Breast Cancer Tumor Stroma: Cellular Components, Phenotypic Heterogeneity, Intercellular Communication, Prognostic Implications and Therapeutic Opportunities
Abstract
:1. Introduction
2. Tumor Stroma Components
2.1. Cancer-Associated Fibroblasts (CAFs)
2.2. Immune Cells
2.3. Endothelial Cells
2.4. Mesenchymal Stromal Cells
3. Stroma Phenotype Associated with Tumor Metastasis
3.1. Metalloproteases and Their Inhibitors
3.2. Cytokines
3.3. Toll-Like Receptors (TLRs)
3.4 Integrins
4. Intercellular Communication System in the Tumor Microenvironment
4.1. Soluble Factors
4.2. Exosomes
4.3. Tumor-Derived Exosomes (T-D-EXs)
4.4. MSC-Derived Exosomes (MSC-D-EXs)
4.5. Integrins and Cancer
4.6. Possible Role of Integrins to Better Characterize the Tumor Stroma Phenotype in Breast Cancer
4.7. Tumor Stroma and Therapeutic Opportunities
5. Conclusions and Future Perspectives
Author Contributions
Funding
Conflicts of Interest
References
- Balkwill, F.R.; Capasso, M.; Hagemann, T. The tumor microenvironment at a glance. J. Cell Sci. 2012, 125, 5591–5596. [Google Scholar] [CrossRef]
- Anderberg, C.; Pietras, K. On the origin of cancer-associated fibroblasts. Cell Cycle 2009, 8, 1461–1462. [Google Scholar] [CrossRef]
- Zhu, C.Q.; Popova, S.N.; Brown, E.R.; Barsyte-Lovejoy, D.; Navab, R.; Shih, W.; Li, M.; Lu, M.; Jurisica, I.; Penn, L.Z.; et al. Integrin alpha 11 regulates IGF2 expression in fibroblasts to enhance tumorigenicity of human non-small-cell lung cancer cells. Proc. Natl. Acad. Sci. USA 2007, 104, 11754–11759. [Google Scholar] [CrossRef]
- Nakagawa, H.; Liyanarachchi, S.; Davuluri, R.V.; Auer, H.; Martin, E.W., Jr.; de la Chapelle, A.; Frankel, W.L. Role of cancer-associated stromal fibroblasts in metastatic colon cancer to the liver and their expression profiles. Oncogene 2004, 23, 7366–7377. [Google Scholar] [CrossRef] [PubMed]
- Walter, K.; Omura, N.; Hong, S.M.; Griffith, M.; Vincent, A.; Borges, M.; Goggins, M. Overexpression of smoothened activates the sonic hedgehog signaling pathway in pancreatic cancer-associated fibroblasts. Clin. Cancer Res. 2010, 16, 1781–1789. [Google Scholar] [CrossRef] [PubMed]
- Glentis, A.; Oertle, P.; Mariani, P.; Chikina, A.; El Marjou, F.; Attieh, Y.; Zaccarini, F.; Lae, M.; Loew, D.; Dingli, F.; et al. Cancer-associated fibroblasts induce metalloprotease-independent cancer cell invasion of the basement membrane. Nat. Commun. 2017, 8, 924. [Google Scholar] [CrossRef]
- Koontongkaew, S. The tumor microenvironment contribution to development, growth, invasion and metastasis of head and neck squamous cell carcinomas. J. Cancer 2013, 4, 66–83. [Google Scholar] [CrossRef]
- Madar, S.; Goldstein, I.; Rotter, V. ‘Cancer associated fibroblasts’—More than meets the eye. Trends Mol. Med. 2013, 19, 447–453. [Google Scholar] [CrossRef]
- Erez, N.; Truitt, M.; Olson, P.; Arron, S.T.; Hanahan, D. Cancer-Associated Fibroblasts Are Activated in Incipient Neoplasia to Orchestrate Tumor-Promoting Inflammation in an NF-kappaB-Dependent Manner. Cancer Cell 2010, 17, 135–147. [Google Scholar] [CrossRef] [PubMed]
- Kalluri, R.; Zeisberg, M. Fibroblasts in cancer. Nat. Rev. Cancer 2006, 6, 392–401. [Google Scholar] [CrossRef] [PubMed]
- Allen, M.; Louise Jones, J. Jekyll and Hyde: The role of the microenvironment on the progression of cancer. J. Pathol. 2011, 223, 162–176. [Google Scholar] [CrossRef]
- Mao, Y.; Keller, E.T.; Garfield, D.H.; Shen, K.; Wang, J. Stromal cells in tumor microenvironment and breast cancer. Cancer Metastasis Rev. 2013, 32, 303–315. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez, L.; Eiro, N.; Fernandez-Garcia, B.; Gonzalez, L.O.; Dominguez, F.; Vizoso, F.J. Gene expression profile of normal and cancer-associated fibroblasts according to intratumoral inflammatory cells phenotype from breast cancer tissue. Mol. Carcinog. 2016, 55, 1489–1502. [Google Scholar] [CrossRef] [PubMed]
- Kogan-Sakin, I.; Cohen, M.; Paland, N.; Madar, S.; Solomon, H.; Molchadsky, A.; Brosh, R.; Buganim, Y.; Goldfinger, N.; Klocker, H.; et al. Prostate stromal cells produce CXCL-1, CXCL-2, CXCL-3 and IL-8 in response to epithelia-secreted IL-1. Carcinogenesis 2009, 30, 698–705. [Google Scholar] [CrossRef] [PubMed]
- Protti, M.P.; De Monte, L. Cross-talk within the tumor microenvironment mediates Th2-type inflammation in pancreatic cancer. Oncoimmunology 2012, 1, 89–91. [Google Scholar] [CrossRef]
- Straussman, R.; Morikawa, T.; Shee, K.; Barzily-Rokni, M.; Qian, Z.R.; Du, J.; Davis, A.; Mongare, M.M.; Gould, J.; Frederick, D.T.; et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 2012, 487, 500–504. [Google Scholar] [CrossRef]
- Sun, X.; Mao, Y.; Wang, J.; Zu, L.; Hao, M.; Cheng, G.; Qu, Q.; Cui, D.; Keller, E.T.; Chen, X.; et al. IL-6 secreted by cancer-associated fibroblasts induces tamoxifen resistance in luminal breast cancer. Oncogene 2014. [Google Scholar] [CrossRef]
- De Visser, K.E.; Eichten, A.; Coussens, L.M. Paradoxical roles of the immune system during cancer development. Nat. Rev. Cancer 2006, 6, 24–37. [Google Scholar] [CrossRef] [PubMed]
- Lin, E.Y.; Pollard, J.W. Role of infiltrated leucocytes in tumour growth and spread. Br. J. Cancer 2004, 90, 2053–2058. [Google Scholar] [CrossRef]
- Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
- Daniel, D.; Chiu, C.; Giraudo, E.; Inoue, M.; Mizzen, L.A.; Chu, N.R.; Hanahan, D. CD4+ T cell-mediated antigen-specific immunotherapy in a mouse model of cervical cancer. Cancer Res. 2005, 65, 2018–2025. [Google Scholar] [CrossRef]
- Sica, A.; Bronte, V. Altered macrophage differentiation and immune dysfunction in tumor development. J. Clin. Investig. 2007, 117, 1155–1166. [Google Scholar] [CrossRef] [PubMed]
- Balkwill, F. Cancer and the chemokine network. Nat. Rev. Cancer 2004, 4, 540–550. [Google Scholar] [CrossRef] [PubMed]
- Le Bitoux, M.A.; Stamenkovic, I. Tumor-host interactions: The role of inflammation. Histochem. Cell Biol. 2008, 130, 1079–1090. [Google Scholar] [CrossRef] [PubMed]
- Bingle, L.; Brown, N.J.; Lewis, C.E. The role of tumour-associated macrophages in tumour progression: Implications for new anticancer therapies. J. Pathol. 2002, 196, 254–265. [Google Scholar] [CrossRef]
- Lewis, C.E.; Pollard, J.W. Distinct role of macrophages in different tumor microenvironments. Cancer Res. 2006, 66, 605–612. [Google Scholar] [CrossRef]
- Alexe, G.; Dalgin, G.S.; Scanfeld, D.; Tamayo, P.; Mesirov, J.P.; DeLisi, C.; Harris, L.; Barnard, N.; Martel, M.; Levine, A.J.; et al. High expression of lymphocyte-associated genes in node-negative HER2+ breast cancers correlates with lower recurrence rates. Cancer Res. 2007, 67, 10669–10676. [Google Scholar] [CrossRef] [PubMed]
- Arnould, L.; Gelly, M.; Penault-Llorca, F.; Benoit, L.; Bonnetain, F.; Migeon, C.; Cabaret, V.; Fermeaux, V.; Bertheau, P.; Garnier, J.; et al. Trastuzumab-based treatment of HER2-positive breast cancer: An antibody-dependent cellular cytotoxicity mechanism? Br. J. Cancer 2006, 94, 259–267. [Google Scholar] [CrossRef]
- Bates, G.J.; Fox, S.B.; Han, C.; Leek, R.D.; Garcia, J.F.; Harris, A.L.; Banham, A.H. Quantification of regulatory T cells enables the identification of high-risk breast cancer patients and those at risk of late relapse. J. Clin. Oncol. 2006, 24, 5373–5380. [Google Scholar] [CrossRef]
- Desmedt, C.; Haibe-Kains, B.; Wirapati, P.; Buyse, M.; Larsimont, D.; Bontempi, G.; Delorenzi, M.; Piccart, M.; Sotiriou, C. Biological processes associated with breast cancer clinical outcome depend on the molecular subtypes. Clin. Cancer Res. 2008, 14, 5158–5165. [Google Scholar] [CrossRef]
- Rody, A.; Holtrich, U.; Pusztai, L.; Liedtke, C.; Gaetje, R.; Ruckhaeberle, E.; Solbach, C.; Hanker, L.; Ahr, A.; Metzler, D.; et al. T-cell metagene predicts a favorable prognosis in estrogen receptor-negative and HER2-positive breast cancers. Breast Cancer Res. 2009, 11, R15. [Google Scholar] [CrossRef]
- Denkert, C.; Loibl, S.; Noske, A.; Roller, M.; Muller, B.M.; Komor, M.; Budczies, J.; Darb-Esfahani, S.; Kronenwett, R.; Hanusch, C.; et al. Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J. Clin. Oncol. 2010, 28, 105–113. [Google Scholar] [CrossRef] [PubMed]
- Lofdahl, B.; Ahlin, C.; Holmqvist, M.; Holmberg, L.; Zhou, W.; Fjallskog, M.L.; Amini, R.M. Inflammatory cells in node-negative breast cancer. Acta Oncol. 2012, 51, 680–686. [Google Scholar] [CrossRef] [PubMed]
- Mahmoud, S.M.; Paish, E.C.; Powe, D.G.; Macmillan, R.D.; Lee, A.H.; Ellis, I.O.; Green, A.R. An evaluation of the clinical significance of FOXP3+ infiltrating cells in human breast cancer. Breast Cancer Res. Treat. 2011, 127, 99–108. [Google Scholar] [CrossRef] [PubMed]
- Mahmoud, S.M.; Lee, A.H.; Paish, E.C.; Macmillan, R.D.; Ellis, I.O.; Green, A.R. The prognostic significance of B lymphocytes in invasive carcinoma of the breast. Breast Cancer Res. Treat. 2012, 132, 545–553. [Google Scholar] [CrossRef] [PubMed]
- Takenaka, M.; Seki, N.; Toh, U.; Hattori, S.; Kawahara, A.; Yamaguchi, T.; Koura, K.; Takahashi, R.; Otsuka, H.; Takahashi, H.; et al. FOXP3 expression in tumor cells and tumor-infiltrating lymphocytes is associated with breast cancer prognosis. Mol. Clin. Oncol. 2013, 1, 625–632. [Google Scholar] [CrossRef] [PubMed]
- Ibrahim, E.M.; Al-Foheidi, M.E.; Al-Mansour, M.M.; Kazkaz, G.A. The prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancer: A meta-analysis. Breast Cancer Res. Treat. 2014, 148, 467–476. [Google Scholar] [CrossRef]
- Yeong, J.; Thike, A.A.; Lim, J.C.; Lee, B.; Li, H.; Wong, S.C.; Hue, S.S.; Tan, P.H.; Iqbal, J. Higher densities of Foxp3(+) regulatory T cells are associated with better prognosis in triple-negative breast cancer. Breast Cancer Res. Treat. 2017, 163, 21–35. [Google Scholar] [CrossRef]
- Eiro, N.; Pidal, I.; Fernandez-Garcia, B.; Junquera, S.; Lamelas, M.L.; del Casar, J.M.; Gonzalez, L.O.; Lopez-Muniz, A.; Vizoso, F.J. Impact of CD68/(CD3+CD20) ratio at the invasive front of primary tumors on distant metastasis development in breast cancer. PLoS ONE 2012, 7, e52796. [Google Scholar] [CrossRef] [PubMed]
- Webb, E.S.; Liu, P.; Baleeiro, R.; Lemoine, N.R.; Yuan, M.; Wang, Y.H. Immune checkpoint inhibitors in cancer therapy. J. Biomed. Res. 2018, 32, 317–326. [Google Scholar] [CrossRef] [PubMed]
- Butler, J.M.; Kobayashi, H.; Rafii, S. Instructive role of the vascular niche in promoting tumour growth and tissue repair by angiocrine factors. Nat. Rev. Cancer 2010, 10, 138–146. [Google Scholar] [CrossRef]
- Butler, J.M.; Rafii, S. Generation of a vascular niche for studying stem cell homeostasis. Methods Mol. Biol. 2012, 904, 221–233. [Google Scholar] [CrossRef]
- Ghajar, C.M.; Peinado, H.; Mori, H.; Matei, I.R.; Evason, K.J.; Brazier, H.; Almeida, D.; Koller, A.; Hajjar, K.A.; Stainier, D.Y.; et al. The perivascular niche regulates breast tumour dormancy. Nat. Cell Biol. 2013, 15, 807–817. [Google Scholar] [CrossRef]
- Ghiabi, P.; Jiang, J.; Pasquier, J.; Maleki, M.; Abu-Kaoud, N.; Rafii, S.; Rafii, A. Endothelial cells provide a notch-dependent pro-tumoral niche for enhancing breast cancer survival, stemness and pro-metastatic properties. PLoS ONE 2014, 9, e112424. [Google Scholar] [CrossRef]
- Folkman, J.; Watson, K.; Ingber, D.; Hanahan, D. Induction of angiogenesis during the transition from hyperplasia to neoplasia. Nature 1989, 339, 58–61. [Google Scholar] [CrossRef]
- Hida, K.; Ohga, N.; Akiyama, K.; Maishi, N.; Hida, Y. Heterogeneity of tumor endothelial cells. Cancer Sci. 2013, 104, 1391–1395. [Google Scholar] [CrossRef]
- Lazennec, G.; Jorgensen, C. Concise review: Adult multipotent stromal cells and cancer: Risk or benefit? Stem Cells 2008, 26, 1387–1394. [Google Scholar] [CrossRef]
- Dominici, M.; Le Blanc, K.; Mueller, I.; Slaper-Cortenbach, I.; Marini, F.; Krause, D.; Deans, R.; Keating, A.; Prockop, D.; Horwitz, E. Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 2006, 8, 315–317. [Google Scholar] [CrossRef]
- Timmers, L.; Lim, S.K.; Arslan, F.; Armstrong, J.S.; Hoefer, I.E.; Doevendans, P.A.; Piek, J.J.; El Oakley, R.M.; Choo, A.; Lee, C.N.; et al. Reduction of myocardial infarct size by human mesenchymal stem cell conditioned medium. Stem Cell Res. 2007, 1, 129–137. [Google Scholar] [CrossRef]
- Ridge, S.M.; Sullivan, F.J.; Glynn, S.A. Mesenchymal stem cells: Key players in cancer progression. Mol. Cancer 2017, 16, 31. [Google Scholar] [CrossRef]
- Lazennec, G.; Lam, P.Y. Recent discoveries concerning the tumor—Mesenchymal stem cell interactions. Biochim. Biophys. Acta 2016, 1866, 290–299. [Google Scholar] [CrossRef]
- Hass, R.; Otte, A. Mesenchymal stem cells as all-round supporters in a normal and neoplastic microenvironment. Cell Commun. Signal 2012, 10, 26. [Google Scholar] [CrossRef]
- El-Haibi, C.P.; Karnoub, A.E. Mesenchymal stem cells in the pathogenesis and therapy of breast cancer. J. Mammary Gland Biol. Neoplasia 2010, 15, 399–409. [Google Scholar] [CrossRef] [PubMed]
- Anthony, B.A.; Link, D.C. Regulation of hematopoietic stem cells by bone marrow stromal cells. Trends Immunol. 2014, 35, 32–37. [Google Scholar] [CrossRef] [PubMed]
- Sanchez, C.A.; Andahur, E.I.; Valenzuela, R.; Castellon, E.A.; Fulla, J.A.; Ramos, C.G.; Trivino, J.C. Exosomes from bulk and stem cells from human prostate cancer have a differential microRNA content that contributes cooperatively over local and pre-metastatic niche. Oncotarget 2016, 7, 3993–4008. [Google Scholar] [CrossRef] [PubMed]
- Zhu, W.; Huang, L.; Li, Y.; Zhang, X.; Gu, J.; Yan, Y.; Xu, X.; Wang, M.; Qian, H.; Xu, W. Exosomes derived from human bone marrow mesenchymal stem cells promote tumor growth in vivo. Cancer Lett. 2012, 315, 28–37. [Google Scholar] [CrossRef] [PubMed]
- Melzer, C.; Yang, Y.; Hass, R. Interaction of MSC with tumor cells. Cell Commun. Signal 2016, 14, 20. [Google Scholar] [CrossRef]
- Szczepanski, M.J.; Szajnik, M.; Welsh, A.; Whiteside, T.L.; Boyiadzis, M. Blast-derived microvesicles in sera from patients with acute myeloid leukemia suppress natural killer cell function via membrane-associated transforming growth factor-beta1. Haematologica 2011, 96, 1302–1309. [Google Scholar] [CrossRef]
- Mandel, K.; Yang, Y.; Schambach, A.; Glage, S.; Otte, A.; Hass, R. Mesenchymal stem cells directly interact with breast cancer cells and promote tumor cell growth in vitro and in vivo. Stem Cells Dev. 2013, 22, 3114–3127. [Google Scholar] [CrossRef] [PubMed]
- Syn, N.; Wang, L.; Sethi, G.; Thiery, J.P.; Goh, B.C. Exosome-Mediated Metastasis: From Epithelial-Mesenchymal Transition to Escape from Immunosurveillance. Trends Pharmacol. Sci. 2016, 37, 606–617. [Google Scholar] [CrossRef]
- Shi, S.; Zhang, Q.; Xia, Y.; You, B.; Shan, Y.; Bao, L.; Li, L.; You, Y.; Gu, Z. Mesenchymal stem cell-derived exosomes facilitate nasopharyngeal carcinoma progression. Am. J. Cancer Res. 2016, 6, 459–472. [Google Scholar]
- Meng, F.; Henson, R.; Wehbe-Janek, H.; Ghoshal, K.; Jacob, S.T.; Patel, T. MicroRNA-21 regulates expression of the PTEN tumor suppressor gene in human hepatocellular cancer. Gastroenterology 2007, 133, 647–658. [Google Scholar] [CrossRef]
- Provenzano, P.P.; Inman, D.R.; Eliceiri, K.W.; Keely, P.J. Matrix density-induced mechanoregulation of breast cell phenotype, signaling and gene expression through a FAK-ERK linkage. Oncogene 2009, 28, 4326–4343. [Google Scholar] [CrossRef] [PubMed]
- Narayanan, R.; Huang, C.C.; Ravindran, S. Hijacking the Cellular Mail: Exosome Mediated Differentiation of Mesenchymal Stem Cells. Stem Cells Int. 2016, 2016, 3808674. [Google Scholar] [CrossRef] [PubMed]
- Cho, J.A.; Park, H.; Lim, E.H.; Lee, K.W. Exosomes from breast cancer cells can convert adipose tissue-derived mesenchymal stem cells into myofibroblast-like cells. Int. J. Oncol. 2012, 40, 130–138. [Google Scholar] [CrossRef]
- Zhang, B.; Yin, Y.; Lai, R.C.; Tan, S.S.; Choo, A.B.; Lim, S.K. Mesenchymal stem cells secrete immunologically active exosomes. Stem Cells Dev. 2014, 23, 1233–1244. [Google Scholar] [CrossRef]
- Mokarizadeh, A.; Delirezh, N.; Morshedi, A.; Mosayebi, G.; Farshid, A.A.; Mardani, K. Microvesicles derived from mesenchymal stem cells: Potent organelles for induction of tolerogenic signaling. Immunol. Lett. 2012, 147, 47–54. [Google Scholar] [CrossRef]
- Del Fattore, A.; Luciano, R.; Pascucci, L.; Goffredo, B.M.; Giorda, E.; Scapaticci, M.; Fierabracci, A.; Muraca, M. Immunoregulatory Effects of Mesenchymal Stem Cell-Derived Extracellular Vesicles on T Lymphocytes. Cell Transplant. 2015, 24, 2615–2627. [Google Scholar] [CrossRef]
- Phinney, D.G.; Di Giuseppe, M.; Njah, J.; Sala, E.; Shiva, S.; St Croix, C.M.; Stolz, D.B.; Watkins, S.C.; Di, Y.P.; Leikauf, G.D.; et al. Mesenchymal stem cells use extracellular vesicles to outsource mitophagy and shuttle microRNAs. Nat. Commun. 2015, 6, 8472. [Google Scholar] [CrossRef]
- Conforti, A.; Scarsella, M.; Starc, N.; Giorda, E.; Biagini, S.; Proia, A.; Carsetti, R.; Locatelli, F.; Bernardo, M.E. Microvescicles derived from mesenchymal stromal cells are not as effective as their cellular counterpart in the ability to modulate immune responses in vitro. Stem Cells Dev. 2014, 23, 2591–2599. [Google Scholar] [CrossRef]
- Chen, W.; Huang, Y.; Han, J.; Yu, L.; Li, Y.; Lu, Z.; Li, H.; Liu, Z.; Shi, C.; Duan, F.; et al. Immunomodulatory effects of mesenchymal stromal cells-derived exosome. Immunol. Res. 2016, 64, 831–840. [Google Scholar] [CrossRef]
- Amarnath, S.; Foley, J.E.; Farthing, D.E.; Gress, R.E.; Laurence, A.; Eckhaus, M.A.; Metais, J.Y.; Rose, J.J.; Hakim, F.T.; Felizardo, T.C.; et al. Bone marrow-derived mesenchymal stromal cells harness purinergenic signaling to tolerize human Th1 cells in vivo. Stem Cells 2015, 33, 1200–1212. [Google Scholar] [CrossRef] [PubMed]
- Ono, M.; Kosaka, N.; Tominaga, N.; Yoshioka, Y.; Takeshita, F.; Takahashi, R.U.; Yoshida, M.; Tsuda, H.; Tamura, K.; Ochiya, T. Exosomes from bone marrow mesenchymal stem cells contain a microRNA that promotes dormancy in metastatic breast cancer cells. Sci. Signal. 2014, 7, ra63. [Google Scholar] [CrossRef]
- Ohyashiki, J.H.; Umezu, T.; Ohyashiki, K. Exosomes promote bone marrow angiogenesis in hematologic neoplasia: The role of hypoxia. Curr. Opin. Hematol. 2016, 23, 268–273. [Google Scholar] [CrossRef]
- Allavena, P.; Sica, A.; Garlanda, C.; Mantovani, A. The Yin-Yang of tumor-associated macrophages in neoplastic progression and immune surveillance. Immunol. Rev. 2008, 222, 155–161. [Google Scholar] [CrossRef]
- Chen, H.W.; Chen, H.Y.; Wang, L.T.; Wang, F.H.; Fang, L.W.; Lai, H.Y.; Chen, H.H.; Lu, J.; Hung, M.S.; Cheng, Y.; et al. Mesenchymal stem cells tune the development of monocyte-derived dendritic cells toward a myeloid-derived suppressive phenotype through growth-regulated oncogene chemokines. J. Immunol. 2013, 190, 5065–5077. [Google Scholar] [CrossRef] [PubMed]
- Waterman, R.S.; Henkle, S.L.; Betancourt, A.M. Mesenchymal stem cell 1 (MSC1)-based therapy attenuates tumor growth whereas MSC2-treatment promotes tumor growth and metastasis. PLoS ONE 2012, 7, e45590. [Google Scholar] [CrossRef] [PubMed]
- Egeblad, M.; Werb, Z. New functions for the matrix metalloproteinases in cancer progression. Nat. Rev. Cancer 2002, 2, 161–174. [Google Scholar] [CrossRef] [PubMed]
- Noe, V.; Fingleton, B.; Jacobs, K.; Crawford, H.C.; Vermeulen, S.; Steelant, W.; Bruyneel, E.; Matrisian, L.M.; Mareel, M. Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J. Cell Sci. 2001, 114, 111–118. [Google Scholar]
- Rundhaug, J.E. Matrix metalloproteinases and angiogenesis. J. Cell. Mol. Med. 2005, 9, 267–285. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Y.; Goldberg, I.D.; Shi, Y.E. Complex roles of tissue inhibitors of metalloproteinases in cancer. Oncogene 2002, 21, 2245–2252. [Google Scholar] [CrossRef]
- Wurtz, S.O.; Schrohl, A.S.; Sorensen, N.M.; Lademann, U.; Christensen, I.J.; Mouridsen, H.; Brunner, N. Tissue inhibitor of metalloproteinases-1 in breast cancer. Endocr. Relat. Cancer 2005, 12, 215–227. [Google Scholar] [CrossRef]
- Jiang, X.; Dutton, C.M.; Qi, W.N.; Block, J.A.; Garamszegi, N.; Scully, S.P. siRNA mediated inhibition of MMP-1 reduces invasive potential of a human chondrosarcoma cell line. J. Cell. Physiol. 2005, 202, 723–730. [Google Scholar] [CrossRef]
- Jones, J.L.; Walker, R.A. Control of matrix metalloproteinase activity in cancer. J. Pathol. 1997, 183, 377–379. [Google Scholar] [CrossRef]
- Nakopoulou, L.; Panayotopoulou, E.G.; Giannopoulou, I.; Alexandrou, P.; Katsarou, S.; Athanassiadou, P.; Keramopoulos, A. Stromelysin-3 protein expression in invasive breast cancer: Relation to proliferation, cell survival and patients’ outcome. Mod. Pathol. 2002, 15, 1154–1161. [Google Scholar] [CrossRef]
- Vizoso, F.J.; Gonzalez, L.O.; Corte, M.D.; Rodriguez, J.C.; Vazquez, J.; Lamelas, M.L.; Junquera, S.; Merino, A.M.; Garcia-Muniz, J.L. Study of matrix metalloproteinases and their inhibitors in breast cancer. Br. J. Cancer 2007, 96, 903–911. [Google Scholar] [CrossRef]
- Eiro, N.; Fernandez-Garcia, B.; Vazquez, J.; Del Casar, J.M.; Gonzalez, L.O.; Vizoso, F.J. A phenotype from tumor stroma based on the expression of metalloproteases and their inhibitors, associated with prognosis in breast cancer. Oncoimmunology 2015, 4, e992222. [Google Scholar] [CrossRef]
- Eiró, N.; Fernandez-Garcia, B.; González, L.O.; Vizoso, F.J. Clinical Relevance of Matrix Metalloproteases and their Inhibitors in Breast Cancer. J. Carcinog. Mutagen. 2013, s13. [Google Scholar] [CrossRef]
- Gonzalez, L.O.; Gonzalez-Reyes, S.; Marin, L.; Gonzalez, L.; Gonzalez, J.M.; Lamelas, M.L.; Merino, A.M.; Rodriguez, E.; Pidal, I.; del Casar, J.M.; et al. Comparative analysis and clinical value of the expression of metalloproteases and their inhibitors by intratumour stromal mononuclear inflammatory cells and those at the invasive front of breast carcinomas. Histopathology 2010, 57, 862–876. [Google Scholar] [CrossRef]
- Del Casar, J.M.; Gonzalez, L.O.; Alvarez, E.; Junquera, S.; Marin, L.; Gonzalez, L.; Bongera, M.; Vazquez, J.; Vizoso, F.J. Comparative analysis and clinical value of the expression of metalloproteases and their inhibitors by intratumor stromal fibroblasts and those at the invasive front of breast carcinomas. Breast Cancer Res. Treat. 2009, 116, 39–52. [Google Scholar] [CrossRef]
- Gonzalez, L.O.; Corte, M.D.; Junquera, S.; Gonzalez-Fernandez, R.; del Casar, J.M.; Garcia, C.; Andicoechea, A.; Vazquez, J.; Perez-Fernandez, R.; Vizoso, F.J. Expression and prognostic significance of metalloproteases and their inhibitors in luminal A and basal-like phenotypes of breast carcinoma. Hum. Pathol. 2009, 40, 1224–1233. [Google Scholar] [CrossRef]
- Del Casar, J.M.; Gonzalez-Reyes, S.; Gonzalez, L.O.; Gonzalez, J.M.; Junquera, S.; Bongera, M.; Garcia, M.F.; Andicoechea, A.; Serra, C.; Vizoso, F.J. Expression of metalloproteases and their inhibitors in different histological types of breast cancer. J. Cancer Res. Clin. Oncol. 2010, 136, 811–819. [Google Scholar] [CrossRef] [PubMed]
- Eiro, N.; Gonzalez, L.; Gonzalez, L.O.; Fernandez-Garcia, B.; Lamelas, M.L.; Marin, L.; Gonzalez-Reyes, S.; del Casar, J.M.; Vizoso, F.J. Relationship between the inflammatory molecular profile of breast carcinomas and distant metastasis development. PLoS ONE 2012, 7, e49047. [Google Scholar] [CrossRef] [PubMed]
- Eiro, N.; Fernandez-Garcia, B.; Gonzalez, L.O.; Vizoso, F.J. Cytokines related to MMP-11 expression by inflammatory cells and breast cancer metastasis. Oncoimmunology 2013, 2, e24010. [Google Scholar] [CrossRef]
- Manicone, A.M.; McGuire, J.K. Matrix metalloproteinases as modulators of inflammation. Semin. Cell Dev. Biol. 2008, 19, 34–41. [Google Scholar] [CrossRef]
- Cid, S.; Eiro, N.; Gonzalez, L.O.; Beridze, N.; Vazquez, J.; Vizoso, F.J. Expression and Clinical Significance of Metalloproteases and Their Inhibitors by Endothelial Cells From Invasive Breast Carcinomas. Clin. Breast Cancer 2016, 16, e83–e91. [Google Scholar] [CrossRef] [PubMed]
- Qi, J.H.; Ebrahem, Q.; Moore, N.; Murphy, G.; Claesson-Welsh, L.; Bond, M.; Baker, A.; Anand-Apte, B. A novel function for tissue inhibitor of metalloproteinases-3 (TIMP3): Inhibition of angiogenesis by blockage of VEGF binding to VEGF receptor-2. Nat. Med. 2003, 9, 407–415. [Google Scholar] [CrossRef]
- Cruz-Munoz, W.; Kim, I.; Khokha, R. TIMP-3 deficiency in the host, but not in the tumor, enhances tumor growth and angiogenesis. Oncogene 2006, 25, 650–655. [Google Scholar] [CrossRef]
- Qi, J.H.; Anand-Apte, B. Tissue inhibitor of metalloproteinase-3 (TIMP3) promotes endothelial apoptosis via a caspase-independent mechanism. Apoptosis 2015, 20, 523–534. [Google Scholar] [CrossRef]
- Zamarron, B.F.; Chen, W. Dual roles of immune cells and their factors in cancer development and progression. Int. J. Biol. Sci. 2011, 7, 651–658. [Google Scholar] [CrossRef]
- Landskron, G.; De la Fuente, M.; Thuwajit, P.; Thuwajit, C.; Hermoso, M.A. Chronic inflammation and cytokines in the tumor microenvironment. J. Immunol. Res. 2014, 2014, 149185. [Google Scholar] [CrossRef]
- Esquivel-Velazquez, M.; Ostoa-Saloma, P.; Palacios-Arreola, M.I.; Nava-Castro, K.E.; Castro, J.I.; Morales-Montor, J. The role of cytokines in breast cancer development and progression. J. Interferon Cytokine Res. 2015, 35, 1–16. [Google Scholar] [CrossRef]
- Lin, S.; Gan, Z.; Han, K.; Yao, Y.; Min, D. Interleukin-6 as a prognostic marker for breast cancer: A meta-analysis. Tumori 2015, 101, 535–541. [Google Scholar] [CrossRef] [PubMed]
- Tripsianis, G.; Papadopoulou, E.; Anagnostopoulos, K.; Botaitis, S.; Katotomichelakis, M.; Romanidis, K.; Kontomanolis, E.; Tentes, I.; Kortsaris, A. Coexpression of IL-6 and TNF-alpha: Prognostic significance on breast cancer outcome. Neoplasma 2014, 61, 205–212. [Google Scholar] [CrossRef] [PubMed]
- Fernandez-Garcia, B.; Eiro, N.; Miranda, M.A.; Cid, S.; Gonzalez, L.O.; Dominguez, F.; Vizoso, F.J. Prognostic significance of inflammatory factors expression by stroma from breast carcinomas. Carcinogenesis 2016, 37, 768–776. [Google Scholar] [CrossRef] [PubMed]
- Hamidullah; Changkija, B.; Konwar, R. Role of interleukin-10 in breast cancer. Breast Cancer Res. Treat. 2012, 133, 11–21. [Google Scholar] [CrossRef]
- Murugaiyan, G.; Saha, B. Protumor vs antitumor functions of IL-17. J. Immunol. 2009, 183, 4169–4175. [Google Scholar] [CrossRef]
- Martin-Orozco, N.; Muranski, P.; Chung, Y.; Yang, X.O.; Yamazaki, T.; Lu, S.; Hwu, P.; Restifo, N.P.; Overwijk, W.W.; Dong, C. T helper 17 cells promote cytotoxic T cell activation in tumor immunity. Immunity 2009, 31, 787–798. [Google Scholar] [CrossRef]
- Karczewska, A.; Nawrocki, S.; Breborowicz, D.; Filas, V.; Mackiewicz, A. Expression of interleukin-6, interleukin-6 receptor, and glycoprotein 130 correlates with good prognoses for patients with breast carcinoma. Cancer 2000, 88, 2061–2071. [Google Scholar] [CrossRef]
- Benchetrit, F.; Ciree, A.; Vives, V.; Warnier, G.; Gey, A.; Sautes-Fridman, C.; Fossiez, F.; Haicheur, N.; Fridman, W.H.; Tartour, E. Interleukin-17 inhibits tumor cell growth by means of a T-cell-dependent mechanism. Blood 2002, 99, 2114–2121. [Google Scholar] [CrossRef]
- Kawai, T.; Akira, S. TLR signaling. Cell Death Differ. 2006, 13, 816–825. [Google Scholar] [CrossRef]
- Sato, Y.; Goto, Y.; Narita, N.; Hoon, D.S. Cancer Cells Expressing Toll-like Receptors and the Tumor Microenvironment. Cancer Microenviron. 2009, 2 (Suppl. S1), 205–214. [Google Scholar] [CrossRef]
- Huang, B.; Zhao, J.; Unkeless, J.C.; Feng, Z.H.; Xiong, H. TLR signaling by tumor and immune cells: A double-edged sword. Oncogene 2008, 27, 218–224. [Google Scholar] [CrossRef]
- Rakoff-Nahoum, S.; Medzhitov, R. Role of toll-like receptors in tissue repair and tumorigenesis. Biochemistry 2008, 73, 555–561. [Google Scholar] [CrossRef]
- Gonzalez-Reyes, S.; Marin, L.; Gonzalez, L.; Gonzalez, L.O.; del Casar, J.M.; Lamelas, M.L.; Gonzalez-Quintana, J.M.; Vizoso, F.J. Study of TLR3, TLR4 and TLR9 in breast carcinomas and their association with metastasis. BMC Cancer 2010, 10, 665. [Google Scholar] [CrossRef]
- Murad, Y.M.; Clay, T.M.; Lyerly, H.K.; Morse, M.A. CPG-7909 (PF-3512676, ProMune): Toll-like receptor-9 agonist in cancer therapy. Expert Opin. Biol. Ther. 2007, 7, 1257–1266. [Google Scholar] [CrossRef]
- Sagiv-Barfi, I.; Czerwinski, D.K.; Levy, S.; Alam, I.S.; Mayer, A.T.; Gambhir, S.S.; Levy, R. Eradication of spontaneous malignancy by local immunotherapy. Sci. Transl. Med. 2018, 10. [Google Scholar] [CrossRef]
- Lambert, A.W.; Ozturk, S.; Thiagalingam, S. Integrin signaling in mammary epithelial cells and breast cancer. ISRN Oncol. 2012, 2012, 493283. [Google Scholar] [CrossRef]
- Campbell, I.D.; Humphries, M.J. Integrin structure, activation, and interactions. Cold Spring Harb. Perspect. Biol. 2011, 3. [Google Scholar] [CrossRef]
- Mousa, S.A. Cell adhesion molecules: Potential therapeutic & diagnostic implications. Mol. Biotechnol. 2008, 38, 33–40. [Google Scholar] [CrossRef]
- Calderwood, D.A.; Campbell, I.D.; Critchley, D.R. Talins and kindlins: Partners in integrin-mediated adhesion. Nat. Rev. Mol. Cell Biol. 2013, 14, 503–517. [Google Scholar] [CrossRef]
- Desgrosellier, J.S.; Cheresh, D.A. Integrins in cancer: Biological implications and therapeutic opportunities. Nat. Rev. Cancer 2010, 10, 9–22. [Google Scholar] [CrossRef]
- Legate, K.R.; Montanez, E.; Kudlacek, O.; Fassler, R. ILK, PINCH and parvin: The tIPP of integrin signalling. Nat. Rev. Mol. Cell Biol. 2006, 7, 20–31. [Google Scholar] [CrossRef]
- Hynes, R.O. Integrins: Bidirectional, allosteric signaling machines. Cell 2002, 110, 673–687. [Google Scholar] [CrossRef]
- Bianconi, D.; Unseld, M.; Prager, G.W. Integrins in the Spotlight of Cancer. Int. J. Mol. Sci. 2016, 17, 2037. [Google Scholar] [CrossRef]
- Winograd-Katz, S.E.; Fassler, R.; Geiger, B.; Legate, K.R. The integrin adhesome: From genes and proteins to human disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 273–288. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
- Yuan, Y.; Jiang, Y.C.; Sun, C.K.; Chen, Q.M. Role of the tumor microenvironment in tumor progression and the clinical applications (Review). Oncol. Rep. 2016, 35, 2499–2515. [Google Scholar] [CrossRef]
- Derbal, Y. Perspective on the dynamics of cancer. Theor. Biol. Med. Model. 2017, 14, 18. [Google Scholar] [CrossRef]
- Gandellini, P.; Andriani, F.; Merlino, G.; D’Aiuto, F.; Roz, L.; Callari, M. Complexity in the tumour microenvironment: Cancer associated fibroblast gene expression patterns identify both common and unique features of tumour-stroma crosstalk across cancer types. Semin. Cancer Biol. 2015, 35, 96–106. [Google Scholar] [CrossRef]
- Harper, J.; Sainson, R.C. Regulation of the anti-tumour immune response by cancer-associated fibroblasts. Semin. Cancer Biol. 2014, 25, 69–77. [Google Scholar] [CrossRef]
- Popivanova, B.K.; Kostadinova, F.I.; Furuichi, K.; Shamekh, M.M.; Kondo, T.; Wada, T.; Egashira, K.; Mukaida, N. Blockade of a chemokine, CCL2, reduces chronic colitis-associated carcinogenesis in mice. Cancer Res. 2009, 69, 7884–7892. [Google Scholar] [CrossRef]
- Roca, H.; Varsos, Z.S.; Sud, S.; Craig, M.J.; Ying, C.; Pienta, K.J. CCL2 and interleukin-6 promote survival of human CD11b+ peripheral blood mononuclear cells and induce M2-type macrophage polarization. J. Biol. Chem. 2009, 284, 34342–34354. [Google Scholar] [CrossRef]
- Sumimoto, H.; Imabayashi, F.; Iwata, T.; Kawakami, Y. The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J. Exp. Med. 2006, 203, 1651–1656. [Google Scholar] [CrossRef]
- Shchors, K.; Shchors, E.; Rostker, F.; Lawlor, E.R.; Brown-Swigart, L.; Evan, G.I. The Myc-dependent angiogenic switch in tumors is mediated by interleukin 1beta. Genes Dev. 2006, 20, 2527–2538. [Google Scholar] [CrossRef]
- Eiro, N.; Gonzalez, L.; Martinez-Ordonez, A.; Fernandez-Garcia, B.; Gonzalez, L.O.; Cid, S.; Dominguez, F.; Perez-Fernandez, R.; Vizoso, F.J. Cancer-associated fibroblasts affect breast cancer cell gene expression, invasion and angiogenesis. Cell. Oncol. 2018, 41, 369–378. [Google Scholar] [CrossRef]
- Cocucci, E.; Meldolesi, J. Ectosomes and exosomes: Shedding the confusion between extracellular vesicles. Trends Cell Biol. 2015, 25, 364–372. [Google Scholar] [CrossRef]
- Abels, E.R.; Breakefield, X.O. Introduction to Extracellular Vesicles: Biogenesis, RNA Cargo Selection, Content, Release, and Uptake. Cell. Mol. Neurobiol. 2016, 36, 301–312. [Google Scholar] [CrossRef]
- Boyiadzis, M.; Whiteside, T.L. The emerging roles of tumor-derived exosomes in hematological malignancies. Leukemia 2017, 31, 1259–1268. [Google Scholar] [CrossRef]
- Muller, L.; Mitsuhashi, M.; Simms, P.; Gooding, W.E.; Whiteside, T.L. Tumor-derived exosomes regulate expression of immune function-related genes in human T cell subsets. Sci. Rep. 2016, 6, 20254. [Google Scholar] [CrossRef]
- Muller, L.; Simms, P.; Hong, C.S.; Nishimura, M.I.; Jackson, E.K.; Watkins, S.C.; Whiteside, T.L. Human tumor-derived exosomes (TEX) regulate Treg functions via cell surface signaling rather than uptake mechanisms. Oncoimmunology 2017, 6, e1261243. [Google Scholar] [CrossRef] [PubMed]
- Mulcahy, L.A.; Pink, R.C.; Carter, D.R. Routes and mechanisms of extracellular vesicle uptake. J. Extracell. Vesicles 2014, 3. [Google Scholar] [CrossRef]
- Ludwig, S.; Floros, T.; Theodoraki, M.N.; Hong, C.S.; Jackson, E.K.; Lang, S.; Whiteside, T.L. Suppression of Lymphocyte Functions by Plasma Exosomes Correlates with Disease Activity in Patients with Head and Neck Cancer. Clin. Cancer Res. 2017, 23, 4843–4854. [Google Scholar] [CrossRef]
- Hong, C.S.; Funk, S.; Muller, L.; Boyiadzis, M.; Whiteside, T.L. Isolation of biologically active and morphologically intact exosomes from plasma of patients with cancer. J. Extracell. Vesicles 2016, 5, 29289. [Google Scholar] [CrossRef] [PubMed]
- Boyiadzis, M.; Whiteside, T.L. Information transfer by exosomes: A new frontier in hematologic malignancies. Blood Rev. 2015, 29, 281–290. [Google Scholar] [CrossRef] [PubMed]
- Whiteside, T.L. Exosomes carrying immunoinhibitory proteins and their role in cancer. Clin. Exp. Immunol. 2017, 189, 259–267. [Google Scholar] [CrossRef] [PubMed]
- Di Trapani, M.; Bassi, G.; Midolo, M.; Gatti, A.; Kamga, P.T.; Cassaro, A.; Carusone, R.; Adamo, A.; Krampera, M. Differential and transferable modulatory effects of mesenchymal stromal cell-derived extracellular vesicles on T, B and NK cell functions. Sci. Rep. 2016, 6, 24120. [Google Scholar] [CrossRef] [PubMed]
- Marote, A.; Teixeira, F.G.; Mendes-Pinheiro, B.; Salgado, A.J. MSCs-Derived Exosomes: Cell-Secreted Nanovesicles with Regenerative Potential. Front. Pharmacol. 2016, 7, 231. [Google Scholar] [CrossRef]
- Lener, T.; Gimona, M.; Aigner, L.; Borger, V.; Buzas, E.; Camussi, G.; Chaput, N.; Chatterjee, D.; Court, F.A.; Del Portillo, H.A.; et al. Applying extracellular vesicles based therapeutics in clinical trials—An ISEV position paper. J. Extracell. Vesicles 2015, 4, 30087. [Google Scholar] [CrossRef]
- Burnstock, G.; Di Virgilio, F. Purinergic signalling and cancer. Purinergic Signal. 2013, 9, 491–540. [Google Scholar] [CrossRef]
- Lai, R.C.; Tan, S.S.; Teh, B.J.; Sze, S.K.; Arslan, F.; de Kleijn, D.P.; Choo, A.; Lim, S.K. Proteolytic Potential of the MSC Exosome Proteome: Implications for an Exosome-Mediated Delivery of Therapeutic Proteasome. Int. J. Proteom. 2012, 2012, 971907. [Google Scholar] [CrossRef]
- Chen, T.S.; Lai, R.C.; Lee, M.M.; Choo, A.B.; Lee, C.N.; Lim, S.K. Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs. Nucleic Acids Res. 2010, 38, 215–224. [Google Scholar] [CrossRef]
- Whiteside, T.L. Exosome and mesenchymal stem cell cross-talk in the tumor microenvironment. Semin. Immunol. 2018, 35, 69–79. [Google Scholar] [CrossRef] [PubMed]
- Dostert, G.; Mesure, B.; Menu, P.; Velot, E. How Do Mesenchymal Stem Cells Influence or Are Influenced by Microenvironment through Extracellular Vesicles Communication? Front. Cell Dev. Biol. 2017, 5, 6. [Google Scholar] [CrossRef]
- Sharma, A. Bioinformatic analysis revealing association of exosomal mRNAs and proteins in epigenetic inheritance. J. Theor. Biol. 2014, 357, 143–149. [Google Scholar] [CrossRef]
- Lindoso, R.S.; Collino, F.; Camussi, G. Extracellular vesicles derived from renal cancer stem cells induce a pro-tumorigenic phenotype in mesenchymal stromal cells. Oncotarget 2015, 6, 7959–7969. [Google Scholar] [CrossRef]
- Seguin, L.; Desgrosellier, J.S.; Weis, S.M.; Cheresh, D.A. Integrins and cancer: Regulators of cancer stemness, metastasis, and drug resistance. Trends Cell Biol. 2015, 25, 234–240. [Google Scholar] [CrossRef] [PubMed]
- Hamidi, H.; Pietila, M.; Ivaska, J. The complexity of integrins in cancer and new scopes for therapeutic targeting. Br. J. Cancer 2016, 115, 1017–1023. [Google Scholar] [CrossRef]
- Guo, W.; Giancotti, F.G. Integrin signalling during tumour progression. Nat. Rev. Mol. Cell Biol. 2004, 5, 816–826. [Google Scholar] [CrossRef]
- Hehlgans, S.; Haase, M.; Cordes, N. Signalling via integrins: Implications for cell survival and anticancer strategies. Biochim. Biophys. Acta 2007, 1775, 163–180. [Google Scholar] [CrossRef]
- Kuphal, S.; Bauer, R.; Bosserhoff, A.K. Integrin signaling in malignant melanoma. Cancer Metastasis Rev. 2005, 24, 195–222. [Google Scholar] [CrossRef] [PubMed]
- Glukhova, M.A.; Streuli, C.H. How integrins control breast biology. Curr. Opin. Cell Biol. 2013, 25, 633–641. [Google Scholar] [CrossRef] [PubMed]
- Hamidi, H.; Ivaska, J. Every step of the way: Integrins in cancer progression and metastasis. Nat. Rev. Cancer 2018, 18, 533–548. [Google Scholar] [CrossRef] [PubMed]
- Cagnet, S.; Faraldo, M.M.; Kreft, M.; Sonnenberg, A.; Raymond, K.; Glukhova, M.A. Signaling events mediated by alpha3beta1 integrin are essential for mammary tumorigenesis. Oncogene 2014, 33, 4286–4295. [Google Scholar] [CrossRef]
- White, D.E.; Kurpios, N.A.; Zuo, D.; Hassell, J.A.; Blaess, S.; Mueller, U.; Muller, W.J. Targeted disruption of beta1-integrin in a transgenic mouse model of human breast cancer reveals an essential role in mammary tumor induction. Cancer Cell 2004, 6, 159–170. [Google Scholar] [CrossRef] [PubMed]
- Stoletov, K.; Kato, H.; Zardouzian, E.; Kelber, J.; Yang, J.; Shattil, S.; Klemke, R. Visualizing extravasation dynamics of metastatic tumor cells. J. Cell Sci. 2010, 123, 2332–2341. [Google Scholar] [CrossRef]
- Dos Santos, P.B.; Zanetti, J.S.; Ribeiro-Silva, A.; Beltrao, E.I. Beta 1 integrin predicts survival in breast cancer: A clinicopathological and immunohistochemical study. Diagn. Pathol. 2012, 7, 104. [Google Scholar] [CrossRef]
- Jahangiri, A.; Aghi, M.K.; Carbonell, W.S. beta1 integrin: Critical path to antiangiogenic therapy resistance and beyond. Cancer Res. 2014, 74, 3–7. [Google Scholar] [CrossRef] [PubMed]
- Sun, Q.; Zhou, C.; Ma, R.; Guo, Q.; Huang, H.; Hao, J.; Liu, H.; Shi, R.; Liu, B. Prognostic value of increased integrin-beta 1 expression in solid cancers: A meta-analysis. OncoTargets Ther. 2018, 11, 1787–1799. [Google Scholar] [CrossRef]
- Friedrichs, K.; Ruiz, P.; Franke, F.; Gille, I.; Terpe, H.J.; Imhof, B.A. High expression level of alpha 6 integrin in human breast carcinoma is correlated with reduced survival. Cancer Res. 1995, 55, 901–906. [Google Scholar]
- Hu, T.; Zhou, R.; Zhao, Y.; Wu, G. Integrin alpha6/Akt/Erk signaling is essential for human breast cancer resistance to radiotherapy. Sci. Rep. 2016, 6, 33376. [Google Scholar] [CrossRef] [PubMed]
- Huck, L.; Pontier, S.M.; Zuo, D.M.; Muller, W.J. beta1-integrin is dispensable for the induction of ErbB2 mammary tumors but plays a critical role in the metastatic phase of tumor progression. Proc. Natl. Acad. Sci. USA 2010, 107, 15559–15564. [Google Scholar] [CrossRef] [PubMed]
- Ramirez, N.E.; Zhang, Z.; Madamanchi, A.; Boyd, K.L.; O’Rear, L.D.; Nashabi, A.; Li, Z.; Dupont, W.D.; Zijlstra, A.; Zutter, M.M. The alpha(2)beta(1) integrin is a metastasis suppressor in mouse models and human cancer. J. Clin. Investig. 2011, 121, 226–237. [Google Scholar] [CrossRef] [PubMed]
- Lanzafame, S.; Emmanuele, C.; Torrisi, A. Correlation of alpha 2 beta 1 integrin expression with histological type and hormonal receptor status in breast carcinomas. Pathol. Res. Pract. 1996, 192, 1031–1038. [Google Scholar] [CrossRef]
- Gonzalez, M.A.; Pinder, S.E.; Wencyk, P.M.; Bell, J.A.; Elston, C.W.; Nicholson, R.I.; Robertson, J.F.; Blamey, R.W.; Ellis, I.O. An immunohistochemical examination of the expression of E-cadherin, alpha- and beta/gamma-catenins, and alpha2- and beta1-integrins in invasive breast cancer. J. Pathol. 1999, 187, 523–529. [Google Scholar] [CrossRef]
- Petricevic, B.; Vrbanec, D.; Jakic-Razumovic, J.; Brcic, I.; Rabic, D.; Badovinac, T.; Ozimec, E.; Bali, V. Expression of Toll-like receptor 4 and beta 1 integrin in breast cancer. Med. Oncol. 2012, 29, 486–494. [Google Scholar] [CrossRef] [PubMed]
- Berry, M.G.; Gui, G.P.; Wells, C.A.; Carpenter, R. Integrin expression and survival in human breast cancer. Eur. J. Surg. Oncol. 2004, 30, 484–489. [Google Scholar] [CrossRef]
- Attieh, Y.; Clark, A.G.; Grass, C.; Richon, S.; Pocard, M.; Mariani, P.; Elkhatib, N.; Betz, T.; Gurchenkov, B.; Vignjevic, D.M. Cancer-associated fibroblasts lead tumor invasion through integrin-beta3-dependent fibronectin assembly. J. Cell Biol. 2017, 216, 3509–3520. [Google Scholar] [CrossRef]
- Cavaco, A.C.M.; Rezaei, M.; Caliandro, M.F.; Lima, A.M.; Stehling, M.; Dhayat, S.A.; Haier, J.; Brakebusch, C.; Eble, J.A. The Interaction between Laminin-332 and alpha3beta1 Integrin Determines Differentiation and Maintenance of CAFs, and Supports Invasion of Pancreatic Duct Adenocarcinoma Cells. Cancers 2018, 11, 14. [Google Scholar] [CrossRef]
- Erdogan, B.; Ao, M.; White, L.M.; Means, A.L.; Brewer, B.M.; Yang, L.; Washington, M.K.; Shi, C.; Franco, O.E.; Weaver, A.M.; et al. Cancer-associated fibroblasts promote directional cancer cell migration by aligning fibronectin. J. Cell Biol. 2017, 216, 3799–3816. [Google Scholar] [CrossRef]
- Pontiggia, O.; Sampayo, R.; Raffo, D.; Motter, A.; Xu, R.; Bissell, M.J.; Joffe, E.B.; Simian, M. The tumor microenvironment modulates tamoxifen resistance in breast cancer: A role for soluble stromal factors and fibronectin through beta1 integrin. Breast Cancer Res. Treat. 2012, 133, 459–471. [Google Scholar] [CrossRef]
- Dittmer, A.; Dittmer, J. Long-term exposure to carcinoma-associated fibroblasts makes breast cancer cells addictive to integrin beta1. Oncotarget 2018, 9, 22079–22094. [Google Scholar] [CrossRef]
- Zhou, W.; Ke, S.Q.; Huang, Z.; Flavahan, W.; Fang, X.; Paul, J.; Wu, L.; Sloan, A.E.; McLendon, R.E.; Li, X.; et al. Periostin secreted by glioblastoma stem cells recruits M2 tumour-associated macrophages and promotes malignant growth. Nat. Cell Biol. 2015, 17, 170–182. [Google Scholar] [CrossRef]
- Kale, S.; Raja, R.; Thorat, D.; Soundararajan, G.; Patil, T.V.; Kundu, G.C. Osteopontin signaling upregulates cyclooxygenase-2 expression in tumor-associated macrophages leading to enhanced angiogenesis and melanoma growth via alpha9beta1 integrin. Oncogene 2014, 33, 2295–2306. [Google Scholar] [CrossRef]
- Yakubenko, V.P.; Lobb, R.R.; Plow, E.F.; Ugarova, T.P. Differential induction of gelatinase B (MMP-9) and gelatinase A (MMP-2) in T lymphocytes upon alpha(4)beta(1)-mediated adhesion to VCAM-1 and the CS-1 peptide of fibronectin. Exp. Cell Res. 2000, 260, 73–84. [Google Scholar] [CrossRef]
- Bank, I.; Book, M.; Ware, R. Functional role of VLA-1 (CD49A) in adhesion, cation-dependent spreading, and activation of cultured human T lymphocytes. Cell. Immunol. 1994, 156, 424–437. [Google Scholar] [CrossRef]
- Park, E.J.; Peixoto, A.; Imai, Y.; Goodarzi, A.; Cheng, G.; Carman, C.V.; von Andrian, U.H.; Shimaoka, M. Distinct roles for LFA-1 affinity regulation during T-cell adhesion, diapedesis, and interstitial migration in lymph nodes. Blood 2010, 115, 1572–1581. [Google Scholar] [CrossRef]
- Bertoni, A.; Alabiso, O.; Galetto, A.S.; Baldanzi, G. Integrins in T Cell Physiology. Int. J. Mol. Sci. 2018, 19, 485. [Google Scholar] [CrossRef]
- Holash, J.; Maisonpierre, P.C.; Compton, D.; Boland, P.; Alexander, C.R.; Zagzag, D.; Yancopoulos, G.D.; Wiegand, S.J. Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF. Science 1999, 284, 1994–1998. [Google Scholar] [CrossRef]
- Munshi, H.G.; Stack, M.S. Reciprocal interactions between adhesion receptor signaling and MMP regulation. Cancer Metastasis Rev. 2006, 25, 45–56. [Google Scholar] [CrossRef]
- Silletti, S.; Kessler, T.; Goldberg, J.; Boger, D.L.; Cheresh, D.A. Disruption of matrix metalloproteinase 2 binding to integrin alpha vbeta 3 by an organic molecule inhibits angiogenesis and tumor growth in vivo. Proc. Natl. Acad. Sci. USA 2001, 98, 119–124. [Google Scholar] [CrossRef]
- Rolli, M.; Fransvea, E.; Pilch, J.; Saven, A.; Felding-Habermann, B. Activated integrin alphavbeta3 cooperates with metalloproteinase MMP-9 in regulating migration of metastatic breast cancer cells. Proc. Natl. Acad. Sci. USA 2003, 100, 9482–9487. [Google Scholar] [CrossRef]
- Nisato, R.E.; Hosseini, G.; Sirrenberg, C.; Butler, G.S.; Crabbe, T.; Docherty, A.J.; Wiesner, M.; Murphy, G.; Overall, C.M.; Goodman, S.L.; et al. Dissecting the role of matrix metalloproteinases (MMP) and integrin alpha(v)beta3 in angiogenesis in vitro: Absence of hemopexin C domain bioactivity, but membrane-Type 1-MMP and alpha(v)beta3 are critical. Cancer Res. 2005, 65, 9377–9387. [Google Scholar] [CrossRef]
- Moritz, M.N.O.; Eustaquio, L.M.S.; Micocci, K.C.; Nunes, A.C.C.; Dos Santos, P.K.; de Castro Vieira, T.; Selistre-de-Araujo, H.S. Alternagin-C binding to alpha2beta1 integrin controls matrix metalloprotease-9 and matrix metalloprotease-2 in breast tumor cells and endothelial cells. J. Venom. Anim. Toxins Incl. Trop. Dis. 2018, 24, 13. [Google Scholar] [CrossRef]
- Bjorklund, M.; Heikkila, P.; Koivunen, E. Peptide inhibition of catalytic and noncatalytic activities of matrix metalloproteinase-9 blocks tumor cell migration and invasion. J. Biol. Chem. 2004, 279, 29589–29597. [Google Scholar] [CrossRef]
- Barkan, D.; Kleinman, H.; Simmons, J.L.; Asmussen, H.; Kamaraju, A.K.; Hoenorhoff, M.J.; Liu, Z.Y.; Costes, S.V.; Cho, E.H.; Lockett, S.; et al. Inhibition of metastatic outgrowth from single dormant tumor cells by targeting the cytoskeleton. Cancer Res. 2008, 68, 6241–6250. [Google Scholar] [CrossRef]
- Shibue, T.; Weinberg, R.A. Integrin beta1-focal adhesion kinase signaling directs the proliferation of metastatic cancer cells disseminated in the lungs. Proc. Natl. Acad. Sci. USA 2009, 106, 10290–10295. [Google Scholar] [CrossRef]
- Wu, P.H.; Onodera, Y.; Ichikawa, Y.; Rankin, E.B.; Giaccia, A.J.; Watanabe, Y.; Qian, W.; Hashimoto, T.; Shirato, H.; Nam, J.M. Targeting integrins with RGD-conjugated gold nanoparticles in radiotherapy decreases the invasive activity of breast cancer cells. Int. J. Nanomed. 2017, 12, 5069–5085. [Google Scholar] [CrossRef]
- Nam, J.M.; Ahmed, K.M.; Costes, S.; Zhang, H.; Onodera, Y.; Olshen, A.B.; Hatanaka, K.C.; Kinoshita, R.; Ishikawa, M.; Sabe, H.; et al. beta1-Integrin via NF-kappaB signaling is essential for acquisition of invasiveness in a model of radiation treated in situ breast cancer. Breast Cancer Res. 2013, 15, R60. [Google Scholar] [CrossRef]
- Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef]
- Paget, S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 1989, 8, 98–101. [Google Scholar] [PubMed]
- Prada, I.; Meldolesi, J. Binding and Fusion of Extracellular Vesicles to the Plasma Membrane of Their Cell Targets. Int. J. Mol. Sci. 2016, 17, 1296. [Google Scholar] [CrossRef] [PubMed]
- Hoshino, A.; Costa-Silva, B.; Shen, T.L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour exosome integrins determine organotropic metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef] [PubMed]
- Alderton, G.K. Metastasis: Directions to metastatic sites. Nat. Rev. Cancer 2015, 15, 696–697. [Google Scholar] [CrossRef] [PubMed]
- Paolillo, M.; Schinelli, S. Integrins and Exosomes, a Dangerous Liaison in Cancer Progression. Cancers 2017, 9, 95. [Google Scholar] [CrossRef]
- Kristensen, T.B.; Knutsson, M.L.; Wehland, M.; Laursen, B.E.; Grimm, D.; Warnke, E.; Magnusson, N.E. Anti-vascular endothelial growth factor therapy in breast cancer. Int. J. Mol. Sci. 2014, 15, 23024–23041. [Google Scholar] [CrossRef]
- Sun, Y. Translational horizons in the tumor microenvironment: Harnessing breakthroughs and targeting cures. Med. Res. Rev. 2015, 35, 408–436. [Google Scholar] [CrossRef] [PubMed]
- Slany, A.; Bileck, A.; Muqaku, B.; Gerner, C. Targeting breast cancer-associated fibroblasts to improve anti-cancer therapy. Breast 2015, 24, 532–538. [Google Scholar] [CrossRef] [PubMed]
- Correia, A.L.; Bissell, M.J. The tumor microenvironment is a dominant force in multidrug resistance. Drug Resist. Updat. 2012, 15, 39–49. [Google Scholar] [CrossRef]
- Kerbel, R.S. A cancer therapy resistant to resistance. Nature 1997, 390, 335–336. [Google Scholar] [CrossRef] [PubMed]
- Tuder, R.M.; Lara, A.R.; Thannickal, V.J. Lactate, a novel trigger of transforming growth factor-beta activation in idiopathic pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2012, 186, 701–703. [Google Scholar] [CrossRef]
- Zhang, J.; Liu, J. Tumor stroma as targets for cancer therapy. Pharmacol. Ther. 2013, 137, 200–215. [Google Scholar] [CrossRef]
- Khan, G.J.; Sun, L.; Khan, S.; Yuan, S.; Nongyue, H. Versatility of Cancer Associated Fibroblasts: Commendable Targets for Anti-tumor Therapy. Curr. Drug Targets 2018, 19, 1573–1588. [Google Scholar] [CrossRef]
- Dorff, T.B.; Goldman, B.; Pinski, J.K.; Mack, P.C.; Lara, P.N., Jr.; Van Veldhuizen, P.J., Jr.; Quinn, D.I.; Vogelzang, N.J.; Thompson, I.M., Jr.; Hussain, M.H. Clinical and correlative results of SWOG S0354: A phase II trial of CNTO328 (siltuximab), a monoclonal antibody against interleukin-6, in chemotherapy-pretreated patients with castration-resistant prostate cancer. Clin. Cancer Res. 2010, 16, 3028–3034. [Google Scholar] [CrossRef]
- Alas, S.; Emmanouilides, C.; Bonavida, B. Inhibition of interleukin 10 by rituximab results in down-regulation of bcl-2 and sensitization of B-cell non-Hodgkin’s lymphoma to apoptosis. Clin. Cancer Res. 2001, 7, 709–723. [Google Scholar]
- Migali, C.; Milano, M.; Trapani, D.; Criscitiello, C.; Esposito, A.; Locatelli, M.; Minchella, I.; Curigliano, G. Strategies to modulate the immune system in breast cancer: Checkpoint inhibitors and beyond. Ther. Adv. Med. Oncol. 2016, 8, 360–374. [Google Scholar] [CrossRef]
- Maffey, A.; Storini, C.; Diceglie, C.; Martelli, C.; Sironi, L.; Calzarossa, C.; Tonna, N.; Lovchik, R.; Delamarche, E.; Ottobrini, L.; et al. Mesenchymal stem cells from tumor microenvironment favour breast cancer stem cell proliferation, cancerogenic and metastatic potential, via ionotropic purinergic signalling. Sci. Rep. 2017, 7, 13162. [Google Scholar] [CrossRef]
- Klopp, A.H.; Gupta, A.; Spaeth, E.; Andreeff, M.; Marini, F., 3rd. Concise review: Dissecting a discrepancy in the literature: Do mesenchymal stem cells support or suppress tumor growth? Stem Cells 2011, 29, 11–19. [Google Scholar] [CrossRef]
- Attar-Schneider, O.; Zismanov, V.; Drucker, L.; Gottfried, M. Secretome of human bone marrow mesenchymal stem cells: An emerging player in lung cancer progression and mechanisms of translation initiation. Tumor Biol. 2016, 37, 4755–4765. [Google Scholar] [CrossRef]
- Marcus, H.; Attar-Schneider, O.; Dabbah, M.; Zismanov, V.; Tartakover-Matalon, S.; Lishner, M.; Drucker, L. Mesenchymal stem cells secretomes’ affect multiple myeloma translation initiation. Cell. Signal. 2016, 28, 620–630. [Google Scholar] [CrossRef]
- Onzi, G.R.; Ledur, P.F.; Hainzenreder, L.D.; Bertoni, A.P.; Silva, A.O.; Lenz, G.; Wink, M.R. Analysis of the safety of mesenchymal stromal cells secretome for glioblastoma treatment. Cytotherapy 2016, 18, 828–837. [Google Scholar] [CrossRef]
- Eiro, N.; Sendon-Lago, J.; Seoane, S.; Bermudez, M.A.; Lamelas, M.L.; Garcia-Caballero, T.; Schneider, J.; Perez-Fernandez, R.; Vizoso, F.J. Potential therapeutic effect of the secretome from human uterine cervical stem cells against both cancer and stromal cells compared with adipose tissue stem cells. Oncotarget 2014, 5, 10692–10708. [Google Scholar] [CrossRef]
- Karnoub, A.E.; Dash, A.B.; Vo, A.P.; Sullivan, A.; Brooks, M.W.; Bell, G.W.; Richardson, A.L.; Polyak, K.; Tubo, R.; Weinberg, R.A. Mesenchymal stem cells within tumour stroma promote breast cancer metastasis. Nature 2007, 449, 557–563. [Google Scholar] [CrossRef]
- Muehlberg, F.L.; Song, Y.H.; Krohn, A.; Pinilla, S.P.; Droll, L.H.; Leng, X.; Seidensticker, M.; Ricke, J.; Altman, A.M.; Devarajan, E.; et al. Tissue-resident stem cells promote breast cancer growth and metastasis. Carcinogenesis 2009, 30, 589–597. [Google Scholar] [CrossRef]
- Galie, M.; Konstantinidou, G.; Peroni, D.; Scambi, I.; Marchini, C.; Lisi, V.; Krampera, M.; Magnani, P.; Merigo, F.; Montani, M.; et al. Mesenchymal stem cells share molecular signature with mesenchymal tumor cells and favor early tumor growth in syngeneic mice. Oncogene 2008, 27, 2542–2551. [Google Scholar] [CrossRef]
- Vizoso, F.J.; Eiro, N.; Cid, S.; Schneider, J.; Perez-Fernandez, R. Mesenchymal Stem Cell Secretome: Toward Cell-Free Therapeutic Strategies in Regenerative Medicine. Int. J. Mol. Sci. 2017, 18, 1852. [Google Scholar] [CrossRef] [PubMed]
- Schneider, J.; Eiro, N.; Perez-Fernandez, R.; Martinez-Ordonez, A.; Vizoso, F. Human Uterine Cervical Stromal Stem Cells (hUCESCs): Why and How they Exert their Antitumor Activity. Cancer Genom. Proteom. 2016, 13, 331–337. [Google Scholar]
- Curley, G.P.; Blum, H.; Humphries, M.J. Integrin antagonists. Cell Mol. Life Sci. 1999, 56, 427–441. [Google Scholar] [CrossRef] [PubMed]
- Raab-Westphal, S.; Marshall, J.F.; Goodman, S.L. Integrins as Therapeutic Targets: Successes and Cancers. Cancers 2017, 9, 110. [Google Scholar] [CrossRef] [PubMed]
Name of Class | MMP | Enzyme Name | Substrates |
---|---|---|---|
Collagenases | MMP-1 | Collagenase-1 | Collagens (I, II, III, VII and X), proteoglycans, entactin, ovostatin, MMP-2, MMP-9 |
MMP-8 | Collagenase-2/neutrophil collagenase | Collagens (I, II, III, VII, VIII and X), fibronectin, proteoglycans | |
MMP-13 | Collagenase-3 | Collagens (I, II, III, VII, VIII and X), tenascin, plasminogen, aggrecan, fibronectin, osteonectin, MMP-9 | |
MMP-18 | Collagenase-4 | Type I collagen | |
Gelatinases | MMP-2 | Gelatinase-A | Gelatin, collagen (IV, V, VII VI, IX and X), elastin, fibronectin |
MMP-9 | Gelatinase-A | Collagens (IV, V, VII, X and XIV), gelatin, entactin, elastin, fibronectin, osteonectin, plasminogen, proteoglycans | |
Stromelysins | MMP-3 | Stromelysin-1 | Collagens (IV, V and IX), gelatin, aggrecan, laminin, elastin, casein, osteonectin, fibronectin, ovostatin, entactin, plasminogen |
MMP-10 | Stromelysin2 | Collagens (I, II, IV and V), gelatin, casein, elastin, fibronectin | |
MMP-11 | Stromelysin2 | Collagens (IV, V, IX and X), laminin, elastin, fibronectin, casein, proteoglycans | |
MMP-17 | Homology tostromelysin-2 | Pro-MMP2, fibrin/fibrinogen, gelatin | |
Matrilysins | MMP-7 | Matrilysin | Collagens IV, gelatin, fibronectin, laminin, elastin, casein, transferrin |
MMP-26 | Matrilysin-2 | Collagen IV, fibronectin, fibrinogen, gelatin, pro-MMP9 | |
MT-MMP (membrane type-MMP) | MMP-14 | MT1-MMP | Collagens (I, II, III), gelatin, fibronectin, laminin, vitronectin, entactin, pro-MMP2 |
MMP-15 | MT2-MMP | Fibronectin, gelatine, vitronectin, entactin, laminin, pro-MMP-2 | |
MMP-16 | MT3-MMP | Collagen III, gelatin, casein, fibronectin, pro- MMP-2 | |
MMP-17 | MT4-MMP | Pro-MMP2, fibrinogen, gelatin | |
MMP-24 | MT5-MMP | Fibronectin, pro-MMP2, proteoglycans, gelatin | |
MMP-25 | MT6-MMP | Pro-MMP2, pro-MMP9, collagen IV, gelatine, fibronectin, Proteinase A | |
Other enzymes | MMP-12 | Macrophage metalloelastase | Collagen IV, gelatin, elastin, casein, fibronectin, vitronectin, laminin, entactin, fibrin/fibrinogen |
MMP-19 | RASI-1 | Collagen (I, IV) gelatin, fibronectin, laminin | |
MMP-20 | Enamelysin | Amelogenin, aggrecan | |
MMP-21 | |||
MMP-22 | |||
MMP-23 | Gelatin | ||
MMP-28 | Epilysin | ||
MMP-29 | Unnamed |
Gene Symbol | Gene Name | Main Role |
---|---|---|
S100A4 | S100 calcium binding protein A4 | Invasion |
FGF7 | Fibroblast growth factor 7 | Cell growth/Invasion |
PDGFB | Platelet-derived growth factor beta | Angiogenesis |
VEGFA | Vascular endothelial growth factor A | Angiogenesis |
TGFβ | Transforming growth factor beta | Inflammation/Invasion |
IL6 | Interleukin 6 | Inflammation |
IL8 | Interleukin 8 | Inflammation |
uPA | Urokinase-type plasminogen activator | ECM remodeling |
MMP2 | Matrix metalloproteases 2 | ECM remodeling |
MMP11 | Matrix metalloproteases 11 | ECM remodeling |
TIMP1 | Tissue inhibitor of metalloproteases 1 | ECM remodeling |
© 2019 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
Eiro, N.; Gonzalez, L.O.; Fraile, M.; Cid, S.; Schneider, J.; Vizoso, F.J. Breast Cancer Tumor Stroma: Cellular Components, Phenotypic Heterogeneity, Intercellular Communication, Prognostic Implications and Therapeutic Opportunities. Cancers 2019, 11, 664. https://doi.org/10.3390/cancers11050664
Eiro N, Gonzalez LO, Fraile M, Cid S, Schneider J, Vizoso FJ. Breast Cancer Tumor Stroma: Cellular Components, Phenotypic Heterogeneity, Intercellular Communication, Prognostic Implications and Therapeutic Opportunities. Cancers. 2019; 11(5):664. https://doi.org/10.3390/cancers11050664
Chicago/Turabian StyleEiro, Noemi, Luis O. Gonzalez, María Fraile, Sandra Cid, Jose Schneider, and Francisco J. Vizoso. 2019. "Breast Cancer Tumor Stroma: Cellular Components, Phenotypic Heterogeneity, Intercellular Communication, Prognostic Implications and Therapeutic Opportunities" Cancers 11, no. 5: 664. https://doi.org/10.3390/cancers11050664
APA StyleEiro, N., Gonzalez, L. O., Fraile, M., Cid, S., Schneider, J., & Vizoso, F. J. (2019). Breast Cancer Tumor Stroma: Cellular Components, Phenotypic Heterogeneity, Intercellular Communication, Prognostic Implications and Therapeutic Opportunities. Cancers, 11(5), 664. https://doi.org/10.3390/cancers11050664