the count and activity of cells such as platelets, leukocytes, endothelial cells, as well as erythrocytes are increased, which can increase the risk of thrombosis through rising intercellular interactions, expression of surface markers, and stimulation of platelet aggregation. In addition to these factors, genetic polymorphisms in hematopoietic stem cells (HSCs), including mutations in JAK2, CALR, MPL, or genetic abnormalities in other genes associated with the hemostatic system may be associated with increased risk of thrombotic events. Moreover, disruption of coagulant factors can pave the way for thrombogeneration. Therefore, the identification of markers related to cell activation, genetic abnormalities, or alternation in the coagulant system can be used together as diagnostic and prognostic markers for the occurrence of thrombosis among ET patients. Thus, because thrombotic complications are the main factors of mortality in ET patients, a hemostatic viewpoint and risk assessment of cellular, genetic, and coagulation factors can have prognostic value and contribute to the choice of effective treatment and prevention of thrombosis.
Collapse
|
26
|
Hauschner H, Bokstad Horev M, Misgav M, Nagar M, Seligsohn U, Rosenberg N, Koren‐Michowitz M. Platelets from Calreticulin mutated essential thrombocythemia patients are less reactive than JAK2 V617F mutated platelets. Am J Hematol 2020; 95:379-386. [PMID: 31868244 DOI: 10.1002/ajh.25713] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/13/2019] [Accepted: 12/20/2019] [Indexed: 01/09/2023]
Abstract
Both JAK2V617F and calreticulin (CALR) mutated essential thrombocythemia (ET) patients have different clinical characteristics, with lower thrombosis risk in patients with CALR mutations. To elucidate the mechanism for this lower risk we studied platelet function in ET patients with either JAK2V617F or a CALR mutation. Platelet activation state was similar in ET and healthy controls at baseline using P-selectin and PAC1 flow cytometry analysis. However, CALR mutated platelets were significantly less activated following ADP stimulation, compared to control or JAK2 mutated platelets (P < .001). In live-cell imaging of platelet attachment to immobilized fibrinogen by Interference Reflection Microscopy (IRM), the number of attached CALR mutated platelets was lower compared to control and JAK2 mutated platelets, with lower fractions of platelets achieving the fully spread state (90%, 78% and 54% of adherent cells for control, JAK2 and CALR mutated subjects, respectively). Compared to controls, ET patients, regardless of the mutation type, had increased numbers of immature platelets (IP) and leukocyte platelet aggregates (LPA), as well as plasma sP-selectin. These were all correlated with the platelet count and not to the state of platelet activation. We also found that intracellular free Ca2+ was increased in resting ET compared to control platelets. Note, CALR had a more dispersed localization in activated ET platelets compared to healthy controls, and mutated CALR interact physically with TpoR in CALR mutated platelets. We hypothesize that defects in platelet activation and spreading in CALR mutated patients can explain, at least in part, the lower thrombotic tendency in CALR mutated ET patients.
Collapse
Affiliation(s)
- Hagit Hauschner
- Scientific Equipment Center, The Mina & Everard Goodman Faculty of Life Sciences Bar‐Ilan University Ramat‐Gan Israel
| | - Melanie Bokstad Horev
- Department of Molecular Cell Biology The Weizmann Institute of Science Rehovot Israel
| | - Modi Misgav
- Sackler Faculty of Medicine Tel Aviv University Tel‐Aviv Israel
- The Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center Tel Hashomer Israel
| | - Meital Nagar
- Molecular Hematology Laboratory Sheba Medical Center Tel Hashomer Israel
| | - Uri Seligsohn
- Sackler Faculty of Medicine Tel Aviv University Tel‐Aviv Israel
- The Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center Tel Hashomer Israel
| | - Nurit Rosenberg
- Sackler Faculty of Medicine Tel Aviv University Tel‐Aviv Israel
- The Amalia Biron Research Institute of Thrombosis and Hemostasis, Sheba Medical Center Tel Hashomer Israel
| | - Maya Koren‐Michowitz
- Sackler Faculty of Medicine Tel Aviv University Tel‐Aviv Israel
- Department of Hematology Shamir Medical Center (Assaf Harofeh) Zerifin Israel
| |
Collapse
|
27
|
Mornet C, Galinat H, Mingant F, Ianotto JC, Lippert E. [Thrombosis and platelet dysfunction in myeloproliferative neoplasms]. Rev Med Interne 2020; 41:319-324. [PMID: 32008800 DOI: 10.1016/j.revmed.2019.12.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 12/23/2019] [Indexed: 12/15/2022]
Abstract
Myeloproliferative neoplasms are acquired hematological malignancies, mainly affecting the adult and whose morbidity and mortality stems from haemostasis disorders. The most frequently encountered complications include thrombosis, affecting preferentially the arterial territory, but also atypical locations such as splanchnic vein thrombosis. The pathophysiology of these thromboses is complex and involves different actors: blood cells, endothelium and flow conditions. Numerous studies have been conducted to identify risk factors for thrombosis. To date, only two risk factors have been validated through prospective studies (age over 60 years old, history of thrombotic events) and allow classification of patients as "low risk" and "high risk" as the basis for current treatment recommendations. Haemorrhagic manifestations, less frequent than thrombosis, are mainly related to an alteration of primary haemostasis and are therefore manifested by mucocutaneous bleeding. In these patients, platelet dysfunctions and/or acquired Willebrand syndromes can be found. The pathophysiology of thrombosis and platelet dysfunction during myeloproliferative neoplasms remains to date partially unknown. In this review, we offer to focus on physiopathological mechanisms as well as the latest advances in their understanding.
Collapse
Affiliation(s)
- C Mornet
- Hématologie biologique, CHU de Brest, Brest, France
| | - H Galinat
- Hématologie biologique, CHU de Brest, Brest, France
| | - F Mingant
- Hématologie biologique, CHU de Brest, Brest, France
| | - J C Ianotto
- Hématologie clinique et thérapie cellulaire, CHU de Brest, Brest, France
| | - E Lippert
- Hématologie biologique, CHU de Brest, Brest, France; Inserm, EFS, UMR 1078, GGB, Université Brest, Brest, France.
| |
Collapse
|
28
|
Pedersen RK, Andersen M, Knudsen TA, Sajid Z, Gudmand-Hoeyer J, Dam MJB, Skov V, Kjaer L, Ellervik C, Larsen TS, Hansen D, Pallisgaard N, Hasselbalch HC, Ottesen JT. Data-driven analysis of JAK2V617F kinetics during interferon-alpha2 treatment of patients with polycythemia vera and related neoplasms. Cancer Med 2020; 9:2039-2051. [PMID: 31991066 PMCID: PMC7064092 DOI: 10.1002/cam4.2741] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 12/22/2022] Open
Abstract
Treatment with PEGylated interferon-alpha2 (IFN) of patients with essential thrombocythemia and polycythemia vera induces major molecular remissions with a reduction in the JAK2V617F allele burden to undetectable levels in a subset of patients. A favorable response to IFN has been argued to depend upon the tumor burden, implying that institution of treatment with IFN should be as early as possible after the diagnosis. However, evidence for this statement is not available. We present a thorough analysis of unique serial JAK2V617F measurements in 66 IFN-treated patients and in 6 untreated patients. Without IFN treatment, the JAK2V617F allele burden increased exponentially with a period of doubling of 1.4 year. During monotherapy with IFN, the JAK2V617F allele burden decreased mono- or bi-exponentially for 33 responders of which 28 patients satisfied both descriptions. Bi-exponential description improved the fits in 19 cases being associated with late JAK2V617F responses. The decay of the JAK2V617F allele burden during IFN treatment was estimated to have half-lives of 1.6 year for the monoexponential response and 1.0 year in the long term for the bi-exponential response. In conclusion, through data-driven analysis of the JAK2V617F allele burden, we provide novel information regarding the JAK2V617F kinetics during IFN-treatment, arguing for early intervention.
Collapse
Affiliation(s)
- Rasmus K Pedersen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Morten Andersen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Trine A Knudsen
- Department of Haematology, Zealand University Hospital, Roskilde, Denmark
| | - Zamra Sajid
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | | | - Marc J B Dam
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Vibe Skov
- Department of Haematology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjaer
- Department of Haematology, Zealand University Hospital, Roskilde, Denmark
| | - Christina Ellervik
- Department of Production, Research, and Innovation, Region Zealand, Sorø, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Pathology, Harvard Medical School, Boston, FL, USA.,Department of Laboratory Medicine, Boston Children's Hospital, Boston, FL, USA
| | - Thomas S Larsen
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Dennis Hansen
- Department of Hematology, Odense University Hospital, Odense, Denmark
| | - Niels Pallisgaard
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Hans C Hasselbalch
- Department of Haematology, Zealand University Hospital, Roskilde, Denmark
| | - Johnny T Ottesen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| |
Collapse
|
29
|
Hamdan F, Mahmood I, Al-Tameemi W. Heat-shock protein 70 and pentraxin-3 inflammatory biomarkers: Implication for thrombosis in polycythemia vera. IRAQI JOURNAL OF HEMATOLOGY 2020. [DOI: 10.4103/ijh.ijh_3_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
30
|
Guy A, James C. [Pathogenesis of thrombosis in JAK2V617F myeloproliferative neoplasms]. Med Sci (Paris) 2019; 35:651-658. [PMID: 31532377 DOI: 10.1051/medsci/2019133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BCR-ABL negative myeloproliferative neoplasms are acquired hematologic diseases characterized by blood cell proliferation and that include polycythemia vera (PV), essential thrombocytemia (ET) and primary myelofibrosis (PMF). Occurring of venous and arterial thrombosis is the main complication of these diseases. Risk factors for thrombosis are individuals older than 60 and history of thrombosis. The mechanisms leading to thrombosis are complex and involve several blood compartments, plasmatic factors and endothelial cells. Over the last years, new actors of thrombosis have been discovered.
Collapse
Affiliation(s)
- Alexandre Guy
- CHU de Bordeaux, Laboratoire d'hématologie, 1, avenue de Magellan, F-33600, Pessac, France. - Univ. Bordeaux, Inserm, UMR 1034, Biologie des maladies cardio-vasculaires, 1, avenue de Magellan, F-33600, Pessac, France
| | - Chloé James
- CHU de Bordeaux, Laboratoire d'hématologie, 1, avenue de Magellan, F-33600, Pessac, France. - Univ. Bordeaux, Inserm, UMR 1034, Biologie des maladies cardio-vasculaires, 1, avenue de Magellan, F-33600, Pessac, France
| |
Collapse
|
31
|
Impact of angiotensin-converting enzyme inhibition on platelet tissue factor expression in stroke-prone rats. J Hypertens 2019; 36:1360-1371. [PMID: 29470366 PMCID: PMC6037281 DOI: 10.1097/hjh.0000000000001702] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Supplemental Digital Content is available in the text Objective: Hypertension is a well known risk factor for thrombotic events such as myocardial infarction and stroke. Platelets express tissue factor (TF), the key activator of blood coagulation and thrombus formation. The number of TF-positive platelets increases in pathological conditions characterized by thrombotic complications but whether this occurs in hypertension is unknown. Here we investigated whether platelet TF expression is increased in a hypertensive status through a mechanism acting on megakaryocytes; the phenomenon could be modulated by antihypertensive drug as captopril; angiotensin (AngII) influences platelet TF expression. Methods: Spontaneously hypertensive stroke prone (SHRSP) rats received standard diet (StD) or a Japanese high-salt permissive diet (JpD). After 3 weeks, JpD animals were randomized to receive captopril or vehicle. Normotensive Wistar Kyoto (WKY) rats were used as controls. Cell-associated TF expression and activity were analyzed by flow cytometry and calibrated automated thrombogram, respectively. Results: Hypertensive StD-SHRSP showed an increased number of TF-positive platelets compared with normotensive WKY. After JpD administration, SHRSP developed severe hypertension and renal damage; the number of TF-positive megakaryocytes significantly increased compared with StD-SHRSP resulting in a higher number of TF-positive platelets with a faster kinetic of thrombin generation. These effects were reverted by captopril. Ex-vivo stimulation of platelets, isolated from normotensive WKY and from healthy individuals, with AngII induced a concentration-dependent increase of surface-associated TF expression. Conclusion: The current study shows for the first time that in hypertension the number of TF-positive megakaryocytes increases thus releasing in the circulation more platelets carrying a functionally active TF. AngII stimulates platelets to express TF.
Collapse
|
32
|
Marin Oyarzún CP, Heller PG. Platelets as Mediators of Thromboinflammation in Chronic Myeloproliferative Neoplasms. Front Immunol 2019; 10:1373. [PMID: 31258539 PMCID: PMC6587101 DOI: 10.3389/fimmu.2019.01373] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 05/30/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic myeloproliferative neoplasms (MPN) are stem cell disorders driven by mutations in JAK2, CALR, or MPL genes and characterized by myeloid proliferation and increased blood cell counts. They encompass three closely related conditions, including essential thrombocythemia, polycythemia vera, and primary myelofibrosis. Elevated levels of cytokines released by clonal and non-clonal cells generate a chronic proinflammatory state that contributes to disease pathogenesis. Thrombosis represents the most common cause of morbidity and mortality in MPN, although paradoxically, patients may also present with a bleeding diathesis. The mechanisms leading to thrombosis are complex and multiple and include increased blood cells together with qualitative abnormalities of red cells, leukocytes, and platelets that favor a prothrombotic activated phenotype. The functional interplay between blood cells, the clotting cascade, and dysfunctional endothelium contributes to hypercoagulability and this process is perpetuated by the effect of inflammatory cytokines. In addition to their well-known function in hemostasis, platelets contribute to innate immunity and inflammation and play a key role in MPN thromboinflammatory state. In vivo platelet activation leads to platelet aggregate formation and exposure of adhesion molecules which favor their interaction with activated neutrophils and monocytes leading to circulating platelet-leukocyte heterotypic aggregates. Platelets are recruited to the activated endothelium further enhancing the reciprocal activation of both cell types. Crosstalk between activated cells drives cytokine production, further fuelling the self-reinforcing thromboinflammatory loop. In addition, MPN platelets provide a procoagulant scaffold which triggers the coagulation cascade and platelet-derived microparticles amplify this response. Markers of platelet, leukocyte, endothelial and coagulation activation are increased in MPN patients although prospective studies are required to determine the potential value of these parameters for identifying patients at increased thrombotic risk. Thrombosis remains the main complication of MPN patients, with a high risk of recurrence despite adequate cytoreductive and antithrombotic treatment. Deeper insight into the mechanism favoring thrombosis development in this setting may lead to novel therapeutic approaches for MPN thrombosis. Considering the critical role of inflammation in the vascular risk, concomitant targeting of inflammatory pathways could potentially impact on primary or secondary prevention strategies.
Collapse
Affiliation(s)
- Cecilia P Marin Oyarzún
- Department of Hematology Research, National Scientific and Technical Research Council (CONICET), Institute of Medical Research (IDIM) "Dr. Alfredo Lanari", University of Buenos Aires, Buenos Aires, Argentina
| | - Paula G Heller
- Department of Hematology Research, National Scientific and Technical Research Council (CONICET), Institute of Medical Research (IDIM) "Dr. Alfredo Lanari", University of Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
33
|
Babu S, Nagarajan SK, Madhavan T. Investigation of Empirical and Semi‐Empirical Charges to Study the Effects of Partial Charges on Quality and Prediction Accuracy in 3D‐QSAR. ChemistrySelect 2019. [DOI: 10.1002/slct.201803387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Sathya Babu
- Computational Biology LabDepartment of Genetic EngineeringSchool of BioengineeringSRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chennai 603203 India
| | - Santhosh Kumar Nagarajan
- Computational Biology LabDepartment of Genetic EngineeringSchool of BioengineeringSRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chennai 603203 India
| | - Thirumurthy Madhavan
- Computational Biology LabDepartment of Genetic EngineeringSchool of BioengineeringSRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chennai 603203 India
| |
Collapse
|
34
|
Lussana F, Femia EA, Pugliano M, Podda G, Razzari C, Maugeri N, Lecchi A, Caberlon S, Gerli G, Cattaneo M. Evaluation of platelet function in essential thrombocythemia under different analytical conditions. Platelets 2019; 31:179-186. [PMID: 30892978 DOI: 10.1080/09537104.2019.1584668] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background. Studies of platelet aggregation (PA) in essential thrombocythemia (ET) reported contrasting results, likely due to differences in analytical conditions.Objective. We investigated platelet aggregation using different techniques and analytical conditions.Patients and Methods. PA was studied by light-transmission aggregometry (LTA) in platelet-rich plasma (PRP) and impedance aggregometry in PRP and whole blood (WB). ADP, collagen, thrombin receptor activating peptide (TRAP-14) and adrenaline were used as agonists. Since ET patients (n = 41) were on treatment with aspirin (100 mg/d), healthy controls (n = 29) were given aspirin (100 mg/d) for 5 days before testing: therefore, thromboxane A2-independent PA was tested in all subjects. Blood samples were collected in citrate (C) [low Ca2+] or lepirudin (L) [physiological Ca2+]; platelet count was adjusted to 250 x 109/L in a set of C-PRP (adjusted C-PRP) and left unmodified in the other samples.Results. Results of PA in 17 ET patients who were poor responders to aspirin (high serum thromboxane B2 levels) were not included in the analysis. With LTA, PA in ET was lower than in controls in adjusted C-PRP and normal in native C-PRP and L-PRP. With impedance aggregometry, PA in L-PRP and L-WB tended to be higher in ET than in controls. Platelet serotonin and ADP contents were reduced in ET. The percentages of circulating platelets expressing P-selectin and platelet-leukocyte hetero-aggregates were higher in ET.Conclusions. Analytical conditions dramatically affect in vitro PA of ET patients, which appears defective under the least physiological conditions and normal/supranormal under conditions that are closer to the physiological.
Collapse
Affiliation(s)
- Federico Lussana
- Medicina II, ASST Santi Paolo e Carlo - Dipartimento di Scienze della Salute, Università degli Studi di Milano,Milano Italy
| | - Eti Alessandra Femia
- Medicina II, ASST Santi Paolo e Carlo - Dipartimento di Scienze della Salute, Università degli Studi di Milano,Milano Italy
| | - Mariateresa Pugliano
- Medicina II, ASST Santi Paolo e Carlo - Dipartimento di Scienze della Salute, Università degli Studi di Milano,Milano Italy
| | - Gianmarco Podda
- Medicina II, ASST Santi Paolo e Carlo - Dipartimento di Scienze della Salute, Università degli Studi di Milano,Milano Italy
| | - Cristina Razzari
- Medicina II, ASST Santi Paolo e Carlo - Dipartimento di Scienze della Salute, Università degli Studi di Milano,Milano Italy
| | - Norma Maugeri
- Unità di Autoimmunità ed Infiammazione Vascolare, Divisione di Immunologia, Trapianti e Malattie Infettive, Istituto Scientifico San Raffaele, Università Vita-Salute San Raffaele, Milano, Italy
| | - Anna Lecchi
- Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Angelo Bianchi Bonomi Haemophilia and Thrombosis Center, Milano, Italy
| | - Sabrina Caberlon
- Medicina II, ASST Santi Paolo e Carlo - Dipartimento di Scienze della Salute, Università degli Studi di Milano,Milano Italy
| | - Giancarla Gerli
- Medicina II, ASST Santi Paolo e Carlo - Dipartimento di Scienze della Salute, Università degli Studi di Milano,Milano Italy
| | - Marco Cattaneo
- Medicina II, ASST Santi Paolo e Carlo - Dipartimento di Scienze della Salute, Università degli Studi di Milano,Milano Italy
| |
Collapse
|
35
|
Gadomska G, Ziołkowska K, Boinska J, Filipiak J, Rość D. Activation of TF-Dependent Blood Coagulation Pathway and VEGF-A in Patients with Essential Thrombocythemia. ACTA ACUST UNITED AC 2019; 55:medicina55020054. [PMID: 30781507 PMCID: PMC6409549 DOI: 10.3390/medicina55020054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 02/11/2019] [Accepted: 02/11/2019] [Indexed: 01/05/2023]
Abstract
Background and objectives: Recent studies suggest that a vascular endothelial growth factor (VEGF-A) may be involved in the thrombotic process by stimulating the expression of tissue factor in vascular endothelial cells. Tissue factor (TF) can also stimulate the transcription of the gene encoding VEGF-A. The relationship between coagulation and angiogenesis in myeloproliferative neoplasms is not fully understood. The aim of this study was to evaluate the concentration of TF in relation to VEGF-A in the blood of patients with essential thrombocythemia (ET). Patients and methods: The study group consisted of 130, newly diagnosed patients with ET (mean age 61 years). The control group consisted of 35 healthy volunteers (mean age 51 years). Concentrations of VEGF-A, TF, and tissue factor pathway inhibitor (TFPI) were analysed using immunoenzymatic methods. TF and TFPI activities were performed using chromogenic assays. Results: The median concentration of TF Ag was 3-fold higher and the TF activity was more than 15-fold higher in ET patients than in normal individuals. There were no statistically significant differences in the TFPI concentration and activity between groups. VEGF-A was significantly increased in patients with ET (p < 0.000001). Analysis of correlations revealed a positive correlation between VEGF-A and TF Ag as well as a positive correlation between VEGF-A and TFPI activity. Conclusions: The simultaneous increase of TF concentration and activity, VEGF-A in the blood of patients with ET, as well as a positive correlation between the concentration of TF and VEGF-A demonstrates the coexistence of TF-dependent coagulation and activation of angiogenesis.
Collapse
Affiliation(s)
- Grażyna Gadomska
- Department of Hematology and Malignant Diseases of Hematopoietic System, Faculty of Medicine, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-168 Bydgoszcz, Poland.
| | - Katarzyna Ziołkowska
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland.
| | - Joanna Boinska
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland.
| | - Jan Filipiak
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland.
| | - Danuta Rość
- Department of Pathophysiology, Faculty of Pharmacy, Nicolaus Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, 85-094 Bydgoszcz, Poland.
| |
Collapse
|
36
|
Stefanou MI, Richter H, Härtig F, Wang Y, Örgel A, Bender B, Mengel A, Ziemann U, Poli S. Recurrent ischaemic cerebrovascular events as presenting manifestations of myeloproliferative neoplasms. Eur J Neurol 2019; 26:903-e64. [PMID: 30629793 DOI: 10.1111/ene.13907] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 01/08/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND PURPOSE Myeloproliferative neoplasms (MPNs) - polycythemia vera, essential thrombocythemia and primary myelofibrosis - are associated with increased risk for ischaemic cerebrovascular events (ICVEs). Due to their low prevalence, MPNs often remain undiagnosed as the cause of ICVEs. METHODS Case records at the University of Tübingen between 2014 and 2017 were screened to identify patients with MPN-related ICVEs. Clinical features, brain imaging, laboratory findings, applied treatments and neurological outcomes were assessed. RESULTS In all, 3318 patients with ICVEs were identified, and amongst them 17 patients with MPN-related ICVEs were included in a retrospective study. In 58% of these patients, ICVEs were the first manifestation of the underlying MPN; 24% presented with transient ischaemic attack and 76% with ischaemic stroke. Potentially concurrent ICVE etiologies were noted in 70% of the patients. The majority (94%) of patients were positive for the JAK2 V617F mutation, whilst in 29% recurrent ICVEs (range two to three) were noted prior to MPN diagnosis. Early MPN diagnosis and management was the only significant prognostic factor for ICVE recurrence (P < 0.001). DISCUSSION Evidence is provided that, although rare, MPNs represent an underdiagnosed cause of recurrent ICVEs. High clinical awareness is warranted to identify an underlying MPN in patients presenting with sustained, abnormal blood count findings. Clinical algorithms for prompt MPN diagnosis and initiation of MPN treatment (e.g. cytoreductive therapy, phlebotomy) are required. As MPN management comprises a significant protective factor against ICVE recurrence, induction of MPN treatment should be regarded as an integral component of secondary stroke prevention in MPN-associated ICVEs.
Collapse
Affiliation(s)
- M I Stefanou
- Department of Neurology and Stroke, Hertie Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - H Richter
- Department of Neurology and Stroke, Hertie Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - F Härtig
- Department of Neurology and Stroke, Hertie Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - Y Wang
- Department of Neurology and Stroke, Hertie Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - A Örgel
- Department of Diagnostic and Interventional Neuroradiology, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - B Bender
- Department of Diagnostic and Interventional Neuroradiology, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - A Mengel
- Department of Neurology and Stroke, Hertie Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - U Ziemann
- Department of Neurology and Stroke, Hertie Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - S Poli
- Department of Neurology and Stroke, Hertie Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
37
|
Understanding the structural features of JAK2 inhibitors: a combined 3D-QSAR, DFT and molecular dynamics study. Mol Divers 2019; 23:845-874. [PMID: 30617940 DOI: 10.1007/s11030-018-09913-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/27/2018] [Indexed: 12/11/2022]
Abstract
JAK2 plays a critical role in JAK/STAT signaling pathway and in patho-mechanism of myeloproliferative disorders and autoimmune diseases. Thus, effective JAK2 inhibitors provide a promising opportunity for the pharmaceutical intervention of many diseases. In this work, 3D-QSAR study was performed on a series of 1-amino-5H-pyrido-indole-4-carboxamide derivatives as JAK2 inhibitors to obtain reliable comparative molecular field analysis (CoMFA) and comparative molecular similarity analysis (CoMSIA) models with three different alignment methods. Among the different alignment methods, ligand-based (CoMFA: q2 = 0.676, r2 = 0.979; CoMSIA: q2 = 0.700, r2 = 0.953) and pharmacophore-based alignment (CoMFA: q2 = 0.710, r2 = 0.982; CoMSIA: q2 = 0.686, r2 = 0.960) has produced better statistical results when compared to receptor-based alignment (CoMFA: q2 = 0.507, r2 = 0.979; CoMSIA: q2 = 0.544, r2 = 0.917). Statistical parameters indicated that data are well fitted and have high predictive ability. The presence of electrostatic and hydrophobic field is highly desirable for potent inhibitory activity, and the steric field plays a minor role in modulating the activity. The contour analysis indicates ARG980, ASN981, ASP939 and LEU937 have more possibility of interacting with bulky, hydrophobic groups in pyrido and positive and negative groups in pyrazole ring. Based on our findings, we have designed sixteen molecules and predicted its activity and drug-like properties. Subsequently, molecular docking, molecular dynamics and DFT calculations were performed to evaluate its potency.
Collapse
|
38
|
Hamdan F, Mahmood I, Al-Tameemi W. Blood cell activation as an indicator of prothrombogenesis in polycythemia vera: Case control study. IRAQI JOURNAL OF HEMATOLOGY 2019. [DOI: 10.4103/ijh.ijh_10_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
39
|
Tawil N, Chennakrishnaiah S, Bassawon R, Johnson R, D'Asti E, Rak J. Single cell coagulomes as constituents of the oncogene-driven coagulant phenotype in brain tumours. Thromb Res 2018; 164 Suppl 1:S136-S142. [PMID: 29703472 DOI: 10.1016/j.thromres.2018.01.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 02/07/2023]
Abstract
Molecular profiling of human cancers revealed a startling diversity in disease-causing mechanisms superseding histological and anatomical commonalities. The emerging molecular subtypes and disease entities are often driven by distinct oncogenic pathways and their effectors, including those acting extracellularly on the vascular and coagulation systems. Indeed, several oncogenic mutations such as those affecting protein-coding genes (RAS, EGFR, PTEN, TP53) and non-coding RNA (microRNA) regulate multiple effectors of the coagulation system (coagulome), including tissue factor, protease activated receptors, clotting factors, mediators of platelet function and fibrinolysis. This is exemplified by differential coagulome profiles in the molecular subtypes of glioblastoma, medulloblastoma and other human tumours. There is mounting clinical evidence that the mutational status of cancer driver genes such as KRAS or IDH1 may influence the risk of venous thromboembolism in patients with colorectal, lung or brain cancers. Notably, single cell sequencing in glioblastoma revealed a remarkable intra-tumoural heterogeneity of cancer cell populations with regard to their individual coagulomes, suggesting a combinatorial and dynamic nature of the global pro-thrombotic phenotype. We suggest that the cellular complexity of specific cancers may define their mechanisms of interactions with the coagulation system, and the risks of thrombosis. Thus, more biologically- based, disease-specific and personalized approaches may be needed to diagnose and manage cancer-related thrombosis.
Collapse
Affiliation(s)
- Nadim Tawil
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada
| | | | - Rayhaan Bassawon
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada
| | - Radia Johnson
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada
| | - Esterina D'Asti
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada
| | - Janusz Rak
- McGill University, Montreal Children's Hospital, RI MUHC, Montreal, Quebec, Canada.
| |
Collapse
|
40
|
Shahrabi S, Ehsanpour A, Heidary S, Shahjahani M, Behzad MM. Expression of CD markers in JAK2 V617F positive myeloproliferative neoplasms: Prognostic significance. Oncol Rev 2018; 12:373. [PMID: 30405895 PMCID: PMC6199554 DOI: 10.4081/oncol.2018.373] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 06/20/2018] [Indexed: 01/08/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are clonal stem cell disorders characterized by the presence of JAK2V617F mutation. Thrombohemorrhagic as well as autoimmune or inflammatory phenomena are common clinical outcomes of these disorders. Recent studies have shown that abnormality in frequency and function of blood cells manifested by an alteration in CD markers' expression patterns play a key role in these complications. So, there may be a relationship between CD markers' expressions and prognosis of JAK2V617F positive MPNs. Therefore, in this review, we have focused on these abnormalities from the perspective of changing expressions of CD markers and assessment of the relationship between these changes with prognosis of JAK2V617F positive MPNs. It can be stated that the abnormal expression of a large number of CD markers can be used as a prognostic biomarker for clinical outcomes including thrombohememorrhagic events, as well as autoimmune and leukemic transformation in JAK2V617F positive MPNs. Considering the possible role of CD markers' expressions in JAK2V617F MPNs prognosis, further studies are needed to confirm the relationship between the expression of CD markers with prognosis to be able to find an appropriate therapeutic approach via targeting CD markers.
Collapse
Affiliation(s)
- Saeid Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan
| | - Ali Ehsanpour
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Somayyeh Heidary
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Shahjahani
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Masumeh Maleki Behzad
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
41
|
Hultcrantz M, Björkholm M, Dickman PW, Landgren O, Derolf ÅR, Kristinsson SY, Andersson TML. Risk for Arterial and Venous Thrombosis in Patients With Myeloproliferative Neoplasms: A Population-Based Cohort Study. Ann Intern Med 2018; 168:317-325. [PMID: 29335713 PMCID: PMC7533681 DOI: 10.7326/m17-0028] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Patients with myeloproliferative neoplasms (MPNs) are reported to be at increased risk for thrombotic events. However, no population-based study has estimated this excess risk compared with matched control participants. OBJECTIVE To assess risk for arterial and venous thrombosis in patients with MPNs compared with matched control participants. DESIGN Matched cohort study. SETTING Population-based setting in Sweden from 1987 to 2009, with follow-up to 2010. PATIENTS 9429 patients with MPNs and 35 820 matched control participants. MEASUREMENTS The primary outcomes were rates of arterial and venous thrombosis. Flexible parametric models were used to calculate hazard ratios (HRs) and cumulative incidence with 95% CIs. RESULTS The HRs for arterial thrombosis among patients with MPNs compared with control participants at 3 months, 1 year, and 5 years were 3.0 (95% CI, 2.7 to 3.4), 2.0 (CI, 1.8 to 2.2), and 1.5 (CI, 1.4 to 1.6), respectively. The corresponding HRs for venous thrombosis were 9.7 (CI, 7.8 to 12.0), 4.7 (CI, 4.0 to 5.4), and 3.2 (CI, 2.9 to 3.6). The rate was significantly elevated across all age groups and was similar among MPN subtypes. The 5-year cumulative incidence of thrombosis in patients with MPNs showed an initial rapid increase followed by gentler increases during follow-up. The HR for venous thrombosis decreased during more recent calendar periods. LIMITATION No information on individual laboratory results or treatment. CONCLUSION Patients with MPNs across all age groups have a significantly increased rate of arterial and venous thrombosis compared with matched control participants, with the highest rates at and shortly after diagnosis. Decreases in the rate of venous thrombosis over time likely reflect advances in clinical management. PRIMARY FUNDING SOURCE The Cancer Research Foundations of Radiumhemmet, Blodcancerfonden, the Swedish Research Council, the regional agreement on medical training and clinical research between Stockholm County Council and Karolinska Institutet, the Adolf H. Lundin Charitable Foundation, and Memorial Sloan Kettering Cancer Center.
Collapse
Affiliation(s)
- Malin Hultcrantz
- Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden, and Memorial Sloan Kettering Cancer Center, New York, New York (M.H.)
| | - Magnus Björkholm
- Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden (M.B., Å.R.D.)
| | - Paul W Dickman
- Karolinska Institutet, Stockholm, Sweden (P.W.D., T.M.A.)
| | - Ola Landgren
- Memorial Sloan Kettering Cancer Center, New York, New York (O.L.)
| | - Åsa R Derolf
- Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden (M.B., Å.R.D.)
| | - Sigurdur Y Kristinsson
- Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden, and University of Iceland and Landspítali National University Hospital, Reykjavik, Iceland (S.Y.K.)
| | | |
Collapse
|
42
|
De Stefano V, Carobbio A, Di Lazzaro V, Guglielmelli P, Iurlo A, Finazzi MC, Rumi E, Cervantes F, Elli EM, Randi ML, Griesshammer M, Palandri F, Bonifacio M, Hernandez-Boluda JC, Cacciola R, Miroslava P, Carli G, Beggiato E, Ellis MH, Musolino C, Gaidano G, Rapezzi D, Tieghi A, Lunghi F, Loscocco GG, Cattaneo D, Cortelezzi A, Betti S, Rossi E, Finazzi G, Censori B, Cazzola M, Bellini M, Arellano-Rodrigo E, Bertozzi I, Sadjadian P, Vianelli N, Scaffidi L, Gomez M, Cacciola E, Vannucchi AM, Barbui T. Benefit-risk profile of cytoreductive drugs along with antiplatelet and antithrombotic therapy after transient ischemic attack or ischemic stroke in myeloproliferative neoplasms. Blood Cancer J 2018. [PMID: 29535299 PMCID: PMC5849668 DOI: 10.1038/s41408-018-0048-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
We analyzed 597 patients with myeloproliferative neoplasms (MPN) who presented transient ischemic attacks (TIA, n = 270) or ischemic stroke (IS, n = 327). Treatment included aspirin, oral anticoagulants, and cytoreductive drugs. The composite incidence of recurrent TIA and IS, acute myocardial infarction (AMI), and cardiovascular (CV) death was 4.21 and 19.2%, respectively at one and five years after the index event, an estimate unexpectedly lower than reported in the general population. Patients tended to replicate the first clinical manifestation (hazard ratio, HR: 2.41 and 4.41 for recurrent TIA and IS, respectively); additional factors for recurrent TIA were previous TIA (HR: 3.40) and microvascular disturbances (HR: 2.30); for recurrent IS arterial hypertension (HR: 4.24) and IS occurrence after MPN diagnosis (HR: 4.47). CV mortality was predicted by age over 60 years (HR: 3.98), an index IS (HR: 3.61), and the occurrence of index events after MPN diagnosis (HR: 2.62). Cytoreductive therapy was a strong protective factor (HR: 0.24). The rate of major bleeding was similar to the general population (0.90 per 100 patient-years). In conclusion, the long-term clinical outcome after TIA and IS in MPN appears even more favorable than in the general population, suggesting an advantageous benefit-risk profile of antithrombotic and cytoreductive treatment.
Collapse
Affiliation(s)
- Valerio De Stefano
- Institute of Hematology, Catholic University, Fondazione Policlinico Universitario A. Gemelli, Roma, Italy
| | | | - Vincenzo Di Lazzaro
- Unit of Neurology, Neurophysiology, Neurobiology, Department of Medicine, Università Campus Biomedico di Roma, Rome, Italy
| | - Paola Guglielmelli
- CRIMM-Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, and Departmentt Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Alessandra Iurlo
- Hematology Division, IRCCS Ca' Granda - Maggiore Policlinico Hospital Foundation, and University of Milan, Milan, Italy
| | | | - Elisa Rumi
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Elena Maria Elli
- Hematology Division, Ospedale San Gerardo, ASST Monza, Monza, Italy
| | | | - Martin Griesshammer
- University Clinic for Hematology and Oncology Minden, University of Bochum, Bochum, Germany
| | - Francesca Palandri
- Institute of Hematology "L. and A. Seràgnoli", S. Orsola-Malpighi Hospital, Bologna, Italy
| | | | | | - Rossella Cacciola
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Palova Miroslava
- Department of Hemato-Oncology, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
| | - Giuseppe Carli
- Hematology Department, Ospedale San Bortolo, Vicenza, Italy
| | - Eloise Beggiato
- Unit of Hematology, Department of Oncology, University of Torino, Torino, Italy
| | - Martin H Ellis
- Hematology Institute and Blood Bank, Meir Medical Center, Kfar Saba, and Sackler School of Medicine Tel Aviv University, Tel Aviv, Israel
| | - Caterina Musolino
- Division of Hematology, Dipartimento di Patologia Umana dell'Adulto e dell'Età Evolutiva, Policlinico G Martino, University of Messina, Messina, Italy
| | - Gianluca Gaidano
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont, Novara, Italy
| | - Davide Rapezzi
- S.C. Ematologia, Azienda Ospedaliera S. Croce e Carle, Cuneo, Italy
| | - Alessia Tieghi
- Divisione di Ematologia, Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, Italy
| | - Francesca Lunghi
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milano, Italy
| | - Giuseppe Gaetano Loscocco
- CRIMM-Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, and Departmentt Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Daniele Cattaneo
- Hematology Division, IRCCS Ca' Granda - Maggiore Policlinico Hospital Foundation, and University of Milan, Milan, Italy
| | - Agostino Cortelezzi
- Hematology Division, IRCCS Ca' Granda - Maggiore Policlinico Hospital Foundation, and University of Milan, Milan, Italy
| | - Silvia Betti
- Institute of Hematology, Catholic University, Fondazione Policlinico Universitario A. Gemelli, Roma, Italy
| | - Elena Rossi
- Institute of Hematology, Catholic University, Fondazione Policlinico Universitario A. Gemelli, Roma, Italy
| | - Guido Finazzi
- Hematology Division, Papa Giovanni XXIII hospital, Bergamo, Italy
| | - Bruno Censori
- Neurology Division, Papa Giovanni XXIII hospital, Bergamo, Italy
| | - Mario Cazzola
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.,Department of Hematology Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Marta Bellini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Irene Bertozzi
- Department of Medicine - DIMED, University of Padua, Padova, Italy
| | - Parvis Sadjadian
- University Clinic for Hematology and Oncology Minden, University of Bochum, Bochum, Germany
| | - Nicola Vianelli
- Institute of Hematology "L. and A. Seràgnoli", S. Orsola-Malpighi Hospital, Bologna, Italy
| | - Luigi Scaffidi
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Montse Gomez
- Hematology Department, Hospital Clínico Universitario, Valencia, Spain
| | - Emma Cacciola
- Department of Medical, Surgical and Advanced Technologies Sciences "G.F. Ingrassia", University of Catania, Catania, Italy
| | - Alessandro M Vannucchi
- CRIMM-Center of Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, and Departmentt Experimental and Clinical Medicine, University of Florence, Firenze, Italy
| | - Tiziano Barbui
- FROM Research Foundation, Papa Giovanni XXIII hospital, Bergamo, Italy.
| |
Collapse
|
43
|
Horowitz NA, Lavi N, Nadir Y, Brenner B. Haematological malignancies in pregnancy: An overview with an emphasis on thrombotic risks. Thromb Haemost 2017; 116:613-7. [DOI: 10.1160/th16-02-0099] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 06/29/2016] [Indexed: 12/13/2022]
Abstract
SummaryWith increase of maternal age, the incidence of haematological malignancies during pregnancy is rising and posing diagnostic and treatment challenges. Lymphoma is the fourth most common malignancy diagnosed in pregnancy; Hodgkin lymphoma is more frequent in pregnant women than non-Hodgkin lymphoma (NHL). The proportion of highly aggressive lymphomas in pregnant women is significantly higher than in non-pregnant women of reproductive age. Reproductive organ involvement is observed in almost half of pregnant women with NHL. The association of acute leukaemia and pregnancy is infrequent and it is assumed that pregnancy does not accelerate the disease course. Both cancer and pregnancy induce a procoagulant state which can lead to maternal venous thromboembolism (VTE) and placental occlusion. Pregnancy in woman with myeloproliferative neoplasms (MPN) promotes thrombotic environment, associating with an augmented risk of placental thrombosis, intrauterine growth retardation or loss and maternal thrombotic events.Haematological malignancies during pregnancy often require urgent diagnosis and management and are associated with potential adverse fetal outcomes. Most chemotherapeutic agents are teratogenic and should be avoided during the first trimester. Their use during the second and third trimesters may cause intrauterine growth restriction, premature birth and intrauterine fetal death. All chemotherapeutic drugs should be administered only after a detailed discussion with the patient and with close fetal monitoring. Chemotherapy and biological agents might also augment thrombotic risk. Guidelines for VTE prophylaxis in pregnant women with hematologic malignancies, apart from MPN, are currently unavailable, and therefore, clinical judgment should be made in each case.
Collapse
|
44
|
Lussana F, Rambaldi A. Inflammation and myeloproliferative neoplasms. J Autoimmun 2017; 85:58-63. [DOI: 10.1016/j.jaut.2017.06.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 06/23/2017] [Indexed: 01/14/2023]
|
45
|
Canzano P, Rossetti L, Ferri N, Balduini A, Abbonante V, Boselli D, De Marco L, Di Minno M, Toschi V, Corsini A, Tremoli E, Brambilla M, Facchinetti L, Camera M. Human megakaryocytes confer tissue factor to a subset of shed platelets to stimulate thrombin generation. Thromb Haemost 2017; 114:579-92. [DOI: 10.1160/th14-10-0830] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Accepted: 04/11/2015] [Indexed: 11/05/2022]
Abstract
SummaryTissue factor (TF), the main activator of the blood coagulation cascade, has been shown to be expressed by platelets. Despite the evidence that both megakaryocytes and platelets express TF mRNA, and that platelets can make de novo protein synthesis, the main mechanism thought to be responsible for the presence of TF within platelets is through the uptake of TF positive microparticles. In this study we assessed 1) whether human megakaryocytes synthesise TF and transfer it to platelets and 2) the contribution of platelet-TF to the platelet hemostatic capacity. In order to avoid the cross-talk with circulating microparticles, we took advantage from an in vitro cultured megakaryoblastic cell line (Meg-01) able to differentiate into megakaryocytes releasing platelet-like particles. We show that functionally active TF is expressed in human megakaryoblasts, increased in megakaryocytes, and is transferred to a subset of shed platelets where it contributes to clot formation. These data were all confirmed in human CD34pos- derived megakaryocytes and in their released platelets. The effect of TF silencing in Meg-megakaryoblasts resulted in a significant reduction of TF expression in these cells and also in Meg-megakaryocytes and Meg-platelets. Moreover, the contribution of platelet-TF to the platelet hemostatic capacity was highlighted by the significant delay in the kinetic of thrombin formation observed in platelets released by TF-silenced megakaryocytes. These findings provide evidences that TF is an endogenously synthesised protein that characterises megakaryocyte maturation and that it is transferred to a subset of newly-released platelets where it is functionally active and able to trigger thrombin generation.
Collapse
|
46
|
Assessing the thrombotic risk of patients with essential thrombocythemia in the genomic era. Leukemia 2017; 31:1845-1854. [PMID: 28529308 DOI: 10.1038/leu.2017.150] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/22/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023]
Abstract
The molecular characterization of myeloproliferative neoplasms, including essential thrombocythemia (ET), has enabled deeper understanding of their pathogenesis. A driver lesion, namely, Janus kinase (JAK)2V617F, calreticulin (CALR) or myeloproliferative leukemia (MPL) gene mutation can be identified in the vast majority of patients. Each of these mutations is associated with distinct clinical features and may modulate the patients' clinical course, risk of complications, including vascular events, and survival. JAK2V617F appears to be a risk-modifying mutation and has been shown to increase the likelihood of thrombotic events in patients with ET across studies. As such, it has been included in prognostic models and its presence may influence treatment decisions. The association of CALR and MPL mutations with the incidence of vascular events has been less clear. Even more limited information is available on the contribution of additional non-driver lesions to the thrombotic risk. In this review we discuss the available evidence on the role of recurrent mutations in the risk of thrombotic complications in patients with ET and how these mutations weigh into modern prognostic scores.
Collapse
|
47
|
Mihaila RG. Thrombin generation - a potentially useful biomarker of thrombotic risk in Philadelphia-negative myeloproliferative neoplasms. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2017; 161:50-53. [DOI: 10.5507/bp.2016.064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 12/15/2016] [Indexed: 12/25/2022] Open
|
48
|
Lussana F, Carobbio A, Salmoiraghi S, Guglielmelli P, Vannucchi AM, Bottazzi B, Leone R, Mantovani A, Barbui T, Rambaldi A. Driver mutations (JAK2V617F, MPLW515L/K or CALR), pentraxin-3 and C-reactive protein in essential thrombocythemia and polycythemia vera. J Hematol Oncol 2017; 10:54. [PMID: 28228104 PMCID: PMC5322581 DOI: 10.1186/s13045-017-0425-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/17/2017] [Indexed: 11/24/2022] Open
Abstract
Background The driver mutations JAK2V617F, MPLW515L/K and CALR influence disease phenotype of myeloproliferative neoplasms (MPNs) and might sustain a condition of chronic inflammation. Pentraxin 3 (PTX3) and high-sensitivity C-reactive protein (hs-CRP) are inflammatory biomarkers potentially useful for refining prognostic classification of MPNs. Methods We evaluated 305 with essential thrombocythemia (ET) and 172 polycythemia vera (PV) patients diagnosed according to the 2016 WHO criteria and with full molecular characterization for driver mutations. Results PTX3 levels were significantly increased in carriers of homozygous JAK2V617F mutation compared to all the other genotypes and triple negative ET patients, while hs-CRP levels were independent of the mutational profile. The risk of haematological evolution and death from any cause was about 2- and 1.5-fold increased in individuals with high PTX-3 levels, while the thrombosis rate tended to be lower. High hs-CRP levels were associated with risk of haematological evolution, death and also major thrombosis. After sequential adjustment for potential confounders (age, gender, diagnosis and treatments) and the presence of JAK2V617F homozygous status, high hs-CRP levels remained significant for all outcomes, while JAK2V617F homozygous status as well as treatments were the factors independently accounting for adverse outcomes among patients with high PTX3 levels. Conclusions These results provide evidence that JAK2V617F mutation influences MPN-associated inflammation with a strong correlation between allele burden and PTX3 levels. Plasma levels of hs-CRP and PTX3 might be of prognostic value for patients with ET and PV, but their validation in future prospective studies is needed.
Collapse
Affiliation(s)
- Federico Lussana
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII Bergamo, Piazza OMS, 1, 24127, Bergamo, Italy.
| | | | - Silvia Salmoiraghi
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII Bergamo, Piazza OMS, 1, 24127, Bergamo, Italy
| | - Paola Guglielmelli
- Department of Experimental and Clinical Medicine, CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| | - Alessandro Maria Vannucchi
- Department of Experimental and Clinical Medicine, CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| | | | - Roberto Leone
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy.,Humanitas University, Rozzano, Milan, Italy
| | - Tiziano Barbui
- Research Foundation, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII Bergamo, Piazza OMS, 1, 24127, Bergamo, Italy.,Department of Oncology and Hematology, University of Milan, Milan, Italy
| |
Collapse
|
49
|
Saki N, Shirzad R, Rahim F, Saki Malehi A. Estimation of diagnosis and prognosis in ET by assessment of CALR and JAK2 V617F mutations and laboratory findings: a meta-analysis. Clin Transl Oncol 2017; 19:874-883. [PMID: 28205126 DOI: 10.1007/s12094-017-1618-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 01/18/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Essential thrombocythemia (ET) is a benign disease with slow progress in which thrombosis is a cause of mortality. JAK2V617F and calreticulin (CALR) are the most frequent mutations in this disease. In this systematic review and meta-analysis, we compared the prevalence of JAK2V617F and CALR mutations in ET and examined the incidence of thrombosis and other hematologic indices. METHODS After choosing MeSH keywords, including essential thrombocythemia, JAK2V617F, calreticulin, prognosis, and diagnosis, as well as searching Medline/PubMed and Scopus, 12 papers were selected. Data were pooled, and summary prevalence and OR were estimated using either a random-effects model or a fixed-effects model. RESULTS The frequency of JAK2V617F and CALR shows heterogeneity in Caucasian population [JAK2V617 I 2% = 84.3, P < 0.001, 95% CI 0.56 (0.51-0.61)], [CALR I 2% = 96.1, P < 0.001, 95% CI 0.23 (0.15-0.31)]. The prevalence of JAK2V617F and CALR was 0.57 (95% CI 0.53-0.61), I 2% = 79.3 and 0.22 (95% CI 0.16-0.27), I 2% = 94, respectively. JAK2V617F positive ET was associated with increasing odds of thrombosis [OR 2.35 (95% CI 1.83-3.02), P < 0.001]. The incidence of splenomegaly was not statistically different between these two mutations. Hemoglobin, platelet, and WBC count did not affect the risk of thrombosis. CONCLUSIONS Detection of CALR mutation is helpful for molecular diagnosis of ET patients as well as JAK2V617F. Due to reduction of thrombosis in CALR-positive patients, it can be stated that such patients have less thrombotic disorders and better prognosis relative to patients bearing JAK2V617F mutation. Therefore, detection of mutation in CALR and JAK2V617F may contribute to diagnosis and prognosis of ET patients.
Collapse
Affiliation(s)
- N Saki
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - R Shirzad
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - F Rahim
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - A Saki Malehi
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. .,Department of Biostatistics and Epidemiology, School of Public Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
50
|
Falchi L, Bose P, Newberry KJ, Verstovsek S. Approach to patients with essential thrombocythaemia and very high platelet counts: what is the evidence for treatment? Br J Haematol 2016; 176:352-364. [DOI: 10.1111/bjh.14443] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Lorenzo Falchi
- Division of Hematology/Oncology; Columbia University Medical Center; New York NY USA
| | - Prithviraj Bose
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Kate J. Newberry
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston TX USA
| | - Srdan Verstovsek
- Department of Leukemia; The University of Texas MD Anderson Cancer Center; Houston TX USA
| |
Collapse
|