1
|
Shestovskaya MV, Luss AL, Bezborodova OA, Venidiktova YB, Vorontsova MS, Vlaskina ER, Kushnerev KK, Kulikov PP, Makarov VV, Yudin VS, Keskinov AA. Iron Oxide Nanoparticles as Enhancers for Radiotherapy of Tumors. ACS APPLIED BIO MATERIALS 2025; 8:2535-2547. [PMID: 40035223 DOI: 10.1021/acsabm.4c01978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
This research aimed to evaluate the potency of preparation based on heparinized iron oxide nanoparticles (hIONPs) in combination with radiation therapy, including magnetic delivery via the applied magnetic field (AMF), in sarcoma and cervical cancer models. For in vitro studies, cells of rhabdomyosarcoma (RD), fibrosarcoma (HT1080), and cervical cancer (HeLa S3) were treated with hIONPs and analyzed for survival rate and hIONP uptake. Then, cell morphology, cell cycle, increase of reactive oxygen species, mitochondria depolarization, and ability to form colonies were assessed for combined treatment (hIONPs + 3Gy). For in vivo research, hIONPs were administered once in the hybrids of CBAxC57Bl/6j mice, grafted with sarcoma (S37) and cervical cancer (CC5) strains. The ultimate in vivo treatment modes were: (1) i.v. hIONPs (14 μg/kg) + 5 Gy; (2) i.v. hIONPs (14 μg/kg) + AMF + 5 Gy; and (3) i.t. hIONPs (2,8 μg/kg) + 5 Gy. The overall survival rates, increase in life expectancy, inhibition of tumor growth (tumor growth inhibition), and degree of inhibition (T/C) were determined, and pathomorphological changes were assessed in experimental groups. The combined treatment in vitro (hIONPs + 3Gy) promotes multiple tumor cell death with high-severity peroxide effects compared with other groups. The sarcoma cells were more sensitive than cervical cancer cells. For in vivo, an enhancing effect was revealed by the combination of radiotherapy and magnetic-delivered hIONPs. For S37 tumor, the treatment regimen was characterized as having a high antitumor effect, ≪++++ ≫, with a 20% cure rate of mice. For the CC5 tumor, the effect was accompanied by the inhibition of tumor growth, an increase in the life expectancy of animals, and was characterized as a significant antitumor effect, ≪+++/++ ≫. From the data obtained, it can be concluded that the radiosensitizing potential of hIONPs may be taken as a basis of combined radiation treatment protocols.
Collapse
Affiliation(s)
- Maria V Shestovskaya
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia
| | - Anna L Luss
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia
| | - Olga A Bezborodova
- P. Hertsen Moscow Oncology Research Institute - Branch of the National Medical Research Radiological Centre, Ministry of Health of the Russian Federation; Second Botkinskiy p. 3, Moscow 125284, Russia
| | - Yulia B Venidiktova
- P. Hertsen Moscow Oncology Research Institute - Branch of the National Medical Research Radiological Centre, Ministry of Health of the Russian Federation; Second Botkinskiy p. 3, Moscow 125284, Russia
| | - Maria S Vorontsova
- P. Hertsen Moscow Oncology Research Institute - Branch of the National Medical Research Radiological Centre, Ministry of Health of the Russian Federation; Second Botkinskiy p. 3, Moscow 125284, Russia
| | - Elizaveta R Vlaskina
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia
| | - Kirill K Kushnerev
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia
| | - Pavel P Kulikov
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia
| | - Valentin V Makarov
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia
| | - Vladimir S Yudin
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia
| | - Anton A Keskinov
- Federal State Budgetary Institution "Centre for Strategic Planning and Management of Biomedical Health Risks" of the Federal Medical Biological Agency, Schukinskaya st. 5/1, Moscow 119435, Russia
| |
Collapse
|
2
|
Koirala N, Poudel M, Shrivastava AK, Subba RK, Panthi M, Paudel S, Almarhoon ZM, Sharifi-Rad J, Calina D. Multifaceted role of heparin in oncology: from anticoagulation to anticancer mechanisms and clinical implications. Discov Oncol 2025; 16:231. [PMID: 39992596 PMCID: PMC11850695 DOI: 10.1007/s12672-025-01985-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 02/17/2025] [Indexed: 02/26/2025] Open
Abstract
Heparin, traditionally known for its anticoagulant properties, has recently been identified as a potential agent in cancer therapy. Its derivatives, including low-molecular-weight heparin (LMWH) and unfractionated heparin (UFH), are being investigated for their multifaceted roles in oncology. This review focuses on the expanding exploration of heparin's anticancer effects and its possible integration into cancer treatment protocols. The primary aim is to consolidate and analyze current research on the anticancer properties of heparin and its derivatives. It seeks to illuminate the mechanisms by which these compounds influence cancer progression, including their impact on angiogenesis, tumor cell proliferation, immune response modulation, and the inhibition of cancer cell migration and invasion. Additionally, the review aims to evaluate the potential of heparin and its derivatives in complementing existing chemotherapy treatments. An extensive literature review was conducted, encompassing in vitro, in vivo, and clinical studies. Sources included a range of scientific databases, employing keywords related to heparin and oncology. The selected studies were critically reviewed to extract relevant data on the efficacy, mechanisms, and potential clinical applications of heparin in cancer therapy. The results reveals that heparin and its derivatives exhibit significant anticancer activity across various research settings; key findings include the inhibition of angiogenesis, reduction in tumor cell proliferation, stimulation of immune responses, and the limitation of cancer cell migration and invasion. The compounds also show promise as adjuncts to conventional chemotherapy, potentially enhancing the efficacy of existing cancer treatments. This review highlights the burgeoning role of heparin and its derivatives in the realm of cancer therapy, marking a shift from their traditional use as anticoagulants. While promising, the research underscores the need for further comprehensive studies to fully understand the mechanisms of action, optimal dosing, potential side effects, and patient selection criteria. The potential integration of heparin into cancer treatment regimens opens new therapeutic possibilities warranting continued investigation in this rapidly evolving field.
Collapse
Affiliation(s)
- Niranjan Koirala
- Specialized Research Center, Nepal Academy of Science and Technology, Pokhara, Gandaki, Nepal.
| | - Melina Poudel
- Department of Chemical and Biological Sciences, Youngstown State University, 1 Tressel Way, Youngstown, OH, 44555, USA
| | - Amit Kumar Shrivastava
- Arkansas Biosciences Institute, Arkansas State University, Jonesboro, AR, 72401, USA
- Molecular Biosciences Graduate program, Arkansas State University, Jonesboro, AR, 72401, USA
| | - Romit Kumar Subba
- College of Pharmacy, Gachon University, Hambakmoe-ro, Incheon, 21936, Republic of Korea
| | - Mamata Panthi
- College of Pharmacy, Gachon University, Hambakmoe-ro, Incheon, 21936, Republic of Korea
| | - Samrat Paudel
- Department of Biotechnology, Kathmandu University, Dhulikhel, Nepal
| | - Zainab M Almarhoon
- Department of Chemistry, College of Science, King Saud University, P. O. Box 2455, 11451, Riyadh, Saudi Arabia
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, Ecuador.
- Centro de Estudios Tecnológicos y Universitarios del Golfo, Veracruz, Mexico.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
3
|
Malik J, Ahmed S, Momin SS, Shaikh S, Alafnan A, Alanazi J, Said Almermesh MH, Anwar S. Drug Repurposing: A New Hope in Drug Discovery for Prostate Cancer. ACS OMEGA 2023; 8:56-73. [PMID: 36643505 PMCID: PMC9835086 DOI: 10.1021/acsomega.2c05821] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/24/2022] [Indexed: 06/12/2023]
Abstract
Prostate cancer (PCA), the most common cancer in men, accounted for 1.3 million new incidences in 2018. An increase in incidences is an issue of concern that should be addressed. Of all the reported prostate cancers, 85% were detected in stages III and IV, making them difficult to treat. Conventional drugs gradually lose their efficacy due to the developed resistance against them, thus requiring newer therapeutic agents to be used as monotherapy or combination. Recent research regarding treatment options has attained remarkable speed and development. Therefore, in this context, drug repurposing comes into the picture, which is defined as the "investigation of the off-patent, approved and marketed drugs for a novel therapeutic indication" which saves at least 30% of the time and cost, reducing the cost of treatment for patients, which usually runs high in cancer patients. The anticancer property of cardiac glycosides in cancers was tested in the early 1980s. The trend then shifts toward treating prostate cancer by repurposing other cardiovascular drugs. The current review mainly emphasizes the advantageous antiprostate cancer profile of conventional CVS drugs like cardiac glycosides, RAAS inhibitors, statins, heparin, and beta-blockers with underlying mechanisms.
Collapse
Affiliation(s)
- Jonaid
Ahmad Malik
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research, Guwahati 781003, India
- Biomedical
Engineering, Indian Institute of Technology
(IIT), Ropar, Punjab 140001, India
| | - Sakeel Ahmed
- Department
of Pharmacology and Toxicology, National
Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat 382355, India
| | - Sadiya Sikandar Momin
- Department
of Pharmaceutics, Annasaheb Dange College of B. Pharmacy, Ashta, Shivaji University, Sangli, Maharastra 416301, India
| | - Sijal Shaikh
- Sandip Institute
of Pharmaceutical Sciences, Savitribai Phule
Pune University, Nashik, Maharashtra 422213, India
| | - Ahmed Alafnan
- Department
of Pharmacology and Toxicology, University
of Hail, Hail 81422, Saudi Arabia
| | - Jowaher Alanazi
- Department
of Pharmacology and Toxicology, University
of Hail, Hail 81422, Saudi Arabia
| | | | - Sirajudheen Anwar
- Department
of Pharmacology and Toxicology, University
of Hail, Hail 81422, Saudi Arabia
| |
Collapse
|
4
|
Eisele A, Seystahl K, Rushing EJ, Roth P, Le Rhun E, Weller M, Gramatzki D. Venous thromboembolic events in glioblastoma patients: an epidemiological study. Eur J Neurol 2022; 29:2386-2397. [PMID: 35545894 PMCID: PMC9543144 DOI: 10.1111/ene.15404] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 04/04/2022] [Accepted: 05/04/2022] [Indexed: 12/01/2022]
Abstract
Background and purpose Venous thromboembolic events (VTEs) are a major complication in cancer patients, and therefore, also in brain cancer patients, anticoagulants are considered appropriate in the treatment of VTEs. Methods Frequency, risk factors, and treatment of VTEs, as well as associated complications, were assessed in a population‐based cohort of glioblastoma patients in the Canton of Zurich, Switzerland. Correlations between clinical data and survival were retrospectively analyzed using the log‐rank test and Cox regression models. Results Four hundred fourteen glioblastoma patients with isocitrate dehydrogenase wild‐type status were identified. VTEs were documented in 65 patients (15.7%). Median time from tumor diagnosis to the occurrence of a VTE was 1.8 months, and 27 patients were diagnosed with VTEs postoperatively (within 35 days; 42.2%). History of a prior VTE was more common in patients who developed VTEs than in those who did not (p = 0.004). Bevacizumab treatment at any time during the disease course was not associated with occurrence of VTEs (p = 0.593). Most patients with VTEs (n = 61, 93.8%) were treated with therapeutic anticoagulation. Complications occurred in 14 patients (23.0%), mainly intracranial hemorrhages (n = 7, 11.5%). Overall survival did not differ between patients diagnosed with VTEs and those who had no VTE (p = 0.139). Tumor progression was the major cause of death (n = 283, 90.7%), and only three patients (1.0%) died in association with acute VTEs. Conclusions Venous thromboembolic events occurred early in the disease course, suggesting that the implementation of primary venous thromboembolism prophylaxis during first‐line chemoradiotherapy could be explored in a randomized setting.
Collapse
Affiliation(s)
- Amanda Eisele
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Katharina Seystahl
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Elisabeth J Rushing
- Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland
| | - Patrick Roth
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Emilie Le Rhun
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.,Department of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Dorothee Gramatzki
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Wang J, Xiao L, Wang W, Zhang D, Ma Y, Zhang Y, Wang X. The Auxiliary Role of Heparin in Bone Regeneration and its Application in Bone Substitute Materials. Front Bioeng Biotechnol 2022; 10:837172. [PMID: 35646879 PMCID: PMC9133562 DOI: 10.3389/fbioe.2022.837172] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/13/2022] [Indexed: 11/18/2022] Open
Abstract
Bone regeneration in large segmental defects depends on the action of osteoblasts and the ingrowth of new blood vessels. Therefore, it is important to promote the release of osteogenic/angiogenic growth factors. Since the discovery of heparin, its anticoagulant, anti-inflammatory, and anticancer functions have been extensively studied for over a century. Although the application of heparin is widely used in the orthopedic field, its auxiliary effect on bone regeneration is yet to be unveiled. Specifically, approximately one-third of the transforming growth factor (TGF) superfamily is bound to heparin and heparan sulfate, among which TGF-β1, TGF-β2, and bone morphogenetic protein (BMP) are the most common growth factors used. In addition, heparin can also improve the delivery and retention of BMP-2 in vivo promoting the healing of large bone defects at hyper physiological doses. In blood vessel formation, heparin still plays an integral part of fracture healing by cooperating with the platelet-derived growth factor (PDGF). Importantly, since heparin binds to growth factors and release components in nanomaterials, it can significantly facilitate the controlled release and retention of growth factors [such as fibroblast growth factor (FGF), BMP, and PDGF] in vivo. Consequently, the knowledge of scaffolds or delivery systems composed of heparin and different biomaterials (including organic, inorganic, metal, and natural polymers) is vital for material-guided bone regeneration research. This study systematically reviews the structural properties and auxiliary functions of heparin, with an emphasis on bone regeneration and its application in biomaterials under physiological conditions.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lan Xiao
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia
- Australia−China Centre for Tissue Engineering and Regenerative Medicine, Brisbane, Australia
| | - Weiqun Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dingmei Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia
- Australia−China Centre for Tissue Engineering and Regenerative Medicine, Brisbane, Australia
| |
Collapse
|
6
|
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
|
7
|
Ma SN, Mao ZX, Wu Y, Liang MX, Wang DD, Chen X, Chang PA, Zhang W, Tang JH. The anti-cancer properties of heparin and its derivatives: a review and prospect. Cell Adh Migr 2021; 14:118-128. [PMID: 32538273 PMCID: PMC7513850 DOI: 10.1080/19336918.2020.1767489] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Heparin, including unfractionated heparin (UFH), low-molecular-weight heparin (LMWH) and heparin derivatives, are commonly used in venous thromboembolism treatment and reportedly have beneficial effects on cancer survival. Heparin can affect the proliferation, adhesion, angiogenesis, migration and invasion of cancer cells via multiple mechanisms. The main mechanisms involve inhibition of heparanase, P-/L-selectin, angiogenesis, and interference with the CXCL12-CXCR4 axis. Here we summarize the current experimental evidence regarding the anti-cancer role of heparin and its derivatives, and conclude that there is evidence to support heparin’s role in inhibiting cancer progression, making it a promising anti-cancer agent.
Collapse
Affiliation(s)
- Sai-Nan Ma
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China.,Department of Oncology, The Affiliated Suqian Hospital of Xuzhou Medical University , Suqian, P.R.China
| | - Zhi-Xiang Mao
- Department of Oncology, Affiliated Hospital of Xuzhou Medical University , Xuzhou, P.R. China
| | - Yang Wu
- Core Facility, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China
| | - Ming-Xing Liang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China
| | - Dan-Dan Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China
| | - Xiu Chen
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China
| | - Ping-An Chang
- Urinary Surgery, Dongtai People's Hospital , Dongtai, P.R. China
| | - Wei Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China
| | - Jin-Hai Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University , Nanjing, P.R. China
| |
Collapse
|
8
|
Fang L, Xu Q, Qian J, Zhou JY. Aberrant Factors of Fibrinolysis and Coagulation in Pancreatic Cancer. Onco Targets Ther 2021; 14:53-65. [PMID: 33442266 PMCID: PMC7797325 DOI: 10.2147/ott.s281251] [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: 09/09/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022] Open
Abstract
Aberrant factors associated with fibrinolysis and thrombosis are found in many cancer patients, which can promote metastasis and are associated with poor prognosis. The relationship between tumor-associated fibrinolysis and thrombosis is poorly understood in pancreatic cancer. This review provides a brief highlight of existing studies that the fibrinolysis and coagulation systems were activated in pancreatic cancer patients, along with aberrant high concentrations of tissue plasminogen activator (t-PA), urine plasminogen activator (u-PA), D-dimer, fibrinogen, or platelets. These factors cooperate with each other, propelling tumor cell shedding, localization, adhesion to distant metastasis. The relationship between thrombosis or fibrinolysis and cancer immune escape is also investigated. In addition, the potential prevention and therapy strategies of pancreatic cancer targeting factors in fibrinolysis and coagulation systems are also been discussed, in which we highlight two effective agents aspirin and low-molecular weight heparin (LMWH). Summarily, this review provides new directions for the research and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Lianghua Fang
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, People's Republic of China
| | - Qing Xu
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210029, People's Republic of China
| | - Jun Qian
- Department of Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, People's Republic of China
| | - Jin-Yong Zhou
- Central Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, People's Republic of China
| |
Collapse
|
9
|
Chen X, Ji H, Wang J, Zhao G, Zheng B, Niu Z, He W. Prognostic Value of the Preoperative Plasma D-Dimer Levels in Patients with Upper Tract Urothelial Carcinoma in a Retrospective Cohort Study. Onco Targets Ther 2020; 13:5047-5055. [PMID: 32606727 PMCID: PMC7292253 DOI: 10.2147/ott.s254514] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/13/2020] [Indexed: 12/22/2022] Open
Abstract
Purpose Elevated plasma D-dimer levels were thought to be associated with decreasing survival in various cancers. The relationship between plasma D-dimer levels and clinicopathology and the optimal D-dimer cutoff as a prognostic predictor has not been determined in patients with upper tract urothelial carcinoma (UTUC). We aimed to investigate the prognostic value of preoperative plasma D-dimer levels as a predictor of patient outcomes in UTUC following radical nephroureterectomy. Patients and Methods We retrospectively reviewed data for 232 patients. The D-dimer cutoff value was set at 0.36 mg/L, and we used the Kaplan-Meier method and Cox's proportional hazards regression models to analyze the association between D-dimer levels and oncological outcomes. Multivariate Cox regression was used to develop a nomogram, which we evaluated for accuracy using a receiver operating characteristic curve, calibration plot, and decision curve analysis. Results Plasma D-dimer levels ≥0.36 mg/L were significantly associated with advanced tumor status regarding size, location, hydronephrosis, tumor grade, lymph node involvement, grade, and stage (all p < 0.05). The Kaplan-Meier analysis showed that plasma D-dimer levels ≥0.36 mg/L predicted worse oncological outcomes vs levels <0.36 mg/L (all p < 0.001). Univariate and multivariate analyses showed that elevated preoperative plasma D-dimer level was an independent predictor of recurrence-free survival (hazard ratio (HR): 1.67, 95% confidence interval (CI): 1.07-2.63; p = 0.025), cancer-specific survival (HR: 2.34, 95% CI: 1.30-4.19; p = 0.004), and overall survival (HR: 1.98, 95% CI: 1.18-3.34; p = 0.010). We also developed a nomogram predicting 3- and 5-year overall survival probability. Conclusion D-dimer levels may be a useful prognostic predictor of survival and improve risk stratification and precisely individualize treatment for patients with UTUC.
Collapse
Affiliation(s)
- Xiaoxu Chen
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Haiyong Ji
- The Third Department of Surgery, Ningjin People's Hospital, Dezhou, Shandong, People's Republic of China
| | - Jianwei Wang
- Department of Urology, Shandong Provincial ENT Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Guiting Zhao
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Bin Zheng
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Zhihong Niu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| | - Wei He
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People's Republic of China.,Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, People's Republic of China
| |
Collapse
|
10
|
Farge D, Bournet B, Conroy T, Vicaut E, Rak J, Zogoulous G, Barkun J, Ouaissi M, Buscail L, Frere C. Primary Thromboprophylaxis in Pancreatic Cancer Patients: Why Clinical Practice Guidelines Should Be Implemented. Cancers (Basel) 2020; 12:E618. [PMID: 32155940 PMCID: PMC7139861 DOI: 10.3390/cancers12030618] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 02/27/2020] [Accepted: 03/04/2020] [Indexed: 12/13/2022] Open
Abstract
Exocrine pancreatic ductal adenocarcinoma, simply referred to as pancreatic cancer (PC) has the worst prognosis of any malignancy. Despite recent advances in the use of adjuvant chemotherapy in PC, the prognosis remains poor, with fewer than 8% of patients being alive at 5 years after diagnosis. The prevalence of PC has steadily increased over the past decades, and it is projected to become the second-leading cause of cancer-related death by 2030. In this context, optimizing and integrating supportive care is important to improve quality of life and survival. Venous thromboembolism (VTE) is a common but preventable complication in PC patients. VTE occurs in one out of five PC patients and is associated with significantly reduced progression-free survival and overall survival. The appropriate use of primary thromboprophylaxis can drastically and safely reduce the rates of VTE in PC patients as shown from subgroup analysis of non-PC targeted placebo-controlled randomized trials of cancer patients and from two dedicated controlled randomized trials in locally advanced PC patients receiving chemotherapy. Therefore, primary thromboprophylaxis with a Grade 1B evidence level is recommended in locally advanced PC patients receiving chemotherapy by the International Initiative on Cancer and Thrombosis clinical practice guidelines since 2013. However, its use and potential significant clinical benefit continues to be underrecognized worldwide. This narrative review aims to summarize the main recent advances in the field including on the use of individualized risk assessment models to stratify the risk of VTE in each patient with individual available treatment options.
Collapse
Affiliation(s)
- Dominique Farge
- Institut Universitaire d’Hématologie, Université de Paris, EA 3518, F-75010 Paris, France
- Assistance Publique Hôpitaux de Paris, Saint-Louis Hospital, Internal Medicine, Autoimmune and Vascular Disease Unit, F-75010 Paris, France
- Department of Medicine, McGill University, Montreal, Québec, QC H4A 3J1, Canada
| | - Barbara Bournet
- University of Toulouse, F-31059 Toulouse, France; (B.B.); (L.B.)
- CHU de Toulouse, Department of Gastroenterology and Pancreatology, F-31059 Toulouse, France
| | - Thierry Conroy
- Institut de Cancérologie de Lorraine, Department of Medical Oncology, Université de Lorraine, APEMAC, EA4360, F-54519 Vandoeuvre-lès-Nancy, France;
| | - Eric Vicaut
- Department of Biostatistics, Université de Paris, F-75010 Paris, France;
- Assistance Publique Hôpitaux de Paris, Department of Biostatistics, Fernand Widal Hospital, F-75010 Paris, France
| | - Janusz Rak
- McGill University and the Research Institute of the McGill University Health Centre, Montreal, Québec, QC H4A 3J1, Canada; (J.R.)
| | - George Zogoulous
- McGill University and the Research Institute of the McGill University Health Centre, Montreal, Québec, QC H4A 3J1, Canada; (J.R.)
| | - Jefferey Barkun
- McGill University and the Research Institute of the McGill University Health Centre, Montreal, Québec, QC H4A 3J1, Canada; (J.R.)
| | - Mehdi Ouaissi
- Department of Digestive, Oncological, Endocrine, and Hepatic Surgery, and Hepatic Transplantation, Trousseau Hospital, CHRU Trousseau, F-37170 Chambray-les-Tours, France;
| | - Louis Buscail
- University of Toulouse, F-31059 Toulouse, France; (B.B.); (L.B.)
- CHU de Toulouse, Department of Gastroenterology and Pancreatology, F-31059 Toulouse, France
| | - Corinne Frere
- Institute of Cardiometabolism and Nutrition, Sorbonne Université, INSERM UMRS_1166, GRC 27 GRECO, F-75013 Paris, France;
- Assistance Publique Hôpitaux de Paris, Department of Haematology, Pitié-Salpêtrière Hospital, F-75013 Paris, France
| |
Collapse
|
11
|
Bokas A, Papakotoulas P, Sarantis P, Papadimitropoulou A, Papavassiliou AG, Karamouzis MV. Mechanisms of the Antitumor Activity of Low Molecular Weight Heparins in Pancreatic Adenocarcinomas. Cancers (Basel) 2020; 12:432. [PMID: 32069809 PMCID: PMC7072375 DOI: 10.3390/cancers12020432] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 02/01/2020] [Accepted: 02/10/2020] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors have revolutionized cancer treatment in the last decade. Despite the progress in immunotherapy, most pancreatic cancer patients still do not derive benefit when receiving immune-based therapies. Recently, resistance mechanisms to immune therapies have been mainly focused on tumor microenvironment properties. Pancreatic cancer is considered one of the most lethal and difficult to treat tumors due to its highly immunosuppressive and desmoplastic microenvironment. Low molecular weight heparins (LMWHs) have been used for the treatment and prevention of thromboembolic disease in these patients. However, many nonanticoagulant properties attributed to LMWHs have been described. Exploiting LMWH properties in a combined treatment modality with immune checkpoint inhibition and chemotherapy could provide a new approach in the management of pancreatic adenocarcinoma patients. The ability of LMWH to interfere with various aspects of the tumor microenvironment could result in both the alleviation of immunosuppression and improvement in drug delivery within the tumor, leading to higher cancer cell destruction rates and more potent immune system activity that would, ultimately, lead to better patient outcomes.
Collapse
Affiliation(s)
- Alexandros Bokas
- 1st Department of Medical Oncology, Theagenion Hospital, 54007 Thessaloniki, Greece; (A.B.); (P.P.)
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (A.G.P.)
| | - Pavlos Papakotoulas
- 1st Department of Medical Oncology, Theagenion Hospital, 54007 Thessaloniki, Greece; (A.B.); (P.P.)
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (A.G.P.)
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (A.G.P.)
| | - Adriana Papadimitropoulou
- Center of Basic Research, Biomedical Research Foundation of the Academy 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; (P.S.); (A.G.P.)
| | - Michalis V Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (A.G.P.)
- First Department of Internal Medicine, ‘Laiko’ General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
12
|
Yamada Y, Kawaguchi R, Iwai K, Niiro E, Morioka S, Tanase Y, Kobayashi H. Preoperative plasma D-dimer level is a useful prognostic marker in ovarian cancer. J OBSTET GYNAECOL 2019; 40:102-106. [PMID: 31335252 DOI: 10.1080/01443615.2019.1606176] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
A high pre-treatment plasma D-dimer level was recently identified as a poor prognostic factor in several malignancies. The aim of this study was to evaluate the prognostic significance of plasma D-dimer levels in epithelial ovarian cancer (EOC). Data of 199 patients were retrospectively analysed. The relationships between pre-treatment D-dimer levels and other clinical parameters and prognosis were evaluated. Univariate analysis identified age, pre-treatment plasma D-dimer level, massive ascites, residual tumours, pre-treatment CA125 level, histological type, and FIGO stage as predictors of overall survival. The multivariate analysis showed that a high pre-treatment plasma D-dimer level (p=.017), residual tumours (p < .001), and FIGO stage (p = .036) were independent risk factors of overall survival. Venous thromboembolism (VTE) did not influence overall survival (p=.091). High pre-treatment D-dimer levels are associated with a poor prognosis independent of VTE status in EOC patients, and might be a useful prognostic biomarker.Impact statementWhat is already known on this subject? In recent years, a high pre-treatment plasma D-dimer level has been identified as a prognostic factor in several malignancies, but only a handful of studies have assessed the role of pre-treatment plasma D-dimer levels in patients with EOC patients. Thus, the clinical significance and prognostic value of the plasma D-dimer level in EOC remain controversial, and there is also debate related to the association of the higher mortality rate among cancer patients with elevated D-dimer levels with VTE.What do the results of this study add? In our study, high pre-treatment D-dimer levels are associated with a poor prognosis independently of VTE in EOC patients.What are the implications of these findings for clinical practice and/or further research? The D-dimer level might emerge as a valuable prognostic biomarker, which will help doctors in the choice of initiating a more aggressive therapy, the combination of chemotherapy with anticoagulation therapy.
Collapse
Affiliation(s)
- Yuki Yamada
- Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Ryuji Kawaguchi
- Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Kana Iwai
- Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Emiko Niiro
- Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Sachiko Morioka
- Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Yasuhito Tanase
- Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| | - Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Nara, Japan
| |
Collapse
|
13
|
Meyer G, Besse B, Doubre H, Charles-Nelson A, Aquilanti S, Izadifar A, Azarian R, Monnet I, Lamour C, Descourt R, Oliviero G, Taillade L, Chouaid C, Giraud F, Falcoz PE, Revel MP, Westeel V, Dixmier A, Tredaniel J, Dehette S, Decroisette C, Prevost A, Pichon E, Fabre E, Soria JC, Friard S, Stern JB, Jabot L, Dennewald G, Pavy G, Petitpretz P, Tourani JM, Alifano M, Chatellier G, Girard P. Anti-tumour effect of low molecular weight heparin in localised lung cancer: a phase III clinical trial. Eur Respir J 2018; 52:13993003.01220-2018. [DOI: 10.1183/13993003.01220-2018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 08/06/2018] [Indexed: 11/05/2022]
Abstract
The anti-tumour and anti-metastatic properties of heparins have not been tested in patients with early stage cancer. Whether adjuvant low molecular weight heparin (LMWH) tinzaparin impacts the survival of patients with resected non-small cell lung cancer (NSCLC) was investigated.Patients with completely resected stage I, II or IIIA NSCLC were randomly allocated to receive subcutaneous tinzaparin 100 IU·kg−1 once a day for 12 weeks or no treatment in addition to standard of care. The trial was open-label with blinded central adjudication of study outcomes. The primary outcome was overall survival.In 549 patients randomised to tinzaparin (n=269) or control (n=280), mean±sd age was 61.6±8.9 years, 190 (34.6%) patients had stage II−III disease, and 220 (40.1%) patients received adjuvant chemotherapy. Median follow-up was 5.7 years. There was no significant difference in overall survival between groups (hazard ratio (HR) 1.24, 95% CI 0.92–1.68; p=0.17). There was no difference in the cumulative incidence of recurrence between groups (subdistribution HR 0.94, 95% CI 0.68–1.30; p=0.70).Adjuvant tinzaparin had no detectable impact on overall and recurrence-free survival of patients with completely resected stage I−IIIA NSCLC. These results do not support further clinical evaluation of LMWHs as anti-tumour agents.
Collapse
|
14
|
Gieseler F, Plattfaut C, Quecke T, Freund A, Ungefroren H, Ender F. Heterogeneity of microvesicles from cancer cell lines under inflammatory stimulation with TNF-α. Cell Biol Int 2018; 42:1533-1544. [PMID: 30080276 DOI: 10.1002/cbin.11040] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/29/2018] [Indexed: 12/11/2022]
Abstract
Microvesicles (MVs) represent a subgroup of extracellular vesicles (EVs) emerging from various cells by blebbing of their outer membrane. Therefore, they share features such as membrane composition and antigenicity with their parental cells. Released by many immune and tumor cells, MVs act as intercellular messengers, account for horizontal gene transfer and can activate the coagulation system. With the aim to investigate their relevance for tumor cell biology, we characterized MVs released by human tumor cell lines of various origins in the absence or presence of TNF-α. After stimulation, we used the combination of low and high-speed centrifugation to enrich MVs from cell culture supernatants. We analyzed the presentation of phosphatidylserine (PS) and tissue factor (TF) activity on the cell surface and investigated their potency to induce tumor cell migration. In all tumor cell lines, TNF-α stimulation enhanced the release of MVs. While the expression of PS was universally increased, an elevated activity of procoagulant TF could be detected on MVs from lung, pancreatic, and colon carcinoma, but not from breast and ovarian cancer cell lines. Functionally, TNF-α stimulation significantly increased the potency of MVs to induce tumor cell migration. In conclusion, inflammatory conditions promote the release of MVs with increased procoagulant activity from tumor cell lines in vitro. PS-containing and TF-expressing MVs may account for systemic activation of the coagulation system as seen in cancer patients and, since they induce tumor cell migration, they may serve as biomarkers for tumor progression.
Collapse
Affiliation(s)
- Frank Gieseler
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Luebeck, Luebeck, 23538, Germany
| | - Corinna Plattfaut
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Luebeck, Luebeck, 23538, Germany
| | - Tabea Quecke
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Luebeck, Luebeck, 23538, Germany
| | - Annika Freund
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Luebeck, Luebeck, 23538, Germany
| | - Hendrik Ungefroren
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Luebeck, Luebeck, 23538, Germany.,Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, Kiel, 24105, Germany
| | - Fanny Ender
- Section Experimental Oncology, University Hospital and Medical School (UKSH), University of Luebeck, Luebeck, 23538, Germany
| |
Collapse
|
15
|
Le Rhun E, Genbrugge E, Stupp R, Chinot OL, Nabors LB, Cloughesy T, Reardon DA, Wick W, Gorlia T, Weller M. Associations of anticoagulant use with outcome in newly diagnosed glioblastoma. Eur J Cancer 2018; 101:95-104. [PMID: 30036741 DOI: 10.1016/j.ejca.2018.06.029] [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: 05/21/2018] [Revised: 06/16/2018] [Accepted: 06/19/2018] [Indexed: 01/13/2023]
Abstract
BACKGROUND To test the hypothesis that despite bleeding risk, anticoagulants improve the outcome in glioblastoma because of reduced incidence of venous thromboembolic events and modulation of angiogenesis, infiltration and invasion. METHODS We assessed survival associations of anticoagulant use from baseline up to the start of temozolomide chemoradiotherapy (TMZ/RT) (period I) and from there to the start of maintenance TMZ chemotherapy (period II) by pooling data of three randomised clinical trials in newly diagnosed glioblastoma including 1273 patients. Progression-free survival (PFS) and overall survival (OS) were compared between patients with anticoagulant use versus no use; therapeutic versus prophylactic versus no use; different durations of anticoagulant use versus no use; anticoagulant use versus use of anti-platelet agents versus neither anticoagulant nor anti-platelet agent use. Cox regression models were stratified by trial and adjusted for baseline prognostic factors. RESULTS Anticoagulant use was documented in 75 patients (5.9%) in period I and in 104 patients (10.2%) in period II. Anticoagulant use during period II, but not period I, was associated with inferior OS than no use on multivariate analysis (p = 0.001, hazard ratio [HR] = 1.52, 95% confidence interval [CI]: 1.18-1.95). No decrease in OS became apparent when only patients with prophylactic anticoagulant use were considered. No survival association was established for anti-platelet agent use. CONCLUSIONS Anticoagulant use was not associated with improved OS. Anticoagulants may not exert relevant anti-tumour properties in glioblastoma.
Collapse
Affiliation(s)
- Emilie Le Rhun
- University of Lille, U-1192, F-59000, Lille, France; Inserm, U-1192, F-59000, Lille, France; CHU Lille, General and Stereotaxic Neurosurgery Service, F-59000, Lille, France; Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland.
| | | | - Roger Stupp
- Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Olivier L Chinot
- Aix-Marseille University, AP-HM, Service de Neuro-Oncologie, CHU Timone, Marseille, France
| | - L Burt Nabors
- University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Wolfgang Wick
- Department of Neurology and Neurooncology Programm at the National Center for Tumor Diseases, University Hospital Heidelberg and German Cancer Research Center, Heidelberg, Germany
| | | | - Michael Weller
- Department of Neurology & Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
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
|
17
|
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
|
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
|
Arlov Ø, Skjåk-Bræk G. Sulfated Alginates as Heparin Analogues: A Review of Chemical and Functional Properties. Molecules 2017; 22:E778. [PMID: 28492485 PMCID: PMC6154561 DOI: 10.3390/molecules22050778] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 05/03/2017] [Accepted: 05/05/2017] [Indexed: 01/22/2023] Open
Abstract
Heparin is widely recognized for its potent anticoagulating effects, but has an additional wide range of biological properties due to its high negative charge and heterogeneous molecular structure. This heterogeneity has been one of the factors in motivating the exploration of functional analogues with a more predictable modification pattern and monosaccharide sequence, that can aid in elucidating structure-function relationships and further be structurally customized to fine-tune physical and biological properties toward novel therapeutic applications and biomaterials. Alginates have been of great interest in biomedicine due to their inherent biocompatibility, gentle gelling conditions, and structural versatility from chemo-enzymatic engineering, but display limited interactions with cells and biomolecules that are characteristic of heparin and the other glycosaminoglycans (GAGs) of the extracellular environment. Here, we review the chemistry and physical and biological properties of sulfated alginates as structural and functional heparin analogues, and discuss how they may be utilized in applications where the use of heparin and other sulfated GAGs is challenging and limited.
Collapse
Affiliation(s)
- Øystein Arlov
- Department of Biotechnology and Nanomedicine, SINTEF Materials and Chemistry, Richard Birkelands vei 3B, 7034 Trondheim, Norway.
| | - Gudmund Skjåk-Bræk
- Department of Biotechnology, Norwegian University of Science and Technology, Sem Sælands vei 6/8, 7034 Trondheim, Norway.
| |
Collapse
|
20
|
Heyman B, Yang Y. Mechanisms of heparanase inhibitors in cancer therapy. Exp Hematol 2016; 44:1002-1012. [PMID: 27576132 DOI: 10.1016/j.exphem.2016.08.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/09/2016] [Accepted: 08/19/2016] [Indexed: 12/26/2022]
Abstract
Heparanase is an endo-β-D-glucuronidase capable of cleaving heparan sulfate side chains contributing to breakdown of the extracellular matrix. Increased expression of heparanase has been observed in numerous malignancies and is associated with a poor prognosis. It has generated significant interest as a potential antineoplastic target because of the multiple roles it plays in tumor growth and metastasis. The protumorigenic effects of heparanase are enhanced by the release of heparan sulfate side chains, with subsequent increase in bioactive fragments and cytokine levels that promote tumor invasion, angiogenesis, and metastasis. Preclinical experiments have found heparanase inhibitors to substantially reduce tumor growth and metastasis, leading to clinical trials with heparan sulfate mimetics. In this review, we examine the role of heparanase in tumor biology and its interaction with heparan surface proteoglycans, specifically syndecan-1, as well as the mechanism of action for heparanase inhibitors developed as antineoplastic therapeutics.
Collapse
Affiliation(s)
- Benjamin Heyman
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Yiping Yang
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University, Durham, North Carolina, USA; Department of Immunology, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
21
|
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
|
22
|
Wang J, Zhu C. Anticoagulation in combination with antiangiogenesis and chemotherapy for cancer patients: evidence and hypothesis. Onco Targets Ther 2016; 9:4737-46. [PMID: 27536135 PMCID: PMC4973715 DOI: 10.2147/ott.s103184] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Hypercoagulable state and disorganized angiogenesis are two conspicuous characteristics during tumor progression. There are a considerable number of clinical trials focusing on the effects of anticoagulant and antiangiogenic drugs on the survival of cancer patients. Favorable outcomes have been observed. Excessive blood coagulation not only causes cancer-associated thrombosis, which is a common complication and is the second leading cause of death in patients, but also decreases intratumoral perfusion rates and drug delivery by reducing the effective cross-sectional area of blood vessels. Meanwhile, structural and functional abnormalities of the tumor microvasculature also compromise convective drug transport and create a hypoxic and acidic microenvironment. Vascular normalization strategy can temporarily recover the abnormal state of tumor vasculature by improving blood density, dilation, and leakiness, resulting in enhanced penetration of chemotherapies and oxygen within a short time window. In this article, we first review the evidence to support the opinion that anticoagulant and antiangiogenic therapy can improve cancer survival through several underlying mechanisms. Next, we speculate on the feasibility and value of the combined strategy and discuss whether such a combination has a synergistic antineoplastic effect in cancer patients by way of increasing blood vessel perfusion and drug distribution.
Collapse
Affiliation(s)
- Ji Wang
- Department of Cardiothoracic Surgery, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, People's Republic of China
| | - Chengchu Zhu
- Department of Cardiothoracic Surgery, Taizhou Hospital, Wenzhou Medical University, Linhai, Zhejiang, People's Republic of China
| |
Collapse
|
23
|
The hypoxia-inducible epigenetic regulators Jmjd1a and G9a provide a mechanistic link between angiogenesis and tumor growth. Mol Cell Biol 2014; 34:3702-20. [PMID: 25071150 DOI: 10.1128/mcb.00099-14] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Hypoxia promotes stem cell maintenance and tumor progression, but it remains unclear how it regulates long-term adaptation toward these processes. We reveal a striking downregulation of the hypoxia-inducible histone H3 lysine 9 (H3K9) demethylase JMJD1A as a hallmark of clinical human germ cell-derived tumors, such as seminomas, yolk sac tumors, and embryonal carcinomas. Jmjd1a was not essential for stem cell self-renewal but played a crucial role as a tumor suppressor in opposition to the hypoxia-regulated oncogenic H3K9 methyltransferase G9a. Importantly, loss of Jmjd1a resulted in increased tumor growth, whereas loss of G9a produced smaller tumors. Pharmacological inhibition of G9a also resulted in attenuation of tumor growth, offering a novel therapeutic strategy for germ cell-derived tumors. Finally, Jmjd1a and G9a drive mutually opposing expression of the antiangiogenic factor genes Robo4, Igfbp4, Notch4, and Tfpi accompanied by changes in H3K9 methylation status. Thus, we demonstrate a novel mechanistic link whereby hypoxia-regulated epigenetic changes are instrumental for the control of tumor growth through coordinated dysregulation of antiangiogenic gene expression.
Collapse
|
24
|
Schlesinger M, Roblek M, Ortmann K, Naggi A, Torri G, Borsig L, Bendas G. The role of VLA-4 binding for experimental melanoma metastasis and its inhibition by heparin. Thromb Res 2014; 133:855-62. [DOI: 10.1016/j.thromres.2014.02.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/31/2014] [Accepted: 02/21/2014] [Indexed: 12/31/2022]
|
25
|
Nugent MA, Zaia J, Spencer JL. Heparan sulfate-protein binding specificity. BIOCHEMISTRY (MOSCOW) 2014; 78:726-35. [PMID: 24010836 DOI: 10.1134/s0006297913070055] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Heparan sulfate (HS) represents a large class of linear polysaccharides that are required for the function of all mammalian physiological systems. HS is characterized by a repeating disaccharide backbone that is subject to a wide range of modifications, making this class of macromolecules arguably the most information dense in all of biology. The majority of HS functions are associated with the ability to bind and regulate a wide range of proteins. Indeed, recent years have seen an explosion in the discovery of new activities for HS where it is now recognized that this class of glycans functions as co-receptors for growth factors and cytokines, modulates cellular uptake of lipoproteins, regulates protease activity, is critical to amyloid plaque formation, is used by opportunistic pathogens to enter cells, and may even participate in epigenetic regulation. This review will discuss the current state of understanding regarding the specificity of HS-protein binding and will describe the concept that protein binding to HS depends on the overall organization of domains within HS rather than fine structure.
Collapse
Affiliation(s)
- M A Nugent
- Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| | | | | |
Collapse
|
26
|
Meyer G. [Venous thromboembolism and cancer]. REVUE DE PNEUMOLOGIE CLINIQUE 2014; 70:91-94. [PMID: 24566034 DOI: 10.1016/j.pneumo.2013.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 11/10/2013] [Indexed: 06/03/2023]
Abstract
Incidence of venous thromboembolism (VTE) is high in patients with cancer and varies with the type, the stage and the histologic type of the cancer but also with some anticancer therapies and patient related risk factors. Therapeutic trials on long-term prophylaxis have not provided convincing results. In patients with established VTE, the risk of recurrence and bleeding is high. Although prolonged treatment with low molecular weight heparin (LMWH) is more efficient than vitamin K antagonists in patients with VTE and cancer, it is already associated with a risk of recurrence which is much higher than in non-cancer patients. The nature of the treatment to be administered after the initial six months, which is often required in this context and the treatment of recurrent VTE during anticoagulant treatment are still debated. Some long-standing data suggest that LMWH could play an adjuvant role as specific anticancer treatment. These data have still not been confirmed in clinical trials but several studies in different cancer types are ongoing.
Collapse
Affiliation(s)
- G Meyer
- Université Paris-Descartes, Sorbonne Paris-Cité, 75006 Paris, France; Service de pneumologie, hôpital européen Georges-Pompidou, AP-HP, 20, rue Leblanc, 75015 Paris, France.
| |
Collapse
|
27
|
Seib FP, Herklotz M, Burke KA, Maitz MF, Werner C, Kaplan DL. Multifunctional silk-heparin biomaterials for vascular tissue engineering applications. Biomaterials 2013; 35:83-91. [PMID: 24099708 DOI: 10.1016/j.biomaterials.2013.09.053] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 09/16/2013] [Indexed: 12/16/2022]
Abstract
Over the past 30 years, silk has been proposed for numerous biomedical applications that go beyond its traditional use as a suture material. Silk sutures are well tolerated in humans, but the use of silk for vascular engineering applications still requires extensive biocompatibility testing. Some studies have indicated a need to modify silk to yield a hemocompatible surface. This study examined the potential of low molecular weight heparin as a material for refining silk properties by acting as a carrier for vascular endothelial growth factor (VEGF) and improving silk hemocompatibility. Heparinized silk showed a controlled VEGF release over 6 days; the released VEGF was bioactive and supported the growth of human endothelial cells. Silk samples were then assessed using a humanized hemocompatibility system that employs whole blood and endothelial cells. The overall thrombogenic response for silk was very low and similar to the clinical reference material polytetrafluoroethylene. Despite an initial inflammatory response to silk, apparent as complement and leukocyte activation, the endothelium was maintained in a resting, anticoagulant state. The low thrombogenic response and the ability to control VEGF release support the further development of silk for vascular applications.
Collapse
Affiliation(s)
- F Philipp Seib
- Tufts University, Department of Biomedical Engineering, 4 Colby Street, Medford, MA 02155, USA
| | | | | | | | | | | |
Collapse
|
28
|
Meyer G, Sanchez O, Planquette B. [Antithrombotic treatment and cancer]. Presse Med 2013; 42:1239-45. [PMID: 23972549 DOI: 10.1016/j.lpm.2013.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 06/12/2013] [Indexed: 01/08/2023] Open
Abstract
Five to 10 % of cancers are complicated by venous thromboembolism. A score is available to classify patients according to their risk of venous thromboembolism. The long-term prophylaxis is not recommended in patients with cancer, even if it reduces the incidence of thromboembolic events, due to the high number of patients to treat to prevent one event. The treatment of venous thromboembolism in cancer is based on the use of low-molecular weight heparin for at least 3 months. The potential role of antithrombotic drugs in the prevention and adjuvant treatment of some cancers is being evaluated.
Collapse
Affiliation(s)
- Guy Meyer
- Université Paris Descartes, Sorbonne Paris Cité, AP-HP, hôpital européen Georges-Pompidou, service de pneumologie et soins intensifs, Inserm UMR S 970, 20, rue Leblanc, 75015 Paris, France.
| | | | | |
Collapse
|
29
|
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
|
30
|
Actions of the Kunitz-type serine protease inhibitor Amblyomin-X on VEGF-A-induced angiogenesis. Toxicon 2012; 60:333-40. [PMID: 22575283 DOI: 10.1016/j.toxicon.2012.04.349] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 04/17/2012] [Accepted: 04/24/2012] [Indexed: 11/22/2022]
Abstract
Amblyomin-X is a Kunitz-type serine protease inhibitor (Kunitz-type SPI) designed from the cDNA library of the Amblyomma cajennense tick, which displays in vivo anti-tumor activities. Here, the mechanisms of actions of Amblyomin-X in vascular endothelial growth factor A (VEGF-A)-induced angiogenesis were characterized. Topical application of Amblyomin-X (10 or 100 ng/10 μl; each 48 h) inhibited VEGF-A-induced (10 ng/10 μl; each 48 h) angiogenesis in the dorsal subcutaneous tissue in male Swiss mice. Moreover, similar effect was observed in the VEGF-A-induced angiogenesis in the chicken chorioallantoic membrane (CAM). Additional in vitro assays in t-End cells showed that Amblyomin-X treatment delayed the cell cycle, by maintaining them in G0/G1 phase, and inhibited cell proliferation and adhesion, tube formation and membrane expression of the adhesion molecule platelet-endothelial cell adhesion molecule-1 (PECAM-1), regardless of mRNA synthesis. Together, results herein reveal the role of Kunitz-type SPI on in vivo VEGF-A-induced angiogenesis, by exerting modulatory actions on endothelial cell proliferation and adhesion, especially on membrane expression of PECAM-1. These data provide further mechanisms of actions of Kunitz-type SPI, corroborating their relevance as scientific tools in the design of therapeutic molecules.
Collapse
|
31
|
Da Pozzo E, Barsotti MC, Bendinelli S, Martelli A, Calderone V, Balbarini A, Martini C, Di Stefano R. Differential effects of fondaparinux and bemiparin on angiogenic and vasculogenesis-like processes. Thromb Res 2012; 130:e113-22. [PMID: 22497885 DOI: 10.1016/j.thromres.2012.03.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 02/14/2012] [Accepted: 03/13/2012] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Conventional therapy for venous thromboembolism or acute coronary syndrome involves the administration of glycoanticoagulants (heparins) or oligosaccharides (fondaparinux). We evaluated the effects of such drugs on angiogenesis and vasculogenesis-like models. MATERIALS AND METHODS Human umbilical vein endothelial cells or human endothelial progenitor cells were treated with bemiparin, fondaparinux or unfractionated heparin, at concentrations reflecting the doses used in clinical practice. After 24h, cell viability, proliferation, tubule formation and angiogenic molecular mechanisms, such as activation of the serine/threonine kinase AKT, were assessed. In vivo angiogenesis was studied using a Matrigel sponge assay in mice. RESULTS Bemiparin gave a significant decrease of in vitro angiogenesis as shown by the reduction of endothelial cell tubule network, while both fondaparinux and unfractionated heparin did not show any significant effect. In assays of Matrigel sponge invasion in mice, unfractionated heparin was able to stimulate angiogenesis and, conversely, bemiparin inhibited angiogenesis. Furthermore, both bemiparin and fondaparinux caused a significant reduction in an in vitro vasculogenesis-like model, as demonstrated by the decrease of tubule network after co-seeding of endothelial progenitor cells and human umbilical vein endothelial cells. In addition, unfractionated heparin but not bemiparin was able to increase AKT phosphorylation. CONCLUSIONS In in vitro experiments, bemiparin was the only drug to show an anti-angiogenic and vasculogenic-like effect, unfractionated heparin showed only a trend to increase in angiogenesis assay and fondaparinux affected only the vasculogenesis-like model. Notably, the in vivo experiments corroborated these data. Such results are important for the choice of a patient-tailored therapy.
Collapse
Affiliation(s)
- Eleonora Da Pozzo
- Department of Psychiatry, Neurobiology, Pharmacology and Biotechnology, University of Pisa, Pisa, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Inhibitory effect of non-anticoagulant heparin (S-NACH) on pancreatic cancer cell adhesion and metastasis in human umbilical cord vessel segment and in mouse model. Clin Exp Metastasis 2012; 29:431-9. [PMID: 22415710 DOI: 10.1007/s10585-012-9461-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2011] [Accepted: 02/18/2012] [Indexed: 01/28/2023]
Abstract
Metastasis is the most devastating aspect of cancer and it is the main cause of morbidity and mortality in cancer patients. Tumor cell adhesion to the vascular endothelial cell lining is an important step in metastatic progression and is prompted by platelets. Mucin 1 is over-expressed and aberrantly glycosylated in more than 60% of pancreatic ductal adeno-carcinomas, which mediate adhesion of pancreatic cancer cells to platelets via P-selectin. The anticoagulant low molecular weight heparins (LMWHs), which are commonly used in venous Thromboprophylaxis and treatment, appear to have an effect on cancer survival. The aim of this study is to investigate the effect of platelets on human pancreatic cancer MPanc96 cell adhesion to the endothelial cell vessel wall, and to examine the effect of heparin derivatives on MPanc96 adhesion using a novel, in vitro model of human umbilical cord vein. The modified heparin S-NACH (sulfated non-anticoagulant heparin), which is devoid of antithrombin (AT) binding and devoid of inhibition of systemic AT-dependent coagulation factors such as factor Xa and IIa, and the LMWH tinzaparin both potently reduced adhesion and invasion of fluorescence-labeled MPanc96 cancer cells to the endothelial layer of umbilical cord vein in a dose-dependent manner. S-NACH effectively inhibited P-selectin mediated MPanc96 cell adhesion, and inhibited cell adhesion and invasion similar to tinzaparin, indicating that systemic anticoagulation is not a necessary component for heparin attenuation of cancer cell adhesion, invasion, and metastasis. Also, S-NACH and tinzaparin versus unfractionated heparin, heparin derivatives enoxaparin, deltaparin, fraxiparin, and fondaparinux were evaluated for their effect on platelet-cancer cell adhesion. An in vivo anti-metastatic S-NACH-treated nude mouse model of MPanc96 pancreatic cancer cell metastasis demonstrated potent anti-metastasis efficacy as evidenced by IVIS imaging and histological staining.
Collapse
|
33
|
Meyer G. [Cancer and venous thromboembolism]. ACTA ACUST UNITED AC 2011; 36 Suppl 1:S42-7. [PMID: 22177769 DOI: 10.1016/s0398-0499(11)70008-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cancer and venous thrombo-embolism is a frequent clinical association. Tumour cells activate the coagulation cascade. Surgery, chemotherapy and antiangiogenic agents are also associated with an increased risk of thrombosis. Current evidence does not suggest that a systematic screening for cancer after an unexplained thrombosis is associated with a clinical benefit. Risk factors for thrombosis specific to the cancer population have been identified. Recent studies suggest that prophylactic treatment may reduce the incidence of venous thrombo-embolism in patients with cancer. These results need to be confirmed. Treatment of venous thrombo-embolism in cancer patients is primarily based on low-molecular weight heparin administered for 3 or 6 months. Experimental data suggest that low-molecular weight heparin may also increase the survival of patients with cancer through a direct effect on tumour biology. Several clinical trials are underway to confirm this hypothesis.
Collapse
Affiliation(s)
- G Meyer
- Service de Pneumologie, Université Paris Descartes, Assistance Publique - Hôpitaux de Paris, Hôpital Européen Georges Pompidou, 20, rue Leblanc 75015 Paris, France.
| |
Collapse
|
34
|
Schlesinger M, Schmitz P, Zeisig R, Naggi A, Torri G, Casu B, Bendas G. The inhibition of the integrin VLA-4 in MV3 melanoma cell binding by non-anticoagulant heparin derivatives. Thromb Res 2011; 129:603-10. [PMID: 22099706 DOI: 10.1016/j.thromres.2011.10.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Revised: 10/07/2011] [Accepted: 10/24/2011] [Indexed: 10/15/2022]
Abstract
INTRODUCTION The integrin VLA-4-mediated binding is important for the metastatic dissemination of melanoma cells. Recently we found that heparin possesses a binding capacity to VLA-4. This could contribute to the heparin function to attenuate metastasis in a selectin-dependent manner. Aiming to a purposive, anti-adhesive heparin application, structural requirements of heparin for VLA-4 recognition have to be elucidated. MATERIALS AND METHODS A series of non-anticoagulant heparin derivatives were investigated concerning their inhibitory capacities for VLA-4 mediated binding of human melanoma MV3 cells to VCAM-1 under physiological flow conditions in vitro. A surface acoustic wave biosensor was applied to detect kinetic constants of selected derivatives binding to both, VLA-4 or P- and L-selectin. RESULTS Experimental metastasis of MV3 cells in mice confirmed the relevance of VLA-4 for metastatic dissemination. LMWHs (enoxaparin, tinzaparin) efficiently blocked VLA-4 cell binding, dominantly via the integrin`s α-chain. Desulfation at 2-O-position, N-acetylation or a size smaller than tetradecasaccharide disfavoured VLA-4 inhibition. Glycol-splitting of heparin and thus higher chain flexibility is a tolerable parameter. A derivative with 50% 6-O-desulfation appeared promising and exceeded tinzaparin in VLA-4 inhibition, both compounds displayed binding affinities to VLA-4 in the low micromolar range. CONCLUSIONS These findings provide structure-activity relationships for heparin VLA-4 binding, which partly differ from P- and L-selectin requirements. The data confirm that anti-coagulative and anti-adhesive function of heparin can be distinguished favouring applications of non-anticoagulant heparins in antimetastatic approaches without the risk of bleeding complications. The 50% 6-O-desulfated heparin-derivative appears promising to further evaluate the interference with selectin and VLA-4 binding functions in vivo.
Collapse
Affiliation(s)
- Martin Schlesinger
- Department of Pharmacy, Rheinische Friedrich Wilhelms University Bonn, 53121 Bonn, Germany.
| | | | | | | | | | | | | |
Collapse
|
35
|
Falanga A, Vignoli A, Diani E, Marchetti M. Comparative assessment of low-molecular-weight heparins in cancer from the perspective of patient outcomes and survival. Patient Relat Outcome Meas 2011; 2:175-88. [PMID: 22915978 PMCID: PMC3417933 DOI: 10.2147/prom.s10099] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Indexed: 11/23/2022] Open
Abstract
Patients with cancer are at high risk of developing venous thromboembolism (VTE), including deep venous thrombosis and pulmonary embolism. Compared to non-cancer patients, VTE in cancer is more frequently associated with clinical consequences, including recurrent VTE, bleeding, and an increase in the risk of death. Low-molecular-weight heparins (LMWHs) are commonly recommended for the prevention and treatment of VTE in cancer patients because of their favorable risk-to-benefit profile. Indeed, compared with vitamin K antagonists, LMWHs are characterized by a reduced need for coagulation monitoring, few major bleeding episodes, and once-daily dosing, which make these drugs more suitable in the cancer setting. Guidelines have been published recently with the aim to improve the clinical outcomes in cancer patients at risk of VTE and its complications. Coagulation activation in cancer may have a role not only in thrombosis but also in tumor growth and dissemination. Hence, inhibition of fibrin formation has been considered a possible tool against the progression of malignant disease. Clinical studies show that anticoagulant drugs may have a beneficial effect on survival in cancer patients, with a major role for LMWHs. Recently a number of prospective randomized clinical trials to test LMWHs to improve cancer survival as a primary endpoint in cancer patients have been conducted. Although the results are controversial, the interest in this research area remains high.
Collapse
Affiliation(s)
- Anna Falanga
- Division of Immunohematology and Transfusion Medicine, Ospedali Riuniti, Bergamo, Italy
| | - Alfonso Vignoli
- Division of Immunohematology and Transfusion Medicine, Ospedali Riuniti, Bergamo, Italy
| | - Erika Diani
- Division of Immunohematology and Transfusion Medicine, Ospedali Riuniti, Bergamo, Italy
| | - Marina Marchetti
- Division of Immunohematology and Transfusion Medicine, Ospedali Riuniti, Bergamo, Italy
| |
Collapse
|
36
|
Meyer G, Belmont L. [Cancer and venous thromboembolism]. Rev Mal Respir 2011; 28:443-52. [PMID: 21549900 DOI: 10.1016/j.rmr.2011.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Accepted: 09/29/2010] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Cancer and venous thromboembolism are frequently associated. STATE OF THE ART Venous thromboembolism is associated with a worse prognosis in patients with cancer. Thrombosis in cancer patients is related to the expression of tissue factor and other procoagulants by tumour cells. Surgery, chemotherapy and antiangiogenic agents are also associated with an increased risk of thrombosis. Venous thromboembolism may be the first manifestation of cancer, the risk being especially increased during the first six months following an unexplained episode of idiopathic thrombosis. Current evidence does not suggest that a systematic screening for cancer after an unexplained thrombosis is associated with a clinical benefit. Risk factors for thrombosis specific to the cancer population have been identified. A recent controlled trial suggests that low-molecular weight heparin may reduce the incidence of venous thromboembolism in patients with cancer. These results need to be confirmed. Treatment of venous thromboembolism in cancer patients is primarily based on low-molecular weight heparin administered for three or six months. PERSPECTIVES Low-molecular weight heparin may increase the survival of patients with cancer through a direct effect on tumour biology. Several clinical trials are underway to confirm this hypothesis. CONCLUSION Thrombosis in cancer patients is a frequent and difficult to treat condition. The role of long-term prophylaxis remains to be defined. The treatment of venous thromboembolism in cancer patients is primarily based on low-molecular weight heparin. Large clinical trials are currently assessing the effect of low-molecular weight heparin on the long-term survival of patients with cancer.
Collapse
Affiliation(s)
- G Meyer
- Service de pneumologie-soins intensifs, hôpital Européen Georges-Pompidou, AP-HP, faculté de médecine, université Paris-Descartes, 20, rue Leblanc, 75015 Paris, France.
| | | |
Collapse
|
37
|
Meyer G, Besse B, Friard S, Girard P, Corbi P, Azarian R, Monnet I, Alifano M, Chouaid C, Descourt R, Dennewald G, Taillade L, De Luca K, Giraud F, Pichon E, Chatellier G. Effet de la tinzaparine sur la mortalité du cancer bronchique non à petites cellules opéré. Rev Mal Respir 2011; 28:654-9. [DOI: 10.1016/j.rmr.2011.03.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 11/18/2010] [Indexed: 10/18/2022]
|
38
|
Stavik B, Skretting G, Sletten M, Sandset PM, Iversen N. Overexpression of both TFPIα and TFPIβ induces apoptosis and expression of genes involved in the death receptor pathway in breast cancer cells. Mol Carcinog 2010; 49:951-63. [PMID: 20886581 DOI: 10.1002/mc.20679] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Thrombosis is a major complication and an important cause of death in cancer patients. Tumor cells may trigger coagulation and induce a prothrombotic phenotype, which in return may enhance angiogenesis, tumor growth, and metastasis. Tissue factor pathway inhibitor (TFPI) has been reported to reduce tumor growth and metastasis in vivo and to induce apoptosis and inhibit proliferation in normal cells in vitro. However, no effect has so far been observed in cancer cells. We therefore aimed to characterize the functional effects of ectopic overexpression and endogenous downregulation of TFPI in cancer cells, and to elucidate possible mechanisms involved. The tumor derived breast cancer cells SK-BR-3 and Sum102 were used to construct stable cell lines overexpressing TFPIα and TFPIβ, and with TFPI knocked down, respectively. Effects of altered TFPI expression were evaluated by measuring apoptosis and proliferation of the cells, and gene expressions were analyzed using PCR arrays. Increased DNA fragmentation and Caspase 3 activity was observed in SK-BR-3 cells overexpressing TFPIα and TFPIβ, while a decrease in apoptosis was seen in Sum102 cells with TFPI expression knocked down. An increase and reduction in expression of pro- and anti-apoptotic genes, respectively, were seen in TFPI overexpressing cells, and the majority of the upregulated genes encoded proteins involved in the death receptor pathway, among them the death receptor ligand TNF-α. In conclusion, TFPIα and TFPIβ induced apoptosis in breast cancer cells and increased expression of apoptotic genes indicating a possible involvement of the death receptor pathway.
Collapse
Affiliation(s)
- Benedicte Stavik
- Department of Medical Genetics, Oslo University Hospital, Norway
| | | | | | | | | |
Collapse
|
39
|
Wang WJ. Acurhagin-C, an ECD disintegrin, inhibits integrin alphavbeta3-mediated human endothelial cell functions by inducing apoptosis via caspase-3 activation. Br J Pharmacol 2010; 160:1338-51. [PMID: 20590625 DOI: 10.1111/j.1476-5381.2010.00781.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND AND PURPOSE Acurhagin, a member of versatile metalloproteinase disintegrins from Agkistrodon acutus venom, has been identified as a platelet aggregation inhibitor, previously. Here, acurhagin-C, the C-terminal Glu-Cys-Asp (ECD)-containing fragment of acurhagin, was evaluated for its biological activities and potential applications in anti-angiogenic therapy. EXPERIMENTAL APPROACH Human umbilical vein endothelial cells (HUVECs) were treated with acurhagin-C to assay effects on viability, apoptosis, adhesion, migration, invasion, proliferation and angiogenesis. The recognition site and signalling involved for the interactions of acurhagin-C with HUVEC were determined using flow cytometric, electrophoresis and immunoblotting analyses. KEY RESULTS Acurhagin-C decreased viability and induced apoptosis in HUVEC. It also dose-dependently inhibited HUVEC adhesion to immobilized extracellular matrices fibronectin, collagen I and vitronectin with respective IC(50) values of approximately 0.6, 0.3 and 0.1 microM. Acurhagin-C prevented migration and invasion of HUVEC through vitronectin- and Matrigel-coated barriers respectively. Furthermore, acurhagin-C attenuated fibroblast growth factor-2-primed angiogenesis both in vitro and in vivo, and specifically blocked the binding of anti-alphavbeta3 monoclonal antibody 23C6 to HUVEC in an ECD-dependent manner. However, purified alphavbeta3 also dose-dependently bound to immobilized acurhagin and acurhagin-C with a saturable pattern. Interference with integrin alphavbeta3-mediated functions and promotion of caspase-3 activation by acurhagin-C affected morphology of HUVEC and induced apoptosis. CONCLUSIONS AND IMPLICATIONS Acurhagin-C elicited endothelial anoikis via disruption of alphavbeta3/focal adhesion kinase/phosphatidylinositol 3-kinase/Akt survival cascade and subsequent initiation of the procaspase-3 apoptotic signalling pathway.
Collapse
Affiliation(s)
- Wen-Jeng Wang
- Department of Nutrition and Health Sciences, Chang-Gung Institute of Technology, Kwei-Shan, Tao-Yuan, Taiwan.
| |
Collapse
|
40
|
Debergh I, Van Damme N, Pattyn P, Peeters M, Ceelen WP. The low-molecular-weight heparin, nadroparin, inhibits tumour angiogenesis in a rodent dorsal skinfold chamber model. Br J Cancer 2010; 102:837-43. [PMID: 20125158 PMCID: PMC2833243 DOI: 10.1038/sj.bjc.6605535] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 12/10/2009] [Accepted: 12/16/2009] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Recently, low-molecular-weight heparins (LMWHs) were found to confer a survival advantage in cancer patients. The mechanism underlying this observation is unclear, but may involve inhibition of tumour angiogenesis. We aimed to examine the effects of nadroparin on tumour angiogenesis using a dorsal skinfold window chamber model in the Syrian hamster. METHODS AMel-3 and HAP-T1 tumours were grown in donor animals and fragments implanted in the window chambers. Animals (N=46) were treated with 200 IU of nadroparin or saline for 10 days. Repeated intravital fluorescence microscopy was performed to calculate functional microcirculatory parameters: number (N) and length (L) of microvessels, vascular area fraction (AF), and red blood cell velocity (V). Microvessel density (MVD), fractal dimension, and pericyte coverage were assessed histologically. RESULTS Active angiogenesis was observed in control animals, resulting in a significant increase in N, L, and AF. In nadroparin-treated animals, however, N and L did not increase whereas AF decreased significantly. Both groups showed an initial increase in V, but nadroparin treatment resulted in an earlier decrease in red blood cell velocity over time. Compared with control animals, nadroparin-treated animals showed a significantly lower MVD and fractal dimension but significantly higher pericyte coverage index (PCI). CONCLUSIONS Taken together, these results suggest that the LMWH nadroparin inhibits tumour angiogenesis and results in microvessel normalisation.
Collapse
Affiliation(s)
- I Debergh
- Department of Surgery, University Hospital, Ghent B-9000, Belgium
| | - N Van Damme
- Department of Gastroenterology, University Hospital, Ghent B-9000, Belgium
| | - P Pattyn
- Department of Surgery, University Hospital, Ghent B-9000, Belgium
| | - M Peeters
- Department of Gastroenterology, University Hospital, Ghent B-9000, Belgium
| | - W P Ceelen
- Department of Surgery, University Hospital, Ghent B-9000, Belgium
- Senior Clinical Investigator of the Research Foundation – Flanders (Belgium) (FWO)
| |
Collapse
|
41
|
Robert F. The potential benefits of low-molecular-weight heparins in cancer patients. J Hematol Oncol 2010; 3:3. [PMID: 20074349 PMCID: PMC2830957 DOI: 10.1186/1756-8722-3-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 01/14/2010] [Indexed: 01/23/2023] Open
Abstract
Cancer patients are at increased risk of venous thromboembolism due to a range of factors directly related to their disease and its treatment. Given the high incidence of post-surgical venous thromboembolism in cancer patients and the poor outcomes associated with its development, thromboprophylaxis is warranted. A number of evidence-based guidelines delineate anticoagulation regimens for venous thromboembolism treatment, primary and secondary prophylaxis, and long-term anticoagulation in cancer patients. However, many give equal weight to several different drugs and do not make specific recommendations regarding duration of therapy. In terms of their efficacy and safety profiles, practicality of use, and cost-effectiveness the low-molecular-weight heparins are at least comparable to, and offer several advantages over, other available antithrombotics in cancer patients. In addition, data are emerging that the antithrombotics, and particularly low-molecular-weight heparins, may exert an antitumor effect which could contribute to improved survival in cancer patients when given for long-term prophylaxis. Such findings reinforce the importance of thromboprophylaxis with low-molecular-weight heparin in cancer patients.
Collapse
Affiliation(s)
- Francisco Robert
- Department of Medicine, Division of Hematology/Oncology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 352943300, USA.
| |
Collapse
|
42
|
The influence of unfractionated and low-molecular weight heparins on the properties of human umbilical vein endothelial cells (HUVEC). Folia Histochem Cytobiol 2009; 47:17-23. [DOI: 10.2478/v10042-009-0008-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
43
|
Borensztajn K, Aberson H, Peppelenbosch MP, Spek CA. FXa-induced intracellular signaling links coagulation to neoangiogenesis: potential implications for fibrosis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:798-805. [PMID: 19339215 DOI: 10.1016/j.bbamcr.2009.01.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Revised: 01/14/2009] [Accepted: 01/15/2009] [Indexed: 02/06/2023]
Abstract
Fibrosis represents the end-stage of a broad range of disorders affecting organ function. These disorders are often associated with aberrant angiogenesis, but whether vascular abnormalities during fibrosis are characterized by excessive or diminished neo-vascularization remains questionable. Strikingly, activation of the coagulation cascade is frequently observed in association with the progression of fibroproliferative disorders. As we recently showed that coagulation factor (F)Xa induced fibrotic responses in fibroblasts, we hypothesized that FXa might indirectly induce angiogenesis by triggering fibroblasts to secrete proangiogenic factors. In the present study, we show that although FXa induces p42/44 MAP Kinase phosphorylation in endothelial cells, it has no direct effect on endothelial cell proliferation, protein synthesis and tube formation. In contrast, conditioned medium of fibroblasts stimulated with FXa enhanced endothelial cell proliferation, extra cellular matrix synthesis, wound healing and endothelial tube formation. FXa induced VEGF production by fibroblasts and a VEGF neutralizing antibody blocked the indirect effect of FXa on proliferation and realignment of endothelial cells identifying VEGF as a crucial player in angiogenesis during coagulation factor-induced fibrosis. Overall, our results establish a link between the coagulation cascade and angiogenesis during fibrosis.
Collapse
Affiliation(s)
- Keren Borensztajn
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, A. Deusinglaan 1, NL-9713 AV Groningen, The Netherlands.
| | | | | | | |
Collapse
|
44
|
Borensztajn K, Peppelenbosch MP, Spek CA. Factor Xa: at the crossroads between coagulation and signaling in physiology and disease. Trends Mol Med 2008; 14:429-40. [DOI: 10.1016/j.molmed.2008.08.001] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Revised: 08/01/2008] [Accepted: 08/01/2008] [Indexed: 01/16/2023]
|
45
|
Heineke J, Auger-Messier M, Xu J, Oka T, Sargent MA, York A, Klevitsky R, Vaikunth S, Duncan SA, Aronow BJ, Robbins J, Crombleholme TM, Cromblehol TM, Molkentin JD. Cardiomyocyte GATA4 functions as a stress-responsive regulator of angiogenesis in the murine heart. J Clin Invest 2008; 117:3198-210. [PMID: 17975667 DOI: 10.1172/jci32573] [Citation(s) in RCA: 190] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Accepted: 09/12/2007] [Indexed: 12/20/2022] Open
Abstract
The transcription factor GATA4 is a critical regulator of cardiac gene expression, modulating cardiomyocyte differentiation and adaptive responses of the adult heart. We report what we believe to be a novel function for GATA4 in murine cardiomyocytes as a nodal regulator of cardiac angiogenesis. Conditional overexpression of GATA4 within adult cardiomyocytes increased myocardial capillary and small conducting vessel densities and increased coronary flow reserve and perfusion-dependent cardiac contractility. Coculture of HUVECs with either GATA4-expressing cardiomyocytes or with myocytes expressing a dominant-negative form of GATA4 enhanced or reduced HUVEC tube formation, respectively. Expression of GATA4 in skeletal muscle by adenoviral gene transfer enhanced capillary densities and hindlimb perfusion following femoral artery ablation. Deletion of Gata4 specifically from cardiomyocytes reduced myocardial capillary density and prevented pressure overload-augmented angiogenesis in vivo. GATA4 induced the angiogenic factor VEGF-A, directly binding the Vegf-A promoter and enhancing transcription. GATA4-overexpressing mice showed increased levels of cardiac VEGF-A, while Gata4-deleted mice demonstrated decreased VEGF-A levels. The induction of HUVEC tube formation in GATA4-overexpressing cocultured myocytes was blocked with a VEGF receptor antagonist. Pressure overload-induced dysfunction in Gata4-deleted hearts was partially rescued by adenoviral gene delivery of VEGF and angiopoietin-1. To our knowledge, these results demonstrate [corrected] a previously unrecognized function for GATA4 as a regulator of cardiac angiogenesis through a nonhypoxic, load, and/or disease-responsive mechanism.
Collapse
Affiliation(s)
- Joerg Heineke
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio 45229, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hostettler N, Naggi A, Torri G, Ishai-Michaeli R, Casu B, Vlodavsky I, Borsig L. P-selectin- and heparanase-dependent antimetastatic activity of non-anticoagulant heparins. FASEB J 2007; 21:3562-72. [PMID: 17557930 DOI: 10.1096/fj.07-8450com] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Vascular cell adhesion molecules, P- and L-selectins, facilitate metastasis of cancer cells in mice by mediating interactions with platelets, endothelium, and leukocytes. Heparanase is an endoglycosidase that degrades heparan sulfate of extracellular matrix, thereby promoting tumor invasion and metastasis. Heparin is known to efficiently attenuate metastasis in different tumor models. Here we identified modified, nonanticoagulant species of heparin that specifically inhibit selectin-mediated cell-cell interactions, heparanase enzymatic activity, or both. We show that selective inhibition of selectin interactions or heparanase with specific heparin derivatives in mouse models of MC-38 colon carcinoma and B16-BL6 melanoma attenuates metastasis. Selectin-specific heparin derivatives attenuated metastasis of MC-38 carcinoma, but heparanase-specific derivatives had no effect, in accordance with the virtual absence of heparanase activity in these cells. Heparin derivatives had no further effect on metastasis in mice deficient in P- and L-selectin, indicating that selectins are the primary targets of heparin antimetastatic activity. Selectin-specific and heparanase-specific derivatives attenuated metastasis of B16-BL6 melanomas to a similar extent. When mice were injected with a derivative containing both heparanase and selectin inhibitory activity, no additional attenuation of metastasis could be observed. Thus, selectin-specific heparin derivatives efficiently attenuated metastasis of both tumor cell types whereas inhibition of heparanase led to reduction of metastasis only in tumor cells producing heparanase.
Collapse
Affiliation(s)
- Nina Hostettler
- Zürich Center for Integrative Human Physiology, Institute of Physiology, University of Zürich, Zürich, Switzerland
| | | | | | | | | | | | | |
Collapse
|
47
|
Niers TMH, Klerk CPW, DiNisio M, Van Noorden CJF, Büller HR, Reitsma PH, Richel DJ. Mechanisms of heparin induced anti-cancer activity in experimental cancer models. Crit Rev Oncol Hematol 2006; 61:195-207. [PMID: 17074500 DOI: 10.1016/j.critrevonc.2006.07.007] [Citation(s) in RCA: 145] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2006] [Revised: 07/28/2006] [Accepted: 07/28/2006] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Retrospective analyses of clinical trials and prospective clinical studies have suggested that heparins may have an effect on cancer survival. This putative anti-cancer activity of heparins is supported by data from studies in animal tumour models. OBJECTIVE To clarify the various potential mechanisms of heparin anti-cancer activity we evaluated the data from pre-clinical studies in which heparins have been tested as anti-cancer therapy. METHODS Pre-clinical studies, published between 1960 and 2005 were assessed. Data were collected on the type and dose of heparin used, duration of exposure to heparin, interval between heparin administration and cancer cell inoculation, and the animal tumour model used. In addition, a distinction was made in the analysis between heparin effects on the primary tumour or on established metastases and effects on the metastatic potential of infused cells. RESULTS Heparins seemed to affect the formation of metastasis rather than the growth of primary tumours. Chemically modified heparins with no or limited anticoagulant activity also showed anti-metastatic properties. Possible mechanisms to explain the effects on the process of metastases include inhibition of blood coagulation, inhibition of cancer cell-platelet and -endothelial interactions by selectin inhibition and inhibition of cell invasion and angiogenesis. CONCLUSION The anti-cancer activity of heparins depends more on inhibition of metastasis formation than on the effects on primary tumour growth. These effects are probably related to both coagulation and non-coagulation dependent factors. For a definitive proof of the anti-cancer activity of heparins in the clinic, prospective randomized trials especially in patients with early metastatic disease or in the adjuvant setting are urgently needed.
Collapse
Affiliation(s)
- T M H Niers
- Department of Medical Oncology, Academic Medical Centre, University of Amsterdam F4-223, Meibergdreef 9, 1100 DE Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
48
|
Dahm AEA, Iversen N, Birkenes B, Ree AH, Sandset PM. Estrogens, selective estrogen receptor modulators, and a selective estrogen receptor down-regulator inhibit endothelial production of tissue factor pathway inhibitor 1. BMC Cardiovasc Disord 2006; 6:40. [PMID: 17029634 PMCID: PMC1609184 DOI: 10.1186/1471-2261-6-40] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2006] [Accepted: 10/09/2006] [Indexed: 11/10/2022] Open
Abstract
Background Hormone therapy, oral contraceptives, and tamoxifen increase the risk of thrombotic disease. These compounds also reduce plasma content of tissue factor pathway inhibitor-1 (TFPI), which is the physiological inhibitor of the tissue factor pathway of coagulation. The current aim was to study if estrogens and estrogen receptor (ER) modulators may inhibit TFPI production in cultured endothelial cells and, if so, identify possible mechanisms involved. Methods Human endothelial cell cultures were treated with 17β-estradiol (E2), 17α-ethinylestradiol (EE2), tamoxifen, raloxifene, or fulvestrant. Protein levels of TFPI in cell media and cell lysates were measured by an enzyme-linked immunosorbent assay, and TFPI mRNA levels were assessed by quantitative PCR. Expression of ERα was analysed by immunostaining. Results All compounds (each in a concentration of 10 nM) reduced TFPI in cell medium, by 34% (E2), 21% (EE2), 16% (tamoxifen), and 28% (raloxifene), respectively, with identical inhibitory effects on cellular TFPI levels. Expression of TFPI mRNA was principally unchanged. Treatment with fulvestrant, which was also associated with down-regulation of secreted TFPI (9% with 10 nM and 26% with 1000 nM), abolished the TFPI-inhibiting effect of raloxifene, but not of the other compounds. Notably, the combination of 1000 nM fulvestrant and 10 nM raloxifene increased TFPI secretion, and, conversely, 10 nM of either tamoxifen or raloxifene seemed to partly (tamoxifen) or fully (raloxifene) counteract the inhibitory effect of 1000 nM fulvestrant. The cells did not express the regular nuclear 66 kDa ERα, but instead a 45 kDa ERα, which was not regulated by estrogens or ER modulators. Conclusion E2, EE2, tamoxifen, raloxifene, and fulvestrant inhibited endothelial production of TFPI by a mechanism apparently independent of TFPI transcription.
Collapse
Affiliation(s)
- Anders E A Dahm
- Department of Haematology, Ullevål University Hospital, 0407 Oslo, Norway.
| | | | | | | | | |
Collapse
|
49
|
Mousa SA, Feng X, Xie J, Du Y, Hua Y, He H, O'Connor L, Linhardt RJ. Synthetic oligosaccharide stimulates and stabilizes angiogenesis: structure-function relationships and potential mechanisms. J Cardiovasc Pharmacol 2006; 48:6-13. [PMID: 16954815 PMCID: PMC4140568 DOI: 10.1097/01.fjc.0000238591.90062.62] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
To determine the proangiogenesis effect of series of saccharides and a synthetic oligosaccharide and potential mechanisms, an in vitro 3-dimensional endothelial cell sprouting (3D-ECS) assay and the chick chorioallantoic membrane (CAM) model were used. We demonstrated that a sulfated oligosaccharide significantly promotes the endothelial capillary network initiated by vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (b-FGF). Furthermore, although the capillary network initiated by VEGF and b-FGF lasts no more than 7 days, addition of a sulfated oligosaccharide significantly amplifies angiogenesis and stabilizes the capillary network of new blood vessels. In the CAM model, sulfated oligosaccharide also stimulated angiogenesis. In both the CAM and the 3D-ECS assay, structure-function studies reveal that increased saccharide chain length up to the hexa- to decasaccharide show optimal proangiogenesis efficacy. In addition, the sulfation and molecular shape (branched vs linear) of oligosaccharide are important for sustained proangiogenesis efficacy. Data indicate that chemically defined synthetic oligosaccharides can play an important role in regulation of capillary structure and stability, which may contribute to future advances in therapeutic angiogenesis. The proangiogenesis efficacy of an oligosaccharide is mediated via integrin alphavbeta3 and involves mitogen-activated protein kinase signaling mechanisms.
Collapse
Affiliation(s)
- S A Mousa
- Pharmaceutical Research Institute at Albany and Albany College of Pharmacy, Albany, NY 12208, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Negaard HFS, Eilertsen AL, Dahm A, Iversen PO, Østenstad B, Sandset PM. Decreased Lung Cancer Survival With Hormone-Replacement Therapy: Caused by a Decreased Tissue Factor Pathway Inhibitor Level? J Clin Oncol 2006; 24:2683-4; author reply 2684. [PMID: 16763285 DOI: 10.1200/jco.2006.06.4345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|