1
|
Bayrak O, Alper M, Basbinar Y, Bayrak S. The role of thrombin in the paradoxical interplay of cancer metastasis and the vascular system: A driving dynamic. Biomed Pharmacother 2025; 186:118031. [PMID: 40215647 DOI: 10.1016/j.biopha.2025.118031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
The coagulation system plays a complex role in cancer therapy. Endothelial damage and tissue factor increased by chemotherapy initiate the coagulation cascade, producing active FXa and releasing thrombin. Thrombin triggers tumor growth and metastasis, leading to severe thromboembolic events in cancer patients. Direct thrombin inhibitors do not have the expected anti-metastatic effect as PAR-2 remains active and increases the risk of bleeding. Therefore, dual inhibition of thrombin by FXa inhibition and plasmin inhibition, which converts fibrin to fibrinogen, is targeted. Clinical studies show that the use of tranexamic acid in patients on NOAC therapy may be beneficial without increasing the risk of bleeding. This approach offers a promising strategy to provide an anti-metastatic effect in cancer treatment.
Collapse
Affiliation(s)
- Ozge Bayrak
- Dokuz Eylul University, Institute of Health Sciences, Department of Oncology, Izmir, Turkey
| | - Meltem Alper
- Dokuz Eylul University, Institute of Oncology, Department of Translational Oncology, Izmir, Turkey
| | - Yasemin Basbinar
- Dokuz Eylul University, Institute of Oncology, Department of Translational Oncology, Izmir, Turkey
| | - Serdar Bayrak
- Dokuz Eylul University, Institute of Oncology, Department of Translational Oncology, Izmir, Turkey; Dokuz Eylul University, Faculty of Medicine, Department of Cardiovascular Surgery, Izmir, Turkey.
| |
Collapse
|
2
|
Al-Azzawi HMA, Hamza SA, Paolini R, Arshad F, Patini R, O'Reilly L, McCullough M, Celentano A. Towards an emerging role for anticoagulants in cancer therapy: a systematic review and meta-analysis. FRONTIERS IN ORAL HEALTH 2024; 5:1495942. [PMID: 39568788 PMCID: PMC11576436 DOI: 10.3389/froh.2024.1495942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 11/22/2024] Open
Abstract
Background Anticoagulants, renowned for their role in preventing blood clot formation, have captivated researchers' attention for the exploitation of their potential to inhibit cancer in pre-clinical models. Objectives To undertake a systematic review and meta-analysis of the effects of anticoagulants in murine cancer research models. Further, to present a reference tool for anticoagulant therapeutic modalities relating to future animal pre-clinical models of cancer and their translation into the clinic. Methods Four databases were utilized including Medline (Ovid), Embase (Ovid), Web of science, and Scopus databases. We included studies relating to any cancer conducted in murine models that assessed the effect of traditional anticoagulants (heparin and its derivatives and warfarin) and newer oral anticoagulants on cancer. Results A total of 6,158 articles were identified in an initial multi-database search. A total of 157 records were finally included for data extraction. Studies on heparin species and warfarin demonstrated statistically significant results in favour of tumour growth and metastasis inhibition. Conclusion Our findings constitute a valuable reference guide for the application of anticoagulants in cancer research and explore the promising utilization of non-anticoagulants heparin in preclinical cancer research. Systematic Review Registration PROSPERO [CRD42024555603].
Collapse
Affiliation(s)
| | - Syed Ameer Hamza
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Rita Paolini
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Fizza Arshad
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Romeo Patini
- Head and Neck Department, "Fondazione Policlinico Universitario A. Gemelli-IRCCS" School of Dentistry, Catholic University of Sacred Heart-Rome Largo A. Gemelli, Rome, Italy
| | - Lorraine O'Reilly
- Clinical Translation Centre, Cancer Biology and Stem Cells Division and Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Michael McCullough
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| | - Antonio Celentano
- Melbourne Dental School, The University of Melbourne, Carlton, VIC, Australia
| |
Collapse
|
3
|
Yousefian Naeini Z, Esfandiari N, Hashemi M, Hushmandi K, Arbabian S, Entezari M. Introduced the ITGB1-DT as a novel biomarker associated with five potential drugs using bioinformatics analysis of breast cancer proteomics data and RT-PCR. Mol Cell Probes 2023; 71:101930. [PMID: 37690573 DOI: 10.1016/j.mcp.2023.101930] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/28/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Breast cancer (BC) has been identified as a significant contributor to the rising number of female cancer deaths. As, it has become clear that breast cancer development depends on the interplay of several biological factors against a single molecule. This research aimed to use proteomics to gain a regulatory and metabolic understanding of BC pathophysiology. METHOD For the study, a breast cancer proteomics dataset was downloaded from ProteomeXchange and then analyzed by employing MaxQuant and Perseus. Functional enrichment analysis through Metascape and Cytoscape software showed DEPs related biomedical phenomena with potential abruption. The expression of selected lncRNA in terms of the highest connectivity parameters was then quantitatively assessed through RT-PCR in 30 tumor tissues of breast cancer patients, as compared to the adjacent healthy ones. RESULT The results indicated that among the 3048 identified proteins, 1149 were differentially expressed, which could be mainly enriched in several key terms. Furthermore, the obtained findings revealed that ITGB1-DT was significantly overexpressed in tumor tissues. Moreover, we found five potential compounds that could be attributed to ITGB1-DT targets (ATN-161, Firategrast, SB-683698, dabigatran-etexilate, and tranexamic-acid). CONCLUSION These analyses proposed that ITGB1-DT could be employed as a differentiated factor to identify breast tumor tissues in healthy samples. Besides this, Firategrast could be introduced as a potential remedial agent for breast cancer patients. Overall, from the analysis of a proteomics dataset, an integrative map was generated, and a novel biomarker that may have been implicated in the early detection of BC was introduced.
Collapse
Affiliation(s)
- Zahra Yousefian Naeini
- Department of Cellular and Molecular, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Negin Esfandiari
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Sedighe Arbabian
- Department of Cellular and Molecular, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
4
|
Russo V, Falco L, Tessitore V, Mauriello A, Catapano D, Napolitano N, Tariq M, Caturano A, Ciccarelli G, D’Andrea A, Giordano A. Anti-Inflammatory and Anticancer Effects of Anticoagulant Therapy in Patients with Malignancy. Life (Basel) 2023; 13:1888. [PMID: 37763292 PMCID: PMC10532829 DOI: 10.3390/life13091888] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Optimizing the anticoagulation therapy is of pivotal importance in patients with a malignant tumor, as venous thromboembolism (VTE) has become the second-leading cause of death in this population. Cancer can highly increase the risk of thrombosis and bleeding. Consequently, the management of cancer-associated VTE is complex. In recent years, translational research has intensified, and several studies have highlighted the role of inflammatory cytokines in cancer growth and progression. Simultaneously, the pleiotropic effects of anticoagulants currently recommended for VTE have emerged. In this review, we describe the anti-inflammatory and anticancer effects of both direct oral anticoagulants (DOACs) and low-molecular-weight heparins (LWMHs).
Collapse
Affiliation(s)
- Vincenzo Russo
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Luigi Falco
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
| | - Viviana Tessitore
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
| | - Alfredo Mauriello
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
| | - Dario Catapano
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
| | - Nicola Napolitano
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
| | - Moiz Tariq
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, NA, Italy (A.D.)
| | - Giovanni Ciccarelli
- Cardiology Unit, Department of Medical Translational Science, University of Campania “Luigi Vanvitelli”—Monaldi Hospital, 80126 Naples, NA, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Antonello D’Andrea
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, NA, Italy (A.D.)
- Cardiology Unit, Umberto I Hospital, 84014 Nocera Inferiore, SA, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
5
|
Yao X, Hu Q, Liu X, Ling Q, Leng Y, Zhao H, Yu P, Ma J, Zhao Y, Liu M, Yang R. Atrial fibrillation and breast cancer—Vicious twins? A systematic review and meta-analysis. Front Cardiovasc Med 2023; 10:1113231. [PMID: 36970342 PMCID: PMC10036368 DOI: 10.3389/fcvm.2023.1113231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 02/13/2023] [Indexed: 03/12/2023] Open
Abstract
BackgroundEpidemiological studies suggest a bidirectional association between atrial fibrillation and breast cancer. This study aimed to conduct a meta-analysis to elucidate the prevalence of atrial fibrillation among breast cancer patients, and the bidirectional association between atrial fibrillation and breast cancer.MethodsPubMed, the Cochrane Library, and Embase were searched to identify studies reporting the prevalence, incidence, and bidirectional association between atrial fibrillation and breast cancer. The study was registered with PROSPERO (CRD42022313251). Levels of evidence and recommendations were assessed by the Grading of Recommendations Assessment, Development and Evaluation (GRADE).ResultsTwenty-three studies (17 retrospective cohort studies, 5 case-control studies and 1 cross-sectional study) involving 8,537,551 participants were included. Among patients with breast cancer, the prevalence of atrial fibrillation was 3% (11 studies; 95% CI: 0.6 to 7.1%) and the incidence was 2.7% (6 studies; 95% CI: 1.1 to 4.9%). Breast cancer was associated with increased risk of atrial fibrillation (5 studies; hazard ratio [HR]: 1.43, 95% CI: 1.12 to 1.82, I2 = 98%). Atrial fibrillation was also significantly associated elevated risk of breast cancer (5 studies HR: 1.18, 95% CI: 1.14 to 1.22, I2 = 0%). Grade assessment shown low certainty of the evidence for the risk of atrial fibrillation and moderate certainty of the evidence for the risk of breast cancer.ConclusionAtrial fibrillation is not uncommon in patients with breast cancer and vice versa. There is a bidirectional association between atrial fibrillation (low certainty) and breast cancer (moderate certainty).
Collapse
Affiliation(s)
- Xiongda Yao
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qingwen Hu
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiao Liu
- Department of Cardiology, Sun Yat-Sen Memorial Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Guangzhou, Guangdong, China
| | - Qing Ling
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yurong Leng
- The Affiliated Stomatological Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Huilei Zhao
- Department of Anesthesiology, The Third Hospital of Nanchang, Nanchang, Jiangxi, China
| | - Peng Yu
- Department of Endocrine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jianyong Ma
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Yujie Zhao
- Department of Cardiology, Seventh People's Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Menglu Liu
- Department of Cardiology, Seventh People's Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Renqiang Yang
- Department of Cardiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- *Correspondence: Renqiang Yang
| |
Collapse
|
6
|
|
7
|
Commonly Prescribed Anticoagulants Exert Anticancer Effects in Oral Squamous Cell Carcinoma Cells In Vitro. BIOLOGY 2022; 11:biology11040596. [PMID: 35453795 PMCID: PMC9027634 DOI: 10.3390/biology11040596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/09/2022] [Accepted: 04/11/2022] [Indexed: 11/24/2022]
Abstract
Simple Summary Oral squamous cell carcinoma (OSCC) is the sixth most common cancer worldwide with 840,000 new cases and 420,000 deaths in 2020. Anticoagulants are widely prescribed medications routinely administered to help prevent blood clots. Despite the great relevance of these two topics, there is complete lack of knowledge regarding the potential effects that these drugs could exert on oral cancer patients. In this in vitro study, we comprehensively investigated the effect of anticoagulants on OSCC activity. This includes the effect of these drugs on cancer cell ability to survive, migrate to colonise distant sites, and resist treatment with conventional chemotherapy. We have demonstrated for the first time that various anticoagulants have anticancer effects on OSCC. Moreover, some of the anticoagulants tested were able to reduce the migratory ability of cancer cells. Finally, the great majority of anticoagulants studied reduced the effectiveness of the tested chemotherapeutic agent, allowing an increase in cancer cell proliferation. Our results highlight the need for urgent further research in the field, to improve the anticoagulant strategies in patients with oral cancer, and in turn their prognosis. Abstract Oral squamous cell carcinoma (OSCC) is the most common head and neck cancer. With anticoagulant usage on the rise, it is important to elucidate their potential effects on tumour biology and interactions with chemotherapeutics. The aim of the present study was to investigate the effects of anticoagulants on OSCC cell lines and their interactions with the drug 5-fluorouracil (5-FU). Cell proliferation was assessed using an MTS in vitro assay in two human OSCC cell lines (H357/H400) and in normal oral keratinocytes (OKF6) treated with the 5-FU (0.2/1/5/10 μg/mL), conventional anticoagulants warfarin (1/5/10/20 μM) and heparin (5/20/80 U), as well as four new oral anticoagulants, dabigatran (5/10/20 μM), rivaroxaban (5/10/20 μM), apixaban (0.1/1/5 μg/mL), and edoxaban (5/10/20 μM). Cell migration was assessed at 3 h intervals up to18 h using a wound healing assay. Our results clearly demonstrate, for the first time, that commonly prescribed anticoagulants exert in vitro antiproliferative effects on OSCC cells. Furthermore, treatment with some anticoagulants reduced the migration of OSCC cell lines. Nevertheless, most of the anticoagulants tested reduced the effectiveness of the chemotherapeutic agent tested, 5-FU, highlighting potential flaws in the current pharmacological management of these patients. Our findings showed the need for the immediate translation of this research to preclinical animal models.
Collapse
|
8
|
Smeda M, Stojak M, Przyborowski K, Sternak M, Suraj-Prazmowska J, Kus K, Derszniak K, Jasztal A, Kij A, Kurpinska A, Kieronska-Rudek A, Wojnar-Lason K, Buczek E, Mohaissen T, Chlopicki S. Direct Thrombin Inhibitor Dabigatran Compromises Pulmonary Endothelial Integrity in a Murine Model of Breast Cancer Metastasis to the Lungs; the Role of Platelets and Inflammation-Associated Haemostasis. Front Pharmacol 2022; 13:834472. [PMID: 35295330 PMCID: PMC8918823 DOI: 10.3389/fphar.2022.834472] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/24/2022] [Indexed: 11/13/2022] Open
Abstract
Activation of the coagulation cascade favours metastatic spread, but antithrombotic therapy might also have detrimental effects on cancer progression. In this study, we characterized the effects of dabigatran, a direct reversible thrombin inhibitor, on the pulmonary endothelial barrier and metastatic spread in a murine model of breast cancer metastasis. Dabigatran etexilate (100 mg kg−1) was administered to mice twice daily by oral gavage. Pulmonary metastasis, pulmonary endothelium permeability in vivo, and platelet reactivity were evaluated after intravenous injection of 4T1 breast cancer cells into BALB/c mice. The effect of dabigatran on platelet-dependent protection of pulmonary endothelial barrier in the presence of an inflammatory stimulus was also verified in vitro using human lung microvascular endothelial cell (HLMVEC) cultures. Dabigatran-treated mice harbored more metastases in their lungs and displayed increased pulmonary endothelium permeability after cancer cell injection. It was not associated with altered lung fibrin deposition, changes in INFγ, or complement activation. In the in vitro model of the pulmonary endothelial barrier, dabigatran inhibited platelet-mediated protection of pulmonary endothelium. In a murine model of breast cancer metastasis, dabigatran treatment promoted pulmonary metastasis by the inhibition of platelet-dependent protection of pulmonary endothelial barrier integrity.
Collapse
Affiliation(s)
- Marta Smeda
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- *Correspondence: Marta Smeda, ; Stefan Chlopicki,
| | - Marta Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Kamil Przyborowski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Magdalena Sternak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Joanna Suraj-Prazmowska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Kamil Kus
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Katarzyna Derszniak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Agnieszka Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Kurpinska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Kieronska-Rudek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Department of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Kamila Wojnar-Lason
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Department of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
| | - Elzbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Tasnim Mohaissen
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
- Department of Pharmacology, Jagiellonian University Medical College, Krakow, Poland
- *Correspondence: Marta Smeda, ; Stefan Chlopicki,
| |
Collapse
|
9
|
Abdel-Bakky MS, Amin E, Ewees MG, Mahmoud NI, Mohammed HA, Altowayan WM, Abdellatif AAH. Coagulation System Activation for Targeting of COVID-19: Insights into Anticoagulants, Vaccine-Loaded Nanoparticles, and Hypercoagulability in COVID-19 Vaccines. Viruses 2022; 14:228. [PMID: 35215822 PMCID: PMC8876839 DOI: 10.3390/v14020228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 01/06/2022] [Accepted: 01/21/2022] [Indexed: 01/08/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), also known as COVID-19, is currently developing into a rapidly disseminating and an overwhelming worldwide pandemic. In severe COVID-19 cases, hypercoagulability and inflammation are two crucial complications responsible for poor prognosis and mortality. In addition, coagulation system activation and inflammation overlap and produce life-threatening complications, including coagulopathy and cytokine storm, which are associated with overproduction of cytokines and activation of the immune system; they might be a lead cause of organ damage. However, patients with severe COVID-19 who received anticoagulant therapy had lower mortality, especially with elevated D-dimer or fibrin degradation products (FDP). In this regard, the discovery of natural products with anticoagulant potential may help mitigate the numerous side effects of the available synthetic drugs. This review sheds light on blood coagulation and its impact on the complication associated with COVID-19. Furthermore, the sources of natural anticoagulants, the role of nanoparticle formulation in this outbreak, and the prevalence of thrombosis with thrombocytopenia syndrome (TTS) after COVID-19 vaccines are also reviewed. These combined data provide many research ideas related to the possibility of using these anticoagulant agents as a treatment to relieve acute symptoms of COVID-19 infection.
Collapse
Affiliation(s)
- Mohamed S. Abdel-Bakky
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Qassim 52471, Saudi Arabia;
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
| | - Elham Amin
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt;
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 52471, Saudi Arabia;
| | - Mohamed G. Ewees
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt; (M.G.E.); (N.I.M.)
| | - Nesreen I. Mahmoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Nahda University, Beni-Suef 11787, Egypt; (M.G.E.); (N.I.M.)
| | - Hamdoon A. Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, Qassim University, Qassim 52471, Saudi Arabia;
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Cairo 11884, Egypt
| | - Waleed M. Altowayan
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Qassim 52471, Saudi Arabia;
| | - Ahmed A. H. Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Qasssim 52471, Saudi Arabia
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| |
Collapse
|
10
|
Lei Z, Guo D. Significant difference between coagulation parameters and clinicopathological characteristics in breast cancer. Blood Coagul Fibrinolysis 2021; 32:572-577. [PMID: 34545042 DOI: 10.1097/mbc.0000000000001084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The purpose of this research is to investigate the correlation between coagulation parameters and survival on patients with breast cancer. A total of 244 patients with breast cancer and 84 patients with metastatic advanced breast cancer admitted to the Second Affiliated Hospital of Chongqing Medical University from January 2017 to February 2019 were selected as the research objects. Their age, tumor size, status of estrogen receptor and progesterone receptor status, status of Her-2 receptor and lymph node were evaluated. Additional 99 cases of benign breast tumors were selected as the control group. All patients were newly diagnosed. PTA, PT ratio, PT, APTT, APTT ratio, TT, TT ratio, PT-INR, FIB, and FDP were detected on all patients. There were significant differences in APTT, APTT ratio, FIB, TT, TT ratio, and FDP between breast cancer and benign tumor control group (P < 0.05). The changes of FIB were correlated with age (P = 0.007), PR status (P = 0.017), and Her-2 status (P = 0.008). The older the age, the higher the FIB level. The FIB level of Er negative state is higher than that of positive state, and the FIB level of PR negative state is also higher than that of positive state. Survival analysis showed that PR negative status, the elevated level of APTT, APTT ratio, TT increased, and TT ratio were the indicators affecting 3-year DFS of patients (P < 0.05). Multivariate COX regression analysis showed that APTT and TT were independent prognostic factors affecting 3 years DFS (P < 0.05). Blood hypercoagulable state in patients with breast cancer, abnormal coagulation system plays an important role in the progress of breast cancer.
Collapse
Affiliation(s)
- Zhengwu Lei
- Health management center, Second Affiliated Hospital of Chongqing Medical University
| | - Dan Guo
- Department of Breast and Thyroid Surgery, Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Castle J, Blower E, Kirwan CC. Update on the role of circulating tumour cells in cancer-associated thrombosis. THROMBOSIS UPDATE 2021. [DOI: 10.1016/j.tru.2021.100066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
12
|
Izuegbuna OO, Agodirin OS, Olawumi HO, Olatoke SA. Plasma D-Dimer and Fibrinogen Levels Correlates with Tumor Size and Disease Progression in Nigerian Breast Cancer Patients. Cancer Invest 2021; 39:597-606. [PMID: 33843402 DOI: 10.1080/07357907.2021.1909059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Fourty-five breast cancer patients and 50 apparently healthy sex-matched controls from the University of Ilorin Teaching Hospital were enrolled in this study. Plasma D-dimer and fibrinogen were found to be significantly higher than controls; APTT was significantly shorter than the controls. D-dimer and fibrinogen were also significantly positively correlated with ECOG, disease stage, lymph node involvement, and tumor size. On multivariate analysis, D-dimer and fibrinogen were found to be independently related to lymph node involvement. This study shows that plasma D-dimer and fibrinogen levels are elevated in breast cancer patients, and both are markers of disease progression.
Collapse
Affiliation(s)
- Ogochukwu O Izuegbuna
- Department of Haematology and Blood Transfusion, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Olayide S Agodirin
- Department of Surgery, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Hannah O Olawumi
- Department of Haematology and Blood Transfusion, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Samuel A Olatoke
- Department of Surgery, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| |
Collapse
|
13
|
Goh CY, Patmore S, Smolenski A, Howard J, Evans S, O'Sullivan J, McCann A. The role of von Willebrand factor in breast cancer metastasis. Transl Oncol 2021; 14:101033. [PMID: 33571850 PMCID: PMC7876567 DOI: 10.1016/j.tranon.2021.101033] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/12/2021] [Accepted: 01/28/2021] [Indexed: 01/16/2023] Open
Abstract
VWF plays an important role in breast tumour progression and metastasis. Patients with metastatic breast cancer have significantly elevated plasma VWF. Increased levels of highly adhesive VWF may regulate platelet-tumour interactions. VWF may protect disseminated tumour cells from chemotherapy.
Breast cancer is the most common female cancer globally, with approximately 12% of patients eventually developing metastatic disease. Critically, limited effective treatment options exist for metastatic breast cancer. Recently, von Willebrand factor (VWF), a haemostatic plasma glycoprotein, has been shown to play an important role in tumour progression and metastasis. In breast cancer, a significant rise in the plasma levels of VWF has been reported in patients with malignant disease compared to benign conditions and healthy controls, with an even greater increase seen in patients with disseminated disease. Direct interactions between VWF, tumour cells, platelets and endothelial cells may promote haematogenous dissemination and thus the formation of metastatic foci. Intriguingly, patients with metastatic disease have unusually large VWF multimers. This observation has been proposed to be a result of a dysfunctional or deficiency of VWF-cleaving protease activity, ADAMTS-13 activity, which may then regulate the platelet-tumour adhesive interactions in the metastatic process. In this review, we provide an overview of VWF in orchestrating the pathological process of breast cancer dissemination, and provide supporting evidence of the role of VWF in mediating metastatic breast cancer.
Collapse
Affiliation(s)
- Chia Yin Goh
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Dublin 4, Ireland; UCD School of Medicine, College of Health and Agricultural Sciences (CHAS), University College Dublin, Belfield, Dublin, Dublin 4, Ireland.
| | - Sean Patmore
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Dublin 2, Ireland
| | - Albert Smolenski
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Dublin 4, Ireland; UCD School of Medicine, College of Health and Agricultural Sciences (CHAS), University College Dublin, Belfield, Dublin, Dublin 4, Ireland
| | - Jane Howard
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Dublin 4, Ireland
| | - Shane Evans
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Dublin 4, Ireland; UCD School of Medicine, College of Health and Agricultural Sciences (CHAS), University College Dublin, Belfield, Dublin, Dublin 4, Ireland
| | - Jamie O'Sullivan
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Dublin 2, Ireland
| | - Amanda McCann
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin, Dublin 4, Ireland; UCD School of Medicine, College of Health and Agricultural Sciences (CHAS), University College Dublin, Belfield, Dublin, Dublin 4, Ireland
| |
Collapse
|
14
|
Metelli A, Wu BX, Riesenberg B, Guglietta S, Huck JD, Mills C, Li A, Rachidi S, Krieg C, Rubinstein MP, Gewirth DT, Sun S, Lilly MB, Wahlquist AH, Carbone DP, Yang Y, Liu B, Li Z. Thrombin contributes to cancer immune evasion via proteolysis of platelet-bound GARP to activate LTGF-β. Sci Transl Med 2021; 12:12/525/eaay4860. [PMID: 31915300 DOI: 10.1126/scitranslmed.aay4860] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 11/18/2019] [Indexed: 12/17/2022]
Abstract
Cancer-associated thrombocytosis and high concentrations of circulating transforming growth factor-β1 (TGF-β1) are frequently observed in patients with progressive cancers. Using genetic and pharmacological approaches, we show a direct link between thrombin catalytic activity and release of mature TGF-β1 from platelets. We found that thrombin cleaves glycoprotein A repetitions predominant (GARP), a cell surface docking receptor for latent TGF-β1 (LTGF-β1) on platelets, resulting in liberation of active TGF-β1 from the GARP-LTGF-β1 complex. Furthermore, systemic inhibition of thrombin obliterates TGF-β1 maturation in platelet releasate and rewires the tumor microenvironment toward favorable antitumor immunity, which translates into efficient cancer control either alone or in combination with programmed cell death 1-based immune checkpoint blockade therapy. Last, we demonstrate that soluble GARP and GARP-LTGF-β1 complex are present in the circulation of patients with cancer. Together, our data reveal a mechanism of cancer immune evasion that involves thrombin-mediated GARP cleavage and the subsequent TGF-β1 release from platelets. We propose that blockade of GARP cleavage is a valuable therapeutic strategy to overcome cancer's resistance to immunotherapy.
Collapse
Affiliation(s)
- Alessandra Metelli
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Bill X Wu
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Brian Riesenberg
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Silvia Guglietta
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - John D Huck
- Hauptman Woodward Medical Research Institute, Buffalo, NY 14203, USA
| | - Catherine Mills
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Anqi Li
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Saleh Rachidi
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Carsten Krieg
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Mark P Rubinstein
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Daniel T Gewirth
- Hauptman Woodward Medical Research Institute, Buffalo, NY 14203, USA
| | - Shaoli Sun
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Michael B Lilly
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Amy H Wahlquist
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - David P Carbone
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.,Division of Medical Oncology, The Ohio State University, Columbus, OH 43210, USA
| | - Yiping Yang
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.,Division of Hematology, Department of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Bei Liu
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Zihai Li
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA. .,Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.,Division of Medical Oncology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
15
|
Rudzinski JK, Govindasamy NP, Lewis JD, Jurasz P. The role of the androgen receptor in prostate cancer-induced platelet aggregation and platelet-induced invasion. J Thromb Haemost 2020; 18:2976-2986. [PMID: 32692888 DOI: 10.1111/jth.15020] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 07/09/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Metastatic prostate cancer progresses from a hormone sensitive androgen receptor expressing phenotype to a hormone insensitive androgen receptor-independent subtype with low overall survival. Human platelets contribute to metastasis via tumor cell-induced platelet aggregation, which in part enhances cancer cell invasion. Given the more aggressive nature of hormone insensitive prostate cancer, we hypothesized that androgen receptor-negative prostate cancer cells exhibit higher platelet aggregation potency and invasive response compared to cells with androgen receptor. OBJECTIVE To characterize the role of androgen receptors in prostate cancer-induced platelet aggregation and platelet-induced invasion. METHODS Tumor cell-induced platelet aggregation experiments were performed with platelets from healthy human donors and benign prostate (RWPE-1) and prostate cancer cell lines positive (LNCaP) and negative for androgen receptor (DU145 and PC3). Immunoblot measured prostate cancer prothrombin. Modified Boyden chamber invasion assays and zymography were performed to assess the effects of platelets on prostate cancer cell invasion and matrix metalloproteinase (MMP) expression, respectively. RESULTS Androgen receptor-positive prostate cancer cell lines failed to induce platelet aggregation. However, androgen receptor-inhibited and -negative cell lines all induced platelet aggregation, which was abolished by dabigatran. Androgen receptor-inhibited and -negative cell lines demonstrated greater expression of prothrombin than androgen receptor-positive cells. Platelets enhanced invasion and MMP-2 and -9 expression by androgen receptor-inhibited and negative prostate cancer cells, but not that of the androgen receptor-positive cells. CONCLUSIONS Androgen receptor loss within prostate cancer results in increased thrombogenicity due to upregulation of prothrombin expression. Reciprocally, platelets enhance invasion of androgen receptor-negative prostate cancer cells via increased MMP expression.
Collapse
Affiliation(s)
- Jan K Rudzinski
- Division of Urology, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Natasha P Govindasamy
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - John D Lewis
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Paul Jurasz
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Cardiovascular Research Centre, University of Alberta, Edmonton, AB, Canada
- Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
16
|
Shin SJ, Hang HT, Thang BQ, Shimoda T, Sakamoto H, Osaka M, Hiramatsu Y, Yamashiro Y, Yanagisawa H. Role of PAR1-Egr1 in the Initiation of Thoracic Aortic Aneurysm in Fbln4-Deficient Mice. Arterioscler Thromb Vasc Biol 2020; 40:1905-1917. [DOI: 10.1161/atvbaha.120.314560] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Objective:
Remodeling of the extracellular matrix plays a vital role in cardiovascular diseases. Using a mouse model of postnatal ascending aortic aneurysms (termed
Fbln4
SMKO
), we have reported that abnormal mechanosensing led to aneurysm formation in
Fbln4
SMKO
with an upregulation of the mechanosensitive transcription factor, Egr1 (Early growth response 1). However, the role of Egr1 and its upstream regulator(s) in the initiation of aneurysm development and their relationship to an aneurysmal microenvironment are unknown.
Approach and Results:
To investigate the contribution of Egr1 in the aneurysm development, we deleted
Egr1
in
Fbln4
SMKO
mice and generated double knockout mice (
DKO
,
Fbln4
SMKO
;
Egr1
−/−
). Aneurysms were prevented in
DKO
mice (42.8%) and
Fbln4
SMKO
;
Egr1
+/−
mice (26%). Ingenuity Pathway Analysis identified PAR1 (protease-activated receptor 1) as a potential Egr1 upstream gene. Protein and transcript levels of PAR1 were highly increased in
Fbln4
SMKO
aortas at postnatal day 1 before aneurysm formed, together with active thrombin and MMP (matrix metalloproteinase)-9, both of which serve as a PAR1 activator. Concordantly, protein levels of PAR1, Egr1, and thrombin were significantly increased in human thoracic aortic aneurysms. In vitro cyclic stretch assays (1.0 Hz, 20% strain, 8 hours) using mouse primary vascular smooth muscle cells induced marked expression of PAR1 and secretion of prothrombin in response to mechanical stretch. Thrombin was sufficient to induce Egr1 expression in a PAR1-dependent manner.
Conclusions:
We propose that thrombin, MMP-9, and mechanical stimuli in the
Fbln4
SMKO
aorta activate PAR1, leading to the upregulation of Egr1 and initiation of ascending aortic aneurysms.
Collapse
Affiliation(s)
- Seung Jae Shin
- From the Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA) (S.J.S., H.T.H., T.S., Y.Y., H.Y.), University of Tsukuba, Ibaraki, Japan
- Graduate School of Life and Environmental Sciences (S.J.S.), University of Tsukuba, Ibaraki, Japan
| | - Huynh Thuy Hang
- From the Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA) (S.J.S., H.T.H., T.S., Y.Y., H.Y.), University of Tsukuba, Ibaraki, Japan
- Graduate School of Comprehensive Human Sciences (H.T.H.), University of Tsukuba, Ibaraki, Japan
| | - Bui Quoc Thang
- Department of Cardiovascular Surgery (B.Q.T., H.S., M.O., Y.H.), University of Tsukuba, Ibaraki, Japan
| | - Tomonari Shimoda
- From the Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA) (S.J.S., H.T.H., T.S., Y.Y., H.Y.), University of Tsukuba, Ibaraki, Japan
- School of Medicine (T.S.), University of Tsukuba, Ibaraki, Japan
| | - Hiroaki Sakamoto
- Department of Cardiovascular Surgery (B.Q.T., H.S., M.O., Y.H.), University of Tsukuba, Ibaraki, Japan
| | - Motoo Osaka
- Department of Cardiovascular Surgery (B.Q.T., H.S., M.O., Y.H.), University of Tsukuba, Ibaraki, Japan
| | - Yuji Hiramatsu
- Department of Cardiovascular Surgery (B.Q.T., H.S., M.O., Y.H.), University of Tsukuba, Ibaraki, Japan
| | - Yoshito Yamashiro
- From the Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA) (S.J.S., H.T.H., T.S., Y.Y., H.Y.), University of Tsukuba, Ibaraki, Japan
| | - Hiromi Yanagisawa
- From the Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA) (S.J.S., H.T.H., T.S., Y.Y., H.Y.), University of Tsukuba, Ibaraki, Japan
- Division of Biomedical Science, Faculty of Medicine (H.Y.), University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
17
|
Cantrell R, Palumbo JS. The thrombin–inflammation axis in cancer progression. Thromb Res 2020; 191 Suppl 1:S117-S122. [DOI: 10.1016/s0049-3848(20)30408-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/18/2019] [Accepted: 11/21/2019] [Indexed: 02/07/2023]
|
18
|
Wojtukiewicz MZ, Skalij P, Tokajuk P, Politynska B, Wojtukiewicz AM, Tucker SC, Honn KV. Direct Oral Anticoagulants in Cancer Patients. Time for a Change in Paradigm. Cancers (Basel) 2020; 12:cancers12051144. [PMID: 32370207 PMCID: PMC7281117 DOI: 10.3390/cancers12051144] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 04/30/2020] [Accepted: 04/30/2020] [Indexed: 12/31/2022] Open
Abstract
Thrombosis is a more common occurrence in cancer patients compared to the general population and is one of the main causes of death in these patients. Low molecular weight heparin (LMWH) has been the recognized standard treatment for more than a decade, both in cancer-related thrombosis and in its prevention. Direct oral anticoagulants (DOACs) are a new option for anticoagulation therapy. Recently published results of large randomized clinical trials have confirmed that DOAC may be a reasonable alternative to LMWH in cancer patients. The following review summarizes the current evidence on the safety and efficacy of DOAC in the treatment and prevention of cancer-related thrombosis. It also draws attention to the limitations of this group of drugs, knowledge of which will facilitate the selection of optimal therapy.
Collapse
Affiliation(s)
- Marek Z. Wojtukiewicz
- Department of Oncology, Medical University of Białystok, 12 Ogrodowa St., 15-027 Białystok, Poland; (P.S.); (P.T.)
- Department of Clinical Oncology, Comprehensive Cancer Center, 12 OgrodowaSt., 15-369 Białystok, Poland
- Correspondence:
| | - Piotr Skalij
- Department of Oncology, Medical University of Białystok, 12 Ogrodowa St., 15-027 Białystok, Poland; (P.S.); (P.T.)
- Department of Clinical Oncology, Comprehensive Cancer Center, 12 OgrodowaSt., 15-369 Białystok, Poland
| | - Piotr Tokajuk
- Department of Oncology, Medical University of Białystok, 12 Ogrodowa St., 15-027 Białystok, Poland; (P.S.); (P.T.)
- Department of Clinical Oncology, Comprehensive Cancer Center, 12 OgrodowaSt., 15-369 Białystok, Poland
| | - Barbara Politynska
- Department of Philosophy and Human Psychology, Medical University of Białystok, 37 Szpitalna St., 15-295 Białystok, Poland; (B.P.); (A.M.W.)
- Robinson College, University of Cambridge, Cambridge CB3 9AN, UK
| | - Anna M. Wojtukiewicz
- Department of Philosophy and Human Psychology, Medical University of Białystok, 37 Szpitalna St., 15-295 Białystok, Poland; (B.P.); (A.M.W.)
| | - Stephanie C. Tucker
- Bioactive Lipids Research Program, Department of Pathology-School of Medicine, Detroit, MI 48202, USA; (S.C.T.); (K.V.H.)
| | - Kenneth V. Honn
- Bioactive Lipids Research Program, Department of Pathology-School of Medicine, Detroit, MI 48202, USA; (S.C.T.); (K.V.H.)
- Department of Chemistry, Wayne State University, Detroit, MI 48202, USA
- Department of Oncology, Karmanos Cancer Institute, Detroit, MI 48202, USA
| |
Collapse
|
19
|
Najidh S, Versteeg HH, Buijs JT. A systematic review on the effects of direct oral anticoagulants on cancer growth and metastasis in animal models. Thromb Res 2020; 187:18-27. [PMID: 31945588 DOI: 10.1016/j.thromres.2019.12.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/18/2019] [Accepted: 12/28/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Direct oral anticoagulants (DOACs) are now the first choice thromboprophylaxis in cancer patients who do not have a high risk of bleeding. In addition to the anticoagulant effects, potential anti-tumor effects of DOACs have also been studied in animal cancer models. In this study, we summarize the effects of DOACs on cancer growth and metastasis in animal models through a systematic review with a qualitative analysis. METHODS PubMed, EMBASE and Web of Science were systematically searched for original studies that describe animal models of cancer in which one of the experimental groups received DOAC monotherapy, and which reported quantitatively on primary tumor or metastases. RESULTS Nine studies - reporting a total of 19 animal experiments - met the inclusion criteria. These 19 experiments included spontaneous cancer (n = 2), carcinogenicity (n = 2), xenograft (n = 7) and syngeneic (n = 8) models, encompassing orthotopic (n = 7), subcutaneous (n = 5), intraperitoneal (n = 1) and intravenous (n = 2) injection of cancer cells and included treatments with the DOACs ximelagatran (n = 4), dabigatran etexilate (n = 6) and/or rivaroxaban (n = 11). DOAC treatment decreased tumor growth at implanted and metastatic site in 18.8% (3/16) and 20.0% (3/15) of the experiments, respectively. Conversely, DOACs increased tumor growth at implanted and metastatic site in 6.3% (1/16) and 20.0% (3/15) of the experiments, respectively. CONCLUSION DOAC monotherapy resulted in neoplastic changes in a rat carcinogenicity study, showed a lack of effect in mouse xenograft models, while the effect on cancer growth and metastasis in mouse syngeneic models depended on the timing of DOAC treatment and type of cancer model used.
Collapse
Affiliation(s)
- Safa Najidh
- Dept. of Dermatology, Leiden University Medical Center, Leiden, the Netherlands.
| | - Henri H Versteeg
- Einthoven Laboratory for Vascular and Regenerative Medicine, Div. of Thrombosis & Hemostasis, Dept. of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| | - Jeroen T Buijs
- Einthoven Laboratory for Vascular and Regenerative Medicine, Div. of Thrombosis & Hemostasis, Dept. of Internal Medicine, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
20
|
The endothelial barrier and cancer metastasis: Does the protective facet of platelet function matter? Biochem Pharmacol 2020; 176:113886. [PMID: 32113813 DOI: 10.1016/j.bcp.2020.113886] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/24/2020] [Indexed: 12/16/2022]
Abstract
Overwhelming evidence suggests that platelets have a detrimental role in promoting cancer spread via platelet-cancer cell interactions linked to thrombotic mechanisms. On the other hand, a beneficial role of platelets in the preservation of the endothelial barrier in inflammatory conditions has been recently described, a phenomenon that could also operate in cancer-related inflammation. It is tempting to speculate that some antiplatelet strategies to combat cancer metastasis may impair the endogenous platelet-dependent mechanisms preserving endothelial barrier function. If the protective function of platelets is impaired, it may lead to increased endothelial permeability and more efficient cancer cell intravasation in the primary tumor and cancer cell extravasation at metastatic sites. In this commentary, we discuss current evidence that could support this hypothesis.
Collapse
|
21
|
Shaker H, Bundred NJ, Landberg G, Pritchard SA, Albadry H, Nicholson SL, Harries LJ, Heah JYE, Castle J, Kirwan CC. Breast cancer stromal clotting activation (Tissue Factor and thrombin): A pre-invasive phenomena that is prognostic in invasion. Cancer Med 2020; 9:1768-1778. [PMID: 31962001 PMCID: PMC7050075 DOI: 10.1002/cam4.2748] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/13/2019] [Accepted: 11/15/2019] [Indexed: 12/25/2022] Open
Abstract
Background Tumor stroma, of which fibroblasts are the most abundant cell, resembles a non‐healing wound, where a procoagulant environment creates a permissive milieu for cancer growth. We aimed to determine if tumor expression of coagulation factors (procoagulant phenotype), and systemic hypercoagulability, occur at the preinvasive (ductal carcinoma in situ; DCIS) stage and correlate with breast cancer subtype, disease‐free survival (DFS), and overall survival (OS). Methods In a prospective cohort of early breast cancer (DCIS, n = 76; invasive, n = 248) tumor, normal breast and plasma were examined. Fibroblast and epithelial expression of Tissue Factor (TF), thrombin, PAR1, PAR2, and plasma thrombin‐antithrombin (TAT) and D‐dimer were correlated with clinicopathological data, and 5‐year survival. Results Fibroblast expression of TF, thrombin, and PAR1 was increased in DCIS and invasive cancer compared to normal breast fibroblasts (P ≤ .003, all). Fibroblast TF, thrombin, PAR1, and PAR2 was increased in cancers with high Ki67, high grade, ER‐ (vs ER+), and HER2+ (vs HER2‐) (all P < .05). On univariate analysis, fibroblast TF expression was inversely associated with DFS (P = .04) and OS (P = .02). D‐dimer was higher in node positive (507 (CI: 411‐625) ng/mL, n = 68) vs negative patients (428 (CI: 387‐472) ng/mL, n = 171, P = .004) and inversely associated with OS (P = .047). On multivariate analysis, plasma TAT was associated with reduced OS (HR 3.26, CI 1.16‐3.1, P = .02), with a high plasma TAT (≥3.2 ng/mL) associated with > 3‐fold mortality risk compared to low TAT. Conclusion This demonstrates procoagulant phenotypic changes occur in fibroblasts at the preinvasive stage. Fibroblast procoagulant phenotype is associated with aggressive breast cancer subtypes and reduced survival. Coagulation may be a therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Hudhaifah Shaker
- Faculty of Biology, Medicine and Health, Division of Cancer Sciences, School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Nigel J Bundred
- Faculty of Biology, Medicine and Health, Division of Cancer Sciences, School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Göran Landberg
- Department of Pathology, Institute for Biomedicine, Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Susan A Pritchard
- Department of Histopathology, Manchester University NHS Foundation Trust, Wythenshawe, Manchester, UK
| | - Harith Albadry
- Department of Histopathology, Royal Liverpool and Broadgreen University Hospitals NHS Trust, Liverpool, UK
| | - Sarah L Nicholson
- Department of Histopathology, East Lancashire Hospitals NHS Trust, Blackburn, UK
| | - Lauren J Harries
- Department of Histopathology, Manchester University NHS Foundation Trust, Wythenshawe, Manchester, UK
| | - Jing Y E Heah
- The Nightingale Centre and Prevent Breast Cancer Research Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - John Castle
- Faculty of Biology, Medicine and Health, Division of Cancer Sciences, School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester, UK
| | - Cliona C Kirwan
- Faculty of Biology, Medicine and Health, Division of Cancer Sciences, School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Manchester, UK.,The Nightingale Centre and Prevent Breast Cancer Research Centre, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
22
|
Maqsood A, Hisada Y, Garratt KB, Homeister J, Mackman N. Rivaroxaban does not affect growth of human pancreatic tumors in mice. J Thromb Haemost 2019; 17:2169-2173. [PMID: 31393055 PMCID: PMC6893077 DOI: 10.1111/jth.14604] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 08/02/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Some clinical studies have shown that low-molecular-weight heparins (LMWHs) prolong the survival of cancer patients. In addition, various anticoagulants have been shown to reduce growth of tumors in mice. However, there are no studies on the effect of the factor Xa inhibitor rivaroxaban on growth of human pancreatic tumors in nude mice. OBJECTIVES To test the hypothesis that the factor Xa inhibitor rivaroxaban reduces the growth of tissue factor (TF)-positive pancreatic tumors but not TF-negative pancreatic tumors in mice. METHODS The TF-positive human pancreatic cancer cell line BxPc-3 and the TF-negative human pancreatic cancer cell line MIA PaCa-2 were injected subcutaneously into nude mice and tumors grown to a mean volume of ~100 mm3 . Mice were then divided into two groups. One group was fed chow containing rivaroxaban (0.5 g/kg chow) whereas the other group was fed chow without rivaroxaban. RESULTS Rivaroxaban significantly prolonged prothrombin time in tumor-bearing mice. Rivaroxaban did not affect cell proliferation or growth of either BxPc-3 or MIA PaCa-2 tumors grown subcutaneously in nude mice. CONCLUSION Our results indicate that inhibition of factor Xa with rivaroxaban does not affect the growth of two human pancreatic tumors in nude mice.
Collapse
Affiliation(s)
- Anaum Maqsood
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yohei Hisada
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Kenison B. Garratt
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Jonathan Homeister
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Nigel Mackman
- Department of Medicine, Division of Hematology and Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
23
|
Kinnunen PT, Murto MO, Artama M, Pukkala E, Visvanathan K, Murtola TJ. Anticoagulants and Breast Cancer Survival: A Nationwide Cohort Study. Cancer Epidemiol Biomarkers Prev 2019; 29:208-215. [PMID: 31653681 DOI: 10.1158/1055-9965.epi-19-0147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/04/2019] [Accepted: 10/07/2019] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Various components of the coagulation cascade have been linked to breast cancer progression. In vivo results suggest that anticoagulants possess anticancer properties, but there are virtually no studies in human populations. Our nationwide study explored the association between anticoagulant use and breast cancer survival. METHODS All anticoagulants used from 1995 to 2015 in women (n = 73,170) diagnosed with invasive breast cancer in Finland between 1995 and 2013 were identified from the national prescription database; women were identified from the Finnish Cancer Registry. Cox regressions were performed to analyze breast cancer survival as a function of pre- and postdiagnostic anticoagulant use; analyses were conducted for different anticoagulant subtypes and overall. Models were adjusted for age, mammography screening, tumor clinical characteristics, comorbidities, statin use, antidiabetic use, and antihypertensive use. To control for immortal time bias, postdiagnostic anticoagulant use was analyzed as a time-dependent variable. RESULTS At a median of 5.8 years after breast cancer diagnosis, 10,900 (15%) women had died from breast cancer. In total, 25,622 (35%) women had used anticoagulants during the study period. Postdiagnostic anticoagulant use increased the risk of breast cancer death (HR = 1.41; 95% confidence interval, 1.33-1.49). The risk was especially high for low-molecular weight heparin, although the effect disappeared in long-term users. CONCLUSIONS Anticoagulant use provides no clinical benefit for breast cancer survival; however, the association between thrombosis and cancer might mask potential survival benefits. IMPACT Future pharmacoepidemiologic studies should adjust for anticoagulant use. Research should focus on the use of new oral anticoagulants because these are rarely studied and might be associated with improved breast cancer survival.
Collapse
Affiliation(s)
- Pete T Kinnunen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
| | - Mika O Murto
- Department of Surgery, Tampere University Hospital, Tampere, Finland
| | - Miia Artama
- National Institute for Health and Welfare, Tampere, Finland
| | - Eero Pukkala
- Faculty of Social Sciences, University of Tampere, Tampere, Finland.,Finnish Cancer Registry - Institute for Statistical and Epidemiological Cancer Research, Helsinki, Finland
| | - Kala Visvanathan
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Teemu J Murtola
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Department of Urology, Tampere University Hospital, Tampere, Finland.,Seinäjoki Central Hospital, Department of Surgery, Seinäjoki, Finland
| |
Collapse
|
24
|
Michael N, Grigoryan MM, Kilday K, Sumbria RK, Vasilevko V, van Ryn J, Cribbs DH, Paganini-Hill A, Fisher MJ. Effects of Dabigatran in Mouse Models of Aging and Cerebral Amyloid Angiopathy. Front Neurol 2019; 10:966. [PMID: 31611836 PMCID: PMC6776875 DOI: 10.3389/fneur.2019.00966] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 08/23/2019] [Indexed: 01/11/2023] Open
Abstract
Oral anticoagulants are a critical component of stroke prevention, but carry a risk of brain hemorrhage. These hemorrhagic complications tend to occur in elderly individuals, especially those with predisposing conditions such as cerebral amyloid angiopathy (CAA). Clinical evidence suggests that non-vitamin K antagonist oral anticoagulants are safer than traditional oral anticoagulants. We analyzed whether the anticoagulant dabigatran produces cerebral microhemorrhage (the pathological substrate of MRI-demonstrable cerebral microbleeds) or intracerebral hemorrhage in aged mice with and without hemorrhage-predisposing angiopathy. We studied aged (22 months old) Tg2576 (a model of CAA) and wild-type (WT) littermate mice. Mice received either dabigatran etexilate (DE) (Tg N = 7; WT N = 10) or vehicle (Tg N = 9; WT N = 7) by gavage for 4 weeks. Anticoagulation effects of DE were confirmed using thrombin time assay. No mice experienced intracerebral hemorrhage. Cerebral microhemorrhage analysis, performed using Prussian-blue and H&E staining, showed no significant change in either number or size of cerebral microhemorrhage in DE-treated animals. Analysis of biochemical parameters for endothelial activation (ICAM-1), blood-brain barrier disruption (IgG, claudin-5, fibrinogen), microglial activation (Iba-1), or astrocyte activation (GFAP) showed neither exacerbation nor protective effects of DE in either Tg2576 or WT mice. Our study provides histological and biochemical evidence that aged mice, with or without predisposing factors for brain hemorrhage, tolerate anticoagulation with dabigatran. The absence of dabigatran-induced intracerebral hemorrhage or increased frequency of acute microhemorrhage may provide some reassurance for its use in high-risk patient populations.
Collapse
Affiliation(s)
- Neethu Michael
- Department of Neurology, University of California, Irvine, Irvine, CA, United States
| | | | - Kelley Kilday
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| | - Rachita K Sumbria
- Department of Neurology, University of California, Irvine, Irvine, CA, United States.,Department of Biopharmaceutical Sciences, School of Pharmacy and Health Sciences, Keck Graduate Institute, Claremont, CA, United States
| | - Vitaly Vasilevko
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| | - Joanne van Ryn
- Department of Cardiometabolic Research, Boehringer Ingelheim, Hanover, Germany
| | - David H Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, United States
| | - Annlia Paganini-Hill
- Department of Neurology, University of California, Irvine, Irvine, CA, United States
| | - Mark J Fisher
- Department of Neurology, University of California, Irvine, Irvine, CA, United States.,Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA, United States.,Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
25
|
Kirwan CC, Descamps T, Castle J. Circulating tumour cells and hypercoagulability: a lethal relationship in metastatic breast cancer. Clin Transl Oncol 2019; 22:870-877. [PMID: 31473984 PMCID: PMC7188731 DOI: 10.1007/s12094-019-02197-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/28/2019] [Indexed: 12/27/2022]
Abstract
Purpose Circulating tumour cells (CTCs) are a marker of poor prognosis and are associated with increased risk of venous thromboembolism in metastatic breast cancer (MBC). We aimed to determine if the presence of CTCs and plasma markers of hypercoagulability [thrombin–antithrombin III (TAT), fibrinogen and d-dimer] are biomarkers of survival in MBC. Methods/patients In a prospective study of MBC patients, CTC (CellSearch®) enumeration and plasma TAT, fibrinogen and d-dimer measured prior to commencement of treatment for disease progression were correlated to overall survival. Results At study completion, of 50 MBC patients recruited (median age 59 years, range 36–82), 40 patients had died (median survival 417 days, range 58–2141). CTCs (≥ 1/7.5 ml) were identified in 16 patients (median number of cells per 7.5 ml, 3 (range 1–31) and were associated with systemic hypercoagulability (medians TAT: 8.1 vs. 5.2 ng/ml, p = 0.03; fibrinogen: 4.3 vs. 3.1 g/l, p = 0.03; d-dimer: 1327 vs. 683 ng/ml, p = 0.0001). At 1 year, of 16 patients with ≥ 1 CTC, 7 had died (44%), compared to 5 of 26 (19%) patients in the no-CTC group. The presence of ≥ 1 CTC was associated with a trend for reduced overall survival (median 455 days vs. 614 days, p = 0.15). Plasma TAT inversely correlated with survival and was significantly higher in patients dying within 1 year (median 9.8 vs. 5.2 ng/ml, p = 0.004) whilst d-dimer showed a trend for reduced 1-year survival (median 1211 vs. 817 ng/ml, p = 0.06). MBC patients with combined high d-dimer (≥ 895 ng/ml) and CTC positivity (≥ 1/7.5 ml whole peripheral blood) had significantly reduced survival (p = 0.04). Conclusions The correlation between CTCs, hypercoagulability and reduced survival in MBC suggests the coagulation system supports tumour cell metastasis and is, therefore, a potential therapeutic target. Electronic supplementary material The online version of this article (10.1007/s12094-019-02197-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- C C Kirwan
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK. .,Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Southmoor Road, Wythenshawe, Manchester, M23 9LT, UK.
| | - T Descamps
- Centre for Cancer Biomarker Sciences, Cancer Research UK Manchester Institute, University of Manchester, Alderley Park, Macclesfield, SK10 4TG, UK
| | - J Castle
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, School of Medical Sciences, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester, M20 4GJ, UK
| |
Collapse
|
26
|
Buijs JT, Laghmani EH, van den Akker RFP, Tieken C, Vletter EM, van der Molen KM, Crooijmans JJ, Kroone C, Le Dévédec SE, van der Pluijm G, Versteeg HH. The direct oral anticoagulants rivaroxaban and dabigatran do not inhibit orthotopic growth and metastasis of human breast cancer in mice. J Thromb Haemost 2019; 17:951-963. [PMID: 30929299 PMCID: PMC6849835 DOI: 10.1111/jth.14443] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/18/2019] [Indexed: 12/21/2022]
Abstract
Essentials Factor Xa (FXa)-targeting direct oral anticoagulants (DOACs) reduce venous thromboembolism (VTE) The effects of FXa-targeting DOACs on cancer progression remain to be studied In xenograft models, a FXa-targeting DOAC did not inhibit breast cancer growth and metastasis A thrombin-targeting DOAC, dabigatran, also did not inhibit breast cancer growth and metastasis ABSTRACT: Background Factor Xa-targeting DOACs were recently found to reduce recurrent VTE efficiently in cancer patients when compared to the standard treatment with low-molecular-weight heparins (LMWHs). While the anticancer effects of LMWHs have been extensively studied in preclinical cancer models, the effects of FXa-targeting DOACs on cancer progression remain to be studied. Objective We investigated whether the FXa-targeting DOAC rivaroxaban and the thrombin-targeting DOAC dabigatran etexilate (DE) affected human breast cancer growth and metastasis in orthotopic xenograft models. Methods/results Mice that were put on a custom-made chow diet supplemented with rivaroxaban (0.4 or 1.0 mg/g diet) or dabigatran etexilate (DE) (10 mg/g diet) showed prolonged ex vivo coagulation times (prothrombin time [PT] and activated partial thromboplastin time [aPTT] assay, respectively). However, rivaroxaban and DE did not inhibit MDA-MB-231 tumor growth and metastasis formation in lungs or livers of 7-week-old fully immunodeficient NOD/SCID/ƴC-/- (NSG) mice. Comparable data were obtained for rivaroxaban-treated mice when using NOD-SCID mice. Rivaroxaban and DE treatment also did not significantly inhibit tumor growth and metastasis formation when using another human triple negative breast cancer (TNBC) cell line (HCC1806) in NOD-SCID mice. The FXa and thrombin-induced gene expression of the downstream target CXCL8 in both cell lines, but FXa and thrombin, did not significantly stimulate migration, proliferation, or stemness in vitro. Conclusion Although effectively inhibiting coagulation, the DOACs rivaroxaban and DE did not inhibit orthotopic growth and metastasis of human TNBC. It remains to be investigated whether DOACs exert antitumorigenic effects in other types of cancer.
Collapse
Affiliation(s)
- Jeroen T. Buijs
- Einthoven Laboratory for Vascular and Regenerative MedicineDivision of Thrombosis and HemostasisDepartment of Internal MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - El H. Laghmani
- Einthoven Laboratory for Vascular and Regenerative MedicineDivision of Thrombosis and HemostasisDepartment of Internal MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Rob F. P. van den Akker
- Einthoven Laboratory for Vascular and Regenerative MedicineDivision of Thrombosis and HemostasisDepartment of Internal MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Chris Tieken
- Einthoven Laboratory for Vascular and Regenerative MedicineDivision of Thrombosis and HemostasisDepartment of Internal MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Esther M. Vletter
- Einthoven Laboratory for Vascular and Regenerative MedicineDivision of Thrombosis and HemostasisDepartment of Internal MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Kim M. van der Molen
- Einthoven Laboratory for Vascular and Regenerative MedicineDivision of Thrombosis and HemostasisDepartment of Internal MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Juliette J. Crooijmans
- Einthoven Laboratory for Vascular and Regenerative MedicineDivision of Thrombosis and HemostasisDepartment of Internal MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Chantal Kroone
- Einthoven Laboratory for Vascular and Regenerative MedicineDivision of Thrombosis and HemostasisDepartment of Internal MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Sylvia E. Le Dévédec
- Division of Drug Discovery and SafetyLeiden Academic Center for Drug ResearchLeiden UniversityLeidenThe Netherlands
| | | | - Henri H. Versteeg
- Einthoven Laboratory for Vascular and Regenerative MedicineDivision of Thrombosis and HemostasisDepartment of Internal MedicineLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
27
|
Featherby S, Xiao YP, Ettelaie C, Nikitenko LL, Greenman J, Maraveyas A. Low molecular weight heparin and direct oral anticoagulants influence tumour formation, growth, invasion and vascularisation by separate mechanisms. Sci Rep 2019; 9:6272. [PMID: 31000751 PMCID: PMC6472388 DOI: 10.1038/s41598-019-42738-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 04/05/2019] [Indexed: 12/14/2022] Open
Abstract
The bidirectional association between coagulation and cancer has been established. However, anticoagulant therapies have been reported to have beneficial outcomes by influencing the vascularisation of the tumours. In this study the influence of a set of anticoagulants on tumour formation, invasion and vascularisation was examined. WM-266-4 melanoma and AsPC-1 pancreatic cancer cell lines were treated with LMWH (Tinzaparin and Dalteparin), and DOAC (Apixaban and Rivaroxaban) and the rate of tumour formation, growth and invasion were measured in vitro. In addition, the influence of these anticoagulants on vascularisation was examined using the chorioallantoic membrane assay (CAM) model and compared to the outcome of treatment with Bevacizumab. Using this model the influence of pharmacological concentrations of the anticoagulant on the growth, invasion and vascularisation of tumours derived from WM-266-4 and AsPC-1 cells was also measured in vivo. Tinzaparin and Daltepain reduced tumour formation and invasion by the cell lines in vitro, but with dissimilar potencies. In addition, treatment of CAM with LMWH reduced the local vascular density beyond that achievable with Bevacizumab, particularly suppressing the formation of larger-diameter blood vessels. In contrast, treatment with DOAC was largely ineffective. Treatment of CAM-implanted tumours with LMWH also reduced tumour vascularisation, while treatment of tumours with Apixaban reduced tumour growth in vivo. In conclusion, LMWH and DOAC appear to have anti-cancer properties that are exerted through different mechanisms.
Collapse
Affiliation(s)
- Sophie Featherby
- Biomedical Section, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| | - Yu Pei Xiao
- Division of Cancer-Hull York Medical School, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| | - Camille Ettelaie
- Biomedical Section, University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | - Leonid L Nikitenko
- Biomedical Section, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| | - John Greenman
- Biomedical Section, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| | - Anthony Maraveyas
- Division of Cancer-Hull York Medical School, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| |
Collapse
|
28
|
Chu G, Versteeg HH, Verschoor AJ, Trines SA, Hemels MEW, Ay C, Huisman MV, Klok FA. Atrial fibrillation and cancer - An unexplored field in cardiovascular oncology. Blood Rev 2019; 35:59-67. [PMID: 30928168 DOI: 10.1016/j.blre.2019.03.005] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 03/15/2019] [Accepted: 03/22/2019] [Indexed: 12/28/2022]
Abstract
An increasing body of evidence suggests an association between cancer and atrial fibrillation (AF). The exact magnitude and underlying mechanism of this association are however unclear. Cancer-related inflammation, anti-cancer treatment and other cancer-related comorbidities are proposed to affect atrial remodelling, increasing the susceptibility of cancer patients for developing AF. Moreover, cancer is assumed to modify the risk of thromboembolisms and bleeding. A thorough and adequate understanding of these risks is however lacking, as current literature is scarce and show ambiguous results in AF patients. The standardized risk-models that normally aid the clinician in the decision of initiating anticoagulant therapy do not take the presence of malignancy into account. Other factors that complicate risk assessment in AF patients with cancer include drug-drug interactions and other cancer-related comorbidities such as renal impairment. In this review, we highlight the available literature regarding epidemiological association, risk assessment and anticoagulation therapy in AF patients with cancer.
Collapse
Affiliation(s)
- Gordon Chu
- Department of Thrombosis and Haemostasis, Leiden University Medical Centre, the Netherlands.
| | - Henri H Versteeg
- Department of Thrombosis and Haemostasis, Leiden University Medical Centre, the Netherlands
| | - Arie J Verschoor
- Department of Medical Oncology, Leiden University Medical Centre, the Netherlands
| | - Serge A Trines
- Department of Cardiology, Heart-Lung Centre, Leiden University Medical Centre, the Netherlands
| | - Martin E W Hemels
- Department of Cardiology, Rijnstate, Arnhem, the Netherlands; Department of Cardiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cihan Ay
- Clinical Division of Haematology and Haemostaseology, Department of Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, Austria
| | - Menno V Huisman
- Department of Thrombosis and Haemostasis, Leiden University Medical Centre, the Netherlands
| | - Frederikus A Klok
- Department of Thrombosis and Haemostasis, Leiden University Medical Centre, the Netherlands
| |
Collapse
|
29
|
Grandoni F, Alberio L. Direct Oral Anticoagulant Drugs: On the Treatment of Cancer-Related Venous Thromboembolism and their Potential Anti-Neoplastic Effect. Cancers (Basel) 2019; 11:E46. [PMID: 30621261 PMCID: PMC6356803 DOI: 10.3390/cancers11010046] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/24/2018] [Accepted: 12/28/2018] [Indexed: 12/27/2022] Open
Abstract
Cancer patients develop a hypercoagulable state with a four- to seven-fold higher thromboembolic risk compared to non-cancer patients. Thromboembolic events can precede the diagnosis of cancer, but they more often occur at diagnosis or during treatment. After malignancy itself, they represent the second cause of death. Low molecular weight heparins are the backbone of the treatment of cancer-associated thromboembolism. This treatment paradigm is possibly changing, as direct oral anticoagulants (DOACs) may prove to be an alternative therapeutic option. The currently available DOACs were approved during the first and second decades of the 21st century for various clinical indications. Three molecules (apixaban, edoxaban and rivaroxaban) are targeting the activated factor X and one (dabigatran) is directed against the activated factor II, thrombin. The major trials analyzed the effect of these agents in the general population, with only a small proportion of cancer patients. Two published and several ongoing studies are specifically investigating the use of DOACs in cancer-associated thromboembolism. This article will review the current available literature on the use of DOACs in cancer patients. Furthermore, we will discuss published data suggesting potential anti-cancer actions exerted by non-anticoagulant effects of DOACs. As soon as more prospective data becomes available, DOACs are likely to be considered as a potential new therapeutic option in the armamentarium for patients suffering of cancer-associated thromboembolism.
Collapse
Affiliation(s)
- Francesco Grandoni
- Division of Haematology and Haematology Central Laboratory, CHUV, University Hospital of Lausanne, 1011 Lausanne, Switzerland.
| | - Lorenzo Alberio
- Division of Haematology and Haematology Central Laboratory, CHUV, University Hospital of Lausanne, 1011 Lausanne, Switzerland.
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland.
| |
Collapse
|
30
|
Posma JJ, Grover SP, Hisada Y, Owens AP, Antoniak S, Spronk HM, Mackman N. Roles of Coagulation Proteases and PARs (Protease-Activated Receptors) in Mouse Models of Inflammatory Diseases. Arterioscler Thromb Vasc Biol 2019; 39:13-24. [PMID: 30580574 PMCID: PMC6310042 DOI: 10.1161/atvbaha.118.311655] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 10/19/2018] [Indexed: 12/20/2022]
Abstract
Activation of the blood coagulation cascade leads to fibrin deposition and platelet activation that are required for hemostasis. However, aberrant activation of coagulation can lead to thrombosis. Thrombi can cause tissue ischemia, and fibrin degradation products and activated platelets can enhance inflammation. In addition, coagulation proteases activate cells by cleavage of PARs (protease-activated receptors), including PAR1 and PAR2. Direct oral anticoagulants have recently been developed to specifically inhibit the coagulation proteases FXa (factor Xa) and thrombin. Administration of these inhibitors to wild-type mice can be used to determine the roles of FXa and thrombin in different inflammatory diseases. These results can be compared with the phenotypes of mice with deficiencies of either Par1 (F2r) or Par2 (F2rl1). However, inhibition of coagulation proteases will have effects beyond reducing PAR signaling, and a deficiency of PARs will abolish signaling from all proteases that activate these receptors. We will summarize studies that examine the roles of coagulation proteases, particularly FXa and thrombin, and PARs in different mouse models of inflammatory disease. Targeting FXa and thrombin or PARs may reduce inflammatory diseases in humans.
Collapse
Affiliation(s)
- Jens J Posma
- Laboratory for Clinical Thrombosis and Hemostasis, Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Steven P Grover
- Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yohei Hisada
- Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - A. Phillip Owens
- Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, OH, USA
| | - Silvio Antoniak
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Henri M Spronk
- Laboratory for Clinical Thrombosis and Hemostasis, Departments of Internal Medicine and Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Nigel Mackman
- Thrombosis and Hemostasis Program, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
31
|
Mechanisms coupling thrombin to metastasis and tumorigenesis. Thromb Res 2018; 164 Suppl 1:S29-S33. [PMID: 29703481 DOI: 10.1016/j.thromres.2017.12.020] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 12/29/2017] [Indexed: 01/01/2023]
Abstract
The association of malignancy and thrombophilia is bidirectional, as evidenced by four decades of studies in animal models showing that hemostatic system components support cancer progression. Consistent with this view, clinical studies have suggested that anticoagulants not only limit thromboembolic complications associated with cancer, but also improve survival by impeding cancer progression, and may even prevent the development of cancer. In order to fully capitalize on this association, a detailed understanding of the mechanisms coupling hemostatic factors to cancer pathogenesis is required. Multiple studies have shown that thrombin-mediated procoagulant functions strongly promote metastatic potential. In particular, the platelet/fibrin(ogen) axis has been shown to protect newly formed micrometastases from innate immune surveillance, contribute to creation of a metastatic niche by recruitment of prometastatic inflammatory cells, and promote the epithelial to mesenchymal transition of metastatic cells. Thrombin-mediated functions have also been shown to support tumor growth in some contexts, and have even been linked to tumorigenesis in the setting of inflammation-driven colon cancer. Here, local thrombin-mediated extravascular fibrin deposition, and specifically fibrin-αMβ2 integrin interaction, push intestinal inflammatory cells toward a pro-tumorigenic phenotype, resulting in the elaboration of key cytokines and growth factors that support the proliferation and survival of transformed intestinal epithelial cells. These studies reveal that hemostatic factors can serve as a bridge between pathological inflammation and the development of cancer. As a large proportion of cancers are caused by pathological inflammation, these studies suggest that therapies targeting the nexus between hemostasis and inflammation could be used to prevent cancer development.
Collapse
|
32
|
Xu D, Su C, Pan J. Advances and Application of a Novel Oral Anticoagulant in Specific Populations: Dabigatran Etexilate. Curr Drug Metab 2018; 21:106-111. [PMID: 30317993 DOI: 10.2174/1389200219666181011160133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 05/31/2018] [Accepted: 08/06/2018] [Indexed: 11/22/2022]
Abstract
BACKGROUND Dabigatran etexilate (DE) was approved by the FDA in 2010 to reduce the risk of stroke and systemic embolism in adults with Non-valvular Atrial Fibrillation (NVAF). Compared with warfarin, a traditional anticoagulant drug, DE exhibits a shorter half-life, improved dose-effect relationship, fewer food and drug interactions, and can be taken orally without monitoring the conventional coagulation index. DE can also prevent or reduce the severity of adverse events, such as attenuated drug efficacy or bleeding. It is convenient for patients to take DE due to low levels of individual variation. This review aims to application of Dabigatran etexilate in specific populations. METHODS Fifty-five papers were included in the review. RESULTS We review the pharmacological mechanisms, pharmacokinetics and drug interactions, as well as the application of DE for different clinical populations, and provide clinical guidelines. CONCLUSION When using DE, one should consider the risk of bleeding, age, renal function, drug interactions, and other factors.
Collapse
Affiliation(s)
- Delai Xu
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Jiangsu, China
| | - Cujin Su
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Jiangsu, China
| | - Jie Pan
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, Jiangsu, China
| |
Collapse
|
33
|
Antoniak S, Tatsumi K, Schmedes CM, Grover SP, Pawlinski R, Mackman N. Protease-activated receptor 1 activation enhances doxorubicin-induced cardiotoxicity. J Mol Cell Cardiol 2018; 122:80-87. [PMID: 30098988 DOI: 10.1016/j.yjmcc.2018.08.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 08/07/2018] [Accepted: 08/09/2018] [Indexed: 12/21/2022]
Abstract
OBJECTIVE The anti-cancer anthracycline drug Doxorubicin (Dox) causes cardiotoxicity. We investigated the role of protease-activated receptor 1 (PAR-1) in Dox-induced cardiotoxicity. METHODS AND RESULTS In vitro experiments revealed that PAR-1 enhanced Dox-induced mitochondrial dysfunction, reactive oxygen species and cell death of cardiac myocytes and cardiac fibroblasts. The contribution of PAR-1 to Dox-induced cardiotoxicity was investigated by subjecting PAR-1-/- mice and PAR-1+/+ mice to acute and chronic exposure to Dox. Heart function was measured by echocardiography. PAR-1-/- mice exhibited significant less cardiac injury and dysfunction compared to PAR-1+/+ mice after acute and chronic Dox administration. PAR-1-/- mice had reduced levels of nitrotyrosine, apoptosis and inflammation in their heart compared to PAR-1+/+ mice. Furthermore, inhibition of PAR-1 in wild-type mice with vorapaxar significantly reduced the acute Dox-induced cardiotoxicity. CONCLUSION Our results indicate that activation of PAR-1 contributes to Dox-induced cardiotoxicity. Inhibition of PAR-1 may be a new approach to reduce Dox-induced cardiotoxicity in cancer patients.
Collapse
Affiliation(s)
- Silvio Antoniak
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| | - Kohei Tatsumi
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States; Department of Physiology and Regenerative Medicine, Kindai University, Faculty of Medicine, Osaka-sayama, Osaka, Japan
| | - Clare M Schmedes
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States
| | - Steven P Grover
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States
| | - Rafal Pawlinski
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States
| | - Nigel Mackman
- Department of Medicine, Thrombosis and Hemostasis Program, Division of Hematology and Oncology, UNC McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
34
|
Wojtukiewicz MZ, Hempel D, Sierko E, Tucker SC, Honn KV. Antiplatelet agents for cancer treatment: a real perspective or just an echo from the past? Cancer Metastasis Rev 2018; 36:305-329. [PMID: 28752248 PMCID: PMC5557869 DOI: 10.1007/s10555-017-9683-z] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The association between coagulation and cancer development has been observed for centuries. However, the connection between inflammation and malignancy is also well-recognized. The plethora of evidence indicates that among multiple hemostasis components, platelets play major roles in cancer progression by providing surface and granular contents for several interactions as well as behaving like immune cells. Therefore, the anticancer potential of anti-platelet therapy has been intensively investigated for many years. Anti-platelet agents may prevent cancer, decrease tumor growth, and metastatic potential, as well as improve survival of cancer patients. On the other hand, there are suggestions that antiplatelet treatment may promote solid tumor development in a phenomenon described as "cancers follow bleeding." The controversies around antiplatelet agents justify insight into the subject to establish what, if any, role platelet-directed therapy has in the continuum of anticancer management.
Collapse
Affiliation(s)
- Marek Z Wojtukiewicz
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa St., 15-025, Bialystok, Poland.
| | - Dominika Hempel
- Department of Radiotherapy, Comprehensive Cancer Center in Bialystok, Bialystok, Poland
| | - Ewa Sierko
- Department of Clinical Oncology, Comprehensive Cancer Center in Bialystok, Bialystok, Poland
| | - Stephanie C Tucker
- Department of Pathology-School of Medicine, Bioactive Lipids Research Program, Detroit, MI, 48202, USA
| | - Kenneth V Honn
- Department of Pathology-School of Medicine, Bioactive Lipids Research Program, Detroit, MI, 48202, USA.,Departments of Chemistry, Wayne State University, Detroit, MI, 48202, USA.,Department of Oncology, Karmanos Cancer Institute, Detroit, MI, 48202, USA
| |
Collapse
|
35
|
Wojtukiewicz MZ, Hempel D, Sierko E, Tucker SC, Honn KV. Thrombin-unique coagulation system protein with multifaceted impacts on cancer and metastasis. Cancer Metastasis Rev 2017; 35:213-33. [PMID: 27189210 DOI: 10.1007/s10555-016-9626-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The association between blood coagulation and cancer development is well recognized. Thrombin, the pleiotropic enzyme best known for its contribution to fibrin formation and platelet aggregation during vascular hemostasis, may also trigger cellular events through protease-activated receptors, PAR-1 and PAR-4, leading to cancer progression. Our pioneering findings provided evidence that thrombin contributes to cancer metastasis by increasing adhesive potential of malignant cells. However, there is evidence that thrombin regulates every step of cancer dissemination: (1) cancer cell invasion, detachment from primary tumor, migration; (2) entering the blood vessel; (3) surviving in vasculature; (4) extravasation; (5) implantation in host organs. Recent studies have provided new molecular data about thrombin generation in cancer patients and the mechanisms by which thrombin contributes to transendothelial migration, platelet/tumor cell interactions, angiogenesis, and other processes. Though a great deal is known regarding the role of thrombin in cancer dissemination, there are new data for multiple thrombin-mediated events that justify devoting focus to this topic with a comprehensive approach.
Collapse
Affiliation(s)
- Marek Z Wojtukiewicz
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa St., 15-025, Bialystok, Poland. .,Department of Clinical Oncology, Comprehensive Cancer Center in Bialystok, Bialystok, Poland.
| | - Dominika Hempel
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa St., 15-025, Bialystok, Poland.,Department of Radiotherapy, Comprehensive Cancer Center in Bialystok, Bialystok, Poland
| | - Ewa Sierko
- Department of Oncology, Medical University of Bialystok, 12 Ogrodowa St., 15-025, Bialystok, Poland.,Department of Radiotherapy, Comprehensive Cancer Center in Bialystok, Bialystok, Poland
| | - Stephanie C Tucker
- Bioactive Lipids Research Program, Department of Pathology-School of Medicine, Wayne State University, Detroit, MI, USA
| | - Kenneth V Honn
- Bioactive Lipids Research Program, Department of Pathology-School of Medicine, Wayne State University, Detroit, MI, USA.,Department of Chemistry, Wayne State University, Detroit, MI, USA.,Department of Oncology, Karmanos Cancer Institute, Detroit, MI, USA
| |
Collapse
|
36
|
Abstract
Venous thromboembolism (VTE) and cancer are strongly associated, and present a major challenge in cancer patient treatment. Cancer patients have a higher risk of developing VTE, although the risk differs widely between tumour types. VTE prophylaxis is routinely given to cancer patients, in the form of vitamin K antagonists (VKA) or low molecular weight heparin (LMWH). Several studies have reported that cancer patients receiving anticoagulants show prolonged survival and this effect was more pronounced in patients with a good prognosis, although the mechanism is poorly understood. Tissue Factor (TF) is the initiator of extrinsic coagulation, but its non-haemostatic signalling via protease-activated receptors (PARs) is a potent driver of tumour angiogenesis. Furthermore, coagulation activation is strongly implicated in tumour cell migration and metastasis. This review discusses the effects of anticoagulants on cancer progression in patients, tumour cell behaviour, angiogenesis, and metastasis in in vitro and in vivo models. Inhibition of TF signalling shows great promise in curbing angiogenesis and in vivo tumour growth, but whether this translates to patients is not yet known. Furthermore, non-haemostatic properties of coagulation factors in cancer progression are discussed, which provide exciting opportunities on limiting oncologic processes without affecting blood coagulation.
Collapse
Affiliation(s)
- Chris Tieken
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Centre, Leiden, The Netherlands
| | - Henri H Versteeg
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine, Leiden University Medical Centre, Leiden, The Netherlands.
| |
Collapse
|
37
|
Alexander ET, Minton AR, Hayes CS, Goss A, Van Ryn J, Gilmour SK. Thrombin inhibition and cyclophosphamide synergistically block tumor progression and metastasis. Cancer Biol Ther 2016; 16:1802-11. [PMID: 26383051 DOI: 10.1080/15384047.2015.1078025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Cancer is often associated with an increased risk of thrombotic events which are exacerbated by treatment with chemotherapeutics such as cyclosphosphamide (CP). Evidence suggests that thrombin can stimulate tumor progression via formation of fibrin and activation of protease-activated receptors (PARs) and platelets. We examined the effect of co-treatment with CP and dabigatran etexilate, a direct inhibitor of thrombin, using the murine orthotopic 4T1 tumor model. Mice receiving co-treatment with both low dose CP and dabigatran etexilate had significantly smaller mammary tumors and fewer lung metastases than mice treated with CP or dabigratran etexilate alone. Co-treatment with dabigatran etexilate and low dose CP also significantly decreased the number of arginase(+)Gr-1(+)CD11b(+) myeloid derived suppressor cells as well as levels of TGF-β in spleens from tumor bearing mice. 4T1 tumors express procoagulant tissue factor (TF) and spontaneously release TF(+) microparticles which are potent procoagulant factors that promote thrombin generation. Treatment with dabigatran etexilate alone prevented tumor-induced increases in circulating TF(+) microparticles and also decreased the numbers of tumor-induced activated platelets by 40%. These results show that co-treatment with dabigatran etexilate and CP synergistically inhibits growth and metastasis of mammary tumors, suggesting that oral administration of the thrombin inhibitor dabigatran etexilate may be beneficial in not only preventing thrombotic events in cancer patients but also in treating malignant tumors themselves.
Collapse
Affiliation(s)
| | | | - Candace S Hayes
- a Lankenau Institute for Medical Research ; Wynnewood , PA USA
| | - Ashley Goss
- b Boehringer Ingelheim Pharmaceuticals Inc. ; Ridgefield , CT USA
| | - Joanne Van Ryn
- c Boehringer Ingelheim Pharma GmbH & Co. KG ; Biberach an der Riss , Germany
| | - Susan K Gilmour
- a Lankenau Institute for Medical Research ; Wynnewood , PA USA
| |
Collapse
|
38
|
Rikhi R, Wilson EM, Deas O, Svalina MN, Bial J, Mansoor A, Cairo S, Keller C. Murine model of hepatic breast cancer. Biochem Biophys Rep 2016; 8:1-5. [PMID: 28955934 PMCID: PMC5613703 DOI: 10.1016/j.bbrep.2016.07.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 07/27/2016] [Accepted: 07/28/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND AIMS Breast cancer is the most common cancer in women and the second leading cause of cancer-related deaths in this population. Breast cancer related deaths have declined due to screening and adjuvant therapies, yet a driving clinical need exists to better understand the cause of the deadliest aspect of breast cancer, metastatic disease. Breast cancer metastasizes to several distant organs, the liver being the third most common site. To date, very few murine models of hepatic breast cancer exist. METHODS In this study, a novel murine model of liver breast cancer using the MDA-MB-231 cell line is introduced as an experimental (preclinical) model. RESULTS Histological typing revealed consistent hepatic breast cancer tumor foci. Common features of the murine model were vascular invasion, lung metastasis and peritoneal seeding. CONCLUSIONS The novel murine model of hepatic breast cancer established in this study provides a tool to be used to investigate mechanisms of hepatic metastasis and to test potential therapeutic interventions.
Collapse
Key Words
- AFP, Alpha Fetal Protein
- Ad:uPA, Adenovirus Urokinase Plasminogen Activator
- Breast cancer
- DMEM, Dulbecco's Modified Eagle Medium
- FRG™ KO, [ Fah(-/-) R ag2(-/-)Il2r g (-/-)])
- GPC3, Glypican-3
- HCM, Hepatocyte Culture Medium
- HER2, human epidermal growth factor receptor 2
- Hep Par 1, Hepatocyte Paraffin 1
- IACUC, Institutional Animal Care and Use Committee
- Liver metastasis
- NOD, Non-obese diabetic
- Preclinical model
- pfu, plaque forming units
Collapse
Affiliation(s)
- Rishi Rikhi
- Children's Cancer Therapy Development Institute, Fort Collins, CO 80524, USA
| | | | | | - Matthew N Svalina
- Children's Cancer Therapy Development Institute, Fort Collins, CO 80524, USA.,Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| | - John Bial
- Yecuris Corp., Tigard, OR 97062, USA
| | - Atiya Mansoor
- Department of Pathology, Oregon Health & Science University, Portland, OR 97239, USA
| | | | - Charles Keller
- Children's Cancer Therapy Development Institute, Fort Collins, CO 80524, USA.,Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
39
|
Auvergne R, Wu C, Connell A, Au S, Cornwell A, Osipovitch M, Benraiss A, Dangelmajer S, Guerrero-Cazares H, Quinones-Hinojosa A, Goldman SA. PAR1 inhibition suppresses the self-renewal and growth of A2B5-defined glioma progenitor cells and their derived gliomas in vivo. Oncogene 2016; 35:3817-28. [PMID: 26616854 PMCID: PMC4885796 DOI: 10.1038/onc.2015.452] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 09/14/2015] [Accepted: 10/22/2015] [Indexed: 12/15/2022]
Abstract
Glioblastoma (GBM) remains the most common and lethal intracranial tumor. In a comparison of gene expression by A2B5-defined tumor-initiating progenitor cells (TPCs) to glial progenitor cells derived from normal adult human brain, we found that the F2R gene encoding PAR1 was differentially overexpressed by A2B5-sorted TPCs isolated from gliomas at all stages of malignant development. In this study, we asked if PAR1 is causally associated with glioma progression. Lentiviral knockdown of PAR1 inhibited the expansion and self-renewal of human GBM-derived A2B5(+) TPCs in vitro, while pharmacological inhibition of PAR 1 similarly slowed both the growth and migration of A2B5(+) TPCs in culture. In addition, PAR1 silencing potently suppressed tumor expansion in vivo, and significantly prolonged the survival of mice following intracranial transplantation of human TPCs. These data strongly suggest the importance of PAR1 to the self-renewal and tumorigenicity of A2B5-defined glioma TPCs; as such, the abrogation of PAR1-dependent signaling pathways may prove a promising strategy for gliomas.
Collapse
Affiliation(s)
- R Auvergne
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - C Wu
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - A Connell
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - S Au
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - A Cornwell
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - M Osipovitch
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - A Benraiss
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - S Dangelmajer
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - H Guerrero-Cazares
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - A Quinones-Hinojosa
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - SA Goldman
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
- Center for Basic and Translational Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
40
|
Las trombosis venosas profundas distales de los miembros inferiores: un problema controvertido. ANGIOLOGIA 2016. [DOI: 10.1016/j.angio.2015.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
41
|
Adams GN, Rosenfeldt L, Frederick M, Miller W, Waltz D, Kombrinck K, McElhinney KE, Flick MJ, Monia BP, Revenko AS, Palumbo JS. Colon Cancer Growth and Dissemination Relies upon Thrombin, Stromal PAR-1, and Fibrinogen. Cancer Res 2015; 75:4235-43. [PMID: 26238780 DOI: 10.1158/0008-5472.can-15-0964] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/01/2015] [Indexed: 12/13/2022]
Abstract
Thrombin-mediated proteolysis is a major determinant of metastasis, but is not universally important for primary tumor growth. Here, we report that colorectal adenocarcinoma represents one important exception whereby thrombin-mediated functions support both primary tumor growth and metastasis. In contrast with studies of multiple nongastrointestinal cancers, we found that the growth of primary tumors formed by murine and human colon cancer cells was reduced in mice by genetic or pharmacologic reduction of circulating prothrombin. Reduced prothrombin expression was associated with lower mitotic indices and invasion of surrounding tissue. Mechanistic investigations revealed that thrombin-driven colonic adenocarcinoma growth relied upon at least two targets of thrombin-mediated proteolysis, protease-activated receptor-1 (PAR-1) expressed by stromal cells and the extracellular matrix protein, fibrinogen. Colonic adenocarcinoma growth was reduced in PAR-1-deficient mice, implicating stromal cell-associated PAR-1 as one thrombin target important for tumor outgrowth. Furthermore, tumor growth was dramatically impeded in fibrinogen-deficient mice, offering the first direct evidence of a critical functional role for fibrinogen in malignant tumor growth. Tumors harvested from fibrinogen-deficient mice displayed a relative reduction in cell proliferative indices, as well as increased tumor necrosis and decreased tumor vascular density. Collectively, our findings established a functional role for thrombin and its targets PAR-1 and fibrinogen in the pathogenesis of colonic adenocarcinoma, supporting tumor growth as well as local invasion and metastasis.
Collapse
Affiliation(s)
- Gregory N Adams
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Leah Rosenfeldt
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Malinda Frederick
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Whitney Miller
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Dusty Waltz
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Keith Kombrinck
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kathryn E McElhinney
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Matthew J Flick
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Brett P Monia
- Department of Antisense Drug Discovery, ISIS Pharmaceuticals, Inc., Carlsbad, California
| | - Alexey S Revenko
- Department of Antisense Drug Discovery, ISIS Pharmaceuticals, Inc., Carlsbad, California
| | - Joseph S Palumbo
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| |
Collapse
|
42
|
Spek CA, Versteeg HH, Borensztajn KS. Anticoagulant therapy of cancer patients: Will patient selection increase overall survival? Thromb Haemost 2015; 114:530-6. [PMID: 25994568 DOI: 10.1160/th15-02-0124] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/11/2015] [Indexed: 12/20/2022]
Abstract
Already since the early 1800s, it has been recognised that malignancies may provoke thromboembolic complications, and indeed cancer patients are at increased risk of developing venous thrombosis. Interestingly, case control studies of deep-vein thrombosis suggested that low-molecular-weight heparin (LMWH) improved survival of cancer patients. This led to the hypothesis that cancer cells might 'take advantage' of a hypercoagulable state to more efficiently metastasise. Initial randomised placebo control trials showed that LMWH improve overall survival of cancer patients, especially in those patients with a relatively good prognosis. The failure of recent phase III trials, however, tempers enthusiasm for anticoagulant treatment in cancer patients despite an overwhelming body of literature showing beneficial effects of anticoagulants in preclinical models. Instead of discarding LMWH as potential (co)treatment modality in cancer patients, these disappointing recent trials should guide future preclinical research on anticoagulants in cancer biology. Most and for all, the underlying mechanisms by which coagulation drives tumour progression need to be elucidated. This could ultimately allow selection of cancer patients most likely to benefit from anticoagulant treatment and/or from targeted therapy downstream of coagulation factor signalling.
Collapse
Affiliation(s)
- C Arnold Spek
- C. Arnold Spek, H2-157, Academic Medical Center, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands, Tel.: +31 20 5668750, E-mail:
| | | | | |
Collapse
|
43
|
Abstract
The progression of breast cancer from early-stage to metastatic disease results from a series of events during which malignant cells invade and travel within the bloodstream to distant sites, leading to a clonogenic accumulation of tumor cells in non-breast tissue. While mechanistically complex, an emerging literature supports hemostatic elements as an important patient factor that facilitates the metastatic potential of breast cancer. Hemostatic elements involved include platelets, coagulation, and fibrinolysis. Key steps in breast tumor progression, including cellular transformation, proliferation, tumor cell survival, and angiogenesis, can be mediated by components of the hemostatic system. Thus, the hemostatic system provides potential targets for novel therapeutic approaches to breast cancer therapy with drugs in current use and in development. The present article provides a comprehensive overview of the evidence and mechanisms supporting the roles played by platelets, coagulation activation, and the fibrinolytic system in breast cancer progression.
Collapse
|
44
|
The significance of circulating tumour cells in breast cancer: a review. Breast 2014; 23:552-60. [PMID: 25124235 DOI: 10.1016/j.breast.2014.07.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 07/03/2014] [Indexed: 12/15/2022] Open
Abstract
Haematogenous spread of circulating tumour cells (CTCs) is the principle mechanism for development of metastases. Research into the enumeration and characterisation of CTCs, particularly in the last decade, has allowed the introduction of semi-automated CTC assessment in the clinical setting. In breast cancer, CTC enumeration is being used as a prognostic biomarker, a predictive biomarker of treatment response and is being assessed to guide treatment in both the early and metastatic setting. CTC characterisation has the potential to direct targeted therapies, such as HER2 therapies in HER2 negative primary breast tumour patients. However, CTC assessment has considerable challenges. Capture and identification of these very rare cells is currently largely dependent on a presumed homogeneity of phenotype. In addition, high throughput assays are lacking. The clinical significance of CTCs is incompletely understood. A large proportion of CTC positive patients have no evidence of metastases, raising the issue of either inconsequential tumour dormancy or non-viable CTCs. CTCs may have additional clinical sequelae such as promoting venous thrombosis. However CTCs provide a real-time liquid biopsy of the tumour and represent an exciting, minimally invasive method of assessing disease status and also a novel therapeutic target for malignancy.
Collapse
|
45
|
Treatment of patients with acute deep vein thrombosis and/or pulmonary embolism: efficacy and safety of non-VKA oral anticoagulants in selected populations. Thromb Res 2014; 134:227-33. [PMID: 24875390 DOI: 10.1016/j.thromres.2014.05.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/02/2014] [Accepted: 05/07/2014] [Indexed: 12/12/2022]
Abstract
Venous thromboembolism (VTE), comprising deep vein thrombosis (DVT) and pulmonary embolism (PE), presents a large clinical burden. Prompt, effective and sustained anticoagulation is vital because of the risk of recurrent events, including life-threatening PE, and complications such as post-thrombotic syndrome and chronic thromboembolic pulmonary hypertension. Dual-drug standard therapy is effective; however, parenteral low molecular weight heparin, coupled with routine coagulation monitoring and dose adjustment of vitamin K antagonists (VKAs), presents challenges for patients and healthcare providers. Non-VKA oral anticoagulants provide a simplified option for VTE treatment. Phase III studies have investigated rivaroxaban and apixaban as single-drug approaches, and edoxaban and dabigatran in conjunction with initial heparin therapy. These agents demonstrated non-inferiority to standard therapy, and most showed significant reductions in major bleeding. However, clinical information is limited in patient subgroups, e.g. fragile patients or patients with renal impairment or cancer, who may be at higher risk of bleeding and/or VTE. A prespecified pooled analysis of the EINSTEIN DVT and EINSTEIN PE studies (8281 patients), undertaken to evaluate clinical outcomes with rivaroxaban versus standard therapy, confirmed the non-inferiority of rivaroxaban, with significant reductions in major bleeding and fewer intracranial and retroperitoneal bleeding events. Consistent efficacy and safety were observed with rivaroxaban, irrespective of fragility, cancer or clot severity. The introduction of the non-VKA oral anticoagulants and approval of rivaroxaban in the EU, US and Canada for the treatment and secondary prevention of DVT and PE offer the potential for improvements in effective care across a broad spectrum of patients with VTE.
Collapse
|
46
|
Turpin B, Miller W, Rosenfeldt L, Kombrinck K, Flick MJ, Steinbrecher KA, Harmel-Laws E, Mullins ES, Shaw M, Witte DP, Revenko A, Monia B, Palumbo JS. Thrombin drives tumorigenesis in colitis-associated colon cancer. Cancer Res 2014; 74:3020-3030. [PMID: 24710407 DOI: 10.1158/0008-5472.can-13-3276] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The established association between inflammatory bowel disease and colorectal cancer underscores the importance of inflammation in colon cancer development. On the basis of evidence that hemostatic proteases are powerful modifiers of both inflammatory pathologies and tumor biology, gene-targeted mice carrying low levels of prothrombin were used to directly test the hypothesis that prothrombin contributes to tumor development in colitis-associated colon cancer (CAC). Remarkably, imposing a modest 50% reduction in circulating prothrombin in fII+/- mice, a level that carries no significant bleeding risk, dramatically decreased adenoma formation following an azoxymethane/dextran sodium sulfate challenge. Similar results were obtained with pharmacologic inhibition of prothrombin expression or inhibition of thrombin proteolytic activity. Detailed longitudinal analyses showed that the role of thrombin in tumor development in CAC was temporally associated with the antecedent inflammatory colitis. However, direct studies of the antecedent colitis showed that mice carrying half-normal prothrombin levels were comparable to control mice in mucosal damage, inflammatory cell infiltration, and associated local cytokine levels. These results suggest that thrombin supports early events coupled to inflammation-mediated tumorigenesis in CAC that are distinct from overall inflammation-induced tissue damage and inflammatory cell trafficking. That prothrombin is linked to early events in CAC was strongly inferred by the observation that prothrombin deficiency dramatically reduced the formation of very early, precancerous aberrant crypt foci. Given the importance of inflammation in the development of colon cancer, these studies suggest that therapeutic interventions at the level of hemostatic factors may be an effective means to prevent and/or impede colitis-associated colon cancer progression.
Collapse
Affiliation(s)
- Brian Turpin
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| | - Whitney Miller
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| | - Leah Rosenfeldt
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| | - Keith Kombrinck
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| | - Matthew J Flick
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| | - Kris A Steinbrecher
- Divisions of Gastroenterology, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| | - Eleana Harmel-Laws
- Divisions of Gastroenterology, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| | - Eric S Mullins
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| | - Maureen Shaw
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| | - David P Witte
- Pathology, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| | - Alexey Revenko
- Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| | - Brett Monia
- Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| | - Joseph S Palumbo
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati OH, and ISIS Pharmaceuticals, Carlsbad CA
| |
Collapse
|
47
|
Säily VMJ, Pétas A, Joutsi-Korhonen L, Taari K, Lassila R, Rannikko AS. Dabigatran for thromboprophylaxis after robotic assisted laparoscopic prostatectomy: Retrospective analysis of safety profile and effect on blood coagulation. Scand J Urol 2013; 48:153-9. [DOI: 10.3109/21681805.2013.817482] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
48
|
Regulating surgical oncotaxis to improve the outcomes in cancer patients. Surg Today 2013; 44:804-11. [PMID: 23736890 DOI: 10.1007/s00595-013-0627-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 04/30/2013] [Indexed: 01/16/2023]
Abstract
Excessive surgical stress and postoperative complications cause a storm of perioperative cytokine release, which has been shown to enhance tumor metastasis in experimental models. We have named this phenomenon "surgical oncotaxis". The mechanisms that underpin this process are thought to be excessive corticosteroid secretion, coagulopathy in the peripheral vasculature, immune suppression and excessive production of reactive oxygen species. Nuclear factor-kappa B (NFkB) activation plays a key role in these mechanisms. Minimally invasive surgical techniques should be used, and postoperative complications should be avoided whenever possible to lessen the impact of surgical oncotaxis. Furthermore, there may be a role for a small preoperative dose of corticosteroid or the use of free radical scavengers in the perioperative period. Recently, there has been a great deal of interest in omega-3 fatty acid, because it regulates NFkB activation. The use of multimodal treatments that regulate surgical oncotaxis may be as important as chemotherapy for determining the outcome of patients with cancer undergoing surgery.
Collapse
|
49
|
Gómez-Outes A, Suárez-Gea ML, Lecumberri R, Terleira-Fernández AI, Vargas-Castrillón E, Rocha E. Potential role of new anticoagulants for prevention and treatment of venous thromboembolism in cancer patients. Vasc Health Risk Manag 2013; 9:207-28. [PMID: 23674896 PMCID: PMC3652561 DOI: 10.2147/vhrm.s35843] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Venous thromboembolism (VTE), encompassing deep vein thrombosis and pulmonary embolism, represents a major cause of morbidity and mortality in patients with cancer. Low molecular weight heparins are the preferred option for anticoagulation in cancer patients according to current clinical practice guidelines. Fondaparinux may also have a place in prevention of VTE in hospitalized cancer patients with additional risk factors and for initial treatment of VTE. Although low molecular weight heparins and fondaparinux are effective and safe, they require daily subcutaneous administration, which may be problematic for many patients, particularly if long-term treatment is needed. Studying anticoagulant therapy in oncology patients is challenging because this patient group has an increased risk of VTE and bleeding during anticoagulant therapy compared with the population without cancer. Risk factors for increased VTE and bleeding risk in these patients include concomitant treatments (surgery, chemotherapy, placement of central venous catheters, radiotherapy, hormonal therapy, angiogenesis inhibitors, antiplatelet drugs), supportive therapies (ie, steroids, blood transfusion, white blood cell growth factors, and erythropoiesis-stimulating agents), and tumor-related factors (local vessel damage and invasion, abnormalities in platelet function, and number). New anticoagulants in development for prophylaxis and treatment of VTE include parenteral compounds for once-daily administration (ie, semuloparin) or once-weekly dosing (ie, idraparinux and idrabiotaparinux), as well as orally active compounds (ie, dabigatran, rivaroxaban, apixaban, edoxaban, betrixaban). In the present review, we discuss the pharmacology of the new anticoagulants, the results of clinical trials testing these new compounds in VTE, with special emphasis on studies that included cancer patients, and their potential advantages and drawbacks compared with existing therapies.
Collapse
Affiliation(s)
- Antonio Gómez-Outes
- Division of Pharmacology and Clinical Evaluation, Medicines for Human Use, Spanish Agency for Medicines and Medical Devices, Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
50
|
van Ryn J, Goss A, Hauel N, Wienen W, Priepke H, Nar H, Clemens A. The discovery of dabigatran etexilate. Front Pharmacol 2013; 4:12. [PMID: 23408233 PMCID: PMC3569592 DOI: 10.3389/fphar.2013.00012] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 01/23/2013] [Indexed: 01/18/2023] Open
Abstract
Thromboembolic disease is a major cause of mortality and morbidity in the developed world and is caused by an excessive stimulation of coagulation. Thrombin is a key serine protease in the coagulation cascade and numerous efforts have been made to develop safe and effective orally active direct thrombin inhibitors (DTIs). Current anticoagulant therapy includes the use of indirect thrombin inhibitors (e.g., heparins, low-molecular-weight-heparins) and vitamin K antagonists such as warfarin. However there are several caveats in the clinical use of these agents including narrow therapeutic window, parenteral delivery, and food- and drug-drug interactions. Dabigatran is a synthetic, reversible DTI with high affinity and specificity for its target binding both free and clot-bound thrombin, and offers a favorable pharmacokinetic profile. Large randomized clinical trials have demonstrated that dabigatran provides comparable or superior thromboprophylaxis in multiple thromboembolic disease indications compared to standard of care. This minireview will highlight the discovery and development of dabigatran, the first in a class of new oral anticoagulant agents to be licensed worldwide for the prevention of thromboembolism in the setting of orthopedic surgery and stroke prevent in atrial fibrillation.
Collapse
Affiliation(s)
- Joanne van Ryn
- Department of CardioMetabolic Disease Research, Boehringer Ingelheim Pharma GmbH & Co. KGBiberach an der Riss, Baden-Württemberg, Germany
| | - Ashley Goss
- Department of CardioMetabolic Disease Research, Boehringer Ingelheim Pharmaceuticals Inc.Ridgefield, CT, USA
| | - Norbert Hauel
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KGBiberach an der Riss, Baden-Württemberg, Germany
| | - Wolfgang Wienen
- Department of Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KGBiberach an der Riss, Baden-Württemberg, Germany
| | - Henning Priepke
- Department of Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KGBiberach an der Riss, Baden-Württemberg, Germany
| | - Herbert Nar
- Department of Lead Identification and Optimization Support, Boehringer Ingelheim Pharma GmbH & Co. KGBiberach an der Riss, Baden-Württemberg, Germany
| | - Andreas Clemens
- Global Clinical Development and Medical Affairs, Boehringer Ingelheim Pharma GmbH & Co. KGIngelheim, Baden-Württemberg, Germany
| |
Collapse
|