Human Cancer and Platelet Interaction, a Potential Therapeutic Target
Abstract
:1. Introduction
2. Roles of Platelets in Cancer Development and Progression
2.1. Roles of Platelets in Tumor Development
2.2. Roles of Platelets in Cancer Metastasis
2.3. Impact of Platelet on the Anti-Tumor Immunity
3. Cancer Induces Platelet Activation
4. Platelets, a Potential Therapeutic Target and Biomarker for Cancer Treatment
4.1. Platelet Is a Potential Target to Suppresses Cancer Metastasis
4.2. Targeting Platelets Is a Potential Strategy to Overcome Drug Resistance
4.3. Platelets in Anti-Cancer Drug Delivery
5. Conclusions and Future Direction
Acknowledgments
Author Contributions
Conflicts of Interest
Abbreviations
VEGF | Vascular endothelial growth factor |
PDGF | Platelet derived growth factor |
P2Y12 | Platelet factor 4 (PF4), ADP receptor |
CLEC2 | C-type lectin-like receptor 2 |
LPA | Lysophosphatidic acid |
Asm | Acid sphingomyelinase |
Ask1 | Apoptosis signal-regulating kinase 1 |
References
- Gupta, G.P.; Massague, J. Cancer metastasis: Building a framework. Cell 2006, 127, 679–695. [Google Scholar] [CrossRef] [PubMed]
- Ashworth, T.R. A case of cancer in which cells similar to those in the tumors were seen in the blood after death. Aust. Med. J. 1869, 14, 146–149. [Google Scholar]
- Aggarwal, C.; Wang, X.; Ranganathan, A.; Torigian, D.; Troxel, A.; Evans, T.; Cohen, R.B.; Vaidya, B.; Rao, C.; Connelly, M.; et al. Circulating tumor cells as a predictive biomarker in patients with small cell lung cancer undergoing chemotherapy. Lung Cancer 2017, 112, 118–125. [Google Scholar] [CrossRef] [PubMed]
- Tartarone, A.; Lerose, R.; Rodriquenz, M.G.; Mambella, G.; Calderoni, G.; Bozza, G.; Aieta, M. Molecular characterization and prognostic significance of circulating tumor cells in patients with non-small cell lung cancer. J. Thorac. Dis. 2017, 9 (Suppl. 13), S1359–S1363. [Google Scholar] [CrossRef] [PubMed]
- Cristofanilli, M.; Budd, G.T.; Ellis, M.J.; Stopeck, A.; Matera, J.; Miller, M.C.; Reuben, J.M.; Doyle, G.V.; Allard, W.J.; Terstappen, L.W.; et al. Circulating tumor cells, disease progression, and survival in metastatic breast cancer. N. Engl. J. Med. 2004, 351, 781–791. [Google Scholar] [CrossRef] [PubMed]
- Cohen, S.J.; Punt, C.J.; Iannotti, N.; Saidman, B.H.; Sabbath, K.D.; Gabrail, N.Y.; Picus, J.; Morse, M.A.; Mitchell, E.; Miller, M.C.; et al. Prognostic significance of circulating tumor cells in patients with metastatic colorectal cancer. Ann. Oncol. 2009, 20, 1223–1229. [Google Scholar] [CrossRef] [PubMed]
- De Bono, J.S.; Scher, H.I.; Montgomery, R.B.; Parker, C.; Miller, M.C.; Tissing, H.; Doyle, G.V.; Terstappen, L.W.; Pienta, K.J.; Raghavan, D. Circulating tumor cells predict survival benefit from treatment in metastatic castration-resistant prostate cancer. Clin. Cancer Res. 2008, 14, 6302–6309. [Google Scholar] [CrossRef] [PubMed]
- Zhang, S.; Wu, T.; Peng, X.; Liu, J.; Liu, F.; Wu, S.; Liu, S.; Dong, Y.; Xie, S.; Ma, S. Mesenchymal phenotype of circulating tumor cells is associated with distant metastasis in breast cancer patients. Cancer Manag. Res. 2017, 9, 691–700. [Google Scholar] [CrossRef] [PubMed]
- Thiele, J.A.; Bethel, K.; Kralickova, M.; Kuhn, P. Circulating Tumor Cells: Fluid Surrogates of Solid Tumors. Annu. Rev. Pathol. 2017, 12, 419–447. [Google Scholar] [CrossRef] [PubMed]
- Ting, D.T.; Wittner, B.S.; Ligorio, M.; Vincent Jordan, N.; Shah, A.M.; Miyamoto, D.T.; Aceto, N.; Bersani, F.; Brannigan, B.W.; Xega, K.; et al. Single-cell RNA sequencing identifies extracellular matrix gene expression by pancreatic circulating tumor cells. Cell Rep. 2014, 8, 1905–1918. [Google Scholar] [CrossRef] [PubMed]
- Okumura, T.; Yamaguchi, T.; Watanabe, T.; Nagata, T.; Shimada, Y. Flow Cytometric Detection of Circulating Tumor Cells Using a Candidate Stem Cell Marker, p75 Neurotrophin Receptor (p75NTR). Methods Mol. Biol. 2017, 1634, 211–217. [Google Scholar] [PubMed]
- Mirza, S.; Jain, N.; Rawal, R. Evidence for circulating cancer stem-like cells and epithelial-mesenchymal transition phenotype in the pleurospheres derived from lung adenocarcinoma using liquid biopsy. Tumour Biol. 2017, 39. [Google Scholar] [CrossRef] [PubMed]
- Takeishi, N.; Imai, Y.; Yamaguchi, T.; Ishikawa, T. Flow of a circulating tumor cell and red blood cells in microvessels. Phys. Rev. E 2015, 92. [Google Scholar] [CrossRef] [PubMed]
- Hamilton, G.; Rath, B. Circulating tumor cell interactions with macrophages: Implications for biology and treatment. Transl. Lung Cancer Res. 2017, 6, 418–430. [Google Scholar] [CrossRef] [PubMed]
- Gruber, I.; Landenberger, N.; Staebler, A.; Hahn, M.; Wallwiener, D.; Fehm, T. Relationship between circulating tumor cells and peripheral T-cells in patients with primary breast cancer. Anticancer Res. 2013, 33, 2233–2238. [Google Scholar] [PubMed]
- Mego, M.; Gao, H.; Cohen, E.N.; Anfossi, S.; Giordano, A.; Sanda, T.; Fouad, T.M.; De Giorgi, U.; Giuliano, M.; Woodward, W.A.; et al. Circulating Tumor Cells (CTC) Are Associated with Defects in Adaptive Immunity in Patients with Inflammatory Breast Cancer. J. Cancer 2016, 7, 1095–1104. [Google Scholar] [CrossRef] [PubMed]
- Lin, M.; Liang, S.Z.; Shi, J.; Niu, L.Z.; Chen, J.B.; Zhang, M.J.; Xu, K.C. Circulating tumor cell as a biomarker for evaluating allogenic NK cell immunotherapy on stage IV non-small cell lung cancer. Immunol. Lett. 2017, 191, 10–15. [Google Scholar] [CrossRef] [PubMed]
- McCarty, O.J.; Ku, D.; Sugimoto, M.; King, M.R.; Cosemans, J.M.; Neeves, K.B. Dimensional analysis and scaling relevant to flow models of thrombus formation: Communication from the SSC of the ISTH. J. Thromb. Haemost. 2016, 14, 619–622. [Google Scholar] [CrossRef] [PubMed]
- Unsicker, K.; Spittau, B.; Krieglstein, K. The multiple facets of the TGF-beta family cytokine growth/differentiation factor-15/macrophage inhibitory cytokine-1. Cytokine Growth Factor Rev. 2013, 24, 373–384. [Google Scholar] [CrossRef] [PubMed]
- Hamilton, G.; Rath, B.; Klameth, L.; Hochmair, M.J. Small cell lung cancer: Recruitment of macrophages by circulating tumor cells. Oncoimmunology 2016, 5, e1093277. [Google Scholar] [CrossRef] [PubMed]
- Sun, W.W.; Xu, Z.H.; Lian, P.; Gao, B.L.; Hu, J.A. Characteristics of circulating tumor cells in organ metastases, prognosis, and T lymphocyte mediated immune response. Onco Targets Ther. 2017, 10, 2413–2424. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Sun, Q.; Liu, Q.; Wang, C.; Yao, R.; Wang, Y. CTC immune escape mediated by PD-L1. Med. Hypotheses 2016, 93, 138–139. [Google Scholar] [CrossRef] [PubMed]
- Stone, J.P.; Wagner, D.D. P-selectin mediates adhesion of platelets to neuroblastoma and small cell lung cancer. J. Clin. Investig. 1993, 92, 804–813. [Google Scholar] [CrossRef] [PubMed]
- Machlus, K.R.; Thon, J.N.; Italiano, J.E., Jr. Interpreting the developmental dance of the megakaryocyte: A review of the cellular and molecular processes mediating platelet formation. Br. J. Haematol. 2014, 165, 227–236. [Google Scholar] [CrossRef] [PubMed]
- Didelot, M.; Docq, C.; Wahl, D.; Lacolley, P.; Regnault, V.; Lagrange, J. Platelet aggregation impacts thrombin generation assessed by calibrated automated thrombography. Platelets 2017, 1–6. [Google Scholar] [CrossRef] [PubMed]
- Davi, G.; Patrono, C. Platelet activation and atherothrombosis. N. Engl. J. Med. 2007, 357, 2482–2494. [Google Scholar] [CrossRef] [PubMed]
- Versteeg, H.H.; Heemskerk, J.W.; Levi, M.; Reitsma, P.H. New fundamentals in hemostasis. Physiol. Rev. 2013, 93, 327–358. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.; Huang, X.Z.; Song, Y.X.; Gao, P.; Sun, J.X.; Wang, Z.N. High Platelet-to-Lymphocyte Ratio Predicts Poor Prognosis and Clinicopathological Characteristics in Patients with Breast Cancer: A Meta-Analysis. BioMed Res. Int. 2017, 2017. [Google Scholar] [CrossRef] [PubMed]
- Tjon-Kon-Fat, L.A.; Lundholm, M.; Schroder, M.; Wurdinger, T.; Thellenberg-Karlsson, C.; Widmark, A.; Wikstrom, P.; Nilsson, R.J.A. Platelets harbor prostate cancer biomarkers and the ability to predict therapeutic response to abiraterone in castration resistant patients. Prostate 2018, 78, 48–53. [Google Scholar] [CrossRef] [PubMed]
- Stone, R.L.; Nick, A.M.; McNeish, I.A.; Balkwill, F.; Han, H.D.; Bottsford-Miller, J.; Rupairmoole, R.; Armaiz-Pena, G.N.; Pecot, C.V.; Coward, J.; et al. Paraneoplastic thrombocytosis in ovarian cancer. N. Engl. J. Med. 2012, 366, 610–618. [Google Scholar] [CrossRef] [PubMed]
- Rothwell, P.M.; Wilson, M.; Price, J.F.; Belch, J.F.; Meade, T.W.; Mehta, Z. Effect of daily aspirin on risk of cancer metastasis: A study of incident cancers during randomised controlled trials. Lancet 2012, 379, 1591–1601. [Google Scholar] [CrossRef]
- Patrignani, P.; Patrono, C. Aspirin and Cancer. J. Am. Coll. Cardiol. 2016, 68, 967–976. [Google Scholar] [CrossRef] [PubMed]
- Fu, S.; Niu, Y.; Zhang, X.; Zhang, J.R.; Liu, Z.P.; Wang, R.T. Squamous cell carcinoma antigen, platelet distribution width, and prealbumin collectively as a marker of squamous cell cervical carcinoma. Cancer Biomark. 2017, 21, 317–321. [Google Scholar] [CrossRef] [PubMed]
- Liu, W.; Ha, M.; Yin, N. Combination of platelet count and lymphocyte to monocyte ratio is a prognostic factor in patients undergoing surgery for non-small cell lung cancer. Oncotarget 2017, 8, 73198–73207. [Google Scholar] [CrossRef] [PubMed]
- Sheu, J.R.; Fong, T.H.; Liu, C.M.; Shen, M.Y.; Chen, T.L.; Chang, Y.; Lu, M.S.; Hsiao, G. Expression of matrix metalloproteinase-9 in human platelets: Regulation of platelet activation in in vitro and in vivo studies. Br. J. Pharmacol. 2004, 143, 193–201. [Google Scholar] [CrossRef] [PubMed]
- Pintucci, G.; Froum, S.; Pinnell, J.; Mignatti, P.; Rafii, S.; Green, D. 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. 2002, 88, 834–842. [Google Scholar] [CrossRef] [PubMed]
- Ma, L.; Perini, R.; McKnight, W.; Dicay, M.; Klein, A.; Hollenberg, M.D.; Wallace, J.L. Proteinase-activated receptors 1 and 4 counter-regulate endostatin and VEGF release from human platelets. Proc. Natl. Acad. Sci. USA 2005, 102, 216–220. [Google Scholar] [CrossRef] [PubMed]
- Jiang, L.; Luan, Y.; Miao, X.; Sun, C.; Li, K.; Huang, Z.; Xu, D.; Zhang, M.; Kong, F.; Li, N. Platelet releasate promotes breast cancer growth and angiogenesis via VEGF-integrin cooperative signalling. Br. J. Cancer 2017, 117, 695–703. [Google Scholar] [CrossRef] [PubMed]
- Elaskalani, O.; Falasca, M.; Moran, N.; Berndt, M.C.; Metharom, P. The Role of Platelet-Derived ADP and ATP in Promoting Pancreatic Cancer Cell Survival and Gemcitabine Resistance. Cancers 2017, 9, 142. [Google Scholar] [CrossRef] [PubMed]
- Cho, M.S.; Noh, K.; Haemmerle, M.; Li, D.; Park, H.; Hu, Q.; Hisamatsu, T.; Mitamura, T.; Mak, S.L.C.; Kunapuli, S.; et al. Role of ADP receptors on platelets in the growth of ovarian cancer. Blood 2017, 130, 1235–1242. [Google Scholar] [CrossRef] [PubMed]
- Ballerini, P.; Dovizio, M.; Bruno, A.; Tacconelli, S.; Patrignani, P. P2Y12 Receptors in Tumorigenesis and Metastasis. Front. Pharmacol. 2018, 9, 66. [Google Scholar] [CrossRef] [PubMed]
- Waldmann, T.A. Cytokines in Cancer Immunotherapy. Cold Spring Harb. Perspect. Biol. 2017. [Google Scholar] [CrossRef] [PubMed]
- Musolino, C.; Allegra, A.; Innao, V.; Allegra, A.G.; Pioggia, G.; Gangemi, S. Inflammatory and Anti-Inflammatory Equilibrium, Proliferative and Antiproliferative Balance: The Role of Cytokines in Multiple Myeloma. Mediat. Inflamm. 2017, 2017. [Google Scholar] [CrossRef] [PubMed]
- Lee, M.; Rhee, I. Cytokine Signaling in Tumor Progression. Immune Netw. 2017, 17, 214–227. [Google Scholar] [CrossRef] [PubMed]
- Assoian, R.K.; Komoriya, A.; Meyers, C.A.; Miller, D.M.; Sporn, M.B. Transforming growth factor-beta in human platelets. Identification of a major storage site, purification, and characterization. J. Biol. Chem. 1983, 258, 7155–7160. [Google Scholar] [PubMed]
- Hu, Q.; Hisamatsu, T.; Haemmerle, M.; Cho, M.S.; Pradeep, S.; Rupaimoole, R.; Rodriguez-Aguayo, C.; Lopez-Berestein, G.; Wong, S.T.C.; Sood, A.K.; et al. Role of Platelet-Derived Tgfbeta1 in the Progression of Ovarian Cancer. Clin. Cancer Res. 2017, 23, 5611–5621. [Google Scholar] [CrossRef] [PubMed]
- Cho, M.S.; Bottsford-Miller, J.; Vasquez, H.G.; Stone, R.; Zand, B.; Kroll, M.H.; Sood, A.K.; Afshar-Kharghan, V. Platelets increase the proliferation of ovarian cancer cells. Blood 2012, 120, 4869–4872. [Google Scholar] [CrossRef] [PubMed]
- Carr, B.I.; Cavallini, A.; D’Alessandro, R.; Refolo, M.G.; Lippolis, C.; Mazzocca, A.; Messa, C. Platelet extracts induce growth, migration and invasion in human hepatocellular carcinoma in vitro. BMC Cancer 2014, 14, 43. [Google Scholar] [CrossRef] [PubMed]
- He, A.D.; Xie, W.; Song, W.; Ma, Y.Y.; Liu, G.; Liang, M.L.; Da, X.W.; Yao, G.Q.; Zhang, B.X.; Gao, C.J.; et al. Platelet releasates promote the proliferation of hepatocellular carcinoma cells by suppressing the expression of KLF6. Sci. Rep. 2017, 7, 3989. [Google Scholar] [CrossRef] [PubMed]
- Narla, G.; Heath, K.E.; Reeves, H.L.; Li, D.; Giono, L.E.; Kimmelman, A.C.; Glucksman, M.J.; Narla, J.; Eng, F.J.; Chan, A.M.; et al. KLF6, a candidate tumor suppressor gene mutated in prostate cancer. Science 2001, 294, 2563–2566. [Google Scholar] [CrossRef] [PubMed]
- Peterson, J.E.; Zurakowski, D.; Italiano, J.E., Jr.; Michel, L.V.; Connors, S.; Oenick, M.; D’Amato, R.J.; Klement, G.L.; Folkman, J. VEGF, PF4 and PDGF are elevated in platelets of colorectal cancer patients. Angiogenesis 2012, 15, 265–273. [Google Scholar] [CrossRef] [PubMed]
- Zizzo, N.; Patruno, R.; Zito, F.A.; Di Summa, A.; Tinelli, A.; Troilo, S.; Misino, A.; Ruggieri, E.; Goffredo, V.; Gadaleta, C.D.; et al. Vascular endothelial growth factor concentrations from platelets correlate with tumor angiogenesis and grading in a spontaneous canine non-Hodgkin lymphoma model. Leuk. Lymphoma 2010, 51, 291–296. [Google Scholar] [CrossRef] [PubMed]
- Heldin, C.H.; Lennartsson, J.; Westermark, B. Involvement of platelet-derived growth factor ligands and receptors in tumorigenesis. J. Intern. Med. 2018, 283, 16–44. [Google Scholar] [CrossRef] [PubMed]
- Repsold, L.; Pool, R.; Karodia, M.; Tintinger, G.; Joubert, A.M. An overview of the role of platelets in angiogenesis, apoptosis and autophagy in chronic myeloid leukaemia. Cancer Cell Int. 2017, 17, 89. [Google Scholar] [CrossRef] [PubMed]
- Pucci, F.; Rickelt, S.; Newton, A.P.; Garris, C.; Nunes, E.; Evavold, C.; Pfirschke, C.; Engblom, C.; Mino-Kenudson, M.; Hynes, R.O.; et al. PF4 Promotes Platelet Production and Lung Cancer Growth. Cell Rep. 2016, 17, 1764–1772. [Google Scholar] [CrossRef] [PubMed]
- Maione, T.E.; Gray, G.S.; Petro, J.; Hunt, A.J.; Donner, A.L.; Bauer, S.I.; Carson, H.F.; Sharpe, R.J. Inhibition of angiogenesis by recombinant human platelet factor-4 and related peptides. Science 1990, 247, 77–79. [Google Scholar] [CrossRef] [PubMed]
- Yamaguchi, K.; Ogawa, K.; Katsube, T.; Shimao, K.; Konno, S.; Shimakawa, T.; Yoshimatsu, K.; Naritaka, Y.; Yagawa, H.; Hirose, K. Platelet factor 4 gene transfection into tumor cells inhibits angiogenesis, tumor growth and metastasis. Anticancer Res. 2005, 25, 847–851. [Google Scholar] [PubMed]
- Sato, Y.; Waki, M.; Ohno, M.; Kuwano, M.; Sakata, T. Carboxyl-terminal heparin-binding fragments of platelet factor 4 retain the blocking effect on the receptor binding of basic fibroblast growth factor. Jpn. J. Cancer Res. 1993, 84, 485–488. [Google Scholar] [CrossRef] [PubMed]
- Gengrinovitch, S.; Greenberg, S.M.; Cohen, T.; Gitay-Goren, H.; Rockwell, P.; Maione, T.E.; Levi, B.Z.; Neufeld, G. Platelet factor-4 inhibits the mitogenic activity of VEGF121 and VEGF165 using several concurrent mechanisms. J. Biol. Chem. 1995, 270, 15059–15065. [Google Scholar] [CrossRef] [PubMed]
- Michael, J.V.; Wurtzel, J.G.T.; Mao, G.F.; Rao, A.K.; Kolpakov, M.A.; Sabri, A.; Hoffman, N.E.; Rajan, S.; Tomar, D.; Madesh, M.; et al. Platelet microparticles infiltrating solid tumors transfer miRNAs that suppress tumor growth. Blood 2017, 130, 567–580. [Google Scholar] [CrossRef] [PubMed]
- Labelle, M.; Begum, S.; Hynes, R.O. Platelets guide the formation of early metastatic niches. Proc. Natl. Acad. Sci. USA 2014, 111, E3053–E3061. [Google Scholar] [CrossRef] [PubMed]
- Sharma, D.; Brummel-Ziedins, K.E.; Bouchard, B.A.; Holmes, C.E. Platelets in tumor progression: A host factor that offers multiple potential targets in the treatment of cancer. J. Cell. Physiol. 2014, 229, 1005–1015. [Google Scholar] [CrossRef] [PubMed]
- Seyfried, T.N.; Huysentruyt, L.C. On the origin of cancer metastasis. Crit. Rev. Oncog. 2013, 18, 43–73. [Google Scholar] [CrossRef] [PubMed]
- Gasic, G.J.; Gasic, T.B.; Stewart, C.C. Antimetastatic effects associated with platelet reduction. Proc. Natl. Acad. Sci. USA 1968, 61, 46–52. [Google Scholar] [CrossRef] [PubMed]
- Gasic, G.J.; Gasic, T.B.; Galanti, N.; Johnson, T.; Murphy, S. Platelet-tumor-cell interactions in mice. The role of platelets in the spread of malignant disease. Int. J. Cancer 1973, 11, 704–718. [Google Scholar] [CrossRef] [PubMed]
- Mitchell, M.J.; King, M.R. Fluid Shear Stress Sensitizes Cancer Cells to Receptor-Mediated Apoptosis via Trimeric Death Receptors. New J. Phys. 2013, 15. [Google Scholar] [CrossRef] [PubMed]
- Egan, K.; Cooke, N.; Kenny, D. Living in shear: Platelets protect cancer cells from shear induced damage. Clin. Exp. Metast. 2014, 31, 697–704. [Google Scholar] [CrossRef] [PubMed]
- Thiery, J.P.; Acloque, H.; Huang, R.Y.; Nieto, M.A. Epithelial-mesenchymal transitions in development and disease. Cell 2009, 139, 871–890. [Google Scholar] [CrossRef] [PubMed]
- Yeung, K.T.; Yang, J. Epithelial-mesenchymal transition in tumor metastasis. Mol. Oncol. 2017, 11, 28–39. [Google Scholar] [CrossRef] [PubMed]
- Takemoto, A.; Okitaka, M.; Takagi, S.; Takami, M.; Sato, S.; Nishio, M.; Okumura, S.; Fujita, N. A critical role of platelet TGF-beta release in podoplanin-mediated tumour invasion and metastasis. Sci. Rep. 2017, 7, 42186. [Google Scholar] [CrossRef] [PubMed]
- Labelle, M.; Begum, S.; Hynes, R.O. Direct signaling between platelets and cancer cells induces an epithelial-mesenchymal-like transition and promotes metastasis. Cancer Cell 2011, 20, 576–590. [Google Scholar] [CrossRef] [PubMed]
- Tang, M.; Jiang, L.; Lin, Y.; Wu, X.; Wang, K.; He, Q.; Wang, X.; Li, W. Platelet microparticle-mediated transfer of miR-939 to epithelial ovarian cancer cells promotes epithelial to mesenchymal transition. Oncotarget 2017, 8, 97464–97475. [Google Scholar] [CrossRef] [PubMed]
- Fujita, N.; Takagi, S. The impact of Aggrus/podoplanin on platelet aggregation and tumour metastasis. J. Biochem. 2012, 152, 407–413. [Google Scholar] [CrossRef] [PubMed]
- Ruf, W. Tissue factor and cancer. Thromb. Res. 2012, 130 (Suppl. 1), S84–S87. [Google Scholar] [CrossRef] [PubMed]
- Orellana, R.; Kato, S.; Erices, R.; Bravo, M.L.; Gonzalez, P.; Oliva, B.; Cubillos, S.; Valdivia, A.; Ibanez, C.; Branes, J.; et al. Platelets enhance tissue factor protein and metastasis initiating cell markers, and act as chemoattractants increasing the migration of ovarian cancer cells. BMC Cancer 2015, 15, 290. [Google Scholar] [CrossRef] [PubMed]
- Dovizio, M.; Maier, T.J.; Alberti, S.; Di Francesco, L.; Marcantoni, E.; Munch, G.; John, C.M.; Suess, B.; Sgambato, A.; Steinhilber, D.; et al. Pharmacological inhibition of platelet-tumor cell cross-talk prevents platelet-induced overexpression of cyclooxygenase-2 in HT29 human colon carcinoma cells. Mol. Pharmacol. 2013, 84, 25–40. [Google Scholar] [CrossRef] [PubMed]
- Xu, X.R.; Carrim, N.; Neves, M.A.; McKeown, T.; Stratton, T.W.; Coelho, R.M.; Lei, X.; Chen, P.; Xu, J.; Dai, X.; et al. Platelets and platelet adhesion molecules: Novel mechanisms of thrombosis and anti-thrombotic therapies. Thromb. J. 2016, 14 (Suppl. 1), 29. [Google Scholar] [CrossRef] [PubMed]
- Lavergne, M.; Janus-Bell, E.; Schaff, M.; Gachet, C.; Mangin, P.H. Platelet Integrins in Tumor Metastasis: Do They Represent a Therapeutic Target? Cancers 2017, 9, 133. [Google Scholar] [CrossRef] [PubMed]
- Mammadova-Bach, E.; Zigrino, P.; Brucker, C.; Bourdon, C.; Freund, M.; De Arcangelis, A.; Abrams, S.I.; Orend, G.; Gachet, C.; Mangin, P.H. Platelet integrin alpha6beta1 controls lung metastasis through direct binding to cancer cell-derived ADAM9. JCI Insight 2016, 1, e88245. [Google Scholar] [CrossRef] [PubMed]
- Bakewell, S.J.; Nestor, P.; Prasad, S.; Tomasson, M.H.; Dowland, N.; Mehrotra, M.; Scarborough, R.; Kanter, J.; Abe, K.; Phillips, D.; et al. Platelet and osteoclast beta3 integrins are critical for bone metastasis. Proc. Natl. Acad. Sci. USA 2003, 100, 14205–14210. [Google Scholar] [CrossRef] [PubMed]
- Boucharaba, A.; Serre, C.M.; Gres, S.; Saulnier-Blache, J.S.; Bordet, J.C.; Guglielmi, J.; Clezardin, P.; Peyruchaud, O. Platelet-derived lysophosphatidic acid supports the progression of osteolytic bone metastases in breast cancer. J. Clin. Investig. 2004, 114, 1714–1725. [Google Scholar] [CrossRef] [PubMed]
- Chopra, H.; Hatfield, J.S.; Chang, Y.S.; Grossi, I.M.; Fitzgerald, L.A.; O'Gara, C.Y.; Marnett, L.J.; Diglio, C.A.; Taylor, J.D.; Honn, K.V. Role of tumor cytoskeleton and membrane glycoprotein IRGpIIb/IIIa in platelet adhesion to tumor cell membrane and tumor cell-induced platelet aggregation. Cancer Res. 1988, 48, 3787–3800. [Google Scholar] [PubMed]
- Frisch, S.M.; Screaton, R.A. Anoikis mechanisms. Curr. Opin. Cell Biol. 2001, 13, 555–562. [Google Scholar] [CrossRef]
- Buchheit, C.L.; Weigel, K.J.; Schafer, Z.T. Cancer cell survival during detachment from the ECM: Multiple barriers to tumour progression. Nat. Rev. Cancer 2014, 14, 632–641. [Google Scholar] [CrossRef] [PubMed]
- Haemmerle, M.; Taylor, M.L.; Gutschner, T.; Pradeep, S.; Cho, M.S.; Sheng, J.; Lyons, Y.M.; Nagaraja, A.S.; Dood, R.L.; Wen, Y.; et al. Platelets reduce anoikis and promote metastasis by activating YAP1 signaling. Nat. Commun. 2017, 8, 310. [Google Scholar] [CrossRef] [PubMed]
- Ichijo, H.; Nishida, E.; Irie, K.; ten Dijke, P.; Saitoh, M.; Moriguchi, T.; Takagi, M.; Matsumoto, K.; Miyazono, K.; Gotoh, Y. Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science 1997, 275, 90–94. [Google Scholar] [CrossRef] [PubMed]
- Kamiyama, M.; Shirai, T.; Tamura, S.; Suzuki-Inoue, K.; Ehata, S.; Takahashi, K.; Miyazono, K.; Hayakawa, Y.; Sato, T.; Takeda, K.; et al. ASK1 facilitates tumor metastasis through phosphorylation of an ADP receptor P2Y12 in platelets. Cell Death Differ. 2017, 24, 2066–2076. [Google Scholar] [CrossRef] [PubMed]
- Carpinteiro, A.; Becker, K.A.; Japtok, L.; Hessler, G.; Keitsch, S.; Pozgajova, M.; Schmid, K.W.; Adams, C.; Muller, S.; Kleuser, B.; et al. Regulation of hematogenous tumor metastasis by acid sphingomyelinase. EMBO Mol. Med. 2015, 7, 714–734. [Google Scholar] [CrossRef] [PubMed]
- Carpinteiro, A.; Beckmann, N.; Seitz, A.; Hessler, G.; Wilker, B.; Soddemann, M.; Helfrich, I.; Edelmann, B.; Gulbins, E.; Becker, K.A. Role of Acid Sphingomyelinase-Induced Signaling in Melanoma Cells for Hematogenous Tumor Metastasis. Cell. Physiol. Biochem. 2016, 38, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Wargo, J.A.; Schumacher, L.Y.; Comin-Anduix, B.; Dissette, V.B.; Glaspy, J.A.; McBride, W.H.; Butterfield, L.H.; Economou, J.S.; Ribas, A. Natural killer cells play a critical role in the immune response following immunization with melanoma-antigen-engineered dendritic cells. Cancer Gene Ther. 2005, 12, 516–527. [Google Scholar] [CrossRef] [PubMed]
- Shimaoka, H.; Takeno, S.; Maki, K.; Sasaki, T.; Hasegawa, S.; Yamashita, Y. A cytokine signal inhibitor for rheumatoid arthritis enhances cancer metastasis via depletion of NK cells in an experimental lung metastasis mouse model of colon cancer. Oncol. Lett. 2017, 14, 3019–3027. [Google Scholar] [CrossRef] [PubMed]
- Nieswandt, B.; Hafner, M.; Echtenacher, B.; Mannel, D.N. Lysis of tumor cells by natural killer cells in mice is impeded by platelets. Cancer Res. 1999, 59, 1295–1300. [Google Scholar] [PubMed]
- Zitvogel, L.; Tesniere, A.; Kroemer, G. Cancer despite immunosurveillance: Immunoselection and immunosubversion. Nat. Rev. Immunol. 2006, 6, 715–727. [Google Scholar] [CrossRef] [PubMed]
- Placke, T.; Orgel, M.; Schaller, M.; Jung, G.; Rammensee, H.G.; Kopp, H.G.; Salih, H.R. Platelet-derived MHC class I confers a pseudonormal phenotype to cancer cells that subverts the antitumor reactivity of natural killer immune cells. Cancer Res. 2012, 72, 440–448. [Google Scholar] [CrossRef] [PubMed]
- Kopp, H.G.; Placke, T.; Salih, H.R. Platelet-derived transforming growth factor-beta down-regulates NKG2D thereby inhibiting natural killer cell antitumor reactivity. Cancer Res. 2009, 69, 7775–7783. [Google Scholar] [CrossRef] [PubMed]
- Fadok, V.A.; Bratton, D.L.; Konowal, A.; Freed, P.W.; Westcott, J.Y.; Henson, P.M. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-beta, PGE2, and PAF. J. Clin. Investig. 1998, 101, 890–898. [Google Scholar] [CrossRef] [PubMed]
- Haribhai, D.; Luo, X.; Chen, J.; Jia, S.; Shi, L.; Schroeder, J.A.; Weiler, H.; Aster, R.H.; Hessner, M.J.; Hu, J.; et al. TGF-beta1 along with other platelet contents augments Treg cells to suppress anti-FVIII immune responses in hemophilia A mice. Blood Adv. 2016, 1, 139–151. [Google Scholar] [CrossRef] [PubMed]
- Grossman, W.J.; Verbsky, J.W.; Barchet, W.; Colonna, M.; Atkinson, J.P.; Ley, T.J. Human T regulatory cells can use the perforin pathway to cause autologous target cell death. Immunity 2004, 21, 589–601. [Google Scholar] [CrossRef] [PubMed]
- Gondek, D.C.; Lu, L.F.; Quezada, S.A.; Sakaguchi, S.; Noelle, R.J. Cutting edge: Contact-mediated suppression by CD4+CD25+ regulatory cells involves a granzyme B-dependent, perforin-independent mechanism. J. Immunol. 2005, 174, 1783–1786. [Google Scholar] [CrossRef] [PubMed]
- Tran, D.Q.; Andersson, J.; Wang, R.; Ramsey, H.; Unutmaz, D.; Shevach, E.M. GARP (LRRC32) is essential for the surface expression of latent TGF-beta on platelets and activated FOXP3+ regulatory T cells. Proc. Natl. Acad. Sci. USA 2009, 106, 13445–13450. [Google Scholar] [CrossRef] [PubMed]
- Rachidi, S.; Metelli, A.; Riesenberg, B.; Wu, B.X.; Nelson, M.H.; Wallace, C.; Paulos, C.M.; Rubinstein, M.P.; Garrett-Mayer, E.; Hennig, M.; et al. Platelets subvert T cell immunity against cancer via GARP-TGFbeta axis. Sci. Immunol. 2017, 2, 11. [Google Scholar] [CrossRef] [PubMed]
- Silverstein, M.D.; Heit, J.A.; Mohr, D.N.; Petterson, T.M.; O’Fallon, W.M.; Melton, L.J. Trends in the incidence of deep vein thrombosis and pulmonary embolism: A 25-year population-based study. Arch. Intern. Med. 1998, 158, 585–593. [Google Scholar] [CrossRef] [PubMed]
- Gomes, F.G.; Sandim, V.; Almeida, V.H.; Rondon, A.M.R.; Succar, B.B.; Hottz, E.D.; Leal, A.C.; Vercoza, B.R.F.; Rodrigues, J.C.F.; Bozza, P.T.; et al. Breast-cancer extracellular vesicles induce platelet activation and aggregation by tissue factor-independent and -dependent mechanisms. Thromb. Res. 2017, 159, 24–32. [Google Scholar] [CrossRef] [PubMed]
- Ferroni, P.; Santilli, F.; Cavaliere, F.; Simeone, P.; Costarelli, L.; Liani, R.; Tripaldi, R.; Riondino, S.; Roselli, M.; Davi, G.; et al. Oxidant stress as a major determinant of platelet activation in invasive breast cancer. Int. J. Cancer 2017, 140, 696–704. [Google Scholar] [CrossRef] [PubMed]
- Lindeman, J.H.; Hanemaaijer, R.; Mulder, A.; Dijkstra, P.D.; Szuhai, K.; Bromme, D.; Verheijen, J.H.; Hogendoorn, P.C. Cathepsin K is the principal protease in giant cell tumor of bone. Am. J. Pathol. 2004, 165, 593–600. [Google Scholar] [CrossRef]
- Chen, B.; Platt, M.O. Multiplex zymography captures stage-specific activity profiles of cathepsins K, L, and S in human breast, lung, and cervical cancer. J. Transl. Med. 2011, 9, 109. [Google Scholar] [CrossRef] [PubMed]
- Andrade, S.S.; Gouvea, I.E.; Silva, M.C.; Castro, E.D.; de Paula, C.A.; Okamoto, D.; Oliveira, L.; Peres, G.B.; Ottaiano, T.; Facina, G.; et al. Cathepsin K induces platelet dysfunction and affects cell signaling in breast cancer - molecularly distinct behavior of cathepsin K in breast cancer. BMC Cancer 2016, 16, 173. [Google Scholar] [CrossRef] [PubMed]
- Hu, Q.; Wang, M.; Cho, M.S.; Wang, C.; Nick, A.M.; Thiagarajan, P.; Aung, F.M.; Han, X.; Sood, A.K.; Afshar-Kharghan, V. Lipid profile of platelets and platelet-derived microparticles in ovarian cancer. BBA Clin. 2016, 6, 76–81. [Google Scholar] [CrossRef] [PubMed]
- Feng, S.; Kroll, M.H.; Nick, A.M.; Sood, A.K.; Afshar-Kharghan, V. Platelets are not hyperreactive in patients with ovarian cancer. Platelets 2016, 27, 716–718. [Google Scholar] [CrossRef] [PubMed]
- Cuzick, J.; Thorat, M.A.; Bosetti, C.; Brown, P.H.; Burn, J.; Cook, N.R.; Ford, L.G.; Jacobs, E.J.; Jankowski, J.A.; La Vecchia, C.; et al. Estimates of benefits and harms of prophylactic use of aspirin in the general population. Ann. Oncol. 2015, 26, 47–57. [Google Scholar] [CrossRef] [PubMed]
- Vane, J. Towards a better aspirin. Nature 1994, 367, 215–216. [Google Scholar] [CrossRef] [PubMed]
- Guillem-Llobat, P.; Dovizio, M.; Bruno, A.; Ricciotti, E.; Cufino, V.; Sacco, A.; Grande, R.; Alberti, S.; Arena, V.; Cirillo, M.; et al. Aspirin prevents colorectal cancer metastasis in mice by splitting the crosstalk between platelets and tumor cells. Oncotarget 2016, 7, 32462–32477. [Google Scholar] [CrossRef] [PubMed]
- Lichtenberger, L.M.; Fang, D.; Bick, R.J.; Poindexter, B.J.; Phan, T.; Bergeron, A.L.; Pradhan, S.; Dial, E.J.; Vijayan, K.V. Unlocking Aspirin’s Chemopreventive Activity: Role of Irreversibly Inhibiting Platelet Cyclooxygenase-1. Cancer Prev. Res. 2017, 10, 142–152. [Google Scholar] [CrossRef] [PubMed]
- Sandler, R.S.; Halabi, S.; Baron, J.A.; Budinger, S.; Paskett, E.; Keresztes, R.; Petrelli, N.; Pipas, J.M.; Karp, D.D.; Loprinzi, C.L.; et al. A randomized trial of aspirin to prevent colorectal adenomas in patients with previous colorectal cancer. N. Engl. J. Med. 2003, 348, 883–890. [Google Scholar] [CrossRef] [PubMed]
- Fisher, B.; Costantino, J.P.; Wickerham, D.L.; Cecchini, R.S.; Cronin, W.M.; Robidoux, A.; Bevers, T.B.; Kavanah, M.T.; Atkins, J.N.; Margolese, R.G.; et al. Tamoxifen for the prevention of breast cancer: Current status of the National Surgical Adjuvant Breast and Bowel Project P-1 study. J. Natl. Cancer Inst. 2005, 97, 1652–1662. [Google Scholar] [CrossRef] [PubMed]
- Johnson, K.E.; Forward, J.A.; Tippy, M.D.; Ceglowski, J.R.; El-Husayni, S.; Kulenthirarajan, R.; Machlus, K.R.; Mayer, E.L.; Italiano, J.E., Jr.; Battinelli, E.M. Tamoxifen Directly Inhibits Platelet Angiogenic Potential and Platelet-Mediated Metastasis. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 664–674. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Wei, J.; Liu, S.; Wang, J.; Han, X.; Qin, H.; Lang, J.; Cheng, K.; Li, Y.; Qi, Y.; et al. Inhibition of platelet function using liposomal nanoparticles blocks tumor metastasis. Theranostics 2017, 7, 1062–1071. [Google Scholar] [CrossRef] [PubMed]
- Chang, Y.W.; Hsieh, P.W.; Chang, Y.T.; Lu, M.H.; Huang, T.F.; Chong, K.Y.; Liao, H.R.; Cheng, J.C.; Tseng, C.P. Identification of a novel platelet antagonist that binds to CLEC-2 and suppresses podoplanin-induced platelet aggregation and cancer metastasis. Oncotarget 2015, 6, 42733–42738. [Google Scholar] [CrossRef] [PubMed]
- Gresele, P.; Momi, S.; Falcinelli, E. Anti-platelet therapy: Phosphodiesterase inhibitors. Br. J. Clin. Pharmacol. 2011, 72, 634–646. [Google Scholar] [CrossRef] [PubMed]
- Rondina, M.T.; Weyrich, A.S. Targeting phosphodiesterases in anti-platelet therapy. Handb. Exp. Pharmacol. 2012, 210, 225–238. [Google Scholar]
- Uzawa, K.; Kasamatsu, A.; Baba, T.; Usukura, K.; Saito, Y.; Sakuma, K.; Iyoda, M.; Sakamoto, Y.; Ogawara, K.; Shiiba, M.; et al. Targeting phosphodiesterase 3B enhances cisplatin sensitivity in human cancer cells. Cancer Med. 2013, 2, 40–49. [Google Scholar] [CrossRef] [PubMed]
- Tzanakakis, G.N.; Agarwal, K.C.; Vezeridis, M.P. Prevention of human pancreatic cancer cell-induced hepatic metastasis in nude mice by dipyridamole and its analog RA-233. Cancer 1993, 71, 2466–24671. [Google Scholar] [CrossRef]
- Desai, P.B.; Duan, J.; Sridhar, R.; Damle, B.D. Reversal of doxorubicin resistance in multidrug resistant melanoma cells in vitro and in vivo by dipyridamole. Methods Find. Exp. Clin. Pharmacol. 1997, 19, 231–239. [Google Scholar] [PubMed]
- Amirkhosravi, A.; Mousa, S.A.; Amaya, M.; Blaydes, S.; Desai, H.; Meyer, T.; Francis, J.L. Inhibition of tumor cell-induced platelet aggregation and lung metastasis by the oral GpIIb/IIIa antagonist XV454. Thromb. Haemost. 2003, 90, 549–554. [Google Scholar] [CrossRef] [PubMed]
- Ishikawa, S.; Miyashita, T.; Inokuchi, M.; Hayashi, H.; Oyama, K.; Tajima, H.; Takamura, H.; Ninomiya, I.; Ahmed, A.K.; Harman, J.W.; et al. Platelets surrounding primary tumor cells are related to chemoresistance. Oncol. Rep. 2016, 36, 787–794. [Google Scholar] [CrossRef] [PubMed]
- Haslehurst, A.M.; Koti, M.; Dharsee, M.; Nuin, P.; Evans, K.; Geraci, J.; Childs, T.; Chen, J.; Li, J.; Weberpals, J.; et al. EMT transcription factors snail and slug directly contribute to cisplatin resistance in ovarian cancer. BMC Cancer 2012, 12, 91. [Google Scholar] [CrossRef] [PubMed]
- Tsukasa, K.; Ding, Q.; Yoshimitsu, M.; Miyazaki, Y.; Matsubara, S.; Takao, S. Slug contributes to gemcitabine resistance through epithelial-mesenchymal transition in CD133(+) pancreatic cancer cells. Hum. Cell 2015, 28, 167–174. [Google Scholar] [CrossRef] [PubMed]
- Sarkar, S.; Alam, M.A.; Shaw, J.; Dasgupta, A.K. Drug delivery using platelet cancer cell interaction. Pharm. Res. 2013, 30, 2785–2794. [Google Scholar] [CrossRef] [PubMed]
- Xu, P.; Zuo, H.; Zhou, R.; Wang, F.; Liu, X.; Ouyang, J.; Chen, B. Doxorubicin-loaded platelets conjugated with anti-CD22 mAbs: A novel targeted delivery system for lymphoma treatment with cardiopulmonary avoidance. Oncotarget 2017, 8, 58322–58337. [Google Scholar] [CrossRef] [PubMed]
- Xu, P.; Zuo, H.; Chen, B.; Wang, R.; Ahmed, A.; Hu, Y.; Ouyang, J. Doxorubicin-loaded platelets as a smart drug delivery system: An improved therapy for lymphoma. Sci. Rep. 2017, 7, 42632. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Ai, Y.; Wang, L.; Bu, P.; Sharkey, C.C.; Wu, Q.; Wun, B.; Roy, S.; Shen, X.; King, M.R. Targeted drug delivery to circulating tumor cells via platelet membrane-functionalized particles. Biomaterials 2016, 76, 52–65. [Google Scholar] [CrossRef] [PubMed]
Platelet Related Factors | Function | Mechanism | Inhibitors | Ref |
---|---|---|---|---|
TGF-β | Promote primary tumor growth, | TGF-β1 promotes cancer cell proliferation directly | SB431542, decorin | [46,47] |
Enhance EMT phenotype and promote tumor cell extravasation | TGF-β releasing induces the EMT phenotype depending on podoplanin | [70,71] | ||
Platelets and tumor cells contacts activate TGF-β/SMAD and NF-κb pathway | ||||
Downregulate reactivity of NK cell, inhibit antitumor immunity | TGF-β down-regulates the NKG2D expression, the activating immunoreceptor | [95,96] | ||
TGF-β downregulates inflammatory cytokine production | ||||
VEGF | Promote the angiogenesis | Enhance endothelial cell growth | [52] | |
PDGF | Promote the tumorigenesis | Stimulate the cells in tumor stroma and promote angiogenesis | Olaratumab, imatinib, sunitinib, sorafenib, pazopa-nib, nilotinib, cediranib, trapidil | [53] |
Induce EMT markers | Upregulate the expression of COX-2 | [76] | ||
PF4 | Inhibit tumor growth and metastasis | Inhibit endothelial proliferation in vitro and angiogenesis in vivo | [57] | |
Promote Kras-driven tumorigenensis | Promote platelet production and modulate the tumor mocroenvironment to accelerate the tumor growth | [55] | ||
P2Y12 | Promote primary tumor growth | Recruits Gβγ subunits, causing phosphoinositide-3-kinase- dependent Akt phosphorylation and Rap1b activation | clopidogrel, ticagrelor, prasugrel | [40,41] |
Induce ERK1/2 and paxillin Ser83 phosphorylation | ||||
MiRNA 24 | Induce the tumor growth inhibition at early stage | Transfer to tumor cells, then induce the mitochondrial dysfunction and tumor cell apoptosis | [60] | |
MiRNA 939 | promotes epithelial to mesenchymal transition | Transfer to tumor cells, downregulate E-cadherin and up-regulate vimentin | [72] | |
CLEC2 | Promote EMT and tumor extravasation in mouse model | Bind with Aggrus, attenuate Aggrus-induced platelet aggregation | 2A2B10, 2CP | [73] |
Integrin (α6β1, αIIbβ3) | Promote metastasis | Bind with molecular on tumor cell surface, such as ADAM9 | ML464, scFv Ab; A11, 7E3 F(ab’)2 | [79,80] |
LPA | Enhance bone metastasis | enhances the LPA-dependent production of IL-6 and IL-8 to stimulate osteoclast-mediated bone resorption | [81] | |
Asm | Promote tumor cell adhesion and metastasis | Activate α5β1 on melanoma cells | [88] | |
Ask1 | Promote cancer metastasis | Protect the cancer cells from anoikis | [86,87] |
© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Wang, S.; Li, Z.; Xu, R. Human Cancer and Platelet Interaction, a Potential Therapeutic Target. Int. J. Mol. Sci. 2018, 19, 1246. https://doi.org/10.3390/ijms19041246
Wang S, Li Z, Xu R. Human Cancer and Platelet Interaction, a Potential Therapeutic Target. International Journal of Molecular Sciences. 2018; 19(4):1246. https://doi.org/10.3390/ijms19041246
Chicago/Turabian StyleWang, Shike, Zhenyu Li, and Ren Xu. 2018. "Human Cancer and Platelet Interaction, a Potential Therapeutic Target" International Journal of Molecular Sciences 19, no. 4: 1246. https://doi.org/10.3390/ijms19041246
APA StyleWang, S., Li, Z., & Xu, R. (2018). Human Cancer and Platelet Interaction, a Potential Therapeutic Target. International Journal of Molecular Sciences, 19(4), 1246. https://doi.org/10.3390/ijms19041246