Skip to main content Accessibility help
×
Hostname: page-component-7479d7b7d-fwgfc Total loading time: 0 Render date: 2024-07-12T17:06:18.015Z Has data issue: false hasContentIssue false

48 - Role of Platelets and Thrombin in Metastasis

from PART II - CLINICAL RESEARCH

Published online by Cambridge University Press:  05 June 2012

Boris Kobrinsky
Affiliation:
New York University School of Medicine, United States
Simon Karpatkin
Affiliation:
New York University School of Medicine, United States
David L. Green
Affiliation:
New York University School of Medicine, United States
David Lyden
Affiliation:
Weill Cornell Medical College, New York
Danny R. Welch
Affiliation:
Weill Cornell Medical College, New York
Bethan Psaila
Affiliation:
Imperial College of Medicine, London
Get access

Summary

In response to vascular injury, platelets become adherent and undergo activation and aggregation. The formation of the primary hemostatic platelet plug occurs simultaneously with surface activation of coagulation, leading to thrombin generation and fibrin formation. Platelet function also contributes to thrombus formation in pathologic settings, leading to vascular occlusion, usually in the setting of underlying vascular disease. Platelets contain many biologically active mediators that are released upon activation, including growth factors, coagulation factors, adhesive ligands, proteases, heparanase, cytokines, chemokines, and vasoactive lipids. Other functions of platelets include a supportive role in vascular maintenance and regulation of angiogenesis, as well as putative roles in inflammation and immunity. Indeed, platelets are now linked to such diverse physiologic and pathologic processes as wound healing and tissue regeneration, response to microbial infection, inflammatory diseases, atherogenesis, tumorigenesis, and metastasis.

Platelets contribute to tumor cell metastasis by mediating tumor cell adhesion and subsequent extravasation, stabilizing platelet–tumor cell emboli in the circulation, and protecting tumor cells from the host immune system. In the tumor microenvironment, platelets become activated and may release growth factors, chemokines, matrix metalloproteinases (MMPs), and inflammatory mediators, with resulting production and remodeling of the extracellular matrix and tumor angiogenesis. In this chapter, we review the role of platelets and thrombin in metastasis. We review clinical trial data with aspirin and anticoagulants in cancer and cancer prevention, and speculate on the potential effect of thrombin in unmasking tumor dormancy.

Type
Chapter
Information
Cancer Metastasis
Biologic Basis and Therapeutics
, pp. 552 - 562
Publisher: Cambridge University Press
Print publication year: 2011

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Lindenmeyer, F, Legrand, Y, Menashi, S et al. (1997) Upregulation of MMP-9 expression in MDA-MB231 tumor cells by platelet granular membrane. FEBS Lett. 418(1–2): 19–22.CrossRefGoogle ScholarPubMed
Ambrus, JL, Ambrus, CM, Mink, IB, Pickren, JW (1975) Causes of death in cancer patients. J Med. 6(1): 61–4.Google ScholarPubMed
Nierodzik, ML, Karpatkin, S (2006) Thrombin induces tumor growth, metastasis, and angiogenesis: Evidence for a thrombin-regulated dormant tumor phenotype. Cancer Cell. 10(5): 355–62.CrossRefGoogle ScholarPubMed
Prandoni, P, Lensing, AW, Piccioli, A et al. (2002) Recurrent venous thromboembolism and bleeding complications during anticoagulant treatment in patients with cancer and venous thrombosis. Blood. 100(10): 3484–8.CrossRefGoogle ScholarPubMed
Sorensen, HT, Mellemkjaer, L, Olsen, JH, Baron, JA (2000) Prognosis of cancers associated with venous thromboembolism. N Engl J Med. 343(25): 1846–50.CrossRefGoogle ScholarPubMed
Levitan, N, Dowlati, A, Remick, SC et al. (1999) Rates of initial and recurrent thromboembolic disease among patients with malignancy versus those without malignancy. Risk analysis using Medicare claims data. Medicine (Baltimore). 78(5): 285–91.CrossRefGoogle ScholarPubMed
Shulman, S, Lindmarker, P (2000) Incidence of cancer after prophylaxis with warfarin against recurrent venous thromboembolism. Duration of anti-coagulation trial. N Engl J Med. 3432: 1953.CrossRefGoogle Scholar
Trousseau, A (1865) Plegmasie alba dolens. Clinique Medical de Hotel-Dieu de Paris, London. New Syndenham Soc. 3: 94.Google Scholar
Billroth, T (1877–8) Lectures on surgical pathology and therapeutics: a handbook for students and practitioners. New Syndenham Soc.
Lee, AY, Levine, MN (2003) Venous thromboembolism and cancer: risks and outcomes. Circulation. 107(23 Suppl 1): I17–21.CrossRefGoogle ScholarPubMed
Sack, JGH, Levin, J, Bell, WR (1977) Trousseau's syndrome and other manifestations of chronic disseminated coagulopathy in patients with neoplasms: Clinical, pathophysiologic and therapeutic features. Medicine. 56: 1–37.CrossRefGoogle ScholarPubMed
Rickles, F (2006) Mechanisms of cancer-induced thrombosis in cancer. Pathophysiol Haemost Thromb. 35(1–2): 103–10.CrossRefGoogle Scholar
Levin, J, Conley, CL (1964) Thrombocytosis associated with malignant disease. Arch Intern Med. 114: 497–500.CrossRefGoogle ScholarPubMed
Sun, NC, McAfee, WM, Hum, GJ, Weiner, JM (1979) Hemostatic abnormalities in malignancy, a prospective study of one hundred eight patients. Part I. Coagulation studies. Am J Clin Pathol. 71(1): 10–16.CrossRefGoogle ScholarPubMed
Nash, GF, Turner, LF, Scully, MF, Kakkar, AK (2002) Platelets and cancer. Lancet Oncol. 3(10): 591–2.CrossRefGoogle ScholarPubMed
Boneu, B, Bugat, R, Boneu, A et al. (1984) Exhausted platelets in patients with malignant solid tumors without evidence of active consumption coagulopathy. Eur J Cancer Clin Oncol. 20: 899–903.CrossRefGoogle ScholarPubMed
Lyman, GH, Bettigole, RE, Robson, E, Ambrus, JL, Urban, H (1978) Fibrinogen kinetics in patients with neoplastic disease. Cancer. 41(3): 1113–22.3.0.CO;2-D>CrossRefGoogle ScholarPubMed
Rickles, F, Edwards, R (1983) Activation of blood coagulation in cancer: Trousseau's syndrome revisited. Blood. 62: 14–31.Google ScholarPubMed
Gale, AJ, Gordon, SG (2001) Update on tumor cell procoagulant factors. Acta Haematol. 106: 25–32.CrossRefGoogle ScholarPubMed
Hanahan, D, Weinberg, RA (2000) The hallmarks of cancer. Cell. 100(1): 57–70.CrossRefGoogle ScholarPubMed
Sporn, MB (1996) The war on cancer. Lancet. 347(9012): 1377–81.CrossRefGoogle ScholarPubMed
Fidler, I (1970) Metastases: quantitative analysis of distribution and fate of tumor emboli labeled with 125I-5-iodo-2′-deoxyuridine. J Natl Cancer Inst. 45: 773–82.Google Scholar
Gasic, G, Gasic, T, Stewart, C (1968) Antimetastatic effects associated with platelet reduction. Proc Natl Acad Sci USA. 61: 46–52.CrossRefGoogle ScholarPubMed
Pearlstein, E, Ambrogio, C, Karpatkin, S (1984) Effect of anti-platelet antibody on the development of pulmonary metastases following injection of CT26 colon adenocarcinoma, Lewis lung carcinoma and B16 amelanotic melanoma tumor cells in mice. Cancer Res. 44: 3884–7.Google Scholar
Wood, S (1964) Experimental studies of the intravascular dissemination of ascetic V2 carcinoma cells in the rabbit with special reference to fibrinogen and fibrinolytic agents. Bull Schweiz Med Wiss. 20: 92.Google Scholar
Jones, J, Wallace, A, Fraser, E (1971) Sequence of events in experimental and electron microscopic observations. J Natl Cancer Inst. 46: 493–504.Google Scholar
Sindelar, W, Tralka, T, Ketcham, A (1975) Electron microscope observations on formation of pulmonary metastases. J Surg Res. 18: 137–61.CrossRefGoogle Scholar
Gasic, GJ, Gasic, TB, Galanti, N, Johnson, T, Murphy, S (1973) Platelet-tumor-cell interactions in mice. The role of platelets in the spread of malignant disease. Int J Cancer. 11(3): 704–18.CrossRefGoogle ScholarPubMed
Pearlstein, E, Salk, PL, Yogeeswaran, G, Karpatkin, S (1980) Correlation between spontaneous metastatic potential, platelet-aggregating activity of cell surface extracts, and cell surface sialylation in 10 metastatic-variant derivatives of a rat renal sarcoma cell line. Proc Natl Acad Sci USA. 77(7): 4336–9.CrossRefGoogle ScholarPubMed
Camerer, E, Qazi, A, Duong, D, Cornelissen, I, Rommel, A, Coughlin, S (2004) Platelets, protease-activated receptors, and fibrinogen in hematogenous metastasis. Blood. 104: 397–401.CrossRefGoogle ScholarPubMed
Crissman, JD, Hatfield, JS, Menter, DG, Sloane, B, Honn, KV (1988) Morphological study of the interaction of intravascular tumor cells with endothelial cells and subendothelial matrix. Cancer Res. 48(14): 4065–72.Google ScholarPubMed
Nieswandt, B, Hafner, M, Echtenacher, B, Mannel, D (1999) Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res. 59: 1295–300.Google ScholarPubMed
Palumbo, JS, Talmage, KE, Massari, JV et al. (2005) Platelets and fibrin(ogen) increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood. 105(1): 178–85.CrossRefGoogle ScholarPubMed
Mehta, P (1984) Potential role of platelets in the pathogenesis of tumor metastasis. Blood. 63(1): 55–63.Google ScholarPubMed
Karpatkin, S, Ambrogio, C, Pearlstein, E (1984) Lack of effect of in vivo prostacyclin on the development of pulmonary metastases in mice following intravenous injection of CT26 colon carcinoma, Lewis lung carcinoma, or B16 amelanotic melanoma cells. Cancer Res. 44(9): 3880–3.Google ScholarPubMed
Karpatkin, S, Pearlstein, E, Ambrogio, C, Coller, BS (1988) Role of adhesive proteins in platelet tumor interaction in vitro and metastasis formation in vivo. J Clin Invest. 81: 1012–9.CrossRefGoogle ScholarPubMed
Trikha, M, Zhou, Z, Timar, J et al. (2002) Multiple roles for platelet GPIIb/IIIa and alphavbeta3 integrins in tumor growth, angiogenesis, and metastasis. Cancer Res. 62(10): 2824–33.Google Scholar
Bakewell, SJ, Nestor, P, Prasad, S et al. (2003) Platelet and osteoclast beta3 integrins are critical for bone metastasis. Proc Natl Acad Sci USA. 100(24): 14205–10.CrossRefGoogle ScholarPubMed
Morimoto, K, Satoh-Yamaguchi, K, Hamaguchi, A, Inoue, Y et al. (2008) Interaction of cancer cells with platelets mediated by Necl-5/poliovirus receptor enhances cancer cell metastasis to the lungs. Oncogene. 27(3):264–73.CrossRefGoogle ScholarPubMed
Kojima, H, Kanada, H, Shimizu, S, Kasama, E et al. (2003) CD226 mediates platelet and megakaryocytic cell adhesion to vascular endothelial cells. J Biol Chem 278(38):36748–53.CrossRefGoogle ScholarPubMed
Kim, Y, Borsig, L, Varki, N, Varki, A (1998) P-selectin deficiency attenuates tumor growth and metastasis. Proc Natl Acad Sci USA. 95:9325–30.CrossRefGoogle ScholarPubMed
McCarty, O, Mousa, S, Bray, P, Konstantopoulos, K (2000) Immobilized platelets support human colon carcinoma cell tethering, rolling, and firm adhesion under dynamic flow conditions. Blood. 96:1789–97.Google ScholarPubMed
Borsig, L, Wong, R, Feramisco, J, Nadeau, DR, Varki, NM, Varki, A (2001) Heparin and cancer revisited: mechanistic connections involving platelets, P-selectin, carcinoma mucins, and tumor metastasis. Proc Natl Acad Sci USA. 98(6): 3352–7.CrossRefGoogle ScholarPubMed
Biggerstaff, J, Seth, N, Amirkhosravi, A et al. (1999) Soluble fibrin augments platelet/tumor cell adherence in vitro and in vivo, and enhances experimental metastasis. Clin Exper Metastasis. 17: 723–30.CrossRefGoogle ScholarPubMed
Felding-Habermann, B, Habermann, R, Saldivar, E, Ruggeri, ZM (1996) Role of beta3 integrins in melanoma cell adhesion to activated platelets under flow. J Biol Chem. 271: 5892–900.CrossRefGoogle ScholarPubMed
Terranova, V, Williams, J, Liotta, L (1984) Modulation of metastatic activity of melanoma cells by laminin and fibronectin. Science. 226: 982–4.CrossRefGoogle ScholarPubMed
Cheresh, D, Smith, W, Cooper, H et al. (1989) A novel vitronectin receptor integrin (avbx) is responsible for distinct adhesive properties of carcinoma cells. Cell. 57: 59–69.CrossRefGoogle Scholar
Kramer, R, Marks, N (1989) Identification of integrin collagen receptors on human melanoma cells. J Biol Chem. 264: 4684–8.Google ScholarPubMed
Roberts, D, Sherwood, J, Ginsburg, V (1987) Platelet thrombospondin mediates attachment and spreading of human melanoma cells. J Cell Biol. 104: 131–9.CrossRefGoogle ScholarPubMed
Klepfish, A, Greco, MA, Karpatkin, S (1993) Thrombin stimulates melanoma tumor-cell binding to endothelial cells and subendothelial matrix. Int J Cancer. 53(6): 978–82.CrossRefGoogle ScholarPubMed
Carney, D, Stiernberg, J, Fenton, J (1984) Initiation of proliferative events by human α-thrombin requires both receptor binding and enzymatic activity. J Cell Biochem. 26: 181–95.CrossRefGoogle Scholar
Chen, L, Buchanan, J (1975) Mitogenic activity of blood components. I. Thrombin and prothrombin. Proc Natl Acad Sci USA. 72: 131–5.CrossRefGoogle ScholarPubMed
Gospodarowicz, D, Brown, K, Birdwell, C, Zetter, B (1978) Control of proliferation of human vascular endothelial cells. Characterization of the response of human umbilical vein endothelial cells to fibroblast growth factor, epidermal growth factor, and thrombin. J Cell Biol. 77: 774–88.CrossRefGoogle ScholarPubMed
Caunt, M, Huang, YQ, Brooks, PC, Karpatkin, S (2003) Thrombin induces neoangiogenesis in the chick chorioallantoic membrane. J Thromb Haemost. 1(10): 2097–102.CrossRefGoogle ScholarPubMed
Nierodzik, M, Plotkin, A, Kajumo, F, Karpatkin, S (1991) Thrombin stimulates tumor-platelet adhesion in vitro and metastasis in vivo. J Clin Invest. 87: 229–36.CrossRefGoogle ScholarPubMed
Dardik, R, Savion, N, Kaufmann, Y, Varon, D (1998) Thrombin promotes platelet-mediated melanoma cell adhesion to endothelial cells under flow conditions: role of platelet glycoproteins P-selectin and GPIIb-IIIa. Br J Cancer. 77: 2069–75.CrossRefGoogle ScholarPubMed
Liu, TY, Zhao, F, Gu, W et al. (2009) The roles of platelet GPIIb/IIIa and αvβ3 integrins during HeLa cells adhesion, migration, and invasion to monolayer endothelium under static and dynamic shear flow. J Biomed Biotechnol. 2009: 829243. Epub 2009 Oct 28. Published online 2009 October 28. doi: 10.1155/2009/829243.CrossRefGoogle ScholarPubMed
Holmes, CE, Levis, JE, Ornstein, DL (2009) Activated platelets enhance ovarian cancer cell invasion in a cellular model of metastasis. Clin Exp Metastasis. 26: 653–661.CrossRefGoogle Scholar
Nierodzik, ML, Bain, RM, Liu, L-X, Shivji, M, Takeshita, K, Karpatkin, S (1996) Presence of the seven transmembrane thrombin receptor on human tumour cells: effect of activation on tumour adhesion to platelets and tumour tyrosine phosphorylation. Br J Haematol. 92: 452–7.CrossRefGoogle ScholarPubMed
Even-Ram, S, Uziely, B, Cohen, P et al. (1998) Thrombin receptor overexpression in malignant and physiological invasion processes. Nat Med. 4: 909–14.CrossRefGoogle ScholarPubMed
Shi, X, Gangadharan, B, Brass, LF, Ruf, W, Mueller, BM (2004) Protease-activated receptors (PAR1 and PAR2) contribute to tumor cell motility and metastasis. Mol Cancer Res. 2(7): 395–402.Google ScholarPubMed
Nierodzik, M, Chen, K, Takeshita, K et al. (1998) Protease-activated receptor 1 (PAR-1) is required and rate-limiting for thrombin-enhanced experimental pulmonary metastasis. Blood. 92(10): 3694–700.Google ScholarPubMed
Caunt, M, Hu, L, Tang, T, Brooks, PC, Ibrahim, S, Karpatkin, S (2006) Growth-regulated oncogene is pivotal in thrombin-induced angiogenesis. Cancer Res. 66(8): 4125–32.CrossRefGoogle ScholarPubMed
Hu, L, Roth, JM, Brooks, P, Ibrahim, S, Karpatkin, S (2008) Twist is required for thrombin-induced tumor angiogenesis and growth. Cancer Res. 68(11): 4296–302.CrossRefGoogle Scholar
Yang, J, Mani, SA, Donaher, JL et al. (2004) Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 117(7): 927–39.CrossRefGoogle ScholarPubMed
Hu, L, Roth, JM, Brooks, P, Luty, J, Karpatkin, S (2008) Thrombin up-regulates cathepsin D which enhances angiogenesis, growth, and metastasis. Cancer Res. 68(12):4666–73.CrossRefGoogle ScholarPubMed
Hu, L, Lee, M, Campbell, W, Perez-Soler, R, Karpatkin, S (2004) Role of endogenous thrombin in tumor implantation, seeding and spontaneous metastasis. Blood. 104: 2746–51.CrossRefGoogle ScholarPubMed
Mueller, B, Reisfeld, R, Edgington, T, Ruf, W (1992) Expression of tissue factor by melanoma cells promotes efficient hematogenous metastasis. Proc Natl Acad Sci USA. 89: 832–6.CrossRefGoogle ScholarPubMed
Fisher, E, Ruf, W, Mueller, B (1995) Tissue factor-initiated thrombin generation activates the signaling thrombin receptor on malignant melanoma cells. Cancer Res. 55: 1629–32.Google Scholar
Zacharski, L, Memoli, V, Morain, W, Schlaeppi, J-M, Rousseau, S (1995) Cellular localization of enzymatically-active thrombin in intact tissue by hirudin binding. Thromb Haemostas. 73: 793–7.Google ScholarPubMed
Kwaan, HC, McMahon, B (2009) The role of plasminogen-plasmin system in cancer. Cancer Treat Res. 148: 43–66.CrossRefGoogle Scholar
Palumbo, JS, Kombrinck, KW, Drew, AF et al. (2000) Fibrinogen is an important determinant of the metastatic potential of circulating tumor cells. Blood. 96(10): 3302–9.Google ScholarPubMed
Offermanns, S, Toombs, F, Hu, Y-H, Simon, MI (1997) Defective platelet activation in Gαq-deficient mice. Nature. 389: 183–6.CrossRefGoogle Scholar
Terraube, V, Pendu, R, Baruch, D et al. (2006) Increased metastatic potential of tumor cells in von Willebrand factor-deficient mice. J Thromb Haemost. 4: 519–26.CrossRefGoogle ScholarPubMed
Spaet, TH (1952) Vascular factors in the pathogenesis of hemorrhagic syndromes. Blood. 7(6): 641–52.Google ScholarPubMed
Gimbrone, MA Jr, Aster, RH, Cotran, RS, Corkery, J, Jandl, JH, Folkman, J (1969) Preservation of vascular integrity in organs perfused in vitro with a platelet-rich medium. Nature. 222(5188): 33–6.CrossRefGoogle ScholarPubMed
Pintucci, G, Froum, S, Pinnell, J, Mignatti, P, Rafii, S, Green, D (2002) Trophic effects of platelets on cultured endothelial cells are mediated by platelet-associated fibroblast growth factor-2 (FGF-2) and vascular endothelial growth factor (VEGF). Thromb Haemost. 88(5): 834–42.CrossRefGoogle ScholarPubMed
Pipili-Synetos, E, Papadimitriou, E, Maragoudakis, ME (1998) Evidence that platelets promote tube formation by endothelial cells on matrigel. Br J Pharmacol. 125(6): 1252–7.CrossRefGoogle ScholarPubMed
Liu, Y, Wada, R, Yamashita, T et al. (2000) Edg-1, the G protein-coupled receptor for sphingosine-1-phosphate, is essential for vascular maturation. J Clin Invest. 106(8): 951–61.CrossRefGoogle Scholar
Paik, JH, Skoura, A, Chae, SS et al. (2004) Sphingosine 1-phosphate receptor regulation of N-cadherin mediates vascular stabilization. Genes Dev. 18(19): 2392–403.CrossRefGoogle ScholarPubMed
Boucharaba, A, Serre, CM, Gres, S et al. (2004) Platelet-derived lysophosphatidic acid supports the progression of osteolytic bone metastases in breast cancer. J Clin Invest. 114(12): 1714–25.CrossRefGoogle ScholarPubMed
Brill, A, Elinav, H, Varon, D (2004) Differential role of platelet granular mediators in angiogenesis. Cardiovasc Res. 63(2): 226–35.CrossRefGoogle ScholarPubMed
Brill, A, Dashevsky, O, Rivo, J, Gozal, Y, Varon, D (2005) Platelet-derived microparticles induce angiogenesis and stimulate post-ischemic revascularization. Cardiovasc Res. 67(1): 30–8.CrossRefGoogle ScholarPubMed
Ma, L, Elliott, SN, Cirino, G, Buret, A, Ignarro, LJ, Wallace, JL (2001) Platelets modulate gastric ulcer healing: role of endostatin and vascular endothelial growth factor release. Proc Natl Acad Sci USA. 98(11): 6470–5.CrossRefGoogle ScholarPubMed
Italiano, JE Jr, Richardson, JL, Patel-Hett, S, Battinelli, E et al. (2008) Angiogenesis is regulated by a novel mechanism: pro- and antiangiogenic proteins are organized into separate platelet alpha granules and differentially released. Blood. 111(3): 1227–33.CrossRefGoogle ScholarPubMed
Cervi, D, Yip, T-T, Bhattacharya, N, Podust, VN et al. (2008) Platelet-associated PF-4 as a biomarker of early tumor growth. Blood. 111(3): 1201–7.CrossRefGoogle ScholarPubMed
Pinedo, HM, Verheul, HM, D'Amato, RJ, Folkman, J (1998) Involvement of platelets in tumour angiogenesis?Lancet. 352(9142): 1775–7.CrossRefGoogle ScholarPubMed
Verheul, HM, Hoekman, K, Luykx-de Bakker, S et al. (1997) Platelet: transporter of vascular endothelial growth factor. Clin Cancer Res. 3(12 Pt 1): 2187–90.Google ScholarPubMed
Frenette, PS, Johnson, RC, Hynes, RO, Wagner, DD (1995) Platelets roll on stimulated endothelium in vivo: an interaction mediated by endothelial P-selectin. Proc Natl Acad Sci USA. 92(16): 7450–4.CrossRefGoogle ScholarPubMed
Honn, KV, Tang, DG, Chen, YQ (1992) Platelets and cancer metastasis: more than an epiphenomenon. Sem Thromb Hemost. 18: 392–415.CrossRefGoogle ScholarPubMed
Thun, MJ, Namboodiri, MM, Heath, CW Jr (1991) Aspirin use and reduced risk of fatal colon cancer. N Engl J Med. 325(23): 1593–6.CrossRefGoogle ScholarPubMed
Thun, MJ, Namboodiri, MM, Calle, EE, Flanders, WD, Heath, CW Jr (1993) Aspirin use and risk of fatal cancer. Cancer Res. 53(6): 1322–7.Google ScholarPubMed
Bosetti, C, Gallus, S, Vecchia, C (2006) Aspirin and cancer risk: an updated quantitative review to 2005. Cancer Causes Control. 17(7): 871–88.CrossRefGoogle ScholarPubMed
Lebeau, B, Chastang, C, Muir, JF, Vincent, J, Massin, F, Fabre, C (1993) No effect of an antiaggregant treatment with aspirin in small cell lung cancer treated with CCAVP16 chemotherapy. Results from a randomized clinical trial of 303 patients. The “Petites Cellules” Group. Cancer. 71(5): 1741–5.3.0.CO;2-Q>CrossRefGoogle ScholarPubMed
Zacharski, L, Henderson, W, Rickles, F et al. (1984) Effect of warfarin anticoagulation on survival in carcinoma of the lung, colon, head and neck, and prostate. Cancer. 53: 2046–52.3.0.CO;2-F>CrossRefGoogle ScholarPubMed
Chahinian, AP, Propert, KJ, Ware, JH et al. (1989) A randomized trial of anticoagulation with warfarin and of alternating chemotherapy in extensive small-cell lung cancer by the Cancer and Leukemia Group B. J Clin Oncol. 7(8): 993–1002.CrossRefGoogle ScholarPubMed
Lebeau, B, Chastang, C, Brechot, J et al. (1994) Subcutaneous heparin treatment increases survival in small cell lung cancer. Cancer. 74: 38–45.3.0.CO;2-E>CrossRefGoogle ScholarPubMed
Altinbas, M, Coskun, H, Er, O et al. (2004) A randomized clinical trial of combination chemotherapy with and without low-molecular-weight heparin in small cell lung cancer. J Thromb Hemost. 2: 1266–71.CrossRefGoogle ScholarPubMed
Kakkar, AK, Levine, MN, Kadziola, Z et al. (2004) Low molecular weight heparin, therapy with dalteparin, and survival in advanced cancer: the Fragmin Advanced Malignancy Outcome Study (FAMOUS). J Clin Oncol. 22(10): 1944–8.CrossRefGoogle ScholarPubMed
Klerk, C, Smorenburg, S, Otten, H et al. (2005) The effect of low molecular weight heparin on survival in patients with advanced malignancy. J Clin Oncol. 23(10): 2130–5.CrossRefGoogle ScholarPubMed
Lee, A, Rickles, F, Julian, J et al. (2005) Randomized comparison of low molecular weight heparin and coumarin derivatives on the survival of patients with cancer and venous thromboembolism. J Clin Oncol. 23(10): 2123–9.CrossRefGoogle ScholarPubMed
Sideras, K, Schaefer, PL, Okuno, SH et al. (2006) Low-molecular-weight heparin in patients with advanced cancer: a phase 3 clinical trial. Mayo Clin Proc. 81(6): 758–67.CrossRefGoogle ScholarPubMed
Kuderer, NM, Khorana, AA, Lyman, GH, Francis, CW (2007) A meta-analysis and systemic review of the efficacy and safety of anticoagulants as cancer treatment:impact on survival and bleeding complications. Cancer. 110(5): 1149–61.CrossRefGoogle Scholar
Miller, G, Bauer, K, Howarth, D, Cooper, J, Humphries, S, Rosenberg, R (2004) Increased incidence of neoplasia of the digestive tract in men with persistent activation of the coagulant pathway. J Thromb Haemost. 2: 2107–14.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×