1
|
Carrier M, Bertoletti L, Girard P, Laporte S, Mahé I. Preventative and curative treatment of venous thromboembolic disease in cancer patients. Presse Med 2024; 53:104242. [PMID: 39182756 DOI: 10.1016/j.lpm.2024.104242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/27/2024] Open
Abstract
Cancer-associated venous thromboembolism (CAT) is common in patients with cancer and associated with significant morbidity and mortality. The incidence of CAT continues to rise, complicating patient care and burdening healthcare systems. Patients with cancer experiencing VTE face poorer prognoses, making prevention and effective management imperative. This narrative review synthesizes evidence on thromboprophylaxis in ambulatory patients with cancer receiving systemic therapy and acute treatment strategies for CAT. Risk assessment models (e.g., Khorana score) aid in identifying high-risk patients who may benefit from thromboprophylaxis. Pharmacological thromboprophylaxis with low molecular weight heparins (LMWHs) and direct oral anticoagulants (DOACs) has been shown to reduce the risk of CAT without significantly increasing the risk of bleeding complications. However, implementation of risk-based strategies remains limited in clinical practice. For acute CAT management, LMWHs have been the standard of care, but DOACs are increasingly favored due to their convenience and efficacy. However, challenges persist, including bleeding risks and drug interactions. Emerging therapies targeting Factor XI inhibitors present promising alternatives, potentially addressing current limitations in anticoagulation management for CAT.
Collapse
Affiliation(s)
- Marc Carrier
- Department of Medicine, The Ottawa Hospital Research Institute at the University of Ottawa, Ottawa, Canada.
| | - Laurent Bertoletti
- Service de Médecine Vasculaire et Thérapeutique, CHU Saint-Etienne, Hôpital Nord, Saint-Etienne, France; INSERM, CIC-1408, CHU Saint-Etienne, Hôpital Nord, Saint-Etienne, France; SAINBIOSE U1059, Université Jean Monnet, INSERM, Saint-Etienne, France. https://twitter.com/https://twitter.com/LaurentBertole1
| | - Philippe Girard
- Département de Pneumologie, Institut Mutualiste Montsouris, Paris, France; F-CRIN INNOVTE Network, Saint-Etienne, France. https://twitter.com/https://twitter.com/Philipp29279657
| | - Sylvie Laporte
- SAINBIOSE U1059, Université Jean Monnet, INSERM, Saint-Etienne, France; Unité de Recherche Clinique, Innovation, Pharmacologie, CHU Saint-Etienne, Hôpital Nord, Saint-Etienne, France. https://twitter.com/https://twitter.com/SilvyLaporte
| | - Isabelle Mahé
- INSERM UMR_S1140, Innovations Thérapeutiques en Hémostase, Laboratoire de Chirurgie expérimentale, Fondation Alain Carpentier, Paris, France. https://twitter.com/https://twitter.com/Isabellemahe1
| |
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
|
Auer RC, Ott M, Karanicolas P, Brackstone MR, Ashamalla S, Weaver J, Tagalakis V, Boutros M, Stotland P, Marulanda AC, Moloo H, Jayaraman S, Patel S, Le Gal G, Spadafora S, MacLellan S, Trottier D, Jonker D, Asmis T, Mallick R, Pecarskie A, Ramsay T, Carrier M. Efficacy and safety of extended duration to perioperative thromboprophylaxis with low molecular weight heparin on disease-free survival after surgical resection of colorectal cancer (PERIOP-01): multicentre, open label, randomised controlled trial. BMJ 2022; 378:e071375. [PMID: 36100263 PMCID: PMC9468899 DOI: 10.1136/bmj-2022-071375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
OBJECTIVE To determine the efficacy and safety of extended duration perioperative thromboprophylaxis by low molecular weight heparin when assessing disease-free survival in patients undergoing resection for colorectal cancer. DESIGN Multicentre, open label, randomised controlled trial. SETTINGS 12 hospitals in Quebec and Ontario, Canada, between 25 October 2011 and 31 December 2020. PARTICIPANTS 614 adults (age ≥18 years) were eligible with pathologically confirmed invasive adenocarcinoma of the colon or rectum, no evidence of metastatic disease, a haemoglobin concentration of ≥8 g/dL, and were scheduled to undergo surgical resection. INTERVENTIONS Random assignment to extended duration thromboprophylaxis using daily subcutaneous tinzaparin at 4500 IU, beginning at decision to operate and continuing for 56 days postoperatively, compared with in-patient postoperative thromboprophylaxis only. MAIN OUTCOME MEASURES Primary outcome was disease-free survival at three years, defined as survival without locoregional recurrence, distant metastases, second primary (same cancer), second primary (other cancer), or death. Secondary outcomes included venous thromboembolism, postoperative major bleeding complications, and five year overall survival. Analyses were done in the intention-to-treat population. RESULTS The trial stopped recruitment prematurely after the interim analysis for futility. The primary outcome occurred in 235 (77%) of 307 patients in the extended duration group and in 243 (79%) of 307 patients in the in-hospital thromboprophylaxis group (hazard ratio 1.1, 95% confidence interval 0.90 to 1.33; P=0.4). Postoperative venous thromboembolism occurred in five patients (2%) in the extended duration group and in four patients (1%) in the in-hospital thromboprophylaxis group (P=0.8). Major surgery related bleeding in the first postoperative week was reported in one person (<1%) in the extended duration and in six people (2%) in the in-hospital thromboprophylaxis group (P=0.1). No difference was noted for overall survival at five years in 272 (89%) patients in the extended duration group and 280 (91%) patients in the in-hospital thromboprophylaxis group (hazard ratio 1.12; 95% confidence interval 0.72 to 1.76; P=0.1). CONCLUSIONS Extended duration to perioperative anticoagulation with tinzaparin did not improve disease-free survival or overall survival in patients with colorectal cancer undergoing surgical resection compared with in-patient postoperative thromboprophylaxis alone. The incidences of venous thromboembolism and postoperative major bleeding were low and similar between groups. TRIAL REGISTRATION ClinicalTrials.gov NCT01455831.
Collapse
Affiliation(s)
- Rebecca C Auer
- Department of Surgery, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Michael Ott
- Department of Surgery, London Health Sciences Centre, London, ON, Canada
| | - Paul Karanicolas
- Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | | | - Shady Ashamalla
- Department of Surgery, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Joel Weaver
- Department of Surgery, Queensway Carleton Hospital, Ottawa, ON, Canada
| | - Vicky Tagalakis
- Jewish General Hospital, Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada
| | - Marylise Boutros
- Jewish General Hospital, Lady Davis Institute for Medical Research, McGill University, Montreal, QC, Canada
| | - Peter Stotland
- Department of Surgery, North York General Hospital, North York, ON, Canada
| | | | - Husein Moloo
- Department of Surgery, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Shiva Jayaraman
- Department of Surgery, St Joseph's Health Centre, Toronto, ON, Canada
| | - Suni Patel
- Department of Surgery, Kingston Health Sciences Centre, Kingston, ON, Canada
| | - Grégoire Le Gal
- Department of Medicine, Institut du Savoir Montfort, Ottawa, ON, Canada
- Department of Medicine University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Silvana Spadafora
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste Marie, ON, Canada
| | - Steven MacLellan
- Department of Surgery, Humber River Hospital, Toronto, ON, Canada
| | - Daniel Trottier
- Department of Surgery, Montfort Hospital, Ottawa, ON, Canada
| | - Derek Jonker
- Department of Medicine University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Timothy Asmis
- Department of Medicine University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Ranjeeta Mallick
- Clinical Epidemiology Program, Method Centre, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Amanda Pecarskie
- Department of Medicine University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Tim Ramsay
- Department of Surgery, Montfort Hospital, Ottawa, ON, Canada
| | - Marc Carrier
- Department of Medicine University of Ottawa, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| |
Collapse
|
4
|
Amerali M, Politou M. Tinzaparin—a review of its molecular profile, pharmacology, special properties, and clinical uses. Eur J Clin Pharmacol 2022; 78:1555-1565. [PMID: 35871241 PMCID: PMC9308487 DOI: 10.1007/s00228-022-03365-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 07/16/2022] [Indexed: 11/05/2022]
Abstract
Purpose Low molecular weight heparins (LMWHs) are a group of heterogenous moieties, long used in the prevention and treatment of thrombosis. They derive from heparin and since they are prepared by different methods of depolymerization, they differ in pharmacokinetic properties and anticoagulant profiles, and thus are not clinically interchangeable. Methods In this review we provide an overview of tinzaparin's main characteristics and uses. Results Tinzaparin which is produced by the enzymatic depolymerization of unfractionated heparin (UFH) can be used for the treatment and prevention of deep venous thrombosis (DVT) and pulmonary embolism (PE); it has been also used in special populations such as elders, obese, pregnant women, and patients with renal impairment and/or cancer with favorable outcomes in both safety and efficacy, with a once daily dose regimen. Furthermore, LMWHs are extensively used in clinical practice for both thromboprophylaxis and thrombosis treatment of COVID-19 patients. Conclusion Tinzaparin features support the hypothesis for having a role in immunothrombosis treatment (i.e. in the context of cancer ,COVID-19), interfering not only with coagulation cascade but also exhibiting anti-inflammatory potency.
Collapse
|
5
|
Mantziou S, Markopoulos G, Thrasyvoulou S, Noutsopoulos D, Gkartziou F, Vartholomatos G, Tzavaras T. Tinzaparin inhibits VL30 retrotransposition induced by oxidative stress and/or VEGF in HC11 mouse progenitor mammary cells: Association between inhibition of cancer stem cell proliferation and mammosphere disaggregation. Oncol Rep 2021; 46:241. [PMID: 34558648 PMCID: PMC8485018 DOI: 10.3892/or.2021.8192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/15/2021] [Indexed: 02/07/2023] Open
Abstract
Tinzaparin is an anticoagulant and antiangiogenic drug with inhibitory properties against tumor growth. VEGF stimulates angiogenesis, while an association between reactive oxygen species (ROS) and angiogenesis is involved in tumor progression. The present study aimed to investigate the effect of tinzaparin on VL30 retrotransposition-positive mouse HC11 mammary stem-like epithelial cells, previously reported to be associated with induced mammosphere/cancer stem cell (CSC) generation and tumorigenesis. Under 24 h serum starvation, 15.2% nominal retrotransposition frequency was increased to 29%. Additionally, while treatment with 3–12 ng/ml VEGF further induced retrotransposition frequency in a dose-dependent manner (up to 40.3%), pre-incubation with tinzaparin (2 IU/ml) for 0.5–4 h reduced this frequency to 18.3% in a time-dependent manner, confirmed by analogous results in NIH3T3 fibroblasts. Treatment with 10–40 pg/ml glucose oxidase (GO) for 24 h induced HC11 cell retrotransposition in a dose-dependent manner (up to 82.5%), while a 3 h pre-incubation with tinzaparin (1 or 2 IU/ml) elicited a 13.5 or 25.5% reduction in retrotransposition, respectively. Regarding tumorigenic VL30 retrotransposition-positive HC11 cells, treatment with 2 IU/ml tinzaparin for 5 days reduced proliferation rate in a time-dependent manner (up to ~55%), and after 3 weeks, disaggregated soft agar-formed foci, as well as low-adherent mammospheres, producing single mesenchymal-like cells with a ~50% reduced retrotransposition. With respect to the VL30 retrotransposition mechanism: While 12 ng/ml VEGF increased the level of VL30 and endogenous reverse transcriptase (enRT) transcripts ~1.41- and ~1.16-fold, respectively, subsequent tinzaparin treatment reduced both endogenous/ROS- and VEGF-induced levels 1.15- and 0.40-fold (VL30) and 0.60- and 0.52-fold (enRT), respectively. To the best of our knowledge, these data demonstrate for the first time, the novel inhibition activity of tinzaparin against ROS- and VEGF-induced VL30 retrotransposition, and the proliferation and/or aggregation of mouse HC11 mammosphere/tumor-initiating CSCs, thus contributing to the inhibition of VL30 retrotransposition-induced primary tumor growth.
Collapse
Affiliation(s)
- Stefania Mantziou
- Laboratory of General Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Georgios Markopoulos
- Laboratory of General Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Soteroula Thrasyvoulou
- Laboratory of General Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Dimitrios Noutsopoulos
- Laboratory of General Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Foteini Gkartziou
- Laboratory of General Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Georgios Vartholomatos
- Molecular Biology Unit, Hematology Laboratory, University Hospital of Ioannina, 45110 Ioannina, Greece
| | - Theodore Tzavaras
- Laboratory of General Biology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
6
|
Sarantis P, Bokas A, Papadimitropoulou A, Koustas E, Theocharis S, Papakotoulas P, Schizas D, Papalampros A, Felekouras E, Papavassiliou AG, Karamouzis MV. Combinatorial Treatment of Tinzaparin and Chemotherapy Can Induce a Significant Antitumor Effect in Pancreatic Cancer. Int J Mol Sci 2021; 22:ijms22137053. [PMID: 34208987 PMCID: PMC8268558 DOI: 10.3390/ijms22137053] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/22/2021] [Accepted: 06/28/2021] [Indexed: 02/05/2023] Open
Abstract
Pancreatic Cancer (PC) is recognized as a highly thrombogenic tumor; thus, low-molecular-weight heparin (LMWH) such as tinzaparin is routinely used for PC patients. On the basis of combinatorial therapy approaches to treat highly malignant and refractory cancers such as PC, we hypothesized that tinzaparin can augment the effectiveness of traditional chemotherapeutic drugs and induce efficient antitumor activity. PANC-1 and MIAPaCa-2 were incubated alone or in combination with tinzaparin, nab-paclitaxel and gemcitabine. In vivo evaluation of these compounds was performed in a NOD/SCID mouse using a model injected with PANC-1. Tinzaparin enhances the anti-tumor effects of nab-paclitaxel and gemcitabine in mtKRAS PC cell lines via apoptosis in in vitro experiments. The triple combination power acts through the induction of apoptosis, reduction of the proliferative potential and angiogenesis; hence, contributing to a decrease in tumor volume observed in vivo. The triple regimen provided an extra 24.3% tumor reduction compared to the double combination (gemcitabine plus nab-paclitaxel). Combinatorial strategies can create novel therapeutic approaches for the treatment of patients with PC, achieving a better clinical outcome and prolonged survival. Further prospective randomized research is needed and the investigation of various concentrations of tinzaparin above 150 UI/Kg, would potentially provide a valuable synergistic effect to the conventional therapeutic compounds.
Collapse
Affiliation(s)
- Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Alexandros Bokas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Medical Oncology, 'Theageneio' Cancer Hospital, 54639 Thessaloniki, Greece
| | - Adriana Papadimitropoulou
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Evangelos Koustas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Pavlos Papakotoulas
- Department of Medical Oncology, 'Theageneio' Cancer Hospital, 54639 Thessaloniki, Greece
| | - Dimitrios Schizas
- First Department of Surgery, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Alexandros Papalampros
- First Department of Surgery, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Evangelos Felekouras
- First Department of Surgery, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Athanasios G Papavassiliou
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
7
|
Ripsman D, Fergusson DA, Montroy J, Auer RC, Huang JW, Dobriyal A, Wesch N, Carrier M, Lalu MM. A systematic review on the efficacy and safety of low molecular weight heparin as an anticancer therapeutic in preclinical animal models. Thromb Res 2020; 195:103-113. [PMID: 32683148 DOI: 10.1016/j.thromres.2020.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 01/30/2023]
Abstract
OBJECTIVE The therapeutic effects of low molecular weight heparins (LMWH) may extend past thrombosis prevention, with preclinical evidence demonstrating anti-metastatic properties. Clinical evidence on the topic, however, remains controversial. A systematic review of preclinical evidence may help elucidate reasons for this contradictory evidence. The objective of our systematic review is to assess the anti-metastatic properties of LMWHs in solid tumour animal models. METHODS MEDLINE, Embase, Web of Science and PubMed were searched from inception to May 12th, 2020. All articles were screened independently and in duplicate. Studies that compared LMWH to a placebo or no treatment arm in solid tumour animal models were included. The primary outcome was the burden of metastasis. Secondary outcomes included primary tumour growth and mortality. The risk of bias was assessed in duplicate using a modified Cochrane Risk of Bias tool. RESULTS Forty-two studies were included in the review. Administration of a LMWH was associated with a significant decrease in the burden of metastasis (SMD -2.18; 95% CI -2.66 to -1.70). Additionally, the administration of a LMWH was also associated with a significant reduction in primary tumour growth (SMD -1.95; 95% CI -2.56 to -1.34) and risk of death (RR 0.39; 95% CI 0.16-0.97). All included studies were deemed to be at an unclear risk of bias for at least one methodological criterion. CONCLUSIONS Our results demonstrate that LMWH can effectively reduce metastatic burden and reduce tumour growth in preclinical animal models of solid tumour malignancies. Reasons for the contradiction with clinical evidence require further exploration.
Collapse
Affiliation(s)
- David Ripsman
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada; Faculty of Medicine, University of Ottawa, 451 Smyth Rd #2044, Ottawa, ON K1H 8M5, Canada.
| | - Dean A Fergusson
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada; Faculty of Medicine, University of Ottawa, 451 Smyth Rd #2044, Ottawa, ON K1H 8M5, Canada.
| | - Joshua Montroy
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada.
| | - Rebecca C Auer
- Department of Surgery, The Ottawa Hospital, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada; Cancer Therapeutics Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada.
| | - Johnny W Huang
- Faculty of Medicine, University of Ottawa, 451 Smyth Rd #2044, Ottawa, ON K1H 8M5, Canada.
| | - Aditi Dobriyal
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada; Faculty of Medicine, University of Ottawa, 451 Smyth Rd #2044, Ottawa, ON K1H 8M5, Canada.
| | - Neil Wesch
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada
| | - Marc Carrier
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada; Faculty of Medicine, University of Ottawa, 451 Smyth Rd #2044, Ottawa, ON K1H 8M5, Canada.
| | - Manoj M Lalu
- Clinical Epidemiology Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada; Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Room B307, 1053 Carling Avenue, Mail Stop 249, Ottawa, ON K1Y 4E9, Canada; Regenerative Medicine Program, Ottawa Hospital Research Institute, 501 Smyth Box 511, Ottawa, ON K1H 8L6, Canada.
| |
Collapse
|
8
|
Beyond the boundaries of cardiology: Still untapped anticancer properties of the cardiovascular system-related drugs. Pharmacol Res 2019; 147:104326. [DOI: 10.1016/j.phrs.2019.104326] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 02/07/2023]
|
9
|
Randomised phase 3 study of adjuvant chemotherapy with or without nadroparin in patients with completely resected non-small-cell lung cancer: the NVALT-8 study. Br J Cancer 2019; 121:372-377. [PMID: 31337877 PMCID: PMC6738047 DOI: 10.1038/s41416-019-0533-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 06/13/2019] [Accepted: 07/04/2019] [Indexed: 11/17/2022] Open
Abstract
Background Retrospective studies suggest that low molecular weight heparin may delay the development of metastasis in patients with resected NSCLC. Methods Multicentre phase 3 study with patients with completely resected NSCLC who were randomised after surgery to receive chemotherapy with or without nadroparin. The main exclusion criteria were R1/2 and wedge/segmental resection. FDG-PET was required. The primary endpoint was recurrence-free survival (RFS). Results Among 235 registered patients, 202 were randomised (nadroparin: n = 100; control n = 102). Slow accrual enabled a decrease in the number of patients needed from 600 to 202, providing 80% power to compare RFS with 94 events (α = 0.05; 2-sided). There were no differences in bleeding events between the two groups. The median RFS was 65.2 months (95% CI, 36—NA) in the nadroparin arm and 37.7 months (95% CI, 22.7—NA) in the control arm (HR 0.77 (95% CI, 0.53–1.13, P = 0.19). FDG-PET SUVmax ≥10 predicted a greater likelihood of recurrence in the first year (HR 0.48, 95% CI 0.22–0.9, P = 0.05). Conclusions Adjuvant nadroparin did not improve RFS in patients with resected NSCLC. In this study, a high SUVmax predicted a greater likelihood of recurrence in the first year. Clinical trial registration Netherlands Trial registry: NTR1250/1217.
Collapse
|
10
|
Anticoagulants and cancer mortality in the Finnish randomized study of screening for prostate cancer. Cancer Causes Control 2019; 30:877-888. [PMID: 31209595 DOI: 10.1007/s10552-019-01195-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 06/12/2019] [Indexed: 12/21/2022]
Abstract
PURPOSE Anticoagulants may reduce mortality of cancer patients, though the evidence remains controversial. We studied the association between different anticoagulants and cancer death. METHODS All anticoagulant use during 1995-2015 was analyzed among 75,336 men in the Finnish Randomized Study of Screening for Prostate Cancer. Men with prevalent cancer were excluded. Multivariable Cox regression was performed to compare risk of death from any cancer and disease-specific death from 9 specific cancer types between (1) anticoagulant users overall and (2) warfarin users compared to anticoagulant non-users and (3) warfarin or (4) low-molecular-weight heparins (LMWH) compared to users of other anticoagulants. Medication use was analyzed as time-dependent variable to minimize immortal time bias. 1-, 2- and 3-year lag-time analyses were performed. RESULTS During a median follow-up of 17.2 years, a total of 27,233 men died of whom 8033 with cancer as the primary cause of death. In total, 32,628 men (43%) used anticoagulants. Any anticoagulant use was associated with an increased risk of cancer death (HR = 2.50, 95% CI 2.37-2.64) compared to non-users. Risk was similar independent of the amount, duration, or intensity of use. The risk increase was observed both among warfarin and LMWH users, although not as strong in warfarin users. Additionally, cancer-specific risks of death were similar to overall cancer mortality in all anticoagulant categories. CONCLUSION Our study does not support reduced cancer mortality among anticoagulant users. Future studies on drug use and cancer mortality should be adjusted for anticoagulants as they are associated with significantly higher risk of cancer death.
Collapse
|
11
|
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
|
12
|
Abstract
Cancer and cancer therapies might be a risk factor for developing Atrial Fibrillation (AF). It remains unclear if one is the cause or consequence of the other, or if they simply coexist. An unpredictable response to anticoagulation can be expected, as a result of the lack of information in oncology patients. The balance between thromboembolic and bleeding risks of AF in these patients is particularly challenging. Little is known about whether embolic and bleeding risk scores used for the general population can be applied in oncologic patients. Cardiology involvement in the management of these patients seems to be associated with favourable AF-related outcomes.
Collapse
Affiliation(s)
- Ana Pardo Sanz
- Cardiology Department, Ramón y Cajal Hospital Madrid, Spain
| | - José Luis Zamorano Gómez
- Cardiology Department, Ramón y Cajal Hospital Madrid, Spain.,Cardiology Department, University of Alcalá, University Hospital La Zarzuela Spain
| |
Collapse
|
13
|
Alexander M, Ball D, Solomon B, MacManus M, Manser R, Riedel B, Westerman D, Evans SM, Wolfe R, Burbury K. Dynamic Thromboembolic Risk Modelling to Target Appropriate Preventative Strategies for Patients with Non-Small Cell Lung Cancer. Cancers (Basel) 2019; 11:cancers11010050. [PMID: 30625975 PMCID: PMC6356389 DOI: 10.3390/cancers11010050] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 12/29/2018] [Indexed: 12/24/2022] Open
Abstract
Prevention of cancer-associated thromboembolism (TE) remains a significant clinical challenge and priority world-wide safety initiative. In this prospective non-small cell lung cancer (NSCLC) cohort, longitudinal TE risk profiling (clinical and biomarker) was undertaken to develop risk stratification models for targeted TE prevention. These were compared with published models from Khorana, CATS, PROTECHT, CONKO, and CATS/MICA. The NSCLC cohort of 129 patients, median follow-up 22.0 months (range 5.6—31.3), demonstrated a hypercoagulable profile in >75% patients and TE incidence of 19%. High TE risk patients were those receiving chemotherapy with baseline fibrinogen ≥ 4 g/L and d-dimer ≥ 0.5 mg/L; or baseline d-dimer ≥ 1.5 mg/L; or month 1 d-dimer ≥ 1.5 mg/L. The model predicted TE with 100% sensitivity and 34% specificity (c-index 0.67), with TE incidence 27% vs. 0% for high vs. low-risk. A comparison using the Khorana, PROTECHT, and CONKO methods were not discriminatory; TE incidence 17–25% vs. 14–19% for high vs. low-risk (c-index 0.51–0.59). Continuous d-dimer (CATS/MICA model) was also not predictive of TE. Independent of tumour stage, high TE risk was associated with cancer progression (HR 1.9, p = 0.01) and mortality (HR 2.2, p = 0.02). The model was tested for scalability in a prospective gastrointestinal cancer cohort with equipotency demonstrated; 80% sensitivity and 39% specificity. This proposed TE risk prediction model is simple, practical, potent and can be used in the clinic for real-time, decision-making for targeted thromboprophylaxis. Validation in a multicentre randomised interventional study is underway (ACTRN12618000811202).
Collapse
Affiliation(s)
- Marliese Alexander
- Department of Epidemiology and Preventive Medicine Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia.
- Pharmacy Department, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - David Ball
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
- Department of Radiation Oncology Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
| | - Benjamin Solomon
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
- Department of Medical Oncology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
| | - Michael MacManus
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
- Department of Radiation Oncology Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
| | - Renee Manser
- Department of Respiratory and Sleep Disorders Medicine, Royal Melbourne Hospital, Grattan Street, Parkville, VIC 3050, Australia.
| | - Bernhard Riedel
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
- Department of Anaesthesia, Perioperative and Pain Medicine, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
| | - David Westerman
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
- Department of Pathology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
- Department of Haematology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
| | - Sue M Evans
- Department of Epidemiology and Preventive Medicine Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia.
| | - Rory Wolfe
- Department of Epidemiology and Preventive Medicine Monash University, 99 Commercial Road, Melbourne, VIC 3004, Australia.
| | - Kate Burbury
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC 3010, Australia.
- Department of Pathology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
- Department of Haematology, Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia.
| |
Collapse
|
14
|
Abstract
Platelet-derived microvesicles (pMVs) are small, heterogeneous vesicles released from platelet membranes as a result of activation. These microvesicles possess a wide range of properties, including prothrombotic, proatherogenic, proinflammatory, immunomodulatory, and even anticoagulant activity. The elevated release of these microvesicles has been observed in various metabolic, inflammatory, thrombotic, and vascular diseases, including ischemic heart disease, stroke, hypertension, diabetes, and connective tissue disease. Modulation of both pMV generation and the expression of their surface molecules may have beneficial clinical implications and could become a novel therapeutic target. However, mechanisms by which pharmacological agents can modify pMV formation are elusive. The purpose of this review is to discuss the effects of drugs routinely used in primary and secondary prevention of vascular disease on the release of pMV and expression of their surface procoagulant and proinflammatory molecules.
Collapse
Affiliation(s)
- Justyna Rosińska
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland.
| | - Maria Łukasik
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland
| | - Wojciech Kozubski
- Department of Neurology, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland
| |
Collapse
|
15
|
García-Escobar I, Beato-Zambrano C, Muñoz Langa J, Brozos Vázquez E, Obispo Portero B, Gutiérrez-Abad D, Muñoz Martín AJ. Pleiotropic effects of heparins: does anticoagulant treatment increase survival in cancer patients? Clin Transl Oncol 2018; 20:1097-1108. [PMID: 29470777 DOI: 10.1007/s12094-018-1835-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 01/09/2018] [Indexed: 10/18/2022]
Abstract
The association between venous thromboembolism (VTE) and cancer has been recognized for more than 100 years. Numerous studies have been performed to investigate strategies to decrease VTE incidence and to establish whether treating VTE impacts cancer progression and overall survival. Accordingly, it is important to understand the role of the hemostatic system in tumorigenesis and progression, as there is abundant evidence associating it with cell survival and proliferation, tumor angiogenesis, invasion, and dissemination, and metastasis formation. In attempts to further the scientific evidence, several studies examine survival benefits in cancer patients treated with anticoagulant therapy, specifically treatment with vitamin K antagonists, unfractionated heparin, and low-molecular-weight heparin. Several studies and meta-analyses have been conducted with a special focus on brain tumors. However, no definitive conclusions have been obtained, and more well-designed clinical trials are needed.
Collapse
Affiliation(s)
- I García-Escobar
- Medical Oncology, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain.
| | - C Beato-Zambrano
- Medical Oncology GU and Breast Cancer Department, Hospital Universitario Virgen Macarena, Seville, Spain
| | - J Muñoz Langa
- Medical Oncology, Hospital Universitario La Fe, Valencia, Spain
| | - E Brozos Vázquez
- Medical Oncology, Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - B Obispo Portero
- Medical Oncology, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - D Gutiérrez-Abad
- Medical Oncology, Hospital Universitario de Fuenlabrada, Madrid, Spain
| | - A J Muñoz Martín
- Medical Oncology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | |
Collapse
|
16
|
Goertz L, Schneider SW, Desch A, Mayer FT, Koett J, Nowak K, Karampinis I, Bohlmann MK, Umansky V, Bauer AT. Heparins that block VEGF-A-mediated von Willebrand factor fiber generation are potent inhibitors of hematogenous but not lymphatic metastasis. Oncotarget 2018; 7:68527-68545. [PMID: 27602496 PMCID: PMC5356571 DOI: 10.18632/oncotarget.11832] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 08/21/2016] [Indexed: 12/31/2022] Open
Abstract
Von Willebrand factor (VWF) serves as a nidus for platelet aggregation and thrombosis. We hypothesize that VWF fibers contribute to the development of venous thromboembolism (VTE) and to metastasis formation. Here, we show that vascular and lymphatic endothelial cells (ECs) express VWF in vitro and release VWF fibers after activation by tumor cell supernatants. In contrast, an ex vivo analysis of primary mouse tumors revealed the presence of VWF fibers in the blood microvasculature but not in lymphatic vessels. Unlike the anticoagulant Fondaparinux, an inhibitor of thrombin generation, the low-molecular-weight heparin (LMWH) Tinzaparin inhibited VWF fiber formation and vessel occlusion in tumor vessels by blocking thrombin-induced EC activation and vascular endothelial growth factor-A (VEGF-A)-mediated VWF release. Intradermal tumor cell inoculation in VWF- and ADAMTS13-deficient mice did not alter lymph node metastases compared with wild type animals. Interestingly, multiple tumor-free distal organs exhibited hallmarks of malignancy-related VTE, including luminal VWF fibers, platelet-rich thrombi and vessel occlusions. Furthermore, ADAMTS13 deficiency, characterized by prolonged intraluminal VWF network lifetimes, resulted in a severely increased number of metastatic foci in an experimental model of hematogenous lung seeding. Treatment with Tinzaparin inhibited tumor-induced release of VWF multimers, impeded platelet aggregation and decreased lung metastasis. Thus, our data strongly suggest a critical role of luminal VWF fibers in determining the occurrence of thrombosis and cancer metastasis. Moreover, the findings highlight LMWHs as therapeutic strategy to treat thrombotic complications while executing anti-metastatic activities.
Collapse
Affiliation(s)
- Lukas Goertz
- Experimental Dermatology, Department of Dermatology, Venereology, and Allergy, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Stefan Werner Schneider
- Department of Dermatology and Venereology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Desch
- Experimental Dermatology, Department of Dermatology, Venereology, and Allergy, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Frank Thomas Mayer
- Experimental Dermatology, Department of Dermatology, Venereology, and Allergy, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Julian Koett
- Experimental Dermatology, Department of Dermatology, Venereology, and Allergy, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Kai Nowak
- Department of Surgery, Mannheim University Medical Center, Heidelberg University, Mannheim, Germany
| | - Ioannis Karampinis
- Department of Surgery, Mannheim University Medical Center, Heidelberg University, Mannheim, Germany
| | - Michael K Bohlmann
- Department of Obstetrics and Gynaecology, Mannheim University Medical Center, Heidelberg University, Mannheim, Germany
| | - Viktor Umansky
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Alexander Thomas Bauer
- Experimental Dermatology, Department of Dermatology, Venereology, and Allergy, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
17
|
Ping Z, Soni A, Williams LA, Pham HP, Basu MK, Zheng XL. Mutations in Coagulation Factor VIII Are Associated with More Favorable Outcome in Patients with Cutaneous Melanoma. TH OPEN 2017; 1:e113-e121. [PMID: 29152610 PMCID: PMC5690574 DOI: 10.1055/s-0037-1607337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Coagulation factor VIII (FVIII), von Willebrand factor (VWF), and ADAMTS13 (a disintegrin and metalloprotease with thrombospondin type 1 repeats 13) play an important role in the regulation of normal hemostasis. However, little is known about their roles in patients with malignancy, particularly with cutaneous melanoma. Whole genome sequencing data are available for 25,719 cases in 126 cancer genomic studies for analysis. All sequencing data and corresponding pathology findings were obtained from The Cancer Genome Atlas. The cBioPortal bioinformatics tools were used for the data analysis. Our results demonstrated that mutations in genes encoding
FVIII
,
VWF
, and
ADAMTS13
were reported in 92 of 126 cancer genomic studies, and high mutation rates in these three genes were observed in patients with cutaneous melanoma from three independent studies. Moreover, high mutation rates in
FVIII
,
VWF
, and
ADAMTS13
were also found in patients with diffuse large B cell lymphoma (22.9%), lung small cell carcinoma (20.7%), and colon adenocarcinoma (19.4%). Among 366 melanoma cases from TCGA provisional, the somatic mutation rates of
FVIII
,
VWF
, and
ADAMTS13
in tumor cells were 15, 14, and 5%, respectively. There was a strong tendency for coexisting mutations of
FVIII
,
VWF
, and
ADAMTS13
. Kaplan–Meier survival analysis demonstrated that melanoma patients with
FVIII
mutations had a more favorable overall survival rate than those without
FVIII
mutations (
p
= 0.02). These findings suggest, for the first time, that the
FVIII
mutation burden may have a prognostic value for patients with cutaneous melanoma. Further studies are warranted to delineate the molecular mechanisms underlying the favorable prognosis associated with
FVIII
mutations.
Collapse
Affiliation(s)
- Zheng Ping
- Divisions of Laboratory Medicine, The University of Alabama at Birmingham, AL 35249
| | - Abha Soni
- Divisions of Laboratory Medicine, The University of Alabama at Birmingham, AL 35249
| | - Lance A Williams
- Divisions of Laboratory Medicine, The University of Alabama at Birmingham, AL 35249
| | - Huy P Pham
- Divisions of Laboratory Medicine, The University of Alabama at Birmingham, AL 35249
| | - Malay K Basu
- Division of Informatics, Department of Pathology, The University of Alabama at Birmingham, AL 35249
| | - X Long Zheng
- Divisions of Laboratory Medicine, The University of Alabama at Birmingham, AL 35249.,Division of Informatics, Department of Pathology, The University of Alabama at Birmingham, AL 35249
| |
Collapse
|
18
|
Abstract
Current guidelines recommend low-molecular-weight heparin treatment in patients with cancer with established venous thromboembolism (VTE). The aim of this article was to study the pharmacological properties and effectiveness of tinzaparin in patients with cancer as well as its potential anticancer properties. A search of PubMed and ScienceDirect databases up to March 2016 was carried out to identify published studies that detect the properties and use of tinzaparin in oncology. Protamine sulfate partially (60% to 65%) neutralized tinzaparin’s anti-Xa activity. No dose adjustment of tinzaparin is needed even in patients with severe renal impairment and Creatinine Clearance ≥20 mL/min. Tinzaparin demonstrated a statistically significant decline in VTE recurrence at 1 year post the index thromboembolic event. A statistically significant reduction in minor bleeding rates was also described, whereas major bleeding events did not decrease in patients with cancer treated with tinzaparin versus those who received vitamin K antagonists. Tinzaparin treatment in patients suffering from deep vein thrombosis reduced the incidence of postthrombotic syndrome and venous ulcers. Tinzaparin’s ability to prevent both metastatic dissemination of cancer cells and tumor angiogenesis has been delineated in preclinical research. Current data show that tinzaparin is safe and efficacious either for short-term or for long-term treatment of VTE in patients with cancer. Clinical trials are needed in order to examine the utility of tinzaparin in primary prevention of VTE and validate its potential anticancer advantages exhibited in preclinical research.
Collapse
Affiliation(s)
- Evangelos P Dimakakos
- 1 Oncology Unit GPP, Sotiria General Hospital Athens School of Medicine, Athens, Greece
| | - Ioannis Vathiotis
- 1 Oncology Unit GPP, Sotiria General Hospital Athens School of Medicine, Athens, Greece
| | - Konstantinos Syrigos
- 1 Oncology Unit GPP, Sotiria General Hospital Athens School of Medicine, Athens, Greece
| |
Collapse
|
19
|
Mitsis M, Koliou P, Bali C, Ntounousi E, Tatsis V, Nousias V, Lianos GD, Vartholomatos G, Nastos D. In Surgical Colon Cancer Patients Extended-Duration Thromboprophylaxis (30 days) with the Highest Dose of Tinzaparin (4,500 IU s.c./q.d.) Normalizes the Postoperative VEGF Levels. J Cancer 2017; 8:2899-2906. [PMID: 28928880 PMCID: PMC5604440 DOI: 10.7150/jca.20107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 06/30/2017] [Indexed: 01/05/2023] Open
Abstract
Background/Purpose: In colon cancer (CC) patients preoperative (pre-op) levels of VEGF-A165 (VEGF) is a strong predictor for disease recurrence. Elevated postoperative (post-op) VEGF levels could have undesirable effects by enhancing tumor growth and metastasis formation. It has been suggested that thromboprophylaxis with a Low Molecular Weight Heparin (LMWH) in surgical cancer patients, further to thromboembolic protection, may exert some anti-neoplastic properties, as well. The aim of our study was to assess the potential impact of the LMWH Tinzaparin (Innohep® - Leo Pharma, Copenhagen, Denmark), given at different doses and for different perioperative (peri-op) periods, upon the post-op variability of serum VEGF levels in surgical CC patients. Methods: A total of 54 consecutive CC patients who underwent a curative resection were randomized in four groups according to their peri-op thromboprophylaxis scheme, which was based on administrating Tinzaparin in different doses and at different periods, as follows: group I: 3,500 IU for 10 days, group II: 3,500 IU for 30 days, group III: 4,500 IU for 10 days and group IV: 4,500 IU for 30 days. Serum VEGF concentrations were evaluated on the pre-op day (Day 0) and on the 10th and 30th post-op days (Day 10 and Day 30, respectively). For statistical analyses the mixed design ANOVA was used. P < 0.05 was considered significant. Results: On Day 0, VEGF didn't differ between groups I, II, III and IV (p>0.05, for every comparison). On Day 10, VEGF was increased in all groups. Between Day 10 and Day 30, VEGF remained stable in groups I (p=0.031) and II (p=1.000) and increased significantly in group III (p=0.005). On the contrary, VEGF decreased significantly in group IV (p<0.001). The most remarkable finding was observed when we compared VEGF between Day 0 and Day 30: while in groups I, II and III, VEGF remained significantly higher compared to Day 0 (p<0.001, p=0.041 and p<0.001, respectively), on the contrary, in group IV (extended-duration with the highest dose of 4,500 IU of tinzaparin) it was comparable to Day 0 (p=1.000). Conclusions: In surgical CC patients only the recommended thromboprophylaxis scheme with the highest prophylactic dose of Tinzaparin (4,500 IU) for extended-duration (30 days) normalizes VEGF levels at the end of the first post-op month by reducing them to the pre-op levels.
Collapse
Affiliation(s)
- Michail Mitsis
- Department of Surgery, University Hospital of Ioannina, Greece
| | | | - Christina Bali
- Department of Surgery, University Hospital of Ioannina, Greece
| | | | | | | | | | - Georgios Vartholomatos
- Unit of Molecular Biology of the Haematology Laboratory, University Hospital of Ioannina, Greece
| | | |
Collapse
|
20
|
Kevane B, Egan K, Allen S, Maguire P, Neary E, Lennon Á, Ní Áinle F. Endothelial barrier protective properties of low molecular weight heparin: A novel potential tool in the prevention of cancer metastasis? Res Pract Thromb Haemost 2017; 1:23-32. [PMID: 30046671 PMCID: PMC5974908 DOI: 10.1002/rth2.12011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/08/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND One of the key events in the progression of cancer metastasis is the trans-endothelial migration of circulating tumor cells. Moreover, inhibition of tumor-induced vascular permeability has been shown to inhibit metastasis in vivo. Low molecular weight heparin (LMWH) appears to confer a survival benefit in cancer but the underlying mechanisms are poorly understood. OBJECTIVE To characterise LMWH-mediated endothelial barrier protection and to explore strategies to limit the LMWH-associated haemorrhagic risk in this setting. METHODS Endothelial barrier function was assessed using in vitro assays of endothelial permeability and tumor cell trans-endothelial migration. Thrombin-mediated activation of PAR-1 signalling was assessed by flow cytometry and western blotting. LMWH anticoagulant activity was assessed by calibrated automated thrombography and plasma anti-factor Xa activity assay. RESULTS LMWH tinzaparin enhanced endothelial barrier function and reduced tumor cell trans-endothelial migration (73.9±5.7% of baseline; P<.05). Tinzaparin-mediated attenuation of thrombin-induced permeability was not mediated through an inhibition of thrombin proteolytic activity. In addition, fractions of LMWH with diminished anticoagulant activity retained endothelial barrier protective properties and a marked synergistic effect on barrier function was observed using combinations of sub-anticoagulant concentrations of tinzaparin with simvastatin (which exhibits endothelial barrier protective properties in vitro), with almost complete protection against agonist-induced endothelial barrier permeability achieved (7.9±0.2% of baseline; P<.05). CONCLUSION Collectively, these results suggest that LMWH supports endothelial barrier function in a manner which does not appear to be dependent on its anticoagulant activity. If replicated in vivo, these findings could represent a novel therapeutic approach to the suppression of metastasis.
Collapse
Affiliation(s)
- Barry Kevane
- School of Medicine & Medical ScienceUniversity College Dublin (UCD)DublinIreland
- SPHERE Research GroupUCD Conway InstituteDublinIreland
- Department of HaematologyRotunda HospitalDublinIreland
- Department of HaematologyMater Misericordiae University HospitalDublinIreland
| | - Karl Egan
- School of Medicine & Medical ScienceUniversity College Dublin (UCD)DublinIreland
- SPHERE Research GroupUCD Conway InstituteDublinIreland
| | - Seamus Allen
- School of Medicine & Medical ScienceUniversity College Dublin (UCD)DublinIreland
- SPHERE Research GroupUCD Conway InstituteDublinIreland
| | - Patricia Maguire
- SPHERE Research GroupUCD Conway InstituteDublinIreland
- Department of Biomolecular and Biomedical SciencesUCDDublinIreland
| | - Elaine Neary
- Department of NeonatologyRotunda HospitalDublinIreland
| | - Áine Lennon
- Department of HaematologyMater Misericordiae University HospitalDublinIreland
| | - Fionnuala Ní Áinle
- School of Medicine & Medical ScienceUniversity College Dublin (UCD)DublinIreland
- SPHERE Research GroupUCD Conway InstituteDublinIreland
- Department of HaematologyRotunda HospitalDublinIreland
- Department of HaematologyMater Misericordiae University HospitalDublinIreland
| |
Collapse
|
21
|
Sylman JL, Mitrugno A, Tormoen GW, Wagner TH, Mallick P, McCarty OJT. Platelet count as a predictor of metastasis and venous thromboembolism in patients with cancer. CONVERGENT SCIENCE PHYSICAL ONCOLOGY 2017; 3. [PMID: 29081989 DOI: 10.1088/2057-1739/aa6c05] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Platelets are anucleate cells in the blood at concentrations of 150,000 to 400,000 cells/µL and play a key role in hemostasis. Several studies have suggested that platelets contribute to cancer progression and cancer-associated thrombosis. In this review, we provide an overview of the biochemical and biophysical mechanisms by which platelets interact with cancer cells and review the evidence supporting a role for platelet-enhanced metastasis of cancer, and venous thromboembolism (VTE) in patients with cancer. We discuss the potential for and limitations of platelet counts to discriminate cancer disease burden and prognosis. Lastly, we consider more advanced diagnostic approaches to improve studies on the interaction between the hemostatic system and cancer cells.
Collapse
Affiliation(s)
- Joanna L Sylman
- Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR.,VA Palo Alto Health Care System, Palo Alto, CA.,Canary Center at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Annachiara Mitrugno
- Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR
| | - Garth W Tormoen
- Department of Radiation Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR
| | - Todd H Wagner
- VA Palo Alto Health Care System, Palo Alto, CA.,Department of Surgery, Stanford University School of Medicine, Stanford, CA
| | - Parag Mallick
- Canary Center at Stanford, Department of Radiology, Stanford University School of Medicine, Stanford, CA
| | - Owen J T McCarty
- Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR
| |
Collapse
|
22
|
Desch A, Gebhardt C, Utikal J, Schneider SW. D-dimers in malignant melanoma: Association with prognosis and dynamic variation in disease progress. Int J Cancer 2016; 140:914-921. [PMID: 27813063 DOI: 10.1002/ijc.30498] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Revised: 10/07/2016] [Accepted: 10/24/2016] [Indexed: 02/05/2023]
Abstract
Malignant cells elicit a chronic hemostatic activation in disease progress. This procoagulant activity does not only bear a risk for thromboembolism but also facilitates tumor growth and dissemination. An elevated plasma D-dimer level indicates an activated coagulation and fibrinolysis. In this study, the association of D-dimer levels with clinicopathological parameters and patients outcome in melanoma was investigated analyzing in total 533 melanoma patients retrospectively. Using the cut-off point of 0.6 mg/L D-dimer 145 of the total 533 patients (27.2%) were identified with elevated plasma D-dimer levels. This increased D-dimer level positively correlated with tumor thickness (p = 0.0003), lymph node invasion (p = 0.0004) and metastatic state (p <0.0001). To assess the association of D-dimer levels with progression-free survival (PFS) and overall survival (OS), long-rank test and the Cox proportional hazard model was performed. Univariate analyses revealed that elevated D-dimer levels were significantly associated with decreased PFS (HR:2.89, 95% CI (2.07-7.56), p < 0.0001) and OS (HR:2.22, 95% CI (1.06-4.57), p = 0.035). Moreover, multivariate analyses identified elevated D-dimer levels being associated with poor disease outcome (PFS:HR:2.47, 95% CI (1.23-4.98), p = 0.012; OS:HR:2.01, 95% CI (0.09-4.45), p = 0.087). Additionally, D-dimer levels were significantly increased in terminal stage patients when comparing plasma levels 0-8 versus 24-48 weeks before death (p = 0.0003). In summary, this study presents multiple evidence that elevated D-dimer levels in melanoma patients associate with poor prognosis and therefore plasma levels of D-dimers could reveal a more aggressive phenotype of melanoma and may guide the management of anti-melanoma treatment including the concept of an anti-coagulatory therapy in tumor patients.
Collapse
Affiliation(s)
- Anna Desch
- Experimental Dermatology, Department of Dermatology, Venereology, and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Christoffer Gebhardt
- Department of Dermatology, Venereology, and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jochen Utikal
- Department of Dermatology, Venereology, and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany.,Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan W Schneider
- Department of Dermatology and Venerology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
23
|
Chang YW, Hsieh PW, Chang YT, Lu MH, Huang TF, Chong KY, Liao HR, Cheng JC, Tseng CP. Identification of a novel platelet antagonist that binds to CLEC-2 and suppresses podoplanin-induced platelet aggregation and cancer metastasis. Oncotarget 2016; 6:42733-48. [PMID: 26528756 PMCID: PMC4767466 DOI: 10.18632/oncotarget.5811] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 10/17/2015] [Indexed: 12/13/2022] Open
Abstract
Podoplanin (PDPN) enhances tumor metastases by eliciting tumor cell-induced platelet aggregation (TCIPA) through activation of platelet C-type lectin-like receptor 2 (CLEC-2). A novel and non-cytotoxic 5-nitrobenzoate compound 2CP was synthesized that specifically inhibited the PDPN/CLEC-2 interaction and TCIPA with no effect on platelet aggregation stimulated by other platelet agonists. 2CP possessed anti-cancer metastatic activity in vivo and augmented the therapeutic efficacy of cisplatin in the experimental animal model without causing a bleeding risk. Analysis of the molecular action of 2CP further revealed that Akt1/PDK1 and PKCμ were two alternative CLEC-2 signaling pathways mediating PDPN-induced platelet activation. 2CP directly bound to CLEC-2 and, by competing with the same binding pocket of PDPN in CLEC-2, inhibited PDPN-mediated platelet activation. This study provides evidence that 2CP is the first defined platelet antagonist with CLEC-2 binding activity. The augmentation in the therapeutic efficacy of cisplatin by 2CP suggests that a combination of a chemotherapeutic agent and a drug with anti-TCIPA activity such as 2CP may prove clinically effective.
Collapse
Affiliation(s)
- Yao-Wen Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC)
| | - Pei-Wen Hsieh
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC).,Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC)
| | - Yu-Tsui Chang
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC)
| | - Meng-Hong Lu
- Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC)
| | - Tur-Fu Huang
- Graduate Institute of Pharmacology, National Taiwan University College of Medicine, Taipei 104, Taiwan, Republic of China (ROC)
| | - Kowit-Yu Chong
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC).,Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC).,Molecular Medicine Research Center, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC)
| | - Hsiang-Ruei Liao
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC).,Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC)
| | - Ju-Chien Cheng
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 404, Taiwan, Republic of China (ROC)
| | - Ching-Ping Tseng
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC).,Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC).,Molecular Medicine Research Center, Chang Gung University, Taoyuan 333, Taiwan, Republic of China (ROC).,Department of Laboratory Medicine, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan, Republic of China (ROC)
| |
Collapse
|
24
|
Gamperl H, Plattfaut C, Freund A, Quecke T, Theophil F, Gieseler F. Extracellular vesicles from malignant effusions induce tumor cell migration: inhibitory effect of LMWH tinzaparin. Cell Biol Int 2016; 40:1050-61. [PMID: 27435911 DOI: 10.1002/cbin.10645] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 07/17/2016] [Indexed: 01/01/2023]
Abstract
Elevated levels of extracellular vesicles (EVs) have been correlated with inflammatory diseases as well as progressive and metastatic cancer. By presenting tissue factor (TF) on their membrane surface, cellular microparticles (MPs) activate both the coagulation system and cell-signaling pathways such as the PAR/ERK pathway. We have shown before that malignant effusions are a rich source of tumor cell-derived EVs. Here, we used EVs from malignant effusions from three different patients after serial low-speed centrifugation steps as recommended by the ISTH (lsEV). Significant migration of human pancreatic carcinoma cells could be induced by lsEVs and was effectively inhibited by pre-incubation with tinzaparin, a low-molecular-weight heparin. Tinzaparin induced tissue factor pathway inhibitor (TFPI) release from tumor cells, and recombinant TFPI inhibited EV-induced tumor cell migration. EVs also induced ERK phosphorylation, whereas inhibitors of PAR2 and ERK suppressed EV-induced tumor cell migration. LsEVs have been characterized by high-resolution flow cytometry and, after elimination of smaller vesicles including exosomes, by further high-speed centrifugation (hsEV). The remaining population consisting primarily of MPs is indeed the main migration-inducing population with tenase activity. Compared to other LMWHs, tinzaparin is suggested to have high potency to induce TFPI release from epithelial cells. The migration-inhibitory effect of TFPI and the interruption of tumor cell migration by inhibitors of PAR2 and ERK suggest that lsEVs induce tumor cell migration by activating the PAR2 signaling pathway. Tinzaparin might inhibit this process at least partly by inducing the release of TFPI from tumor cells, which blocks PAR-activating TF complexes. The clinical relevance of the results is discussed.
Collapse
Affiliation(s)
- Hans Gamperl
- Experimental Oncology, Ethics and Palliative Care in Oncology, University Hospital and Medical School, UKSH, Luebeck, Germany
| | - Corinna Plattfaut
- Experimental Oncology, Ethics and Palliative Care in Oncology, University Hospital and Medical School, UKSH, Luebeck, Germany
| | - Annika Freund
- Experimental Oncology, Ethics and Palliative Care in Oncology, University Hospital and Medical School, UKSH, Luebeck, Germany
| | - Tabea Quecke
- Experimental Oncology, Ethics and Palliative Care in Oncology, University Hospital and Medical School, UKSH, Luebeck, Germany
| | - Friederike Theophil
- Experimental Oncology, Ethics and Palliative Care in Oncology, University Hospital and Medical School, UKSH, Luebeck, Germany
| | - Frank Gieseler
- Experimental Oncology, Ethics and Palliative Care in Oncology, University Hospital and Medical School, UKSH, Luebeck, Germany.
| |
Collapse
|
25
|
Zhang B, Jiang T, She X, Shen S, Wang S, Deng J, Shi W, Mei H, Hu Y, Pang Z, Jiang X. Fibrin degradation by rtPA enhances the delivery of nanotherapeutics to A549 tumors in nude mice. Biomaterials 2016; 96:63-71. [PMID: 27149664 DOI: 10.1016/j.biomaterials.2016.04.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 11/26/2022]
Abstract
Effective drug delivery to a tumor depends on favorable blood perfusion within the tumor. As an important component of tumor extracellular matrix, fibrin is abundant near tumor vessels. Inspired by the distinct distribution pattern and vessel-dependent production of fibrin, we hypothesized that fibrin depletion in tumors decompresses tumor vessels to improve tumor blood perfusion and accordingly enhance drug delivery to tumors rich in vessels. In the present study, we attempted to employ a clinically used thrombolytic drug, recombinant tissue plasminogen activator (rtPA), to modulate fibrin deposition in tumors. We then combined this drug with a nanoparticle drug delivery system for tumor therapy. RtPA treatment (25 mg/kg/d i. p. administration for two weeks) successfully depleted fibrin deposition and enhanced blood perfusion within A549 tumor xenografts. Furthermore, rtPA treatment also improved the in vivo delivery of 115-nm nanoparticles to tumor tissues. Finally, rtPA combined with therapeutic agent-loaded nanoparticles resulted in the most effective shrinkage of A549 tumor xenografts compared with the control groups. Overall, the present study provides a new strategy to enhance the delivery of nanotherapeutics to tumors rich in vessels.
Collapse
Affiliation(s)
- Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China
| | - Ting Jiang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China
| | - Xiaojian She
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Shun Shen
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Sheng Wang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China
| | - Jun Deng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China
| | - Wei Shi
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, PR China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei, 430022, PR China; Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, PR China.
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China.
| | - Xinguo Jiang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, PR China
| |
Collapse
|
26
|
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
|
27
|
Abstract
A close relationship between cancer and thrombosis does exist, documented by the fact that an overall 7-fold increased risk of venous thromboembolism (VTE) has been reported in patients with malignancy compared to non-malignancy. The potential impact of antithrombotic agents in cancer-associated VTE has long been recognized, and, in particular, several clinical trials in the last 20 years have reported the safety and efficacy of low-molecular-weight heparins (LMWHs) for treatment and prophylaxis of VTE in patients with various types of cancer. More recently, a number of preclinical and clinical studies have suggested that LMWHs may improve survival in cancer patients with mechanisms that are different from its antithrombotic effect but are linked to the ability of influencing directly the tumor biology. This paper reviews the evidence around the potential survival benefits of LMWHs by analyzing the suggested mechanisms and the available clinical data.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital , Mantova , Italy
| | | |
Collapse
|
28
|
Chen YP, Chen C, Mai ZY, Gao J, Shen LJ, Zhao BC, Chen MK, Chen G, Yan F, Huang TY, Xia YF. Pretreatment platelet count as a predictor for survival and distant metastasis in nasopharyngeal carcinoma patients. Oncol Lett 2015; 9:1458-1466. [PMID: 25663931 PMCID: PMC4314978 DOI: 10.3892/ol.2015.2872] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 12/09/2014] [Indexed: 11/12/2022] Open
Abstract
The aim of the present study was to investigate the prognostic value of different pretreatment platelet (PLT) counts on the treatment outcome in nasopharyngeal carcinoma (NPC) patients receiving concurrent chemoradiotherapy (CCRT) or radiotherapy (RT) alone. A total of 1,501 NPC patients, including 412 receiving CCRT and 1,089 receiving RT, were enrolled in the present study. The PLT count cut-off points for the CCRT and RT groups were 150 and 300×109/l, respectively, and the PLT counts were categorized it into three groups: Low (PLT≤150×109/l), moderate (150×109/l<PLT≤300×109/l) and high (PLT>300×109/l). To identify independent predictors of overall survival (OS), the Cox proportional hazards model was used to determine local-regional recurrence-free survival (LRFS) and distant metastasis-free survival (DMFS) rates in the CCRT and RT patients. Furthermore, univariate and multivariate analysis indicated that compared with a moderate PLT count, a low PLT count was an independent unfavorable prognostic factor for OS rate in CCRT patients [hazard ratio (HR), 2.024; 95% confidence interval (CI), 1.165–3.516], and a high PLT count was an independent unfavorable prognostic factor for OS and DMFS rates in CCRT (OS: HR, 1.742; 95% CI, 1.090–2.786; DFMS: HR, 2.110; 95%CI, 1.084–4.108) and RT (OS: HR, 1.740; 95%CI, 1.283–2.362; DMFS: HR, 2.819; 95% CI, 1.766–4.497) patients. Compared with a low PLT count, a high PLT count was significantly and independently associated with a poor DMFS rate in the RT patients (P=0.025; HR, 2.454; 95% CI, 1.121–5.372). Therefore, the present study indicates that low and high PLT counts may be useful indicators of survival and distant metastasis in NPC patients who have undergone radiation treatment.
Collapse
Affiliation(s)
- Yu-Pei Chen
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China ; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Chen Chen
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China ; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Zhuo-Yao Mai
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China ; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Jin Gao
- Department of Radiation Oncology, Anhui Provincial Hospital, Hefei, Anhui, P.R. China
| | - Lu-Jun Shen
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China ; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Bing-Cheng Zhao
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China ; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Meng-Kun Chen
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China ; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Gang Chen
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China ; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Fang Yan
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China ; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Tong-Yi Huang
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China ; Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| | - Yun-Fei Xia
- State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China ; Department of Radiation Oncology, Cancer Center, Sun Yat-Sen University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
29
|
Sanford D, Naidu A, Alizadeh N, Lazo-Langner A. The effect of low molecular weight heparin on survival in cancer patients: an updated systematic review and meta-analysis of randomized trials. J Thromb Haemost 2014; 12:1076-85. [PMID: 24796727 DOI: 10.1111/jth.12595] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Indexed: 12/01/2022]
Abstract
BACKGROUND Tumors may exploit the coagulation system to enhance the survival and dissemination of cancer cells. Some studies have suggested that heparin and low molecular weight heparin (LMWH) have antitumor effects. We reported a previous meta-analysis that suggested a modest improvement in overall survival with the use of LMWH in patients with cancer. Herein, we present the results of an updated systematic review and meta-analysis. OBJECTIVE To evaluate the effect of LMWH as compared with placebo or no anticoagulant on the overall survival in patients with solid cancers. METHODS We conducted a systematic review and meta-analysis of randomized trials evaluating the use of LMWH vs. placebo or no anticoagulant in cancer patients without venous thrombosis. A meta-analysis was conducted with a random-effects model, and data were analyzed by the use of odds ratios (ORs) and relative risks (RRs) calculated for 1-year overall mortality. RESULTS We identified 724 potentially relevant studies, nine of which met our inclusion criteria, and reported data on 1-year overall mortality. Studies were heterogeneous regarding types of cancer and interventions, and included 5987 patients, 98.4% of whom had advanced-stage disease (III and IV). There was no discernible effect on mortality with the use of LMWH (pooled OR 0.87, 95% CI 0.70-1.08; RR 0.94, 95% CI 0.86-1.04). CONCLUSIONS In contrast to the previous study, these results did not show a survival benefit in cancer patients receiving LMWH. The effect of LMWH on overall survival in patients with limited-stage disease still is unknown.
Collapse
Affiliation(s)
- D Sanford
- Division of Hematology, Department of Medicine, London Health Sciences Centre, Western University, London, Ontario, Canada
| | | | | | | |
Collapse
|
30
|
Nikitovic D, Mytilinaiou M, Berdiaki A, Karamanos NK, Tzanakakis GN. Heparan sulfate proteoglycans and heparin regulate melanoma cell functions. Biochim Biophys Acta Gen Subj 2014; 1840:2471-81. [PMID: 24486410 DOI: 10.1016/j.bbagen.2014.01.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 01/17/2014] [Accepted: 01/20/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND The solid melanoma tumor consists of transformed melanoma cells, and the associated stromal cells including fibroblasts, endothelial cells, immune cells, as well as, soluble macro- and micro-molecules of the extracellular matrix (ECM) forming the complex network of the tumor microenvironment. Heparan sulfate proteoglycans (HSPGs) are an important component of the melanoma tumor ECM. Importantly, there appears to be both a quantitative and a qualitative shift in the content of HSPGs, in parallel to the nevi-radial growth phase-vertical growth phase melanoma progression. Moreover, these changes in HSPG expression are correlated to modulations of key melanoma cell functions. SCOPE OF REVIEW This review will critically discuss the roles of HSPGs/heparin in melanoma development and progression. MAJOR CONCLUSIONS We have correlated HSPGs' expression and distribution with melanoma cell signaling and functions as well as angiogenesis. GENERAL SIGNIFICANCE The current knowledge of HSPGs/heparin biology in melanoma provides a foundation we can utilize in the ongoing search for new approaches in designing anti-tumor therapy. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- D Nikitovic
- Department of Anatomy, Histology, Embryology, Medical School, University of Crete, Heraklion 71003, Greece
| | - M Mytilinaiou
- Department of Anatomy, Histology, Embryology, Medical School, University of Crete, Heraklion 71003, Greece
| | - Ai Berdiaki
- Department of Anatomy, Histology, Embryology, Medical School, University of Crete, Heraklion 71003, Greece
| | - N K Karamanos
- Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras 26110, Greece
| | - G N Tzanakakis
- Department of Anatomy, Histology, Embryology, Medical School, University of Crete, Heraklion 71003, Greece.
| |
Collapse
|
31
|
Jeong KW, Jeong MC, Jin B, Kim Y. Relationship between Structural Flexibility and Function in the C-Terminal Region of the Heparin-Binding Domain of VEGF165. Biochemistry 2013; 52:8823-32. [DOI: 10.1021/bi4011682] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Ki-Woong Jeong
- Department of Bioscience
and Biotechnology and Bio/Molecular Informatics
Center, Institute of KU Biotechnology, Konkuk University, Seoul 143-701, Korea
| | - Min-Cheol Jeong
- Department of Bioscience
and Biotechnology and Bio/Molecular Informatics
Center, Institute of KU Biotechnology, Konkuk University, Seoul 143-701, Korea
| | - Bonghwan Jin
- Department of Bioscience
and Biotechnology and Bio/Molecular Informatics
Center, Institute of KU Biotechnology, Konkuk University, Seoul 143-701, Korea
| | - Yangmee Kim
- Department of Bioscience
and Biotechnology and Bio/Molecular Informatics
Center, Institute of KU Biotechnology, Konkuk University, Seoul 143-701, Korea
| |
Collapse
|
32
|
The antineoplastic effect of low-molecular-weight heparins - a literature review. Contemp Oncol (Pozn) 2013; 17:6-13. [PMID: 23788954 PMCID: PMC3685354 DOI: 10.5114/wo.2013.33766] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 04/15/2012] [Accepted: 05/07/2012] [Indexed: 11/17/2022] Open
Abstract
There is some evidence for the antitumor effect of heparins, especially the low-molecular-weight ones. The authors discuss the potential mechanism of this antineoplastic effect and present results from several in vitro and in vivo experiments. The clinical trials concerning the impact of low-molecular-weight heparins on the tumor and on the patients' survival are described. The objective was to find out if heparins could be administered as an antitumor drug, independently of their anticoagulatory properties. The antitumor role of tissue factor, heparinase, chemokines, stromal proteins, cellular interactions as well as angiogenesis and immunology seems certain. The results of the available studies seem promising but large clinical trials are necessary in order to confirm the antineoplastic effect of the low-molecular-weight heparins and to approve them for standard anticancer treatment. It could be a breakthrough in modern oncology.
Collapse
|
33
|
Elit LM, Lee AY, Parpia S, Swystun LL, Liaw PC, Hoskins P, Julian DH, Julian JA, Levine MN. Dalteparin Low Molecular Weight Heparin (LMWH) in ovarian cancer: A phase II randomized study. Thromb Res 2012; 130:894-900. [DOI: 10.1016/j.thromres.2012.09.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 09/13/2012] [Accepted: 09/14/2012] [Indexed: 01/28/2023]
|
34
|
Abstract
Cancer is frequently associated with activation of blood coagulation, which in turn has been suggested to promote tumor growth and metastasis. Indeed, low molecular weight heparin treatment significantly prolongs the survival of a wide variety of patients with cancer. Based on this notion that anticoagulant treatment seems to benefit cancer patients, recent experiments aimed to elucidate the importance of the natural anticoagulant protein C pathways in cancer progression. Interestingly, these experiments showed that the repeated administration of exogenous activated protein C limits cancer cell extravasation in experimental animal models. In line, reducing endogenous activated protein C activity dramatically increased the number of experimental metastasis. These data thus strongly suggest that exogenous activated protein C administration may be a novel therapeutic avenue to limit cancer metastasis thereby prolonging overall survival of cancer patients. The current review provides an overview of recent data on the role of the protein C pathway in cancer metastasis. It discusses the potential of activated protein C as a novel target to reduce cancer progression, it points to several limitations of activated protein C administration in the setting of cancer cell metastasis and it suggest zymogen protein C as an attractive alternative.
Collapse
Affiliation(s)
- C Arnold Spek
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands.
| | | |
Collapse
|
35
|
Mousa SA. Comparative Pharmacodynamic Assessment of the Antiangiogenesis Activity of Heparin and Low-Molecular-Weight Heparin Fractions. Clin Appl Thromb Hemost 2012; 19:48-54. [DOI: 10.1177/1076029611436194] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Shaker A. Mousa
- The Pharmaceutical Research Institute at Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
36
|
Chu AJ. Tissue factor, blood coagulation, and beyond: an overview. Int J Inflam 2011; 2011:367284. [PMID: 21941675 PMCID: PMC3176495 DOI: 10.4061/2011/367284] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Revised: 06/16/2011] [Accepted: 06/18/2011] [Indexed: 12/18/2022] Open
Abstract
Emerging evidence shows a broad spectrum of biological functions of tissue factor (TF). TF classical role in initiating the extrinsic blood coagulation and its direct thrombotic action in close relation to cardiovascular risks have long been established. TF overexpression/hypercoagulability often observed in many clinical conditions certainly expands its role in proinflammation, diabetes, obesity, cardiovascular diseases, angiogenesis, tumor metastasis, wound repairs, embryonic development, cell adhesion/migration, innate immunity, infection, pregnancy loss, and many others. This paper broadly covers seminal observations to discuss TF pathogenic roles in relation to diverse disease development or manifestation. Biochemically, extracellular TF signaling interfaced through protease-activated receptors (PARs) elicits cellular activation and inflammatory responses. TF diverse biological roles are associated with either coagulation-dependent or noncoagulation-mediated actions. Apparently, TF hypercoagulability refuels a coagulation-inflammation-thrombosis circuit in “autocrine” or “paracrine” fashions, which triggers a wide spectrum of pathophysiology. Accordingly, TF suppression, anticoagulation, PAR blockade, or general anti-inflammation offers an array of therapeutical benefits for easing diverse pathological conditions.
Collapse
Affiliation(s)
- Arthur J Chu
- Division of Biological and Physical Sciences, Delta State University, Cleveland, MS 38733, USA
| |
Collapse
|
37
|
Norrby K, Nordenhem A. Dalteparin, a low-molecular-weight heparin, promotes angiogenesis mediated by heparin-binding VEGF-A in vivo. APMIS 2010; 118:949-57. [PMID: 21091776 PMCID: PMC3003896 DOI: 10.1111/j.1600-0463.2010.02635.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Tumors are angiogenesis dependent and vascular endothelial growth factor-A (VEGF-A), a heparin-binding protein, is a key angiogenic factor. As chemotherapy and co-treatment with anticoagulant low-molecular-weight heparin (LMWH) are common in cancer patients, we investigated whether angiogenesis in vivo mediated by VEGF-A is modulated by metronomic-type treatment with: (i) the LMWH dalteparin; (ii) low-dosage cytostatic epirubicin; or (iii) a combination of these two drugs. Using the quantitative rat mesentery angiogenesis assay, in which angiogenesis was induced by intraperitoneal injection of very low doses of VEGF, dalteparin sodium (Fragmin(®) ) and epirubicin (Farmorubicin(®) ) were administered separately or in combination by continuous subcutaneous infusion at a constant rate for 14 consecutive days. Dalteparin was administered at 27, 80, or 240 IU/kg/day, i.e., doses that reflect the clinical usage of this drug, while epirubicin was given at the well-tolerated dosage of 0.4 mg/kg/day. While dalteparin significantly stimulated angiogenesis in an inversely dose-dependent manner, epirubicin did not significantly affect angiogenesis. However, concurrent treatment with dalteparin and epirubicin significantly inhibited angiogenesis. The effect of dalteparin is the first demonstration of a proangiogenic effect of any LMWH in vivo. The fact that co-treatment with dalteparin and epirubicin significantly inhibited angiogenesis suggests a complex drug effect.
Collapse
Affiliation(s)
- Klas Norrby
- Department of Pathology, University of Gothenburg, Stockholm, Sweden.
| | | |
Collapse
|
38
|
van Sluis GL, Büller HR, Spek CA. The role of activated protein C in cancer progression. Thromb Res 2010; 125 Suppl 2:S138-42. [PMID: 20433994 DOI: 10.1016/s0049-3848(10)70032-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Activated protein C (APC) is best known as a natural anticoagulant that also has direct cell signaling properties which (among others) enhance vascular barrier function. We recently established the relevance of APC-induced barrier enhancement by showing that endogenous APC limits cancer cell extravasation. In line with this concept, repeated administration of exogenous APC reduced the number of experimental metastasis. It is thus tempting to speculate that exogenous APC administration would be a novel therapeutic avenue to fight cancer metastasis. The current review summarizes recent data on the role of the protein C pathway in cancer metastasis. It discusses the APC pathway as a potential novel target to influence cancer progression, but it also points to several limitations of APC administration in the setting of cancer cell metastasis.
Collapse
Affiliation(s)
- Geerte L van Sluis
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands.
| | | | | |
Collapse
|
39
|
The mechanism of melanoma-associated thrombin activity and von Willebrand factor release from endothelial cells. J Invest Dermatol 2010; 130:2259-68. [PMID: 20505748 DOI: 10.1038/jid.2010.136] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Activation of the coagulation system in malignancy enables tumor spreading and is thus associated with poor prognosis for the patient. In this study, we analyzed the in vitro mechanisms by which two human metastatic melanoma cell lines, MV3 and WM9, transform the vascular endothelium into a prothrombotic activated state. We show that both melanoma cell lines activate prothrombin due to tissue factor (TF) expression by showing that thrombin generation was blocked with a TF-neutralizing antibody and TF-siRNA. In addition, using the cysteine protease inhibitor E-64, we excluded the formerly described cancer procoagulant (CP) as a major factor contributing to thrombin generation. Furthermore, we describe a direct thrombin-independent response of endothelial cells (ECs) to MV3-derived supernatant as measured by rapid release of VWF. We also show that two clinically approved LMWHs, tinzaparin and enoxaparin, are effective inhibitors of thrombin generation and thrombin activity in plasma. Furthermore, our data indicate a protective effect of heparins on EC activation as shown by reduced VWF release in response to MV3 supernatant. These promising effects of heparins on the melanoma-induced thrombotic conditions justify further clinical investigations in the field of oncology.
Collapse
|
40
|
Borsig L. Antimetastatic activities of heparins and modified heparins. Experimental evidence. Thromb Res 2010; 125 Suppl 2:S66-71. [DOI: 10.1016/s0049-3848(10)70017-7] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
41
|
Barbera L, Thomas G. Erythropoiesis stimulating agents, thrombosis and cancer. Radiother Oncol 2010; 95:269-76. [PMID: 20219259 DOI: 10.1016/j.radonc.2010.02.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Accepted: 02/09/2010] [Indexed: 12/27/2022]
Abstract
Venous thromboembolism (VTE) is common in cancer and is associated with both morbidity and mortality. Erythropoiesis stimulating agents (ESAs) were originally developed to correct anemia. Recent trials in cancer patients however, raise concerns over both increased VTE rates and the possibility of worse tumour outcomes and increased mortality with ESA use. The most common reason offered for explaining the possible negative impact of ESAs on cancer outcomes has been the stimulation of erythropoietin receptors on tumour cells. Despite an extensive literature, it is unlikely that most practicing appreciate the intricate relationship and interaction between the coagulation pathways, angiogenesis and tumour progression and ESA effects. This paper will review these connections and interactions and examine the hypothesis that other mechanisms may underlie the possible negative impact of ESAs on cancer outcomes.
Collapse
Affiliation(s)
- Lisa Barbera
- Department of Radiation Oncology, University of Toronto, Canada
| | | |
Collapse
|
42
|
Abstract
Heparin and its improved version, low-molecular weight heparin (LMWH), are known to exert polypharmacological effects at various levels. Early studies focused on the plasma anti-Xa and anti-IIa pharmacodynamics of different LMWHs. Other important pharmacodynamic parameters for heparin and LMWH, including effects on vascular tissue factor pathway inhibitor (TFPI) release, inhibition of inflammation through NFkappaB, inhibition of key matrix-degrading enzymes, selectin modulation, inhibition of platelet-cancer cell interactions, and inflammatory cell adhesion, help explain the diverse clinical impact of this class of agents in thrombosis and beyond.
Collapse
Affiliation(s)
- Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| |
Collapse
|
43
|
Borsig L. Heparin as an inhibitor of cancer progression. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 93:335-49. [PMID: 20807651 DOI: 10.1016/s1877-1173(10)93014-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Heparin is frequently used in the treatment of cancer-associated thromboembolism. Accumulating clinical evidence indicates that cancer patients treated with unfractionated and low-molecular weight heparin (LMWH) survive longer than patients treated by other anticoagulants, especially patients in the early stage of the disease. Experimental analysis from a number of animal models constantly provides evidence for the ability of heparin to attenuate metastasis. The non-anticoagulant activity of heparin on metastasis includes the ability to inhibit cell-cell-interaction through blocking of P- and L-selectin, to inhibit extracellular matrix protease heparanase, and to inhibit angiogenesis. This chapter summarizes current experimental evidence on the biology of heparin during cancer progression, with the focus on potential mechanism of heparin antimetastatic activity.
Collapse
Affiliation(s)
- Lubor Borsig
- Institute of Physiology, Zürich Center for Integrative Human Physiology, University of Zürich, Switzerland
| |
Collapse
|
44
|
Banga J, Tripathi C. Rapid purification and characterization of a novel heparin degrading enzyme from Acinetobacter calcoaceticus. N Biotechnol 2009; 26:99-104. [DOI: 10.1016/j.nbt.2009.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Revised: 03/04/2009] [Accepted: 04/28/2009] [Indexed: 11/15/2022]
|
45
|
Kuderer NM, Ortel TL, Francis CW. Impact of venous thromboembolism and anticoagulation on cancer and cancer survival. J Clin Oncol 2009; 27:4902-11. [PMID: 19738120 DOI: 10.1200/jco.2009.22.4584] [Citation(s) in RCA: 170] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Changes in the hemostatic system and chronic hemostatic activation are frequently observed in patients with cancer, even in the absence of venous thromboembolism (VTE). VTE is a leading cause of death among patients with cancer and contributes to long-term mortality in patients with early as well as advanced-stage cancer. Mounting evidence suggests that components of the clotting cascade and associated vascular factors play an integral part in tumor progression, invasion, angiogenesis, and metastasis formation. Furthermore, there are intriguing in vitro and animal findings that anticoagulants, in particular the low molecular weight heparins (LMWHs), exert an antineoplastic effect through multiple mechanisms, including interference with tumor cell adhesion, invasion, metastasis formation, angiogenesis, and the immune system. Several relatively small randomized controlled clinical trials of anticoagulation as cancer therapy in patients without a VTE diagnosis have been completed. These comprise studies with LMWH, unfractionated heparin, and vitamin K antagonists, with overall encouraging but nonconclusive results and some limitations. Meta-analyses performed for the American Society of Clinical Oncology VTE Guidelines Committee and the Cochrane Collaboration suggest overall favorable effects of anticoagulation on survival of patients with cancer, mainly with LMWH. However, definitive clinical trials have been elusive and questions remain regarding the importance of tumor type and stage on treatment efficacy, the impact of fatal thromboembolic events, optimal anticoagulation therapy, and safety with differing chemotherapy regimens. Although the LMWHs and related agents hold promise for improving outcomes in patients with cancer, additional studies of their efficacy and safety in this setting are needed.
Collapse
Affiliation(s)
- Nicole M Kuderer
- Division of Hematology, Oncology and Cellular Therapy, Duke Comprehensive Cancer Center, Duke University Medical Center, DUMC 3841, Durham, NC 27710, USA.
| | | | | |
Collapse
|
46
|
Niers TMH, Brüggemann LW, VAN Sluis GL, Liu RD, Versteeg HH, Büller HR, VAN Noorden CJF, Reitsma PH, Spek CA, Richel DJ. Long-term thrombin inhibition promotes cancer cell extravasation in a mouse model of experimental metastasis. J Thromb Haemost 2009; 7:1595-7. [PMID: 19558435 DOI: 10.1111/j.1538-7836.2009.03529.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
47
|
Banga J, Tripathi C. Response surface methodology for optimization of medium components in submerged culture ofAspergillus flavusfor enhanced heparinase production. Lett Appl Microbiol 2009; 49:204-9. [DOI: 10.1111/j.1472-765x.2009.02640.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
48
|
Läubli H, Borsig L. Heparins Attenuate Cancer Metastasis: Are Selectins the Link? Cancer Invest 2009; 27:474-81. [DOI: 10.1080/07357900802647136] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
49
|
Abstract
Both women and men with cancer are at increased risk for developing venous thromboembolism (VTE), a propensity that has been known for many years. Until recently it was assumed, however, that the association between cancer and thrombosis is an epiphenomenon - not causally related to the transforming malignant events. The pathophysiology of thrombosis in patients with cancer is complex involving multiple tumor-related and host-related factors. Several recent studies have provided strong evidence that activation of blood coagulation, perhaps most often mediated by tissue factor (TF)-rich microparticles (MPs), is linked directly to oncogene-induced malignant transformation. In addition, the development of VTE, either before or concurrent with the diagnosis of cancer, appears to predict an aggressive behavior of a tumor, and correlates with increased tumor angiogenesis and early onset of distant metastasis. The regulation of expression of TF in tumor cells is controlled at the molecular level by several oncogenes, as appears to be true for cyclooxygenase-2 (COX-2), an important regulator of platelet function and plasminogen activator inhibitor type 1 (PAI-1), an inhibitor of fibrinolysis. In addition, engagement of protease-activated receptors (PARs) by the TF-factor VIIa complex, factor Xa and/or thrombin, have now been shown to be important for tumor growth, angiogenesis and metastasis. Targeting blood clotting reactions in cancer, therefore, may provide a unique approach to cancer treatment.
Collapse
Affiliation(s)
- Frederick R Rickles
- Departments of Medicine, Pediatrics and Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.
| |
Collapse
|
50
|
Affiliation(s)
- Frederick R Rickles
- Division of Hematology-Oncology, Department of Medicine, The George Washington University, 2150 Pennsylvania Ave NW Suite 3-428, Washington, DC 20037, USA.
| | | |
Collapse
|