1
|
Bonifay A, Cointe S, Plantureux L, Lacroix R, Dignat-George F. Update on Tissue Factor Detection in Blood in 2024: A Narrative Review. Hamostaseologie 2024; 44:368-376. [PMID: 39442509 DOI: 10.1055/a-2381-6854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024] Open
Abstract
Tissue factor (TF) is a transmembrane protein essential for hemostasis. Different forms of active TF circulate in the blood, either as a component of blood cells and extracellular vesicles (EVs) or as a soluble plasma protein. Accumulating experimental and clinical evidence suggests that TF plays an important role in thrombosis. Many in-house and commercially available assays have been developed to measure TF-dependent procoagulant activity or antigen in blood and have shown promising results for the prediction of disease outcomes or the occurrence of thrombosis events in diseases such as cancer or infectious coagulopathies. This review addresses the different assays that have been published for measuring circulating TF antigen and/or activity in whole blood, cell-free plasma, and EVs and discusses the main preanalytical and analytical parameters that impact results and their interpretation, highlighting their strengths and limitations. In the recent decade, EVTF assays have been significantly developed. Among them, functional assays that use a blocking anti-TF antibody or immunocapture to measure EVTF activity have higher specificity and sensitivity than antigen assays. However, there is still a high variability between assays. Standardization and automatization are prerequisites for the measurement of EVTF in clinical laboratories.
Collapse
Affiliation(s)
- Amandine Bonifay
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
- Department of Hematology, Biogénopôle, CHU La Timone, APHM, Marseille, France
| | - Sylvie Cointe
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
- Department of Hematology, Biogénopôle, CHU La Timone, APHM, Marseille, France
| | - Léa Plantureux
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
| | - Romaric Lacroix
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
- Department of Hematology, Biogénopôle, CHU La Timone, APHM, Marseille, France
| | - Françoise Dignat-George
- Aix-Marseille University, C2VN, INSERM 1263, INRAE 1260, Marseille, France
- Department of Hematology, Biogénopôle, CHU La Timone, APHM, Marseille, France
| |
Collapse
|
2
|
Pavlovic D, Niciforovic D, Markovic M, Papic D. Cancer-Associated Thrombosis: Epidemiology, Pathophysiological Mechanisms, Treatment, and Risk Assessment. Clin Med Insights Oncol 2023; 17:11795549231220297. [PMID: 38152726 PMCID: PMC10752082 DOI: 10.1177/11795549231220297] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/28/2023] [Indexed: 12/29/2023] Open
Abstract
Cancer patients represent a growing population with drastically difficult care and a lowered quality of life, especially due to the heightened risk of vast complications. Thus, it is well established so far that one of the most prominent complications in individuals with cancer is venous thromboembolism. Since there are various improved methods for screening and diagnosing cancer and its complications, the incidence of cancer-associated thrombosis has been on the rise in recent years. Therefore, the high mortality and morbidity rates among these patients are not a surprise. Consequently, there is an excruciating need for understanding the mechanisms behind this complex process, as well as the imperative for adequate analysis and application of the most suitable steps for cancer-associated thrombosis prevention. There are various and numerous mechanisms offering potential answers to cancer-associated thrombosis, some of which have already been elucidated in various preclinical and clinical scenarios, yet further and more elaborate studies are crucial to understanding and preventing this complex and harsh clinical entity. This article elaborates on the growing incidence, mortality, morbidity, and risk factors of cancer-associated thrombosis while emphasizing the pathophysiological mechanisms in the light of various types of cancer in patients and summarizes the most novel therapy and prevention guidelines recommendations.
Collapse
Affiliation(s)
- Dragica Pavlovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Danijela Niciforovic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Marina Markovic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Dragana Papic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
3
|
Kapteijn MY, Zwaan S, Ter Linden E, Laghmani EH, van den Akker RFP, Rondon AMR, van der Zanden SY, Neefjes J, Versteeg HH, Buijs JT. Temozolomide and Lomustine Induce Tissue Factor Expression and Procoagulant Activity in Glioblastoma Cells In Vitro. Cancers (Basel) 2023; 15:cancers15082347. [PMID: 37190275 DOI: 10.3390/cancers15082347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
Glioblastoma (GBM) patients have one of the highest risks of venous thromboembolism (VTE), which is even further increased upon treatment with chemotherapy. Tissue factor (TF) is the initiator of the extrinsic coagulation pathway and expressed by GBM cells. In this study, we aimed to examine the effect of routinely used chemotherapeutic agents Temozolomide (TMZ) and Lomustine (LOM) on TF procoagulant activity and expression in GBM cells in vitro. Three human GBM cell lines (U-251, U-87, U-118) were exposed to 100 µM TMZ or 30 µM LOM for 72 h. TF procoagulant activity was assessed via an FXa generation assay and TF gene and protein expression through qPCR and Western blotting. The externalization of phosphatidylserine (PS) was studied using Annexin V flow cytometry. Treatment with TMZ and LOM resulted in increased procoagulant activity in all cell lines. Furthermore, both agents induced procoagulant activity in the supernatant and tumor-cell-secreted extracellular vesicles. In line, TF gene and protein expression were increased upon TMZ and LOM treatment. Additionally, PS externalization and induction of inflammatory-associated genes were observed. Overall, the chemotherapeutic modalities TMZ and LOM induced procoagulant activity and increased TF gene and protein expression in all GBM cell lines tested, which may contribute to the increased VTE risk observed in GBM patients undergoing chemotherapy.
Collapse
Affiliation(s)
- Maaike Y Kapteijn
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis & Hemostasis, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Shanna Zwaan
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis & Hemostasis, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Esther Ter Linden
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - El Houari Laghmani
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis & Hemostasis, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Rob F P van den Akker
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis & Hemostasis, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Araci M R Rondon
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis & Hemostasis, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Sabina Y van der Zanden
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, ONCODE Institute, Leiden University Medical Center, 2333 ZC Leiden, The Netherlands
| | - Henri H Versteeg
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis & Hemostasis, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jeroen T Buijs
- Einthoven Laboratory for Vascular and Regenerative Medicine, Division of Thrombosis & Hemostasis, Department of Internal Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
4
|
Subtype-specific plasma signatures of platelet-related protein releasate in acute pulmonary embolism. Thromb Res 2022; 220:75-87. [DOI: 10.1016/j.thromres.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/21/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022]
|
5
|
Adnani L, Spinelli C, Tawil N, Rak J. Role of extracellular vesicles in cancer-specific interactions between tumour cells and the vasculature. Semin Cancer Biol 2022; 87:196-213. [PMID: 36371024 DOI: 10.1016/j.semcancer.2022.11.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/25/2022] [Accepted: 11/08/2022] [Indexed: 11/11/2022]
Abstract
Cancer progression impacts and exploits the vascular system in several highly consequential ways. Among different types of vascular cells, blood cells and mediators that are engaged in these processes, endothelial cells are at the centre of the underlying circuitry, as crucial constituents of angiogenesis, angiocrine stimulation, non-angiogenic vascular growth, interactions with the coagulation system and other responses. Tumour-vascular interactions involve soluble factors, extracellular matrix molecules, cell-cell contacts, as well as extracellular vesicles (EVs) carrying assemblies of molecular effectors. Oncogenic mutations and transforming changes in the cancer cell genome, epigenome and signalling circuitry exert important and often cancer-specific influences upon pathways of tumour-vascular interactions, including the biogenesis, content, and biological activity of EVs and responses of cancer cells to them. Notably, EVs may carry and transfer bioactive, oncogenic macromolecules (oncoproteins, RNA, DNA) between tumour and vascular cells and thereby elicit unique functional changes and forms of vascular growth and remodeling. Cancer EVs influence the state of the vasculature both locally and systemically, as exemplified by cancer-associated thrombosis. EV-mediated communication pathways represent attractive targets for therapies aiming at modulation of the tumour-vascular interface (beyond angiogenesis) and could also be exploited for diagnostic purposes in cancer.
Collapse
Affiliation(s)
- Lata Adnani
- McGill University and Research Institute of the McGill University Health Centre, Canada
| | - Cristiana Spinelli
- McGill University and Research Institute of the McGill University Health Centre, Canada
| | - Nadim Tawil
- McGill University and Research Institute of the McGill University Health Centre, Canada
| | - Janusz Rak
- McGill University and Research Institute of the McGill University Health Centre, Canada; Department of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada.
| |
Collapse
|
6
|
Moraes ECDS, Martins-Gonçalves R, da Silva LR, Mandacaru SC, Melo RM, Azevedo-Quintanilha I, Perales J, Bozza FA, Souza TML, Castro-Faria-Neto HC, Hottz ED, Bozza PT, Trugilho MRO. Proteomic Profile of Procoagulant Extracellular Vesicles Reflects Complement System Activation and Platelet Hyperreactivity of Patients with Severe COVID-19. Front Cell Infect Microbiol 2022; 12:926352. [PMID: 35937696 PMCID: PMC9354812 DOI: 10.3389/fcimb.2022.926352] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/20/2022] [Indexed: 01/08/2023] Open
Abstract
Background Extracellular vesicles (EVs) are a valuable source of biomarkers and display the pathophysiological status of various diseases. In COVID-19, EVs have been explored in several studies for their ability to reflect molecular changes caused by SARS-CoV-2. Here we provide insights into the roles of EVs in pathological processes associated with the progression and severity of COVID-19. Methods In this study, we used a label-free shotgun proteomic approach to identify and quantify alterations in EV protein abundance in severe COVID-19 patients. We isolated plasma extracellular vesicles from healthy donors and patients with severe COVID-19 by size exclusion chromatography (SEC). Then, flow cytometry was performed to assess the origin of EVs and to investigate the presence of circulating procoagulant EVs in COVID-19 patients. A total protein extraction was performed, and samples were analyzed by nLC-MS/MS in a Q-Exactive HF-X. Finally, computational analysis was applied to signify biological processes related to disease pathogenesis. Results We report significant changes in the proteome of EVs from patients with severe COVID-19. Flow cytometry experiments indicated an increase in total circulating EVs and with tissue factor (TF) dependent procoagulant activity. Differentially expressed proteins in the disease groups were associated with complement and coagulation cascades, platelet degranulation, and acute inflammatory response. Conclusions The proteomic data reinforce the changes in the proteome of extracellular vesicles from patients infected with SARS-CoV-2 and suggest a role for EVs in severe COVID-19.
Collapse
Affiliation(s)
- Emilly Caroline dos Santos Moraes
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Remy Martins-Gonçalves
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Luana Rocha da Silva
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Samuel Coelho Mandacaru
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Reynaldo Magalhães Melo
- Laboratory Protein Chemistry and Biochemistry and Laboratory of Gene Biology, Department of Cell Biology, University of Brasília, Brasília, Brazil
| | | | - Jonas Perales
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Fernando A. Bozza
- National Institute of Infectious Disease Evandro Chagas, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- D’Or Institute for Research and Education, Rio de Janeiro, Brazil
| | - Thiago Moreno Lopes Souza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | | | - Eugenio D. Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Patricia T. Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
| | - Monique R. O. Trugilho
- Laboratory of Toxinology, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, Brazil
- Center for Technological Development in Health, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Hisada Y, Sachetto ATA, Mackman N. Circulating tissue factor-positive extracellular vesicles and their association with thrombosis in different diseases. Immunol Rev 2022; 312:61-75. [PMID: 35708588 DOI: 10.1111/imr.13106] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/27/2022] [Indexed: 12/23/2022]
Abstract
Tissue factor (TF) is a procoagulant protein released from activated host cells, such as monocytes, and tumor cells on extracellular vesicles (EVs). TF + EVs are observed in the circulation of patients with various types of diseases. In this review, we will summarize the association between TF + EVs and activation of coagulation and survival in different types of diseases, including cancer, sepsis, and infections with different viruses, such as human immunodeficiency virus (HIV), influenza A virus (IAV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We will also discuss the source of TF + EVs in various diseases. EVTF activity is associated with thrombosis in pancreatic cancer patients and coronavirus disease 2019 patients (COVID-19) and with disseminated intravascular coagulation in cancer patients. EVTF activity is also associated with worse survival in patients with cancer and COVID-19. Monocytes are the major sources of TF + EVs in sepsis, and viral infections, such as HIV, Ebola virus, and SARS-CoV-2. In contrast, alveolar epithelial cells are the major source of TF + EVs in bronchoalveolar lavage fluid in COVID-19 and influenza A patients. These studies indicate that EVTF activity could be used as a biomarker to identify patients that have an increased risk of coagulopathy and mortality.
Collapse
Affiliation(s)
- Yohei Hisada
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ana Teresa Azevedo Sachetto
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nigel Mackman
- Division of Hematology, Department of Medicine, UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
8
|
Beck S, Hochreiter B, Schmid JA. Extracellular Vesicles Linking Inflammation, Cancer and Thrombotic Risks. Front Cell Dev Biol 2022; 10:859863. [PMID: 35372327 PMCID: PMC8970602 DOI: 10.3389/fcell.2022.859863] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/21/2022] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) being defined as lipid-bilayer encircled particles are released by almost all known mammalian cell types and represent a heterogenous set of cell fragments that are found in the blood circulation and all other known body fluids. The current nomenclature distinguishes mainly three forms: microvesicles, which are formed by budding from the plasma membrane; exosomes, which are released, when endosomes with intraluminal vesicles fuse with the plasma membrane; and apoptotic bodies representing fragments of apoptotic cells. Their importance for a great variety of biological processes became increasingly evident in the last decade when it was discovered that they contribute to intercellular communication by transferring nucleotides and proteins to recipient cells. In this review, we delineate several aspects of their isolation, purification, and analysis; and discuss some pitfalls that have to be considered therein. Further on, we describe various cellular sources of EVs and explain with different examples, how they link cancer and inflammatory conditions with thrombotic processes. In particular, we elaborate on the roles of EVs in cancer-associated thrombosis and COVID-19, representing two important paradigms, where local pathological processes have systemic effects in the whole organism at least in part via EVs. Finally, we also discuss possible developments of the field in the future and how EVs might be used as biomarkers for diagnosis, and as vehicles for therapeutics.
Collapse
Affiliation(s)
- Sarah Beck
- Institute of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- Institute of Experimental Biomedicine, University Hospital Würzburg and Rudolf Virchow Center for Integrative and Translational Bioimaging, University of Würzburg, Würzburg, Germany
- *Correspondence: Sarah Beck, ; Johannes A. Schmid,
| | - Bernhard Hochreiter
- Institute of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Johannes A. Schmid
- Institute of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
- *Correspondence: Sarah Beck, ; Johannes A. Schmid,
| |
Collapse
|
9
|
Abstract
Cancer-associated thrombosis (including venous thromboembolism (VTE) and arterial events) is highly consequential for patients with cancer and is associated with worsened survival. Despite substantial improvements in cancer treatment, the risk of VTE has increased in recent years; VTE rates additionally depend on the type of cancer (with pancreas, stomach and primary brain tumours having the highest risk) as well as on individual patient's and cancer treatment factors. Multiple cancer-specific mechanisms of VTE have been identified and can be classified as mechanisms in which the tumour expresses proteins that alter host systems, such as levels of platelets and leukocytes, and in which the tumour expresses procoagulant proteins released into the circulation that directly activate the coagulation cascade or platelets, such as tissue factor and podoplanin, respectively. As signs and symptoms of VTE may be non-specific, diagnosis requires clinical assessment, evaluation of pre-test probability, and objective diagnostic testing with ultrasonography or CT. Risk assessment tools have been validated to identify patients at risk of VTE. Primary prevention of VTE (thromboprophylaxis) has long been recommended in the inpatient and post-surgical settings, and is now an option in the outpatient setting for individuals with high-risk cancer. Anticoagulant therapy is the cornerstone of therapy, with low molecular weight heparin or newer options such as direct oral anticoagulants. Personalized treatment incorporating risk of bleeding and patient preferences is essential, especially as a diagnosis of VTE is often considered by patients even more distressing than their cancer diagnosis, and can severely affect the quality of life. Future research should focus on current knowledge gaps including optimizing risk assessment tools, biomarker discovery, next-generation anticoagulant development and implementation science.
Collapse
|
10
|
Hisada Y, Mackman N. Tissue Factor and Extracellular Vesicles: Activation of Coagulation and Impact on Survival in Cancer. Cancers (Basel) 2021; 13:cancers13153839. [PMID: 34359742 PMCID: PMC8345123 DOI: 10.3390/cancers13153839] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary The tissue factor (TF)-factor VIIa complex is the major physiological initiator of blood coagulation. Tumors express TF and release TF-positive extracellular vesicles (EVs) into the circulation, and this is associated with the activation of coagulation. Circulating levels of EVTF activity may be a useful biomarker to identify patients at risk for thrombosis. Tumor TF and TF-positive EVs are also associated with reduced survival. Abstract Tissue factor (TF) is a transmembrane glycoprotein that functions as a receptor for FVII/FVIIa and initiates the extrinsic coagulation pathway. Tumors and cancer cells express TF that can be released in the form of TF positive (TF+) extracellular vesicles (EVs). In this review, we summarize the studies of tumor TF and TF + EVs, and their association with activation of coagulation and survival in cancer patients. We also summarize the role of tumor-derived TF + EVs in venous thrombosis in mouse models. Levels of tumor TF and TF + EVs are associated with venous thromboembolism in pancreatic cancer patients. In addition, levels of EVTF activity are associated with disseminated intravascular coagulation in cancer patients. Furthermore, tumor-derived TF + EVs enhance venous thrombosis in mice. Tumor TF and TF + EVs are also associated with worse survival in cancer patients, particularly in pancreatic cancer patients. These studies indicate that EVTF activity could be used as a biomarker to identify pancreatic cancer patients at risk for venous thrombosis and cancer patients at risk for disseminated intravascular coagulation. EVTF activity may also be a useful prognostic biomarker in cancer patients.
Collapse
|
11
|
Pedersen S, Kristensen AF, Falkmer U, Christiansen G, Kristensen SR. Increased activity of procoagulant factors in patients with small cell lung cancer. PLoS One 2021; 16:e0253613. [PMID: 34288927 PMCID: PMC8294523 DOI: 10.1371/journal.pone.0253613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/08/2021] [Indexed: 12/21/2022] Open
Abstract
Small cell lung cancer (SCLC) patients have augmented risk of developing venous thromboembolism, but the mechanisms triggering this burden on the coagulation system remain to be understood. Recently, cell-derived microparticles carrying procoagulant phospholipids (PPL) and tissue factor (TF) in their membrane have attracted attention as possible contributors to the thrombogenic processes in cancers. The aims of this study were to assess the coagulation activity of platelet-poor plasma from 38 SCLC patients and to provide a detailed procoagulant profiling of small and large extracellular vesicles (EVs) isolated from these patients at the time of diagnosis, during and after treatment compared to 20 healthy controls. Hypercoagulability testing was performed by thrombin generation (TG), procoagulant phospholipid (PPL), TF activity, Protein C, FVIII activity and cell-free deoxyribonucleic acid (cfDNA), a surrogate measure for neutrophil extracellular traps (NETs). Our results revealed a coagulation activity that is significantly increased in the plasma of SCLC patients when compared to age-related healthy controls, but no substantial changes in coagulation activity during treatment and at follow-up. Although EVs in the patients revealed an increased PPL and TF activity compared with the controls, the TG profiles of EVs added to a standard plasma were similar for patients and controls. Finally, we found no differences in the coagulation profile of patients who developed VTE to those who did not, i.e. the tests could not predict VTE. In conclusion, we found that SCLC patients display an overall increased coagulation activity at time of diagnosis and during the disease, which may contribute to their higher risk of VTE.
Collapse
Affiliation(s)
- Shona Pedersen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- * E-mail:
| | - Anne Flou Kristensen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Ursula Falkmer
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Oncology, Aalborg University Hospital, Aalborg, Denmark
| | - Gunna Christiansen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Søren Risom Kristensen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
12
|
Obermann WMJ, Brockhaus K, Eble JA. Platelets, Constant and Cooperative Companions of Sessile and Disseminating Tumor Cells, Crucially Contribute to the Tumor Microenvironment. Front Cell Dev Biol 2021; 9:674553. [PMID: 33937274 PMCID: PMC8085416 DOI: 10.3389/fcell.2021.674553] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Although platelets and the coagulation factors are components of the blood system, they become part of and contribute to the tumor microenvironment (TME) not only within a solid tumor mass, but also within a hematogenous micrometastasis on its way through the blood stream to the metastatic niche. The latter basically consists of blood-borne cancer cells which are in close association with platelets. At the site of the primary tumor, the blood components reach the TME via leaky blood vessels, whose permeability is increased by tumor-secreted growth factors, by incomplete angiogenic sprouts or by vasculogenic mimicry (VM) vessels. As a consequence, platelets reach the primary tumor via several cell adhesion molecules (CAMs). Moreover, clotting factor VII from the blood associates with tissue factor (TF) that is abundantly expressed on cancer cells. This extrinsic tenase complex turns on the coagulation cascade, which encompasses the activation of thrombin and conversion of soluble fibrinogen into insoluble fibrin. The presence of platelets and their release of growth factors, as well as fibrin deposition changes the TME of a solid tumor mass substantially, thereby promoting tumor progression. Disseminating cancer cells that circulate in the blood stream also recruit platelets, primarily by direct cell-cell interactions via different receptor-counterreceptor pairs and indirectly by fibrin, which bridges the two cell types via different integrin receptors. These tumor cell-platelet aggregates are hematogenous micrometastases, in which platelets and fibrin constitute a particular TME in favor of the cancer cells. Even at the distant site of settlement, the accompanying platelets help the tumor cell to attach and to grow into metastases. Understanding the close liaison of cancer cells with platelets and coagulation factors that change the TME during tumor progression and spreading will help to curb different steps of the metastatic cascade and may help to reduce tumor-induced thrombosis.
Collapse
Affiliation(s)
| | | | - Johannes A. Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| |
Collapse
|
13
|
Dhami SPS, Patmore S, O'Sullivan JM. Advances in the Management of Cancer-Associated Thrombosis. Semin Thromb Hemost 2021; 47:139-149. [PMID: 33636745 DOI: 10.1055/s-0041-1722863] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The association between cancer and venous thromboembolism (VTE) has been established for more than 150 years. Nevertheless, cancer-associated thrombosis still remains a major clinical challenge and is associated with significant morbidity and mortality for patients with cancer. The clinical presentation of cancer-associated thrombosis can be distinct from that of a patient without an underlying malignancy. Moreover, specific cancer types, including pancreatic cancer and hematological malignancies, as well as advanced stage disease can confer a significant thrombotic risk. This risk is further augmented by specific anticancer treatment modalities. The pathophysiology of cancer-associated thrombosis is complex and multifactorial. However, understanding the biological mechanisms underpinning VTE risk may provide insight into novel targeted prophylaxis in cancer patients. Over the last decade, low-molecular-weight heparin has been the preferred anticoagulant agent for patients with cancer-associated thrombosis due to improved efficacy compared with vitamin K antagonists. However, the advent of direct oral anticoagulants (DOACs) has added to the repertoire of ammunition now at the disposal of clinicians to aid in the management of cancer-associated thrombosis. Several randomized controlled trials have now been published, demonstrating DOAC as a noninferior alternative for both the treatment and prevention of cancer-associated thrombosis. Notwithstanding this, limitations for their widespread use remain, with the potential for increased bleeding risk, drug interactions, and poor DOAC metabolism. This review discusses the evidence base for the incidence and risk factors associated with VTE in cancer, development, and refinement of risk prediction models and novel advances in the therapeutic management of cancer-associated thrombosis.
Collapse
Affiliation(s)
- Sukhraj Pal Singh Dhami
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Sean Patmore
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Jamie M O'Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| |
Collapse
|
14
|
Rosell A, Havervall S, von Meijenfeldt F, Hisada Y, Aguilera K, Grover SP, Lisman T, Mackman N, Thålin C. Patients With COVID-19 Have Elevated Levels of Circulating Extracellular Vesicle Tissue Factor Activity That Is Associated With Severity and Mortality-Brief Report. Arterioscler Thromb Vasc Biol 2021; 41:878-882. [PMID: 33267656 PMCID: PMC7837685 DOI: 10.1161/atvbaha.120.315547] [Citation(s) in RCA: 158] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/15/2020] [Indexed: 01/23/2023]
Abstract
OBJECTIVE Patients with coronavirus disease 2019 (COVID-19) have a high rate of thrombosis. We hypothesized that severe acute respiratory syndrome coronavirus 2 infection leads to induction of TF (tissue factor) expression and increased levels of circulating TF-positive extracellular vesicles (EV) that may drive thrombosis. Approach and Results: We measured levels of plasma EV TF activity in 100 patients with COVID-19 with moderate and severe disease and 28 healthy controls. Levels of EV TF activity were significantly higher in patients with COVID-19 compared with controls. In addition, levels of EV TF activity were associated with disease severity and mortality. Finally, levels of EV TF activity correlated with several plasma markers, including D-dimer, which has been shown to be associated with thrombosis in patients with COVID-19. CONCLUSIONS Our results indicate that severe acute respiratory syndrome coronavirus 2 infection induces the release of TF-positive EVs into the circulation that are likely to contribute to thrombosis in patients with COVID-19. EV TF activity was also associated with severity and mortality.
Collapse
Affiliation(s)
- Axel Rosell
- Division of Internal Medicine, Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden (A.R., S.H., K.A., C.T.)
| | - Sebastian Havervall
- Division of Internal Medicine, Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden (A.R., S.H., K.A., C.T.)
| | - Fien von Meijenfeldt
- Surgical Research Laboratory, Department of Surgery, University of Groningen, University Medical Center Groningen, the Netherlands (F.v.M., T.L.)
| | - Yohei Hisada
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill (Y.H., S.P.G., N.M.)
| | - Katherina Aguilera
- Division of Internal Medicine, Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden (A.R., S.H., K.A., C.T.)
| | - Steven P. Grover
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill (Y.H., S.P.G., N.M.)
| | - Ton Lisman
- Surgical Research Laboratory, Department of Surgery, University of Groningen, University Medical Center Groningen, the Netherlands (F.v.M., T.L.)
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill (Y.H., S.P.G., N.M.)
| | - Charlotte Thålin
- Division of Internal Medicine, Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden (A.R., S.H., K.A., C.T.)
| |
Collapse
|
15
|
Badimon L, Suades R, Vilella-Figuerola A, Crespo J, Vilahur G, Escate R, Padro T, Chiva-Blanch G. Liquid Biopsies: Microvesicles in Cardiovascular Disease. Antioxid Redox Signal 2020; 33:645-662. [PMID: 31696726 DOI: 10.1089/ars.2019.7922] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Significance: Circulating microvesicles (cMV) are small (0.1-1 μm) phospholipid-rich blebs released by almost all cell types, and their release increases with cell activation and injury, thus reflecting the state of the cell from which they are originated. Microvesicles (MV) are found in the bloodstream, and they affect the phenotype of recipient cells, after local or systemic circulation, by intercellular transfer of their molecular content. Recent Advances: Several studies suggest the use of cell-specific MV subpopulations as predictive biomarkers for cardiovascular diseases (CVDs) at different stages and degrees of severity. In this review, we describe the state of the art of cMV as noninvasive surrogate biomarkers of vascular injury and dysfunction correlated with poor clinical outcomes in CVD. Critical Issues: Despite the growing body of evidence supporting the importance of cMV as hallmarks of CVD and their utility as biomarkers of CVD, the specific roles of each phenotype of cMV in CVD burden and prognosis still remain to be elucidated and validated in large cohorts. In addition, the development of standardized and reproducible techniques is required to be used as biomarkers for disease progression in the clinical setting. Future Directions: A multipanel approach with specific cMV phenotypes, added to current biomarkers and scores, will undoubtedly provide unique prognostic information to stratify patients for appropriate therapy on the basis of their risk of atherothrombotic disease and will open a new research area as therapeutic targets for CVD. MV will add to the implementation of precision medicine by helping the cellular and molecular characterization of CVD patients.
Collapse
Affiliation(s)
- Lina Badimon
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rosa Suades
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,Cardiology Unit, Department of Medicine Solna, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Alba Vilella-Figuerola
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain
| | - Javier Crespo
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain
| | - Gemma Vilahur
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Rafael Escate
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Teresa Padro
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Gemma Chiva-Blanch
- Cardiovascular Program ICCC, Institut de Recerca de l'Hospital Santa Creu i Sant Pau-IIB Sant Pau, Barcelona, Spain
| |
Collapse
|
16
|
Lucotti S, Muschel RJ. Platelets and Metastasis: New Implications of an Old Interplay. Front Oncol 2020; 10:1350. [PMID: 33042789 PMCID: PMC7530207 DOI: 10.3389/fonc.2020.01350] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/26/2020] [Indexed: 12/17/2022] Open
Abstract
During the process of hematogenous metastasis, tumor cells interact with platelets and their precursors megakaryocytes, providing a selection driver for the metastatic phenotype. Cancer cells have evolved a plethora of mechanisms to engage platelet activation and aggregation. Platelet coating of tumor cells in the blood stream promotes the successful completion of multiple steps of the metastatic cascade. Along the same lines, clinical evidence suggests that anti-coagulant therapy might be associated with reduced risk of metastatic disease and better prognosis in cancer patients. Here, we review experimental and clinical literature concerning the contribution of platelets and megakaryocytes to cancer metastasis and provide insights into the clinical relevance of anti-coagulant therapy in cancer treatment.
Collapse
Affiliation(s)
- Serena Lucotti
- Children's Cancer and Blood Foundation Laboratories, Departments of Pediatrics, and Cell and Developmental Biology, Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, United States
| | - Ruth J Muschel
- Cancer Research UK and MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
17
|
Cancer cell-derived tissue factor-positive extracellular vesicles: biomarkers of thrombosis and survival. Curr Opin Hematol 2020; 26:349-356. [PMID: 31261175 DOI: 10.1097/moh.0000000000000521] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Tissue factor (TF) is released from cancer cells and tumors in the form of extracellular vesicles (EVs). This review summarizes our current knowledge of the mechanisms of release of TF-positive EVs (TF+EVs) from cancer cells and the effect of these TF+EVs on cultured endothelial cells. In addition, we will summarize the contribution of TF+EVs to thrombosis in mice, and the association between plasma EVTF activity and venous thrombosis as well as survival of cancer patients. RECENT FINDINGS The release of TF+EVs from cancer cells is regulated by multiple factors, including hypoxia, epithelial-mesenchymal transition, and various intracellular signaling pathways. Cancer cell-derived, TF+EVs confer procoagulant activity to endothelial cells and induce the expression of adhesion proteins and IL-8. In addition, they contribute to thrombosis by directly activating the coagulation system and by generating thrombin that activates platelets in mouse models. Finally, there is an association between EVTF activity and venous thrombosis in pancreatic cancer patients as well as mortality in cancer patients. SUMMARY Cancer cell-derived TF+EVs bind to and activate endothelial cells. In addition, they serve as biomarkers of survival of cancer patients and venous thrombosis in pancreatic cancer patients.
Collapse
|
18
|
Sánchez-López V, Gao L, Ferrer-Galván M, Arellano-Orden E, Elías-Hernández T, Jara-Palomares L, Asensio-Cruz MI, Castro-Pérez MJ, Rodríguez-Martorell FJ, Lobo-Beristain JL, Ballaz-Quincoces A, López-Campos JL, Vila-Liante V, Otero-Candelera R. Differential biomarker profiles between unprovoked venous thromboembolism and cancer. Ann Med 2020; 52:310-320. [PMID: 32634035 PMCID: PMC7877930 DOI: 10.1080/07853890.2020.1779956] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/08/2020] [Accepted: 05/31/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The relationship between cancer and venous thromboembolic disease (VTD) are complex because the activated coagulation factors are not only involved in thrombosis but also in malignant processes, such as angiogenesis and metastasis. OBJECTIVE To compare phenotypes of extracellular vesicles (EVs), and levels of D-dimer, soluble P-selectin (sP-selectin) and antigenic tissue factor (TF) between unprovoked VTD patients, who did not develop cancer during one-year follow-up, and those with advanced stage of cancer but not associated with VTD. METHODS A prospective study in which we included 138 unprovoked VTD patients and 67 advanced cancer patients, who did not develop thrombosis. Levels of EVs of different cellular origin (platelet, endothelium and leukocyte), EVs positive for tissue factor (TF) and P-selectin glycoprotein ligand 1 were quantified by flow cytometry. D-dimer, soluble P-selectin (sP-selectin) and antigenic TF were determined by ELISA. RESULTS TF-positive EVs, D-dimer, and sP-selectin were markedly elevated in unprovoked VTD patients compared to cancer patients without association with thrombosis. CONCLUSIONS Levels of TF-positive EVs, D-dimer and sP-selectin are able to discriminate between unprovoked VTD patients not related to cancer and cancer patients not associated with VTD. These results could lead to the application of EVs as biomarkers of both diseases. Key messages: Circulating EVs, specifically TF-positive EVs, in combination with plasmatic markers of hypercoagulable states, such as D-dimer, sP-selectin and antigen TF, are able to discriminate between cancer patients without thrombosis and patients with unprovoked VTD. Research fields could be opened. Future studies will assess if these biomarkers together serve as predicting thrombotic events in cancer populations.
Collapse
Affiliation(s)
- V. Sánchez-López
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Seville, Spain
| | - L. Gao
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Seville, Spain
| | - M. Ferrer-Galván
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
| | - E. Arellano-Orden
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Seville, Spain
| | - T. Elías-Hernández
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
| | - L. Jara-Palomares
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Seville, Spain
| | - M. I. Asensio-Cruz
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
| | - M. J. Castro-Pérez
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
| | - F. J. Rodríguez-Martorell
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
| | | | | | - J. L. López-Campos
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Seville, Spain
| | - V. Vila-Liante
- Instituto de Investigación Sanitaria, Hospital Universitario y Politécnico La Fe-Valencia, Valencia, Spain
| | - R. Otero-Candelera
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocio/CSIC/Universidad de Sevilla, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Seville, Spain
| |
Collapse
|
19
|
Elevated Microparticles, Thrombin-antithrombin and VEGF Levels in Colorectal Cancer Patients Undergoing Chemotherapy. Pathol Oncol Res 2020; 26:2499-2507. [PMID: 32583332 PMCID: PMC7471181 DOI: 10.1007/s12253-020-00854-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 06/11/2020] [Indexed: 12/11/2022]
Abstract
Hypercoagulable state and neoangiogenesis are common phenomena associated with malignancy. Cancer patients have increased levels of circulating endothelium-derived microparticles (EMPs), which have been hypothesized to be involved in numerous pathophysiological processes. Hemostasis and angiogenesis are also activated in colorectal cancer (CRC) patients. The study aimed to investigate potential influence of chemotherapy on EMPs, thrombin anti-thrombin complex (TAT) and vascular endothelial growth factor (VEGF) levels in CRC patients undergoing chemotherapy. The study group consisted of 18 CRC patients: 8 stage III colon cancer (CC) and 10 stage IV rectal cancer (RC) patients. EMPs, TAT and VEGF levels were assessed before chemotherapy and after the third course. Results were compared with 10 healthy subjects. EMP concentration was measured by flow cytometry, while TAT and VEGF concentrations were assayed employing ELISA. Compared to the control group, CC and RC patients had significantly higher levels of tissue factor (TF)-bearing and non-TF-bearing EMPs before and after three courses of chemotherapy. VEGF concentrations in CRC patients were higher than in the control groups and increased following chemotherapy. TAT levels were elevated in CRC patients before chemotherapy compared to healthy subjects and significantly increased after the third course of chemotherapy. No significant correlation was found either between EMP and TAT levels, or between EMP concentrations and VEGF levels in the study group. CRC patients have increased EMPs, and TAT as well as VEGF levels tend to increase during chemotherapy.
Collapse
|
20
|
Lipets EN, Antonova OA, Shustova ON, Losenkova KV, Mazurov AV, Ataullakhanov FI. Use of Thrombodynamics for revealing the participation of platelet, erythrocyte, endothelial, and monocyte microparticles in coagulation activation and propagation. PLoS One 2020; 15:e0227932. [PMID: 32469873 PMCID: PMC7259734 DOI: 10.1371/journal.pone.0227932] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 05/13/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND OBJECTIVE For many pathological states, microparticles are supposed to be one of the causes of hypercoagulation. Although there are some indirect data about microparticles participation in coagulation activation and propagation, the integral hemostasis test Thrombodynamics allows to measure micropaticles participation in these two coagulation phases directly. Demonstrates microparticles participation in coagulation activation by influence on the appearance of coagulation centres in the plasma volume and the rate of clot growth from the surface with immobilized tissue factor.Methods: Microparticles were obtained from platelets and erythrocytes by stimulation with thrombin receptor-activating peptide (SFLLRN) and calcium ionophore (A23187), respectively, from monocytes, endothelial HUVEC culture and monocytic THP cell culture by stimulation with lipopolysaccharides. Microparticles were counted by flow cytometry and titrated in microparticle-depleted normal plasma in the Thrombodynamics test. RESULTS Monocyte microparticles induced the appearance of clotting centres through the TF pathway at concentrations approximately 100-fold lower than platelet and erythrocyte microparticles, which activated plasma by the contact pathway. For endothelial microparticles, both activation pathways were essential, and their activity was intermediate. Monocyte microparticles induced plasma clotting by the appearance of hundreds of clots with an extremely slow growth rate, while erythrocyte microparticles induced the appearance of a few clots with a growth rate similar to that from surface covered with high-density tissue factor. Patterns of clotting induced by platelet and endothelial microparticles were intermediate. Platelet, erythrocyte and endothelial microparticles impacts on the rate of clot growth from the surface with tissue factor did not differ significantly within the 0-200·103/ul range of microparticles concentrations. However, at concentrations greater than 500·103/ul, erythrocyte microparticles increased the stationary clot growth rate to significantly higher levels than do platelet microparticles or artificial phospholipid vesicles consisting of phosphatidylcholine and phosphatidylserine. CONCLUSION Microparticles of different origins demonstrated qualitatively different characteristics related to coagulation activation and propagation.
Collapse
Affiliation(s)
- E. N. Lipets
- Department of Biophysics, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russian Federation
| | - O. A. Antonova
- Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Russian Ministry of Health, Moscow, Russian Federation
| | - O. N. Shustova
- Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Russian Ministry of Health, Moscow, Russian Federation
| | - K. V. Losenkova
- Medicity Research Laboratory, University of Turku, Turku, Finland
| | - A. V. Mazurov
- Institute of Experimental Cardiology, National Medical Research Center for Cardiology, Russian Ministry of Health, Moscow, Russian Federation
| | - F. I. Ataullakhanov
- Department of Biophysics, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation
- Center for Theoretical Problems of Physicochemical Pharmacology, Russian Academy of Sciences, Moscow, Russian Federation
- Faculty of Physics, Lomonosov Moscow State University, Moscow, Russian Federation
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russian Federation
| |
Collapse
|
21
|
Wang M, Huang X, Zhu M, Zhang S, Zhang Y. High concentration of the plasma microparticles for venous thromboembolism associated with lung cancer. CLINICAL RESPIRATORY JOURNAL 2020; 14:481-487. [PMID: 32031323 DOI: 10.1111/crj.13158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 12/05/2019] [Accepted: 02/02/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Limited biomarkers are used for predicting risk of venous thromboembolism (VTE) associated with cancer. Circulating microparticles (MPs), especially tissue factor- positive microparticles (TF + MPs), play an important role in the development of cancer-associated VTE. This study investigated the predictive value of plasma MPs and TF + MPs for VTE in lung cancer. METHODS A case-control study was performed using the Beijing Chao-Yang Hospital Lung Cancer Registry. Cases had VTE occurring 3 months before or after a diagnosis of lung cancer. Controls were patients with lung cancer without VTE matched for age, histology and stage. The proportion of MPs and TF + MPs was evaluated by light-scatter-based flow cytometry. RESULTS Between January 2012 and December 2015, 30 cases with VTE and 60 controls without VTE were included. The proportion of MPs and TF + MPs was significantly more elevated in patients with VTE than in those without VTE (P < 0.05 for both). By multivariate logistic regression analysis, MPs (OR 1.153; 95% CI 1.068-1.245; P < 0.001) and adenocarcinoma (OR 3.223; 95% CI 1.062-9.782; P = 0.039) were significantly associated with VTE. The sensitivity of the proportion of MPs in diagnosing VTE was 93.3%, and the specificity was 70.0%. The sensitivity of the proportion of TF + MPs in diagnosing VTE was 66.7%, and the specificity was 88.3%. The area under the receiver operating characteristic curves for the diagnostic of the proportion of MPs and TF + MPs values were 0.836 (95% CI 0.750-0.922, P < 0.001) and 0.828 (95% CI 0.736-0.920, P < 0.001) respectively. CONCLUSION The elevated proportion of MPs and TF + MPs might help predict VTE associated with lung cancer.
Collapse
Affiliation(s)
- Meiru Wang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, China
| | - Xiaoxi Huang
- Basic Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Min Zhu
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, China
| | - Shu Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, China
| | - Yuhui Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing, China
| |
Collapse
|
22
|
Dou F, Zhang Y, Yi J, Zhu M, Zhang S, Zhang D, Zhang Y. Association of ALK rearrangement and risk of venous thromboembolism in patients with non-small cell lung cancer: A prospective cohort study. Thromb Res 2019; 186:36-41. [PMID: 31864154 DOI: 10.1016/j.thromres.2019.12.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/08/2019] [Accepted: 12/14/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND Isolated reports are inconsistent regarding the risk of venous thromboembolism (VTE) in patients with anaplastic lymphoma kinase (ALK) rearranged non-small cell lung cancer (NSCLC). This study examined whether ALK rearrangement could have an influence on VTE in a prospective cohort. METHODS In a cohort of 836 consecutive patients with NSCLC, patients with epidermal growth factor receptor (EGFR) or kitten rat sarcoma (KRAS) mutations were ruled out for VTE interference. Finally, 341 qualified patients were observed. The median follow up period is 7.5 months (3.1-15.4m). ALK rearrangement was detected by fluorescence in situ hybridization at baseline. RESULTS Overall VTE events occurred in 37 (10.9%) of 341 patients. In multivariable analysis including age, sex, tumor histology, tumor stage, performance status, and ALK status, ALK rearrangement (sub-distribution hazard radio 2.47, 95% confidence interval 1.04-5.90) was associated with the increased risk of VTE. The cumulative incidence of VTE was 26.9% and 9.2% in the patients with and without ALK rearrangement after 6 months. After 1 year the corresponding cumulative incidence was 26.9% and 9.7% respectively (Gray test P = .005). CONCLUSIONS The presence of ALK rearrangement is associated with increased risk of VTE in patients with NSCLC.
Collapse
Affiliation(s)
- Feifei Dou
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing 100020, China
| | - Yuan Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing 100020, China
| | - Jiawen Yi
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing 100020, China
| | - Min Zhu
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing 100020, China
| | - Shu Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing 100020, China
| | - Di Zhang
- Department of Clinical Epidemiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yuhui Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing Institute of Respiratory Medicine, Beijing 100020, China.
| |
Collapse
|
23
|
Kobayashi K, Baba K, Igase M, Primarizky H, Nemoto Y, Shimokawa Miyama T, Kambayashi S, Mizuno T, Okuda M. Tissue factor procoagulant activity in the tumor cell lines and plasma of dogs with various malignant tumors. J Vet Med Sci 2019; 81:1713-1721. [PMID: 31611484 PMCID: PMC6943321 DOI: 10.1292/jvms.19-0400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hypercoagulability is a common paraneoplastic complication in dogs with various malignant tumors. Importantly, tissue factor procoagulant activity (TF-PCA) induced by TF-bearing microparticles (TF-MPs) is associated with hypercoagulability in human patients with cancer. However, TF-PCA in tumor cells and the association between circulating TF-MPs and hypercoagulability in dogs with malignant tumors remain poorly understood. Therefore, the present study was conducted to evaluate the TF-PCA in various types of canine tumor cell lines and plasma in dogs with malignant tumors. Mammary gland tumor, hemangiosarcoma, and malignant melanoma cell lines, but not lymphoma cell lines, expressed TF on their surfaces and showed cellular surface and MP-associated TF-PCA. The plasma TF-PCA was elevated in some dogs that naturally developed such tumors. No significant difference was observed in plasma TF-PCA between the disseminated intravascular coagulation (DIC) group (median: 43.40; range: 3.47-85.19; n=5) and non-DIC group (median: 7.73; range: 1.70-16.13; n=12). However, plasma TF-PCA was remarkably elevated in three of five dogs with DIC. To the best of our knowledge, this is the first study to evaluate plasma TF-PCA in dogs with malignant tumors. Further studies must be conducted to determine the cellular origin of TF-MPs and the efficacy of plasma TF-PCA as a biomarker of DIC in dogs with malignant tumors.
Collapse
Affiliation(s)
- Kosuke Kobayashi
- Laboratory of Veterinary Internal Medicine, The United Graduate School of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8511, Japan
| | - Kenji Baba
- Laboratory of Veterinary Internal Medicine, The United Graduate School of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8511, Japan
| | - Masaya Igase
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi 753-8511, Japan
| | - Hardany Primarizky
- Veterinary Clinical Department, Faculty of Veterinary Medicine, Universitas Airlangga, Campus "C" Unair, Surabaya, East Java 60115, Indonesia
| | - Yuki Nemoto
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi 753-8511, Japan
| | - Takako Shimokawa Miyama
- Laboratory of Veterinary Internal Medicine, The United Graduate School of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8511, Japan
| | - Satoshi Kambayashi
- Laboratory of Veterinary Internal Medicine, The United Graduate School of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8511, Japan
| | - Takuya Mizuno
- Laboratory of Molecular Diagnostics and Therapeutics, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1, Yoshida, Yamaguchi 753-8511, Japan
| | - Masaru Okuda
- Laboratory of Veterinary Internal Medicine, The United Graduate School of Veterinary Science, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8511, Japan
| |
Collapse
|
24
|
Vallier L, Bouriche T, Bonifay A, Judicone C, Bez J, Franco C, Guervilly C, Hisada Y, Mackman N, Houston R, Poncelet P, Dignat-George F, Lacroix R. Increasing the sensitivity of the human microvesicle tissue factor activity assay. Thromb Res 2019; 182:64-74. [PMID: 31450010 DOI: 10.1016/j.thromres.2019.07.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 06/10/2019] [Accepted: 07/14/2019] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The TF-FVIIa complex is the primary activator of coagulation. Elevated levels of microvesicle (MV) bearing tissue factor (TF)-dependent procoagulant activity are detectable in patients with an increased risk of thrombosis. Several methods have been described to measure MV TF activity but they are hampered by limited sensitivity and specificity. The aim of this work was to increase the sensitivity of the MV TF activity assay (called Chapel Hill assay). MATERIAL AND METHODS Improvements of the MV TF activity assay included i/ speed and time of centrifugation, ii/ use of a more potent inhibitory anti-TF antibody iii/ use of FVII and a fluorogenic substrate to increase specificity. RESULTS The specificity of the MV TF activity assay was demonstrated by the absence of activity on MV derived from a knock-out-TF cell line using an anti-human TF monoclonal antibody called SBTF-1, which shows a higher TF inhibitory effect than the anti-human TF monoclonal antibody called HTF-1. Experiments using blood from healthy individuals, stimulated or not by LPS, or plasma spiked with 3 different levels of MV, demonstrated that the new assay was more sensitive and this allowed detection of MV TF activity in platelet free plasma (PFP) samples from healthy individuals. However, the assay was limited by an inter-assay variability, mainly due to the centrifugation step. CONCLUSIONS We have improved the sensitivity of the MV TF activity assay without losing specificity. This new assay could be used to evaluate levels of TF-positive MV as a potential biomarker of thrombotic risk in patients.
Collapse
Affiliation(s)
- Loris Vallier
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France
| | - Tarik Bouriche
- Research and Technology Department, BioCytex, Marseille, France
| | | | - Coralie Judicone
- Research and Technology Department, BioCytex, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | - Jeremy Bez
- Research and Technology Department, BioCytex, Marseille, France
| | - Corentin Franco
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France; Research and Technology Department, BioCytex, Marseille, France
| | | | - Yohei Hisada
- Division of Hematology and Oncology, Thrombosis and Hemostasis Program, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Nigel Mackman
- Division of Hematology and Oncology, Thrombosis and Hemostasis Program, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Reaves Houston
- Division of Hematology and Oncology, Thrombosis and Hemostasis Program, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | | | - Françoise Dignat-George
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France.
| | - Romaric Lacroix
- Aix Marseille Univ, INSERM, INRA, C2VN, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| |
Collapse
|
25
|
Mechanisms of cancer-associated thrombosis. Hemasphere 2019; 3:HemaSphere-2019-0056. [PMID: 35309769 PMCID: PMC8925691 DOI: 10.1097/hs9.0000000000000239] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 03/31/2019] [Indexed: 12/21/2022] Open
|
26
|
Zarà M, Guidetti GF, Camera M, Canobbio I, Amadio P, Torti M, Tremoli E, Barbieri SS. Biology and Role of Extracellular Vesicles (EVs) in the Pathogenesis of Thrombosis. Int J Mol Sci 2019; 20:ijms20112840. [PMID: 31212641 PMCID: PMC6600675 DOI: 10.3390/ijms20112840] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 06/07/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are well-established mediators of cell-to-cell communication. EVs can be released by every cell type and they can be classified into three major groups according to their biogenesis, dimension, density, and predominant protein markers: exosomes, microvesicles, and apoptotic bodies. During their formation, EVs associate with specific cargo from their parental cell that can include RNAs, free fatty acids, surface receptors, and proteins. The biological function of EVs is to maintain cellular and tissue homeostasis by transferring critical biological cargos to distal or neighboring recipient cells. On the other hand, their role in intercellular communication may also contribute to the pathogenesis of several diseases, including thrombosis. More recently, their physiological and biochemical properties have suggested their use as a therapeutic tool in tissue regeneration as well as a novel option for drug delivery. In this review, we will summarize the impact of EVs released from blood and vascular cells in arterial and venous thrombosis, describing the mechanisms by which EVs affect thrombosis and their potential clinical applications.
Collapse
Affiliation(s)
- Marta Zarà
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| | | | - Marina Camera
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milano, Italy.
- Unit of Cell and Molecular Biology in Cardiovascular Diseases, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| | - Ilaria Canobbio
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy.
| | - Patrizia Amadio
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| | - Mauro Torti
- Department of Biology and Biotechnology, University of Pavia, 27100 Pavia, Italy.
| | - Elena Tremoli
- Scientific Direction, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| | - Silvia Stella Barbieri
- Unit of Heart-Brain Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138 Milano, Italy.
| |
Collapse
|
27
|
Riondino S, Ferroni P, Zanzotto FM, Roselli M, Guadagni F. Predicting VTE in Cancer Patients: Candidate Biomarkers and Risk Assessment Models. Cancers (Basel) 2019; 11:cancers11010095. [PMID: 30650562 PMCID: PMC6356247 DOI: 10.3390/cancers11010095] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 12/07/2018] [Accepted: 01/08/2019] [Indexed: 02/07/2023] Open
Abstract
Risk prediction of chemotherapy-associated venous thromboembolism (VTE) is a compelling challenge in contemporary oncology, as VTE may result in treatment delays, impaired quality of life, and increased mortality. Current guidelines do not recommend thromboprophylaxis for primary prevention, but assessment of the patient's individual risk of VTE prior to chemotherapy is generally advocated. In recent years, efforts have been devoted to building accurate predictive tools for VTE risk assessment in cancer patients. This review focuses on candidate biomarkers and prediction models currently under investigation, considering their advantages and disadvantages, and discussing their diagnostic performance and potential pitfalls.
Collapse
Affiliation(s)
- Silvia Riondino
- Interinstitutional Multidisciplinary Biobank, IRCCS San Raffaele Pisana, 00166 Rome, Italy.
- Department of Systems Medicine, Medical Oncology, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Patrizia Ferroni
- Interinstitutional Multidisciplinary Biobank, IRCCS San Raffaele Pisana, 00166 Rome, Italy.
- Department of Human Sciences & Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy.
| | - Fabio Massimo Zanzotto
- Department of Enterprise Engineering, University of Rome "Tor Vergata", 00133 Rome, Italy.
| | - Mario Roselli
- Department of Systems Medicine, Medical Oncology, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Fiorella Guadagni
- Interinstitutional Multidisciplinary Biobank, IRCCS San Raffaele Pisana, 00166 Rome, Italy.
- Department of Human Sciences & Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy.
| |
Collapse
|
28
|
Abstract
Venous thromboembolism is known to be associated with an increase in morbidity and mortality in patients with malignancy. Predictive laboratory biomarkers of venous thromboembolism (VTE) have long been sought after to improve outcomes and help guide clinical decision making. Previously studied biomarkers include C reactive protein (CRP), tissue factor expressing microparticles (TF MP), D-dimer, soluble P-selectin (sP-selectin), plasminogen activator inhibitor 1 (PAI-1), factor VIII, platelet count, and leukocyte counts. This chapter will focus on these possible biomarkers for cancer-associated thrombosis (CAT) with particular emphasis on the pathophysiology behind thrombosis formation as well as data from clinical studies in patients with malignancy. The incorporation of the above biomarkers into risk assessment tools to predict CAT will also be reviewed, as will risk factors for recurrent VTE in patients with malignancy. Further studies are ongoing to develop readily available biomarkers that can be incorporated into future risk assessment models with the goal of reducing morbidity and mortality due to cancer-associated thrombosis.
Collapse
Affiliation(s)
- Anjlee Mahajan
- Division of Hematology and Oncology, UC Davis School of Medicine, UC Davis Cancer Center, 4501 X Street, Sacramento, CA, 95817, USA.
| | - Ted Wun
- Division of Hematology and Oncology, UC Davis School of Medicine, UC Davis Cancer Center, 4501 X Street, Sacramento, CA, 95817, USA
- UC Davis School of Medicine, Clinical and Translational Sciences Center (CTSC), Sacramento, USA
| |
Collapse
|
29
|
Mantia C, Zwicker JI. Anticoagulation in the Setting of Primary and Metastatic Brain Tumors. Cancer Treat Res 2019; 179:179-189. [PMID: 31317488 DOI: 10.1007/978-3-030-20315-3_12] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Venous thromboembolism is commonly diagnosed in patients with primary and secondary brain tumors. Anticoagulation management in the setting of brain tumors is complicated by the high background rate of spontaneous intracranial hemorrhage. Until recently, there was limited evidence to support the decision to administer therapeutic anticoagulation in the setting of brain metastases or primary brain tumors. The current evidence suggests that the safety profile of therapeutic low molecular weight heparin for the treatment of venous thromboembolism is contingent on whether the origin of brain tumor is primary (i.e., glioma) versus secondary. In patients with brain metastases, the rate of intracranial hemorrhage often exceeds 20% but is not influenced by the administration of low molecular weight heparin. In contrast, in primary brain tumors such as glioma, therapeutic anticoagulation is associated with an increased risk of intracranial hemorrhage that can negatively impact survival. This chapter reviews the underlying mechanisms contributing to thrombosis and hemorrhage in brain tumors and summarizes the current evidence and approaches in anticoagulation to treat venous thromboembolism.
Collapse
Affiliation(s)
- Charlene Mantia
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Jeffrey I Zwicker
- Division of Hemostasis and Thrombosis, Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA, 02215, USA.
| |
Collapse
|
30
|
Hisada Y, Mackman N. Measurement of tissue factor activity in extracellular vesicles from human plasma samples. Res Pract Thromb Haemost 2019; 3:44-48. [PMID: 30656275 PMCID: PMC6332748 DOI: 10.1002/rth2.12165] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/13/2018] [Indexed: 11/11/2022] Open
Abstract
Tissue factor (TF) is the cellular receptor for plasma factor (F) VII/FVIIa and triggers blood coagulation. Extracellular vesicles (EVs) are small membrane vesicles that are released from activated cells and tumor cells. Different cell types, including activated monocytes and tumors cells release TF-positive EVs into the circulation. We developed an assay to measure levels of TF activity in EVs isolated from human plasma samples. We and others have used this assay to demonstrate increased levels of EV TF activity in a variety of diseases associated with thrombosis, including cancer. These studies suggest that EV TF can be used as a biomarker of thrombotic risk. The strength of this laboratory assay is that it is relatively sensitive and specific. However, the limitations of the assay are that it is labor intensive and the coefficient of variation is too high for it to be used as a clinical assay.
Collapse
Affiliation(s)
- Yohei Hisada
- Division of Hematology and Oncology, Thrombosis and Hemostasis ProgramDepartment of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| | - Nigel Mackman
- Division of Hematology and Oncology, Thrombosis and Hemostasis ProgramDepartment of MedicineUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
- Department of Pathology and Laboratory MedicineUniversity of North Carolina at Chapel HillChapel HillNorth Carolina
| |
Collapse
|
31
|
Buchanan IA, Lin M, Donoho DA, Patel A, Ding L, Amar AP, Giannotta SL, Mack WJ, Attenello F. Predictors of Venous Thromboembolism After Nonemergent Craniotomy: A Nationwide Readmission Database Analysis. World Neurosurg 2018; 122:e1102-e1110. [PMID: 30465948 DOI: 10.1016/j.wneu.2018.10.237] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/29/2018] [Accepted: 10/31/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Venous thromboembolism (VTE) is responsible for many hospital readmissions each year, particularly among postsurgical cohorts. Because early and indiscriminate VTE prophylaxis carries catastrophic consequences in postcraniotomy cohorts, identifying factors associated with a high risk for thromboembolic complications is important for guiding postoperative management. OBJECTIVE To determine VTE incidence in patients undergoing nonemergent craniotomy and to evaluate for factors that predict 30-day and 90-day readmission with VTE. METHODS The 2010-2014 cohorts of the Nationwide Readmissions Database were used to generate a large heterogeneous craniotomy sample. RESULTS There were 89,450 nonemergent craniotomies that met inclusion criteria. Within 30 days, 1513 patients (1.69%) were readmitted with VTE diagnoses; among them, 678 (44.8%) had a diagnosis of deep vein thrombosis alone, 450 (29.7%) had pulmonary embolism alone, and 385 (25.4%) had both. The corresponding 30-day deep vein thrombosis and pulmonary embolism incidences were 1.19% and 0.93%, respectively. In multivariate analysis, several factors were significantly associated with VTE readmission, namely, craniotomy for tumor, corticosteroids, advanced age, greater length of stay, and discharge to institutional care. CONCLUSIONS Craniotomies for tumor, corticosteroids, advanced age, prolonged length of stay, and discharge to institutional care are significant predictors of VTE readmission. The implication of steroids, coupled with their ubiquity in neurosurgery, makes them a potentially modifiable risk factor and a prime target for VTE reduction in craniotomy cohorts. Furthermore, the fact that dose is proportional to VTE risk in the literature suggests that careful consideration should be given toward decreasing regimens in situations in which use of a lower dose might prove equally sufficient.
Collapse
Affiliation(s)
- Ian A Buchanan
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.
| | - Michelle Lin
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Daniel A Donoho
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA; Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Arati Patel
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Li Ding
- Departments of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Arun P Amar
- Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Steven L Giannotta
- Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - William J Mack
- Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Frank Attenello
- Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
32
|
Cedervall J, Hamidi A, Olsson AK. Platelets, NETs and cancer. Thromb Res 2018; 164 Suppl 1:S148-S152. [PMID: 29703474 DOI: 10.1016/j.thromres.2018.01.049] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/25/2018] [Accepted: 01/28/2018] [Indexed: 12/14/2022]
Abstract
In addition to the central role of platelets in hemostasis, they contribute to pathological conditions such as inflammation and tumor progression. Aberrant expression and/or exposure of pro-coagulant factors in the tumor microenvironment induce platelet activation and subsequent release of growth factors from platelet granules. Cancer patients are commonly affected by thrombotic events, as a result of tumor-induced platelet activation. A novel player potentially contributing to cancer-associated thrombosis is the formation of neutrophil extracellular traps (NETs). NETs are composed of externalized DNA of nuclear or mitochondrial origin, bound to histones and granular proteases such as neutrophil elastase (NE) and myeloperoxidase (MPO). These extracellular traps help neutrophils to catch and kill pathogens such as bacteria, virus and fungi. It is now clear that NETs form also under conditions of sterile inflammation such as cancer and autoimmunity and can promote thrombosis. Recent data show that platelets play a key role in determining when and where NETs should form. This review will highlight our current insight in the role of platelets as regulators of NET formation, both during infection and sterile inflammation.
Collapse
Affiliation(s)
- Jessica Cedervall
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Box 582, SE-751 23 Uppsala, Sweden
| | - Anahita Hamidi
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Box 582, SE-751 23 Uppsala, Sweden
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, Biomedical Center, Box 582, SE-751 23 Uppsala, Sweden.
| |
Collapse
|
33
|
Yeung J, Li W, Holinstat M. Platelet Signaling and Disease: Targeted Therapy for Thrombosis and Other Related Diseases. Pharmacol Rev 2018; 70:526-548. [PMID: 29925522 PMCID: PMC6013590 DOI: 10.1124/pr.117.014530] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Platelets are essential for clotting in the blood and maintenance of normal hemostasis. Under pathologic conditions such as atherosclerosis, vascular injury often results in hyperactive platelet activation, resulting in occlusive thrombus formation, myocardial infarction, and stroke. Recent work in the field has elucidated a number of platelet functions unique from that of maintaining hemostasis, including regulation of tumor growth and metastasis, inflammation, infection, and immune response. Traditional therapeutic targets for inhibiting platelet activation have primarily been limited to cyclooxygenase-1, integrin αIIbβ3, and the P2Y12 receptor. Recently identified signaling pathways regulating platelet function have made it possible to develop novel approaches for pharmacological intervention in the blood to limit platelet reactivity. In this review, we cover the newly discovered roles for platelets as well as their role in hemostasis and thrombosis. These new roles for platelets lend importance to the development of new therapies targeted to the platelet. Additionally, we highlight the promising receptor and enzymatic targets that may further decrease platelet activation and help to address the myriad of pathologic conditions now known to involve platelets without significant effects on hemostasis.
Collapse
Affiliation(s)
- Jennifer Yeung
- Departments of Pharmacology (J.Y., W.L., M.H.) and Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor, Michigan
| | - Wenjie Li
- Departments of Pharmacology (J.Y., W.L., M.H.) and Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor, Michigan
| | - Michael Holinstat
- Departments of Pharmacology (J.Y., W.L., M.H.) and Internal Medicine, Division of Cardiovascular Medicine (M.H.), University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
34
|
Haemmerle M, Stone RL, Menter DG, Afshar-Kharghan V, Sood AK. The Platelet Lifeline to Cancer: Challenges and Opportunities. Cancer Cell 2018; 33:965-983. [PMID: 29657130 PMCID: PMC5997503 DOI: 10.1016/j.ccell.2018.03.002] [Citation(s) in RCA: 392] [Impact Index Per Article: 65.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 01/08/2018] [Accepted: 03/01/2018] [Indexed: 12/21/2022]
Abstract
Besides their function in limiting blood loss and promoting wound healing, experimental evidence has highlighted platelets as active players in all steps of tumorigenesis including tumor growth, tumor cell extravasation, and metastasis. Additionally, thrombocytosis in cancer patients is associated with adverse patient survival. Due to the secretion of large amounts of microparticles and exosomes, platelets are well positioned to coordinate both local and distant tumor-host crosstalk. Here, we present a review of recent discoveries in the field of platelet biology and the role of platelets in cancer progression as well as challenges in targeting platelets for cancer treatment.
Collapse
Affiliation(s)
- Monika Haemmerle
- Department of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Institute of Pathology, Martin Luther University Halle-Wittenberg, 06112 Halle, Germany
| | - Rebecca L Stone
- Department of Obstetrics and Gynecology, Johns Hopkins Hospital, Baltimore, MD 21287-1281, USA
| | - David G Menter
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Benign Hematology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Anil K Sood
- Department of Gynecologic Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for RNA Interference and Non-Coding RNA, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
35
|
Muhsin-Sharafaldine MR, McLellan AD. Apoptotic vesicles: deathly players in cancer-associated coagulation. Immunol Cell Biol 2018; 96:723-732. [PMID: 29738615 DOI: 10.1111/imcb.12162] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/01/2018] [Accepted: 05/02/2018] [Indexed: 12/27/2022]
Abstract
Although cancer is associated with coagulation disorders, it is still unclear how the combination of tumor cell and host factors enhance the hypercoagulable state of cancer patients. Emerging evidence points to a central role for tumor endosomal and plasma membrane-derived vesicular components in the pathogenesis of cancer-related thrombosis. In particular, tumor cell membranes and extracellular vesicles (EV) harbor lipids and proteinaceous coagulation factors able to initiate multiple points within the coagulation matrix. The impact of chemotherapy upon a host already burdened with a hypercoagulable state increases the risk of pathological coagulation. We argue that chemotherapy-induced EV harbor the most active components for cancer related thrombosis and discuss how membrane components of the host and tumor act to initiate coagulation to enhance thrombotic risk in cancer patients.
Collapse
|
36
|
Muhsin-Sharafaldine MR, McLellan AD. Tumor-Derived Apoptotic Vesicles: With Death They Do Part. Front Immunol 2018; 9:957. [PMID: 29780392 PMCID: PMC5952256 DOI: 10.3389/fimmu.2018.00957] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/17/2018] [Indexed: 12/21/2022] Open
Abstract
Tumor cells release lipid particles known as extracellular vesicles (EV) that contribute to cancer metastasis, to the immune response, and to thrombosis. When tumors are exposed to radiation or chemotherapy, apoptotic vesicles (ApoVs) are released in abundance as the plasma membrane delaminates from the cytoskeleton. Recent studies have suggested that ApoVs are distinct from the EVs released from living cells, such as exosomes or microvesicles. Depending on their treatment conditions, tumor-released ApoV have been suggested to either enhance or suppress anti-cancer immunity. In addition, tumor-derived ApoV possess procoagulant activity that could increase the thrombotic state in cancer patients undergoing chemotherapy or radiotherapy. Since ApoVs are one of the least appreciated type of EVs, we focus in this review on the distinctive characterization of tumor ApoVs and their proposed mechanistic effects on cancer immunity, coagulation, and metastasis.
Collapse
Affiliation(s)
| | - Alexander D McLellan
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
37
|
Garcia-Romero N, Esteban-Rubio S, Rackov G, Carrión-Navarro J, Belda-Iniesta C, Ayuso-Sacido A. Extracellular vesicles compartment in liquid biopsies: Clinical application. Mol Aspects Med 2018; 60:27-37. [DOI: 10.1016/j.mam.2017.11.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/08/2017] [Accepted: 11/14/2017] [Indexed: 02/07/2023]
|
38
|
Abstract
INTRODUCTION Increased levels of circulating tissue factor (TF)-positive microvesicles (MVs) may increase the risk of thrombosis. Indeed, TF-positive MVs are detected in plasma of patients with various types of diseases. In this study, we measured levels of MV TF activity in non-cancer severely ill patients and cancer patients. METHODS We used an in-house MV TF activity assay to measure MV TF activity. RESULTS MV TF activity was significantly increased in a population of cancer patients but not in a population of non-cancer severely ill patients compared with healthy controls. However, in the population of severely ill patients, those with infection had significantly elevated levels of MV TF activity compared with controls. Interestingly, patients with adenocarcinoma had higher levels of MV TF activity compared with patients with non-adenocarcinoma tumors. Levels of MV TF activity were not associated with venous thromboembolism in cancer patients. MV TF activity was associated with reduced survival in cancer patients. CONCLUSION Cancer patients as well as severely ill patients with infection have higher levels of MV TF activity compared with healthy controls. Patients with adenocarcinoma have higher levels of MV TF activity compared with patients with other types of cancer. An elevated level of MV TF activity was associated with reduced survival in cancer patients.
Collapse
|
39
|
Grover SP, Mackman N. Tissue Factor: An Essential Mediator of Hemostasis and Trigger of Thrombosis. Arterioscler Thromb Vasc Biol 2018; 38:709-725. [PMID: 29437578 DOI: 10.1161/atvbaha.117.309846] [Citation(s) in RCA: 437] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 01/25/2018] [Indexed: 12/21/2022]
Abstract
Tissue factor (TF) is the high-affinity receptor and cofactor for factor (F)VII/VIIa. The TF-FVIIa complex is the primary initiator of blood coagulation and plays an essential role in hemostasis. TF is expressed on perivascular cells and epithelial cells at organ and body surfaces where it forms a hemostatic barrier. TF also provides additional hemostatic protection to vital organs, such as the brain, lung, and heart. Under pathological conditions, TF can trigger both arterial and venous thrombosis. For instance, atherosclerotic plaques contain high levels of TF on macrophage foam cells and microvesicles that drives thrombus formation after plaque rupture. In sepsis, inducible TF expression on monocytes leads to disseminated intravascular coagulation. In cancer patients, tumors release TF-positive microvesicles into the circulation that may contribute to venous thrombosis. TF also has nonhemostatic roles. For instance, TF-dependent activation of the coagulation cascade generates coagulation proteases, such as FVIIa, FXa, and thrombin, which induce signaling in a variety of cells by cleavage of protease-activated receptors. This review will focus on the roles of TF in protective hemostasis and pathological thrombosis.
Collapse
Affiliation(s)
- Steven P Grover
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Nigel Mackman
- From the Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill.
| |
Collapse
|
40
|
Sun DS, Lee HK, Kim Y, Lee JH, Won HS, Ko YH, Kim H. Circulating Microparticles and Coagulation Profiles in Patients with Advanced Stage Solid Tumors. Ann Lab Med 2018; 38:274-276. [PMID: 29401565 PMCID: PMC5820075 DOI: 10.3343/alm.2018.38.3.274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/12/2017] [Accepted: 12/28/2017] [Indexed: 11/19/2022] Open
Affiliation(s)
- Der Sheng Sun
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hae Kyung Lee
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yonggoo Kim
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Je Hoon Lee
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hye Sung Won
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yoon Ho Ko
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyunjung Kim
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
41
|
Tissue Factor-bearing MPs and the risk of venous thrombosis in cancer patients: A meta-analysis. Sci Rep 2018; 8:1675. [PMID: 29374212 PMCID: PMC5786054 DOI: 10.1038/s41598-018-19889-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 01/09/2018] [Indexed: 01/18/2023] Open
Abstract
Cancer patients with Tissue Factor (TF)-bearing MPs have been presented association with increased risk of venous thromboembolism (VTE), but results of these studies have not been consistent. We aimed to conduct a meta-analysis to assess the relationship between TF-bearing MPs and risk of VTE in patients with cancer. PubMed, Web of Science and EMBASE Databases were systematically retrieved up to1th June 2017. Two case-control studies and four cohort studies met the entry requirements in this analysis. The summary odd ratio (OR) were estimated by a random effect model. The overall OR was 1.76 (95% CI: 1.21-2.56, I2 = 62.0%). The OR of case-control studies was 3.41 (95% CI: 1.45-8.02, I2 = 0.0%) and that of cohort studies was1.53 (95% CI: 1.05-2.24, I2 = 66.1%). The association between TF-bearing MPs and the risk of VTE in cancer patients was found in this meta-analysis. Publication bias testing and sensitivity subgroup analysis suggested that results of this meta-analysis were robustness. In conclusion, TF-bearing MPs were associated with increased risk of VTE in patients with cancer. Whereas, more well-designed studies and more comprehensive adjustments for confounders in further studies are warranted to affirm the association.
Collapse
|
42
|
Hisada Y, Mackman N. Mouse models of cancer-associated thrombosis. Thromb Res 2017; 164 Suppl 1:S48-S53. [PMID: 29306575 DOI: 10.1016/j.thromres.2017.12.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/22/2017] [Accepted: 12/27/2017] [Indexed: 10/18/2022]
Abstract
Cancer patients have an increased risk of venous thromboembolism (VTE) compared with the general population. Mouse models are used to better understand the mechanisms of cancer-associated thrombosis. Several mouse models of cancer-associated thrombosis have been developed that use different mouse strains, tumors, tumor sites and thrombosis models. In this review, we summarize these different models. These models have been used to determine the role of different pathways in cancer-associated thrombosis. For instance, they have revealed roles for tumor-derived tissue factor-positive microvesicles and neutrophil extracellular traps in thrombosis in tumor-bearing mice. A better understanding of the mechanisms of cancer-associated thrombosis may allow the development of new therapies to reduce thrombosis in cancer patients.
Collapse
Affiliation(s)
- Yohei Hisada
- Department of Medicine, Division of Hematology and Oncology, Thrombosis and Hemostasis Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - Nigel Mackman
- Department of Medicine, Division of Hematology and Oncology, Thrombosis and Hemostasis Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
43
|
Bokemeyer C, Langer F. Crosstalk between cancer and haemostasis. Hamostaseologie 2017; 32:95-104. [DOI: 10.5482/ha-1160] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2011] [Accepted: 06/20/2011] [Indexed: 12/14/2022] Open
Abstract
SummaryCancer is characterized by bidirectional interrelations between tumour progression, coagulation activation, and inflammation. Tissue factor (TF), the principal initiator of the coagulation protease cascade, is centrally positioned in this complex triangular network due to its pleiotropic effects in haemostasis, angiogenesis, and haematogenous metastasis. While formation of macroscopic thrombi is the correlate of cancer-associated venous thromboembolism (VTE), a major healthcare burden in clinical haematology and oncology, microvascular thrombosis appears to be critically important to blood-borne tumour cell dissemination. In this regard, expression of TF in malignant tissues as well as shedding of TFbearing microparticles into the circulation are thought to be regulated by defined genetic events relevant to pathological cancer progression, thus directly linking Trousseau’s syndrome to molecular tumourigenesis.Because pharmacological inhibition of the TF pathway in selective tumour types and patient subgroups would be in line with the modern concept of individualized, targeted anti-cancer therapy, this review will focus on the role of TF in tumour biology and cancer-associated VTE.
Collapse
|
44
|
Campello E, Spiezia L, Radu CM, Bon M, Gavasso S, Zerbinati P, Woodhams B, Tormene D, Prandoni P, Simioni P. Circulating microparticles in carriers of factor V Leiden with and without a history of venous thrombosis. Thromb Haemost 2017; 108:633-9. [DOI: 10.1160/th12-05-0280] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 07/03/2012] [Indexed: 11/05/2022]
Abstract
SummaryAlthough factor V Leiden (FVL) is a major determinant of thrombotic risk, the reason why less than 10% of carriers eventually develop venous thromboembolic (VTE) events is unknown. Recent observations suggest that circulating levels of microparticles (MP) may contribute to the thrombogenic profile of FVL carriers. We measured the plasma level of annexin V-MP (AMP) platelet-MP (PMP), endothelial-MP (EMP), leukocyte-MP (LMP) and tissue factor-bearing MP (TF+MP), and the MP procoagulant activity (PPL) in 142 carriers of FVL (of these 30 homozygous and 49 with prior VTE), and in 142 age and gender-matched healthy individuals. The mean (± SD) level of AMP was 2,802 ± 853 MP/ μl in carriers and 1,682 ± 897 in controls (p<0.0001). A statistically significant difference between homozygous and heterozygous carriers of FVL was seen in the level of PMP, EMP and LMP, but not in that of the remaining parameters. When the analysis was confined to carriers with and without a VTE history, the mean level of AMP was 3,110 ± 791 MP/ μl in the former, and 2,615 ± 839 MP/μl in the latter (p<0.005). The mean level of all subtypes of circulating MP showed a similar pattern. The PPL clotting time was 39 ± 9 seconds (sec) in carriers, and 52 ± 15 sec in controls (p=0.003); and was 35 ± 8 sec in carriers with prior thrombosis, and 41 ± 10 sec in thrombosis-free carriers (p<0.005). Our study results suggest that circulating MP may contribute to the development of thrombosis in carriers of FVL mutation.
Collapse
|
45
|
Hernández C, Orbe J, Roncal C, Alvarez-Hernandez M, de Lizarrondo SM, Alves MT, Mata JG, Páramo JA. Tissue factor expressed by microparticles is associated with mortality but not with thrombosis in cancer patients. Thromb Haemost 2017; 110:598-608. [DOI: 10.1160/th13-02-0122] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 06/07/2013] [Indexed: 01/08/2023]
Abstract
SummaryA prothrombotic state is one of the hallmarks of malignancy and a major contributor to morbidity and mortality in cancer patients. Tissue factor (TF) is often overexpressed in malignancy and is a prime candidate in predicting the hypercoagulable state. Moreover, increased number of TF-exposing microparticles (MPs) in cancer patients may contribute to venous thromboembolism (VTE). We have conducted a prospective cohort study to determine whether elevated TF antigen, TF activity and TF associated to MPs (MPs-TF) are predictive of VTE and mortality in cancer patients. The studied population consisted of 252 cancer patients and 36 healthy controls. TF antigen and activity and MPs-TF were determined by ELISA and chromogenic assays. During a median follow-up of 10 months, 40 thrombotic events were recorded in 34 patients (13.5%), and 73 patients (28.9%) died. TF antigen and activity were significantly higher in patients than in controls (p<0.01) mainly in patients with advanced stages, whereas no differences were observed for TF activity of isolated MPs. We did not find a statistically significant association of TF variables with the risk of VTE. Multivariate analysis adjusting for age, sex, type of cancer and other confounding variables showed that TF activity (p<0.01) and MPs-TF activity (p<0.05) were independently associated with mortality. In conclusion, while TF variables were not associated with future VTE in cancer patients, we found a strong association of TF and MPs-TF activity with mortality, thus suggesting they might be good prognostic markers in cancer patients.
Collapse
|
46
|
Hisada Y, Ay C, Auriemma AC, Cooley BC, Mackman N. Human pancreatic tumors grown in mice release tissue factor-positive microvesicles that increase venous clot size. J Thromb Haemost 2017; 15:2208-2217. [PMID: 28834179 DOI: 10.1111/jth.13809] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Indexed: 12/18/2022]
Abstract
Essentials Tumor-bearing mice have larger venous clots than controls. Human tissue factor is present in clots in tumor-bearing mice. Inhibition of human tissue factor reduces clot size in tumor-bearing mice. This new mouse model may be useful to study mechanisms of cancer-associated thrombosis. SUMMARY Background Pancreatic cancer patients have a high rate of venous thromboembolism. Human pancreatic tumors and cell lines express high levels of tissue factor (TF), and release TF-positive microvesicles (TF+ MVs). In pancreatic cancer patients, tumor-derived TF+ MVs are present in the blood, and increased levels are associated with venous thromboembolism and decreased survival. Previous studies have shown that mice with orthotopic human or murine pancreatic tumors have circulating tumor-derived TF+ MVs, an activated clotting system, and increased incidence and mean clot weight in an inferior vena cava stenosis model. These results suggest that TF+ MVs contribute to thrombosis. However, the specific role of tumor-derived TF+ MVs in venous thrombosis in mice has not been determined. Objectives To test the hypothesis that tumor-derived TF+ MVs enhance thrombosis in mice. Methods We determined the contribution of TF+ MVs derived from human pancreatic tumors grown orthotopically in nude mice to venous clot formation by using an anti-human TF mAb. We used an inferior vena cava stasis model of venous thrombosis. Results Tumor-bearing mice had significantly larger clots than control mice. Clots from tumor-bearing mice contained human TF, suggesting the incorporation of tumor-derived MVs. Importantly, administration of an anti-human TF mAb reduced clot size in tumor-bearing mice but did not affect clot size in control mice. Conclusions Our results indicate that TF+ MVs released from orthotopic pancreatic tumors increase venous thrombosis in mice. This new model may be useful for evaluating the roles of different factors in cancer-associated thrombosis.
Collapse
Affiliation(s)
- Y Hisada
- Department of Medicine, Division of Hematology and Oncology, Thrombosis and Hemostasis Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - C Ay
- Department of Medicine, Division of Hematology and Oncology, Thrombosis and Hemostasis Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - A C Auriemma
- Department of Medicine, Division of Hematology and Oncology, Thrombosis and Hemostasis Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - B C Cooley
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - N Mackman
- Department of Medicine, Division of Hematology and Oncology, Thrombosis and Hemostasis Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
47
|
Mege D, Crescence L, Ouaissi M, Sielezneff I, Guieu R, Dignat-George F, Dubois C, Panicot-Dubois L. Fibrin-bearing microparticles: marker of thrombo-embolic events in pancreatic and colorectal cancers. Oncotarget 2017; 8:97394-97406. [PMID: 29228619 PMCID: PMC5722571 DOI: 10.18632/oncotarget.22128] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 08/04/2017] [Indexed: 12/16/2022] Open
Abstract
Background Microparticles (MPs) are plasma membrane-derived extracellular vesicles present in the bloodstream. We have described a specific signature of MPs, called microparticulosome, in colorectal (CRC) and pancreatic (PC) cancers. We observed that levels of fibrin-bearing MPs were significantly increased in patients suffering from PC and CRC in comparison with control groups. Here, we hypothesised that fibrin-MPs may constitute a relevant biomarker of thrombosis associated with cancer. The objective was to compare the microparticulosome signature between patients presenting with thrombo-embolic event and those without. Methods Patients with CRC and PC were prospectively included and divided in those with thrombo-embolic events (Group A) and those without (Group B). MPs were analyzed by flow cytometer, combining the analysis of Annexin V-positive with characterization of their origin and determination of their procoagulant activities. D-dimer levels were measured in the same samples. Results We included 118 patients, divided in 19 patients with thrombo embolic event and 99 patients without. Fibrin-bearing MPs levels were significantly higher in presence of thrombo-embolic events, contrary to D-dimers levels. Fibrin-bearing MPs were more frequently produced by erythrocytes, endothelial cells or Ep-CAM+cells than platelets or leukocytes. Overall survival was shorter in case of thrombo-embolic events than without. The most frequent genes expressed by MPs derived from PC or CRC were implicated in metastatic diffusion of tumor cells, drug resistance, coagulation and inflammation. Conclusion Circulating MPs, particularly fibrin-bearing MPs, could be used as a new biomarker to predict cancer-associated thrombo-embolic events and poor survival.
Collapse
Affiliation(s)
- Diane Mege
- Aix Marseille Univ, INSERM UMR-S1076, VRCM, Marseille, France.,Department of digestive surgery, Timone University Hospital, Marseille, France
| | - Lydie Crescence
- Aix Marseille Univ, INSERM UMR-S1076, VRCM, Marseille, France
| | - Mehdi Ouaissi
- Department of digestive surgery, Timone University Hospital, Marseille, France
| | - Igor Sielezneff
- Aix Marseille Univ, INSERM UMR-S1076, VRCM, Marseille, France.,Department of digestive surgery, Timone University Hospital, Marseille, France
| | - Regis Guieu
- Aix Marseille Univ, UMR MD2, Laboratory of Biochemistry, Timone University Hospital, Marseille, France
| | - Françoise Dignat-George
- Aix Marseille Univ, INSERM UMR-S1076, VRCM, Marseille, France.,Laboratory of Hematology, Conception University Hospital, Marseille, France
| | | | | |
Collapse
|
48
|
The effect of corn trypsin inhibitor, anti-tissue factor pathway inhibitor antibodies and phospholipids on microvesicle-associated thrombin generation in patients with pancreatic cancer and healthy controls. PLoS One 2017; 12:e0184579. [PMID: 28910348 PMCID: PMC5598995 DOI: 10.1371/journal.pone.0184579] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/26/2017] [Indexed: 11/19/2022] Open
Abstract
Circulating microvesicles (MVs) are suggested to be important contributors to cancer-associated thrombosis due to the presence of surface-bound procoagulant molecules like tissue factor (TF) and phosphatidylserine (PS). Pancreatic cancer is considered to be one of the most prothrombotic malignancies. The aim of this study was to describe the impact of analytical variables on MV-associated thrombin generation in patients with pancreatic cancer and in healthy controls. MVs were isolated from citrated plasma and added to pooled normal plasma (PNP). Thrombin generation was measured by the calibrated automated thrombogram. The impact of corn trypsin inhibitor (CTI), anti-tissue factor pathway inhibitor (TFPI) antibodies and phospholipids was described. Antibodies against TF were used to assess TF-dependency, and MV-bound PS activity was measured with the Zymuphen MP-activity kit. MVs from the pancreatic cancer patients displayed higher thrombin generation and higher PS-activity than MVs from the healthy control group, while TF-dependency was observed in only 1 out of 13 patient samples. Adequate thrombin generation-curves were only achieved when CTI was omitted and anti-TFPI antibodies were added to PNP prepared in low contact-activating tubes. Addition of phospholipids reduced the significant differences between the two groups, and should be omitted. This modified thrombin generation assay could be useful for measurement of procoagulant circulating MVs, allowing the contribution from MVs affecting both the intrinsic and the extrinsic pathway to be measured.
Collapse
|
49
|
Fischer D, Büssow J, Meybohm P, Weber CF, Zacharowski K, Urbschat A, Müller MM, Jennewein C. Microparticles from stored red blood cells enhance procoagulant and proinflammatory activity. Transfusion 2017; 57:2701-2711. [DOI: 10.1111/trf.14268] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Dania Fischer
- Department of Anesthesiology; Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt; Frankfurt am Main Germany
| | - Julian Büssow
- Department of Anesthesiology; Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt; Frankfurt am Main Germany
| | - Patrick Meybohm
- Department of Anesthesiology; Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt; Frankfurt am Main Germany
| | - Christian Friedrich Weber
- Department of Anesthesiology; Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt; Frankfurt am Main Germany
| | - Kai Zacharowski
- Department of Anesthesiology; Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt; Frankfurt am Main Germany
| | - Anja Urbschat
- Department of Urology and Pediatric Urology; University Hospital of Marburg, Philipps-University; Marburg Germany
| | - Markus Matthias Müller
- German Red Cross Blood Transfusion Service of Baden-Wuerttemberg-Hessen, Institute of Transfusion Medicine and Immunohematology, University Hospital of Frankfurt; Frankfurt am Main Germany
| | - Carla Jennewein
- Department of Anesthesiology; Intensive Care Medicine, and Pain Therapy, University Hospital Frankfurt; Frankfurt am Main Germany
| |
Collapse
|
50
|
Cohen JG, Prendergast E, Geddings JE, Walts AE, Agadjanian H, Hisada Y, Karlan BY, Mackman N, Walsh CS. Evaluation of venous thrombosis and tissue factor in epithelial ovarian cancer. Gynecol Oncol 2017; 146:146-152. [PMID: 28501328 DOI: 10.1016/j.ygyno.2017.04.021] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/25/2017] [Accepted: 04/27/2017] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Ovarian clear cell carcinoma (OCCC) and high grade serous ovarian cancer (HGSOC) are associated with the highest risk of VTE among patients with epithelial ovarian cancer (EOC). Tissue factor (TF) is a transmembrane glycoprotein which can trigger thrombosis. We sought to evaluate if there is an association between VTE and tumor expression of tissue factor (TF), plasma TF, and microvesicle TF (MV TF) activity in this high-risk population. METHODS We performed a case-control study of OCCC and HGSOC patients with and without VTE. 105 patients who underwent surgery at a tertiary care center between January 1995 and October 2013 were included. Plasma TF was measured with an enzyme-linked immunosorbent assay. A TF-dependent Factor Xa generation assay was used to measure MV TF activity. Immunohistochemical (IHC) analysis was performed to evaluate tumor expression of TF. RESULTS 35 women with OCCC or HGSOC diagnosed with VTE within 9months of surgery were included in the case group. Those with VTE had a worse OS, p<0.0001, with a greater than three-fold increase in risk of death, HR 3.33 (CI 1.75-6.35). There was no significant difference in median plasma TF level or MV TF activity level between patients with and without VTE. OCCC patients had greater expression of TF in their tumors than patients with HGSOC, p<0.0001. CONCLUSIONS TFMV activity and plasma TF level were not predictive of VTE in this patient population. Given the extensive expression of TF in OCCC tumors, it is unlikely IHC expression will be useful in risk stratification for VTE in this population.
Collapse
Affiliation(s)
- Joshua G Cohen
- Division of Gynecologic Oncology, University of California, Los Angeles, 200 Medical Plaza, Suite 220, Los Angeles, CA 90095, USA
| | - Emily Prendergast
- Division of Gynecologic Oncology, Cedars-Sinai Medical Center, 8635 West 3rd Street, Suite 280W, Los Angeles, CA 90048, USA
| | - Julia E Geddings
- Division of Hematology/Oncology, University of North Carolina, Chapel Hill, 2312 MBRB, 111 Mason Farm Rd, CB#7126, Chapel Hill, NC 27599, USA
| | - Ann E Walts
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Hasmik Agadjanian
- Division of Gynecologic Oncology, Cedars-Sinai Medical Center, 8635 West 3rd Street, Suite 280W, Los Angeles, CA 90048, USA
| | - Yohei Hisada
- Division of Hematology/Oncology, University of North Carolina, Chapel Hill, 2312 MBRB, 111 Mason Farm Rd, CB#7126, Chapel Hill, NC 27599, USA; K.G. Jebsen TREC, The Faculty of Health Sciences, UiT- The Arctic University of Tromsø, 9037 Tromsø, Norway
| | - Beth Y Karlan
- Division of Gynecologic Oncology, Cedars-Sinai Medical Center, 8635 West 3rd Street, Suite 280W, Los Angeles, CA 90048, USA
| | - Nigel Mackman
- Division of Hematology/Oncology, University of North Carolina, Chapel Hill, 2312 MBRB, 111 Mason Farm Rd, CB#7126, Chapel Hill, NC 27599, USA; K.G. Jebsen TREC, The Faculty of Health Sciences, UiT- The Arctic University of Tromsø, 9037 Tromsø, Norway
| | - Christine S Walsh
- Division of Gynecologic Oncology, Cedars-Sinai Medical Center, 8635 West 3rd Street, Suite 280W, Los Angeles, CA 90048, USA.
| |
Collapse
|