5,008
Views
74
CrossRef citations to date
0
Altmetric
Special Review Section: Platelets And Inflammation

The pro-inflammatory role of platelets in cancer

& ORCID Icon
Pages 569-573 | Received 30 Nov 2017, Accepted 12 Mar 2018, Published online: 27 Mar 2018

References

  • Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med. 1986;315(26):1650–1659. doi:10.1056/NEJM198612253152606.
  • Ruf W. Tissue factor and cancer. Thromb Res. 2012;130(Suppl 1):SS84–7. doi:10.1016/j.thromres.2012.08.285.
  • Lip GY, Chin BS, Blann AD. Cancer and the prothrombotic state. Lancet Oncol. 2002;3(1):27–34. doi:10.1016/S1470-2045(01)00619-2.
  • Chen N, et al. Increased platelet-lymphocyte ratio closely relates to inferior clinical features and worse long-term survival in both resected and metastatic colorectal cancer: an updated systematic review and meta-analysis of 24 studies. Oncotarget. 2017;8(19):32356–32369.
  • Gu X, et al. Clinicopathological and prognostic significance of platelet to lymphocyte ratio in patients with gastric cancer. Oncotarget. 2016;7(31):49878–49887. doi:10.18632/oncotarget.10490.
  • Wang J, et al. Prognostic role of pretreatment platelet to lymphocyte ratio in urologic cancer. Oncotarget. 2017;8(41):70874–70882.
  • You J, et al. Preoperative platelet to lymphocyte ratio is a valuable prognostic biomarker in patients with colorectal cancer. Oncotarget. 2016;7(18):25516–25527. doi:10.18632/oncotarget.8334.
  • Zhang M, et al. High platelet-to-lymphocyte ratio predicts poor prognosis and clinicopathological characteristics in patients with breast cancer: A meta-analysis. Biomed Res Int. 2017;2017:9503025. doi:10.1155/2017/9503025.
  • Gay LJ, Felding-Habermann B. Contribution of platelets to tumour metastasis. Nat Rev Cancer. 2011;11(2):123–134. doi:10.1038/nrc3004.
  • Labelle M, Begum S, Hynes RO. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell. 2011;20(5):576–590. doi:10.1016/j.ccr.2011.09.009.
  • Gasic GJ, Gasic TB, Stewart CC. Antimetastatic effects associated with platelet reduction. Proc Natl Acad Sci. 1968;61(1):46–52. doi:10.1073/pnas.61.1.46.
  • Nieswandt B, et al. Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res. 1999;59(6):1295–1300.
  • Palumbo JS, et al. Platelets and fibrin(ogen) increase metastatic potential by impeding natural killer cell-mediated elimination of tumor cells. Blood. 2005;105(1):178–185. doi:10.1182/blood-2004-06-2272.
  • Trousseau A. Plegmasia alba dolens. Lectures on clinical medicine, delivered at the Hotel-Dieu, Paris. 1865;5:281–332.
  • Varki A. Trousseau’s syndrome: multiple definitions and multiple mechanisms. Blood. 2007;110(6):1723–1729. doi:10.1182/blood-2006-10-053736.
  • Nourshargh S, Alon R. Leukocyte migration into inflamed tissues. Immunity. 2014;41(5):694–707. doi:10.1016/j.immuni.2014.10.008.
  • Bevilacqua MP, Nelson RM. Selectins. J Clin Invest. 1993;91(2):379–387. doi:10.1172/JCI116210.
  • Lievens D, et al. Platelet CD40L mediates thrombotic and inflammatory processes in atherosclerosis. Blood. 2010;116(20):4317–4327. doi:10.1182/blood-2010-01-261206.
  • Thomas MR, Storey RF. The role of platelets in inflammation. Thromb Haemost. 2015;114(3):449–458. doi:10.1160/TH14-12-1067.
  • Sreeramkumar V, et al. Neutrophils scan for activated platelets to initiate inflammation. Science. 2014;346(6214):1234–1238. doi:10.1126/science.1256478.
  • Matsuo Y, et al. Involvement of p38alpha mitogen-activated protein kinase in lung metastasis of tumor cells. J Biol Chem. 2006;281(48):36767–36775. doi:10.1074/jbc.M604371200.
  • Peinado H, et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat Rev Cancer. 2017;17(5):302–317. doi:10.1038/nrc.2017.6.
  • Aigner S, et al. CD24, a mucin-type glycoprotein, is a ligand for P-selectin on human tumor cells. Blood. 1997;89(9):3385–3395.
  • Brodt P, et al. Liver endothelial E-selectin mediates carcinoma cell adhesion and promotes liver metastasis. Int J Cancer. 1997;71(4):612–619. doi:10.1002/(SICI)1097-0215(19970516)71:4<612::AID-IJC17>3.0.CO;2-D.
  • Iwai K, et al. Importance of E-selectin (ELAM-1) and sialyl Lewis(a) in the adhesion of pancreatic carcinoma cells to activated endothelium. Int J Cancer. 1993;54(6):972–977. doi:10.1002/ijc.2910540618.
  • Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity. 2014;41(1):49–61. doi:10.1016/j.immuni.2014.06.010.
  • Coffelt SB, Wellenstein MD, De Visser KE. Neutrophils in cancer: nNeutral no more. Nat Rev Cancer. 2016;16(7):431–446. doi:10.1038/nrc.2016.52.
  • Nozawa H, Chiu C, Hanahan D. Infiltrating neutrophils mediate the initial angiogenic switch in a mouse model of multistage carcinogenesis. Proc Natl Acad Sci. 2006;103(33):12493–12498. doi:10.1073/pnas.0601807103.
  • Gleissner CA, Von Hundelshausen P, Ley K. Platelet chemokines in vascular disease. Arterioscler Thromb Vasc Biol. 2008;28(11):1920–1927. doi:10.1161/ATVBAHA.108.169417.
  • Lindemann S, et al. Activated platelets mediate inflammatory signaling by regulated interleukin 1beta synthesis. J Cell Biol. 2001;154(3):485–490. doi:10.1083/jcb.200105058.
  • Bevilacqua MP, et al. Interleukin-1 activation of vascular endothelium. Effects on procoagulant activity and leukocyte adhesion. Am J Pathol. 1985;121(3):394–403.
  • Brown GT, et al. Lipopolysaccharide stimulates platelets through an IL-1beta autocrine loop. J Immunol. 2013;191(10):5196–5203. doi:10.4049/jimmunol.1300354.
  • Abdol Razak N, Elaskalani O, Metharom P. Pancreatic cancer-induced neutrophil extracellular traps: a potential contributor to cancer-associated thrombosis. Int J Mol Sci. 2017;18(3). doi:10.3390/ijms18030487.
  • Neuzillet C, et al. Targeting the TGFbeta pathway for cancer therapy. Pharmacol Ther. 2015;147:22–31. doi:10.1016/j.pharmthera.2014.11.001.
  • Italiani P, Boraschi D. From monocytes to M1/M2 macrophages: phenotypical vs. functional differentiation. Front Immunol. 2014;5:514. doi:10.3389/fimmu.2014.00514.
  • Fridlender ZG, et al. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell. 2009;16(3):183–194. doi:10.1016/j.ccr.2009.06.017.
  • Assoian RK, et al. Transforming growth factor-beta in human platelets. Identification of a major storage site, purification, and characterization. J Biol Chem. 1983;258(11):7155–7160.
  • Kaplan RN, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438(7069):820–827. doi:10.1038/nature04186.
  • Labelle M, Begum S, Hynes RO. Platelets guide the formation of early metastatic niches. Proc Natl Acad Sci. 2014;111(30):E3053–61. doi:10.1073/pnas.1411082111.
  • Sun X, et al. CXCL12/CXCR4/CXCR7 chemokine axis and cancer progression. Cancer Metastasis Rev. 2010;29(4):709–722. doi:10.1007/s10555-010-9256-x.
  • Palframan RT, et al. Inflammatory chemokine transport and presentation in HEV: A remote control mechanism for monocyte recruitment to lymph nodes in inflamed tissues. J Exp Med. 2001;194(9):1361–1373. doi:10.1084/jem.194.9.1361.
  • Qian BZ, et al. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature. 2011;475(7355):222–225. doi:10.1038/nature10138.
  • Wolf MJ, et al. Endothelial CCR2 signaling induced by colon carcinoma cells enables extravasation via the JAK2-Stat5 and p38MAPK pathway. Cancer Cell. 2012;22(1):91–105. doi:10.1016/j.ccr.2012.05.023.
  • Laubli H, Spanaus KS, Borsig L. Selectin-mediated activation of endothelial cells induces expression of CCL5 and promotes metastasis through recruitment of monocytes. Blood. 2009;114(20):4583–4591. doi:10.1182/blood-2008-10-186585.
  • Blanchet X, et al. Inflammatory role and prognostic value of platelet chemokines in acute coronary syndrome. Thromb Haemost. 2014;112(6):1277–1287. doi:10.1160/TH14-02-0139.
  • Brinkmann V, et al. Neutrophil extracellular traps kill bacteria. Science. 2004;303(5663):1532–1535. doi:10.1126/science.1092385.
  • Cedervall J, Zhang Y, Olsson AK. Tumor-induced NETosis as a risk factor for metastasis and organ failure. Cancer Res. 2016;76(15):4311–4315. doi:10.1158/0008-5472.CAN-15-3051.
  • Jorch SK, Kubes P. An emerging role for neutrophil extracellular traps in noninfectious disease. Nat Med. 2017;23(3):279–287. doi:10.1038/nm.4294.
  • Demers M, et al. Cancers predispose neutrophils to release extracellular DNA traps that contribute to cancer-associated thrombosis. Proc Natl Acad Sci. 2012;109(32):13076–13081. doi:10.1073/pnas.1200419109.
  • Berger-Achituv S, et al. A proposed role for neutrophil extracellular traps in cancer immunoediting. Front Immunol. 2013;4:48. doi:10.3389/fimmu.2013.00048.
  • Gupta AK, et al. Activated endothelial cells induce neutrophil extracellular traps and are susceptible to NETosis-mediated cell death. FEBS Lett. 2010;584(14):3193–3197. doi:10.1016/j.febslet.2010.06.006.
  • Saffarzadeh M, et al. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: a predominant role of histones. PLoS One. 2012;7(2):e32366. doi:10.1371/journal.pone.0032366.
  • Villanueva E, et al. Netting neutrophils induce endothelial damage, infiltrate tissues, and expose immunostimulatory molecules in systemic lupus erythematosus. J Immunol. 2011;187(1):538–552. doi:10.4049/jimmunol.1100450.
  • Fuchs TA, et al. Extracellular DNA traps promote thrombosis. Proc Natl Acad Sci. 2010;107(36):15880–15885. doi:10.1073/pnas.1005743107.
  • von Bruhl ML, et al. Monocytes, neutrophils, and platelets cooperate to initiate and propagate venous thrombosis in mice in vivo. J Exp Med. 2012;209(4):819–835. doi:10.1084/jem.20112322.
  • Clark SR, et al. Platelet TLR4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nat Med. 2007;13(4):463–469. doi:10.1038/nm1565.
  • Etulain J, et al. P-selectin promotes neutrophil extracellular trap formation in mice. Blood. 2015;126(2):242–246. doi:10.1182/blood-2015-01-624023.
  • Carestia A, Kaufman T, Schattner M. Platelets: new bricks in the building of neutrophil extracellular traps. Front Immunol. 2016;7:271. doi:10.3389/fimmu.2016.00271.
  • Alfaro C, et al. Tumor-produced interleukin-8 attracts human myeloid-derived suppressor cells and elicits extrusion of neutrophil extracellular traps (NETs). Clin Cancer Res. 2016;22(15):3924–3936. doi:10.1158/1078-0432.CCR-15-2463.
  • Cedervall J, et al. Neutrophil extracellular traps accumulate in peripheral blood vessels and compromise organ function in tumor-bearing animals. Cancer Res. 2015;75(13):2653–2662. doi:10.1158/0008-5472.CAN-14-3299.
  • Leal AC, et al. Tumor-derived exosomes induce the formation of neutrophil extracellular traps: implications for the establishment of cancer-associated thrombosis. Sci Rep. 2017;7(1):6438. doi:10.1038/s41598-017-06893-7.
  • Cools-Lartigue J, et al. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. J Clin Invest. 2013;123:3446–3458. doi:10.1172/JCI67484.
  • Park J, et al. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Sci Transl Med. 2016;8(361):361ra138. doi:10.1126/scitranslmed.aag1711.
  • Tohme S, et al. Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer Res. 2016;76(6):1367–1380. doi:10.1158/0008-5472.CAN-15-1591.
  • Thalin C, et al. NETosis promotes cancer-associated arterial microthrombosis presenting as ischemic stroke with troponin elevation. Thromb Res. 2016;139:56–64. doi:10.1016/j.thromres.2016.01.009.
  • Cedervall J, et al. Pharmacological targeting of peptidylarginine deiminase 4 prevents cancer-associated kidney injury in mice. Oncoimmunology. 2017;6(8):e1320009. doi:10.1080/2162402X.2017.1320009.
  • Elalamy I, et al. Long-term treatment of cancer-associated thrombosis: The choice of the optimal anticoagulant. J Thromb Haemost. 2017;15(5):848–857. doi:10.1111/jth.13659.
  • Barni S, et al. The effect of low-molecular-weight heparin in cancer patients: The mirror image of survival? Blood. 2014;124(1):155–156. doi:10.1182/blood-2014-03-561761.
  • Zhang N, et al. Low molecular weight heparin and cancer survival: clinical trials and experimental mechanisms. J Cancer Res Clin Oncol. 2016;142(8):1807–1816. doi:10.1007/s00432-016-2131-6.
  • Gresele P, et al. Platelet-targeted pharmacologic treatments as anti-cancer therapy. Cancer Metastasis Rev. 2017;36:331–355. doi:10.1007/s10555-017-9679-8.
  • Rothwell PM, et al. Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials. Lancet. 2011;377(9759):31–41. doi:10.1016/S0140-6736(10)62110-1.
  • Rothwell PM, et al. Effect of daily aspirin on risk of cancer metastasis: Astudy of incident cancers during randomised controlled trials. Lancet. 2012;379(9826):1591–1601. doi:10.1016/S0140-6736(12)60209-8.
  • Marnett LJ. Aspirin and the potential role of prostaglandins in colon cancer. Cancer Res. 1992;52(20):5575–5589.
  • Cao Y, et al. Population-wide impact of long-term use of aspirin and the risk for cancer. JAMA Oncol. 2016;2(6):762–769. doi:10.1001/jamaoncol.2015.6396.
  • Santilli F, Boccatonda A, Davi G. Aspirin, platelets, and cancer: The point of view of the internist. Eur J Intern Med. 2016;34:11–20. doi:10.1016/j.ejim.2016.06.004.
  • Elaskalani O, et al. Targeting platelets for the treatment of cancer. Cancers (Basel). 2017;9(7). doi:10.3390/cancers9070094.