Influence of Innate Immunity on Cancer Cell Stemness
Abstract
:1. Introduction
1.1. Cancer
1.2. Innate Immune Populations in Cancer
1.3. Cancer Stem Cells
2. Elimination of Tumor Bulk
2.1. Equilibrium and Survival of CSCs
2.2. CSCs Awakening and Immunoediting
2.3. Dissemination and Metastases
3. Immunological Targeting of CSCs
4. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
Abbreviations
ADCC | antibody-dependent cellular cytotoxicity |
APC | Antigen-presenting cell |
APM | Antigen-processing machinery |
BCL | B cell lymphoma |
BMDC | Bone marrow-derived cell |
CAF | Cancer-associated fibroblast |
CCL | Chemokine (C-C motif) ligand |
CSF1 | Colony-stimulating factor 1 |
CXCL | Chemokine (C-X-C motif) ligand |
CXCR | Chemokine (C-X-C motif) receptor |
CRC | Colorectal cancer |
CSC | Cancer stem cell |
CTL | Cytotoxic T cell |
DC | Dendritic cells |
EGF | Epidermal growth factor |
EMT | Epithelial-to-mesenchymal transition |
GM-CSF | Granulocyte-macrophage colony-stimulating factor |
G-MDSC | Granulocytic myeloid-derived suppressor cell |
HCC | Hepatocellular carcinoma |
HLA | Human leukocyte antigen |
HNSCC | Head and neck squamous cell carcinoma |
IFNγ | Interferon gamma |
IL | Interleukin |
iNOS | induced Nitric Oxide synthase |
MDSC | Myeloid-derived suppressor cell |
M-MDSC | Monocytic myeloid-derived suppressor cell |
MHC | Major histocompatibility complex |
NET | Neutrophil extracellular trap |
NK | Natural Killer cells |
NKG2D/A | Natural killer group 2 member D/A |
NKT | Natural killer T cell |
NO | Nitric oxide |
NSCLC | Non-small cell lung cancer |
PBMC | peripheral blood mononuclear cell |
PC | Pancreatic cancer |
PDAC | Pancreatic ductal adenocarcinoma |
PGE2 | Prostaglandin E2 |
SIRP | Signal regulatory protein |
SPARC | Secreted protein acidic and rich in cysteine |
STAT3 | Signal transducer and activator of transcription 3 |
TAA | Tumor-associated antigen |
TAM | Tumor-associated macrophages |
TAN | Tumor-associated neutrophils |
TGF | Transforming growth factor |
Th | T helper cell |
Treg | Regulatory T cell |
TME | Tumor microenvironment |
TNFα | Tumor necrosis factor alpha |
TRAIL | TNF-related apoptosis-inducing ligand |
VEGF | Vascular endothelial growth factor |
XCL1 | X-C Motif Chemokine Ligand 1 |
References
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef] [PubMed]
- Balkwill, F.; Mantovani, A. Inflammation and cancer: Back to virchow? Lancet 2001, 357, 539–545. [Google Scholar] [CrossRef]
- Fridman, W.H.; Zitvogel, L.; Sautès-Fridman, C.; Kroemer, G. The immune contexture in cancer prognosis and treatment. Nat. Rev. Clin. Oncol. 2017, 14, 717–734. [Google Scholar] [CrossRef]
- Coussens, L.M.; Zitvogel, L.; Palucka, A.K. Neutralizing tumor-promoting chronic inflammation: A magic bullet? Science 2013, 339, 286–291. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Hinshaw, D.C.; Shevde, L.A. The tumor microenvironment innately modulates cancer progression. Cancer Res. 2019, 79, 4557–4567. [Google Scholar] [CrossRef] [Green Version]
- Dunn, G.P.; Old, L.J.; Schreiber, R.D. The three es of cancer immunoediting. Annu. Rev. Immunol. 2004, 22, 329–360. [Google Scholar] [CrossRef]
- Schreiber, R.D.; Old, L.J.; Smyth, M.J. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science 2011, 331, 1565–1570. [Google Scholar] [CrossRef] [Green Version]
- Bruttel, V.S.; Wischhusen, J. Cancer stem cell immunology: Key to understanding tumorigenesis and tumor immune escape? Front. Immunol. 2014, 5, 360. [Google Scholar] [CrossRef] [Green Version]
- Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef] [PubMed]
- Bonnet, D.; Dick, J.E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 1997, 3, 730–737. [Google Scholar] [CrossRef] [PubMed]
- Pastò, A.; Bellio, C.; Pilotto, G.; Ciminale, V.; Silic-Benussi, M.; Guzzo, G.; Rasola, A.; Frasson, C.; Nardo, G.; Zulato, E.; et al. Cancer stem cells from epithelial ovarian cancer patients privilege oxidative phosphorylation, and resist glucose deprivation. Oncotarget 2014, 5, 4305–4319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sica, A.; Porta, C.; Amadori, A.; Pastò, A. Tumor-associated myeloid cells as guiding forces of cancer cell stemness. Cancer Immunol. Immunother. 2017, 66, 1025–1036. [Google Scholar] [CrossRef] [PubMed]
- Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
- Bottazzi, B.; Riboli, E.; Mantovani, A. Aging, inflammation and cancer. Semin. Immunol. 2018, 40, 74–82. [Google Scholar] [CrossRef] [PubMed]
- Włodarczyk, M.; Jabłonowska-Lietz, B.; Olejarz, W.; Nowicka, G. Anthropometric and dietary factors as predictors of DNA damage in obese women. Nutrients 2018, 10, 578. [Google Scholar] [CrossRef] [Green Version]
- Włodarczyk, M.; Nowicka, G. Obesity, DNA damage, and development of obesity-related diseases. Int. J. Mol. Sci. 2019, 20, 1146. [Google Scholar] [CrossRef]
- Teng, M.W.L.; Galon, J.; Fridman, W.H.; Smyth, M.J. From mice to humans: Developments in cancer immunoediting. J. Clin. Invest. 2015, 125, 3338–3346. [Google Scholar] [CrossRef] [Green Version]
- Castriconi, R.; Daga, A.; Dondero, A.; Zona, G.; Poliani, P.L.; Melotti, A.; Griffero, F.; Marubbi, D.; Spaziante, R.; Bellora, F.; et al. NK cells recognize and kill human glioblastoma cells with stem cell-like properties. J. Immunol. 2009, 182, 3530–3539. [Google Scholar] [CrossRef] [Green Version]
- Vitale, M.; Della Chiesa, M.; Carlomagno, S.; Pende, D.; Aricò, M.; Moretta, L.; Moretta, A. NK-dependent DC maturation is mediated by TNFα and IFNγ released upon engagement of the NKp30 triggering receptor. Blood 2005, 106, 566–571. [Google Scholar] [CrossRef] [PubMed]
- Guia, S.; Fenis, A.; Vivier, E.; Narni-Mancinelli, E. Activating and inhibitory receptors expressed on innate lymphoid cells. Semin. Immunopathol. 2018, 40, 331–341. [Google Scholar] [CrossRef] [PubMed]
- Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of natural killer cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef] [PubMed]
- Lanier, L.L. On guard—Activating NK cell receptors. Nat. Immunol. 2001, 2, 23–27. [Google Scholar] [CrossRef] [PubMed]
- Dambrauskas, Z.; Svensson, H.; Joshi, M.; Hyltander, A.; Naredi, P.; Iresjö, B.M. Expression of major histocompatibility complex class I-related chain A/B (MICA/B) in pancreatic carcinoma. Int. J. Oncol. 2014, 44, 99–104. [Google Scholar] [CrossRef]
- Gasser, S.; Orsulic, S.; Brown, E.J.; Raulet, D.H. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 2005, 436, 1186–1190. [Google Scholar] [CrossRef] [Green Version]
- Orange, J.S. Formation and function of the lytic NK-cell immunological synapse. Nat. Rev. Immunol. 2008, 8, 713–725. [Google Scholar] [CrossRef] [Green Version]
- Chyuan, I.T.; Tsai, H.F.; Liao, H.J.; Wu, C.S.; Hsu, P.N. An apoptosis-independent role of TRAIL in suppressing joint inflammation and inhibiting T-cell activation in inflammatory arthritis. Cell. Mol. Immunol. 2018, 15, 846–857. [Google Scholar] [CrossRef] [Green Version]
- Screpanti, V.; Wallin, R.P.A.; Ljunggren, H.-G.; Grandien, A. A central role for death receptor-mediated apoptosis in the rejection of tumors by NK cells. J. Immunol. 2001, 167, 2068–2073. [Google Scholar] [CrossRef] [Green Version]
- Kwon, B. IFN-γ in tissue-immune homeostasis and antitumor immunity. Cell. Mol. Immunol. 2018, 15, 531–532. [Google Scholar] [CrossRef]
- O’Sullivan, T.; Saddawi-Konefka, R.; Koebel, W.V.; Arthur, C.; White, J.M.; Uppaluri, R.; Andrews, D.M.; Ngiow, S.F.; Teng, M.W.L.; Smyth, M.J.; et al. Cancer immunoediting by the innate immune system in the absence of adaptive immunity. J. Exp. Med. 2012, 209, 1869–1882. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kelly, J.M.; Darcy, P.K.; Markby, J.L.; Godfrey, D.I.; Takeda, K.; Yagita, H.; Smyth, M.J. Induction of tumor-specific T cell memory by NK cell-mediated tumor rejection. Nat. Immunol. 2002, 3, 83–90. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Bi, J.; Zheng, X.; Chen, Y.; Wang, H.; Wu, W.; Wang, Z.; Wu, Q.; Peng, H.; Wei, H.; et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat. Immunol. 2018, 19, 723–732. [Google Scholar] [CrossRef] [PubMed]
- Finisguerra, V.; Di Conza, G.; Di Matteo, M.; Serneels, J.; Costa, S.; Thompson, A.A.R.; Wauters, E.; Walmsley, S.; Prenen, H.; Granot, Z.; et al. MET is required for the recruitment of anti-tumoural neutrophils. Nature 2015, 522, 349–353. [Google Scholar] [CrossRef]
- Showalter, A.; Limaye, A.; Oyer, J.L.; Igarashi, R.; Kittipatarin, C.; Copik, A.J.; Khaled, A.R. Cytokines in immunogenic cell death: Applications for cancer immunotherapy. Cytokine 2017, 97, 123–132. [Google Scholar] [CrossRef]
- Böttcher, J.P.; Bonavita, E.; Chakravarty, P.; Blees, H.; Cabeza-Cabrerizo, M.; Sammicheli, S.; Rogers, N.C.; Sahai, E.; Zelenay, S.; Reis e Sousa, C. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 2018, 172, 1022–1037. [Google Scholar] [CrossRef] [Green Version]
- Tran Janco, J.M.; Lamichhane, P.; Karyampudi, L.; Knutson, K.L. Tumor-infiltrating dendritic cells in cancer pathogenesis. J. Immunol. 2015, 194, 2985–2991. [Google Scholar] [CrossRef] [Green Version]
- Mildner, A.; Jung, S. Development and function of dendritic cell subsets. Immunity 2014, 40, 642–656. [Google Scholar] [CrossRef] [Green Version]
- Walzer, T.; Dalod, M.; Robbins, S.H.; Zitvogel, L.; Vivier, E. Natural-killer cells and dendritic cells: “L’union fait la force”. Blood 2005, 106, 2252–2258. [Google Scholar] [CrossRef] [Green Version]
- Mantovani, A.; Sica, A. Macrophages, innate immunity and cancer: Balance, tolerance, and diversity. Curr. Opin. Immunol. 2010, 22, 231–237. [Google Scholar] [CrossRef]
- Brown, J.M.; Recht, L.; Strober, S. The promise of targeting macrophages in cancer therapy. Clin. Cancer Res. 2017, 23, 3241–3250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Quail, D.F.; Joyce, J.A. Microenvironmental regulation of tumor progression and metastasis. Nat. Med. 2013, 19, 1423–1437. [Google Scholar] [CrossRef] [PubMed]
- Diefenbach, A.; Jamieson, A.M.; Liu, S.D.; Shastri, N.; Raulet, D.H. Ligands for the murine NKG2D receptor: Expression by tumor cells and activation of NK cells and macrophages. Nat. Immunol. 2000, 1, 119–126. [Google Scholar] [CrossRef] [PubMed]
- Miguel, R.D.V.; Cherpes, T.L.; Watson, L.J.; McKenna, K.C. CTL induction of tumoricidal nitric oxide production by intratumoral macrophages is critical for tumor elimination. J. Immunol. 2010, 185, 6706–6718. [Google Scholar] [CrossRef] [Green Version]
- Pantano, F.; Berti, P.; Guida, F.M.; Perrone, G.; Vincenzi, B.; Amato, M.M.C.; Righi, D.; Dell’Aquila, E.; Graziano, F.; Catalano, V.; et al. The role of macrophages polarization in predicting prognosis of radically resected gastric cancer patients. J. Cell. Mol. Med. 2013, 17, 1415–1421. [Google Scholar] [CrossRef] [Green Version]
- Mantovani, A.; Sozzani, S.; Locati, M.; Allavena, P.; Sica, A. Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002, 23, 549–555. [Google Scholar] [CrossRef]
- Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of Tumor-Associated Neutrophil Phenotype by TGF-β: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef] [Green Version]
- Andzinski, L.; Kasnitz, N.; Stahnke, S.; Wu, C.F.; Gereke, M.; Von Köckritz-Blickwede, M.; Schilling, B.; Brandau, S.; Weiss, S.; Jablonska, J. Type I IFNs induce anti-tumor polarization of tumor associated neutrophils in mice and human. Int. J. Cancer 2016, 138, 1982–1993. [Google Scholar] [CrossRef]
- Bonde, A.K.; Tischler, V.; Kumar, S.; Soltermann, A.; Schwendener, R.A. Intratumoral macrophages contribute to epithelial-mesenchymal transition in solid tumors. BMC Cancer 2012, 12, 35. [Google Scholar] [CrossRef] [Green Version]
- Bao, S.; Wu, Q.; McLendon, R.E.; Hao, Y.; Shi, Q.; Hjelmeland, A.B.; Dewhirst, M.W.; Bigner, D.D.; Rich, J.N. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature 2006, 444, 756–760. [Google Scholar] [CrossRef]
- Blanpain, C.; Mohrin, M.; Sotiropoulou, P.A.; Passegué, E. DNA-damage response in tissue-specific and cancer stem cells. Cell Stem. Cell 2011, 8, 16–29. [Google Scholar] [CrossRef] [PubMed]
- Begicevic, R.R.; Falasca, M. ABC transporters in cancer stem cells: Beyond chemoresistance. Int. J. Mol. Sci. 2017, 18, 2362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- El Khoury, F.; Corcos, L.; Durand, S.; Simon, B.; Le Jossic-Corcos, C. Acquisition of anticancer drug resistance is partially associated with cancer stemness in human colon cancer cells. Int. J. Oncol. 2016, 49, 2558–2568. [Google Scholar] [CrossRef] [PubMed]
- Fletcher, J.I.; Haber, M.; Henderson, M.J.; Norris, M.D. ABC transporters in cancer: More than just drug efflux pumps. Nat. Rev. Cancer 2010, 10, 147–156. [Google Scholar] [CrossRef] [PubMed]
- Di Tomaso, T.; Mazzoleni, S.; Wang, E.; Sovena, G.; Clavenna, D.; Franzin, A.; Mortini, P.; Ferrone, S.; Doglioni, C.; Marincola, F.M.; et al. Immunobiological characterization of cancer stem cells isolated from glioblastoma patients. Clin. Cancer Res. 2010, 16, 800–813. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schatton, T.; Schütte, U.; Frank, N.Y.; Zhan, Q.; Hoerning, A.; Robles, S.C.; Zhou, J.; Hodi, F.S.; Spagnoli, G.C.; Murphy, G.F.; et al. Modulation of T-cell activation by malignant melanoma initiating cells. Cancer Res. 2010, 70, 697–708. [Google Scholar] [CrossRef] [Green Version]
- Volonté, A.; Di Tomaso, T.; Spinelli, M.; Todaro, M.; Sanvito, F.; Albarello, L.; Bissolati, M.; Ghirardelli, L.; Orsenigo, E.; Ferrone, S.; et al. Cancer-initiating cells from colorectal cancer patients escape from T cell–mediated immunosurveillance in vitro through membrane-bound IL-4. J. Immunol. 2014, 192, 523–532. [Google Scholar] [CrossRef] [Green Version]
- Grau, J.J.; Mesía, R.; De La Iglesia-Vicente, M.; Williams, E.S.; Taberna, M.; Caballero, M.; Larque, A.B.; De La Oliva, J.; Cordón-Cardo, C.; Domingo-Domenech, J. Enrichment of cells with cancer stem cell-like markers in relapses of chemoresistant patients with locally advanced head and neck squamous cell carcinoma. Oncology 2016, 90, 267–272. [Google Scholar] [CrossRef]
- MacCalli, C.; Volontè, A.; Cimminiello, C.; Parmiani, G. Immunology of cancer stem cells in solid tumours. A review. Eur. J. Cancer 2014, 50, 649–655. [Google Scholar] [CrossRef]
- Maccalli, C.; Rasul, K.I.; Elawad, M.; Ferrone, S. The role of cancer stem cells in the modulation of anti-tumor immune responses. Semin. Cancer Biol. 2018, 53, 189–200. [Google Scholar] [CrossRef]
- Morrison, B.J.; Steel, J.C.; Morris, J.C. Reduction of MHC-I expression limits T-lymphocyte-mediated killing of Cancer-initiating cells. BMC Cancer 2018, 18, 469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abdullah, L.N.; Chow, E.K.-H. Mechanisms of chemoresistance in cancer stem cells. Clin. Transl. Med. 2013, 2, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ni, J.; Cozzi, P.; Hao, J.; Duan, W.; Graham, P.; Kearsley, J.; Li, Y. Cancer stem cells in prostate cancer chemoresistance. Curr. Cancer Drug Targets 2014, 14, 225–240. [Google Scholar] [CrossRef] [PubMed]
- Qadir, A.S.; Ceppi, P.; Brockway, S.; Law, C.; Mu, L.; Khodarev, N.N.; Kim, J.; Zhao, J.C.; Putzbach, W.; Murmann, A.E.; et al. CD95/fas increases stemness in cancer cells by inducing a STAT1-dependent type I interferon response. Cell Rep. 2017, 18, 2373–2386. [Google Scholar] [CrossRef]
- Ceppi, P.; Hadji, A.; Kohlhapp, F.J.; Pattanayak, A.; Hau, A.; Liu, X.; Liu, H.; Murmann, A.E.; Peter, M.E. CD95 and CD95L promote and protect cancer stem cells. Nat. Commun. 2014, 5, 1–13. [Google Scholar] [CrossRef] [Green Version]
- Shang, B.; Liu, Y.; Jiang, S.J.; Liu, Y. Prognostic value of tumor-infiltrating FoxP3+ regulatory T cells in cancers: A systematic review and meta-analysis. Sci. Rep. 2015, 5, 15179. [Google Scholar] [CrossRef] [Green Version]
- Wang, B.; Wang, Q.; Wang, Z.; Jiang, J.; Yu, S.C.; Ping, Y.F.; Yang, J.; Xu, S.L.; Ye, X.Z.; Xu, C.; et al. Metastatic consequences of immune escape from NK cell cytotoxicity by human breast cancer stem cells. Cancer Res. 2014, 74, 5746–5757. [Google Scholar] [CrossRef] [Green Version]
- Drukker, M.; Katz, G.; Urbach, A.; Schuldiner, M.; Markel, G.; Itskovitz-Eldor, J.; Reubinoff, B.; Mandelboim, O.; Benvenisty, N. Characterization of the expression of MHC proteins in human embryonic stem cells. Proc. Natl. Acad. Sci. USA 2002, 99, 9864–9869. [Google Scholar] [CrossRef] [Green Version]
- Reim, F.; Dombrowski, Y.; Ritter, C.; Buttmann, M.; Häusler, S.; Ossadnik, M.; Krockenberger, M.; Beier, D.; Beier, C.P.; Dietl, J.; et al. Immunoselection of breast and ovarian cancer cells with trastuzumab and natural killer cells: Selective escape of CD44high/CD24 low/HER2low breast cancer stem cells. Cancer Res. 2009, 69, 8058–8066. [Google Scholar] [CrossRef] [Green Version]
- Liu, L.; Zhang, L.; Yang, L.; Li, H.; Li, R.; Yu, J.; Yang, L.; Wei, F.; Yan, C.; Sun, Q.; et al. Anti-CD47 antibody as a targeted therapeutic agent for human lung cancer and cancer stem cells. Front. Immunol. 2017, 8, 404. [Google Scholar] [CrossRef] [Green Version]
- Cioffi, M.; Trabulo, S.; Hidalgo, M.; Costello, E.; Greenhalf, W.; Erkan, M.; Kleeff, J.; Sainz, B.; Heeschen, C. Inhibition of CD47 Effectively targets pancreatic cancer stem cells via dual mechanisms. Clin. Cancer Res. 2015, 21, 2325–2337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, T.K.W.; Cheung, V.C.H.; Lu, P.; Lau, E.Y.T.; Ma, S.; Tang, K.H.; Tong, M.; Lo, J.; Ng, I.O.L. Blockade of CD47-mediated cathepsin S/protease-activated receptor 2 signaling provides a therapeutic target for hepatocellular carcinoma. Hepatology 2014, 60, 179–191. [Google Scholar] [CrossRef] [PubMed]
- Uger, R.; Johnson, L. Blockade of the CD47-SIRPα axis: A promising approach for cancer immunotherapy. Expert Opin. Biol. Ther. 2020, 20, 5–8. [Google Scholar] [CrossRef] [PubMed]
- Ravindran, S.; Rasool, S.; Maccalli, C. The cross talk between cancer stem cells/cancer initiating cells and tumor microenvironment: The missing piece of the puzzle for the efficient targeting of these cells with immunotherapy. Cancer Microenviron. 2019, 12, 133–148. [Google Scholar] [CrossRef] [Green Version]
- Inoda, S.; Hirohashi, Y.; Torigoe, T.; Morita, R.; Takahashi, A.; Asanuma, H.; Nakatsugawa, M.; Nishizawa, S.; Tamura, Y.; Tsuruma, T.; et al. Cytotoxic T lymphocytes efficiently recognize human colon cancer stem-like cells. Am. J. Pathol. 2011, 178, 1805–1813. [Google Scholar] [CrossRef]
- Chikamatsu, K.; Takahashi, G.; Sakakura, K.; Ferrone, S.; Masuyama, K. Immunoregulatory properties of CD44+ cancer stem-like cells in squamous cell carcinoma of the head and neck. Head Neck 2011, 33, 208–215. [Google Scholar] [CrossRef] [Green Version]
- Ames, E.; Canter, R.J.; Grossenbacher, S.K.; Mac, S.; Chen, M.; Smith, R.C.; Hagino, T.; Perez-Cunningham, J.; Sckisel, G.D.; Urayama, S.; et al. NK cells preferentially target tumor cells with a cancer stem cell phenotype. J. Immunol. 2015, 195, 4010–4019. [Google Scholar] [CrossRef] [Green Version]
- Shlush, L.I.; Chapal-Ilani, N.; Adar, R.; Pery, N.; Maruvka, Y.; Spiro, A.; Shouval, R.; Rowe, J.M.; Tzukerman, M.; Bercovich, D.; et al. Cell lineage analysis of acute leukemia relapse uncovers the role of replication-rate heterogeneity and microsatellite instability. Blood 2012, 120, 603–612. [Google Scholar] [CrossRef] [Green Version]
- Vahidian, F.; Duijf, P.H.G.; Safarzadeh, E.; Derakhshani, A.; Baghbanzadeh, A.; Baradaran, B. Interactions between cancer stem cells, immune system and some environmental components: Friends or foes? Immunol. Lett. 2019, 208, 19–29. [Google Scholar] [CrossRef]
- De Vlaeminck, Y.; González-Rascón, A.; Goyvaerts, C.; Breckpot, K. Cancer-associated myeloid regulatory cells. Front. Immunol. 2016, 7, 113. [Google Scholar] [CrossRef] [Green Version]
- Mantovani, A.; Marchesi, F.; Malesci, A.; Laghi, L.; Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 2017, 14, 399–416. [Google Scholar] [CrossRef] [PubMed]
- Ruffell, B.; Chang-Strachan, D.; Chan, V.; Rosenbusch, A.; Ho, C.M.T.; Pryer, N.; Daniel, D.; Hwang, E.S.; Rugo, H.S.; Coussens, L.M. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell 2014, 26, 623–637. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Cañellas, A.; Hernando-Momblona, X.; et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sica, A.; Guarneri, V.; Gennari, A. Myelopoiesis, metabolism and therapy: A crucial crossroads in cancer progression. Cell Stress 2019, 3, 284–294. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Strauss, L.; Sangaletti, S.; Consonni, F.M.; Szebeni, G.; Morlacchi, S.; Totaro, M.G.; Porta, C.; Anselmo, A.; Tartari, S.; Doni, A.; et al. RORC1 regulates tumor-promoting “emergency” granulo-monocytopoiesis. Cancer Cell 2015, 28, 253–269. [Google Scholar] [CrossRef] [Green Version]
- Medina, E.; Hartl, D. Myeloid-derived suppressor cells in infection: A general overview. J. Innate Immun. 2018, 10, 407–413. [Google Scholar] [CrossRef]
- Boettcher, S.; Manz, M.G. Regulation of inflammation- and infection-driven hematopoiesis. Trends Immunol. 2017, 38, 345–357. [Google Scholar] [CrossRef]
- Ueha, S.; Shand, F.H.W.; Matsushima, K. Myeloid cell population dynamics in healthy and tumor-bearing mice. Int. Immunopharmacol. 2011, 11, 783–788. [Google Scholar] [CrossRef]
- Velten, L.; Haas, S.F.; Raffel, S.; Blaszkiewicz, S.; Islam, S.; Hennig, B.P.; Hirche, C.; Lutz, C.; Buss, E.C.; Nowak, D.; et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat. Cell Biol. 2017, 19, 271–281. [Google Scholar] [CrossRef] [Green Version]
- Sica, A.; Bronte, V. Altered macrophage differentiation and immune dysfunction in tumor development. J. Clin. Invest. 2007, 117, 1155–1166. [Google Scholar] [CrossRef] [PubMed]
- Lau, E.Y.T.; Ho, N.P.Y.; Lee, T.K.W. Cancer stem cells and their microenvironment: Biology and therapeutic implications. Stem. Cells Int. 2017. [Google Scholar] [CrossRef] [PubMed]
- Inamoto, S.; Itatani, Y.; Yamamoto, T.; Minamiguchi, S.; Hirai, H.; Iwamoto, M.; Hasegawa, S.; Taketo, M.M.; Sakai, Y.; Kawada, K. Loss of SMAD4 promotes colorectal cancer progression by accumulation of myeloid-derived suppressor cells through the CCL15-CCR1 chemokine axis. Clin. Cancer Res. 2016, 22, 492–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Panni, R.Z.; Sanford, D.E.; Belt, B.A.; Mitchem, J.B.; Worley, L.A.; Goetz, B.D.; Mukherjee, P.; Wang-Gillam, A.; Link, D.C.; Denardo, D.G.; et al. Tumor-induced STAT3 activation in monocytic myeloid-derived suppressor cells enhances stemness and mesenchymal properties in human pancreatic cancer. Cancer Immunol. Immunother. 2014, 63, 513–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Y.; Yin, K.; Tian, J.; Xia, X.; Ma, J.; Tang, X.; Xu, H.; Wang, S. Granulocytic myeloid-derived suppressor cells promote the stemness of colorectal cancer cells through exosomal S100A9. Adv. Sci. 2019, 6, 1901278. [Google Scholar] [CrossRef] [Green Version]
- Cui, T.X.; Kryczek, I.; Zhao, L.; Zhao, E.; Kuick, R.; Roh, M.H.; Vatan, L.; Szeliga, W.; Mao, Y.; Thomas, D.G.; et al. Myeloid-derived suppressor cells enhance stemness of cancer cells by inducing microRNA101 and suppressing the corepressor CTBP2. Immunity 2013, 39, 611–621. [Google Scholar] [CrossRef] [Green Version]
- Kuroda, H.; Mabuchi, S.; Yokoi, E.; Komura, N.; Kozasa, K.; Matsumoto, Y.; Kawano, M.; Takahashi, R.; Sasano, T.; Shimura, K.; et al. Prostaglandin E2 produced by myeloid-derived suppressive cells induces cancer stem cells in uterine cervical cancer. Oncotarget 2018, 9, 36317–36330. [Google Scholar] [CrossRef] [Green Version]
- Peng, D.; Tanikawa, T.; Li, W.; Zhao, L.; Vatan, L.; Szeliga, W.; Wan, S.; Wei, S.; Wang, Y.; Liu, Y.; et al. Myeloid-derived suppressor cells endow stem-like qualities to breast cancer cells through IL6/STAT3 and NO/NOTCH cross-talk signaling. Cancer Res. 2016, 76, 3156–3165. [Google Scholar] [CrossRef] [Green Version]
- Wan, S.; Zhao, E.; Kryczek, I.; Vatan, L.; Sadovskaya, A.; Ludema, G.; Simeone, D.M.; Zou, W.; Welling, T.H. Tumor-associated macrophages produce interleukin 6 and signal via STAT3 to promote expansion of human hepatocellular carcinoma stem cells. Gastroenterology 2014, 147, 1393–1404. [Google Scholar] [CrossRef] [Green Version]
- Jinushi, M.; Chiba, S.; Yoshiyama, H.; Masutomi, K.; Kinoshita, I.; Dosaka-Akita, H.; Yagita, H.; Takaoka, A.; Tahara, H. Tumor-associated macrophages regulate tumorigenicity and anticancer drug responses of cancer stem/initiating cells. Proc. Natl. Acad. Sci. USA 2011, 108, 12425–12430. [Google Scholar] [CrossRef] [Green Version]
- Mitchem, J.B.; Brennan, D.J.; Knolhoff, B.L.; Belt, B.A.; Zhu, Y.; Sanford, D.E.; Belaygorod, L.; Carpenter, D.; Collins, L.; Piwnica-Worms, D.; et al. Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res. 2013, 73, 1128–1141. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Wu, A.; Wei, J.; Kong, L.Y.; Wang, Y.; Priebe, W.; Qiao, W.; Sawaya, R.; Heimberger, A.B. Glioma cancer stem cells induce immunosuppressive macrophages/microglia. Neuro. Oncol. 2010, 12, 1113–1125. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Cai, D.J.; Li, B. Ovarian cancer stem-like cells elicit the polarization of M2 macrophages. Mol. Med. Rep. 2015, 11, 4685–4693. [Google Scholar] [CrossRef] [Green Version]
- Nusblat, L.M.; Carroll, M.J.; Roth, C.M. Crosstalk between M2 macrophages and glioma stem cells. Cell. Oncol. 2017, 40, 471–482. [Google Scholar] [CrossRef]
- Kokubu, Y.; Tabu, K.; Muramatsu, N.; Wang, W.; Murota, Y.; Nobuhisa, I.; Jinushi, M.; Taga, T. Induction of protumoral CD11chigh macrophages by glioma cancer stem cells through GM-CSF. Genes to Cells 2016, 21, 241–251. [Google Scholar] [CrossRef] [Green Version]
- Sainz, B.; Martín, B.; Tatari, M.; Heeschen, C.; Guerra, S. ISG15 is a critical microenvironmental factor for pancreatic cancer stem cells. Cancer Res. 2014, 74, 7309–7320. [Google Scholar] [CrossRef] [Green Version]
- Chen, R.H.; Du, Y.; Han, P.; Wang, H.B.; Liang, F.Y.; Feng, G.K.; Zhou, A.J.; Cai, M.Y.; Zhong, Q.; Zeng, M.S.; et al. ISG15 predicts poor prognosis and promotes cancer stem cell phenotype in nasopharyngeal carcinoma. Oncotarget 2016, 7, 16910–16922. [Google Scholar] [CrossRef] [Green Version]
- Jinushi, M.; Baghdadi, M.; Chiba, S.; Yoshiyama, H. Regulation of cancer stem cell activities by tumor-associated macrophages. Am. J. Cancer Res. 2012, 2, 529–539. [Google Scholar]
- Fan, Q.M.; Jing, Y.Y.; Yu, G.F.; Kou, X.R.; Ye, F.; Gao, L.; Li, R.; Zhao, Q.D.; Yang, Y.; Lu, Z.H.; et al. Tumor-associated macrophages promote cancer stem cell-like properties via transforming growth factor-beta1-induced epithelial-mesenchymal transition in hepatocellular carcinoma. Cancer Lett. 2014, 352, 160–168. [Google Scholar] [CrossRef]
- Plaks, V.; Kong, N.; Werb, Z. The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 2015, 16, 225–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Elaimy, A.L.; Guru, S.; Chang, C.; Ou, J.; Amante, J.J.; Zhu, L.J.; Goel, H.L.; Mercurio, A.M. VEGF-neuropilin-2 signaling promotes stem-like traits in breast cancer cells by TAZ-mediated repression of the Rac GAP β2-chimaerin. Sci. Signal. 2018, 11, eaao6897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mercurio, A.M. VEGF/neuropilin signaling in cancer stem cells. Int. J. Mol. Sci. 2019, 20, 490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raghavan, S.; Mehta, P.; Xie, Y.; Lei, Y.L.; Mehta, G. Ovarian cancer stem cells and macrophages reciprocally interact through the WNT pathway to promote pro-tumoral and malignant phenotypes in 3D engineered microenvironments. J. Immunother. Cancer 2019, 7, 190. [Google Scholar] [CrossRef] [Green Version]
- Lu, H.; Clauser, K.R.; Tam, W.L.; Fröse, J.; Ye, X.; Eaton, E.N.; Reinhardt, F.; Donnenberg, V.S.; Bhargava, R.; Carr, S.A.; et al. A breast cancer stem cell niche supported by juxtacrine signalling from monocytes and macrophages. Nat. Cell Biol. 2014, 16, 1105–1117. [Google Scholar] [CrossRef] [Green Version]
- Gabrusiewicz, K.; Li, X.; Wei, J.; Hashimoto, Y.; Marisetty, A.L.; Ott, M.; Wang, F.; Hawke, D.; Yu, J.; Healy, L.M.; et al. Glioblastoma stem cell-derived exosomes induce M2 macrophages and PD-L1 expression on human monocytes. Oncoimmunology 2018, 7, e1412909. [Google Scholar] [CrossRef]
- Domenis, R.; Cesselli, D.; Toffoletto, B.; Bourkoula, E.; Caponnetto, F.; Manini, I.; Beltrami, A.P.; Ius, T.; Skrap, M.; Di Loreto, C.; et al. Systemic T cells immunosuppression of glioma stem cell-derived exosomes is mediated by monocytic myeloid-derived suppressor cells. PLoS ONE 2017, 12, e0169932. [Google Scholar] [CrossRef] [Green Version]
- Biswas, S.; Mandal, G.; Roy Chowdhury, S.; Purohit, S.; Payne, K.K.; Anadon, C.; Gupta, A.; Swanson, P.; Yu, X.; Conejo-Garcia, J.R.; et al. Exosomes produced by mesenchymal stem cells drive differentiation of myeloid cells into immunosuppressive M2-polarized macrophages in breast cancer. J. Immunol. 2019, 203, 3447–3460. [Google Scholar] [CrossRef]
- Grange, C.; Tapparo, M.; Tritta, S.; Deregibus, M.C.; Battaglia, A.; Gontero, P.; Frea, B.; Camussi, G. Role of HLA-G and extracellular vesicles in renal cancer stem cell-induced inhibition of dendritic cell differentiation. BMC Cancer 2015, 15, 1009. [Google Scholar] [CrossRef] [Green Version]
- Zhang, B.; Yin, Y.; Lai, R.C.; Tan, S.S.; Choo, A.B.H.; Lim, S.K. Mesenchymal stem cells secrete immunologically active exosomes. Stem. Cells Dev. 2014, 23, 1233–1244. [Google Scholar] [CrossRef]
- Hwang, W.L.; Lan, H.Y.; Cheng, W.C.; Huang, S.C.; Yang, M.H. Tumor stem-like cell-derived exosomal RNAs prime neutrophils for facilitating tumorigenesis of colon cancer. J. Hematol. Oncol. 2019, 12, 10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alzahrani, F.A.; El-Magd, M.A.; Abdelfattah-Hassan, A.; Saleh, A.A.; Saadeldin, I.M.; El-Shetry, E.S.; Badawy, A.A.; Alkarim, S. Potential effect of exosomes derived from cancer stem cells and MSCs on progression of DEN-induced HCC in rats. Stem Cells Int. 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, Z.P.; Li, A.Q.; Jia, W.H.; Ye, S.; Van Eps, G.; Yu, J.M.; Yang, W.J. MicroRNA expression profiling in exosomes derived from gastric cancer stem-like cells. Oncotarget 2017, 8, 93839–93855. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grange, C.; Tapparo, M.; Collino, F.; Vitillo, L.; Damasco, C.; Deregibus, M.C.; Tetta, C.; Bussolati, B.; Camussi, G. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res. 2011, 71, 5346–5356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, L.; Yang, G.; Zhao, D.; Wang, J.; Bai, Y.; Peng, Q.; Wang, H.; Fang, R.; Chen, G.; Wang, Z.; et al. CD103-positive CSC exosome promotes EMT of clear cell renal cell carcinoma: Role of remote MiR-19b-3p. Mol. Cancer 2019, 18, 86. [Google Scholar] [CrossRef] [PubMed]
- Hardin, H.; Helein, H.; Meyer, K.; Robertson, S.; Zhang, R.; Zhong, W.; Lloyd, R.V. Thyroid cancer stem-like cell exosomes: Regulation of EMT via transfer of lncRNAs. Lab. Investig. 2018, 98, 1133–1142. [Google Scholar] [CrossRef] [Green Version]
- McAllister, S.S.; Weinberg, R.A. The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis. Nat. Cell Biol. 2014, 16, 717–727. [Google Scholar] [CrossRef]
- Peinado, H.; Lavotshkin, S.; Lyden, D. The secreted factors responsible for pre-metastatic niche formation: Old sayings and new thoughts. Semin. Cancer Biol. 2011, 21, 139–146. [Google Scholar] [CrossRef]
- Fidler, I.J. The pathogenesis of cancer metastasis: The “seed and soil” hypothesis revisited. Nat. Rev. Cancer 2003, 3, 453–458. [Google Scholar] [CrossRef]
- Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Invest. 2009, 119, 1420–1428. [Google Scholar] [CrossRef] [Green Version]
- Dang, H.; Ding, W.; Emerson, D.; Rountree, C.B. Snail1 induces epithelial-to-mesenchymal transition and tumor initiating stem cell characteristics. BMC Cancer 2011, 11, 396. [Google Scholar] [CrossRef] [Green Version]
- Scheel, C.; Weinberg, R.A. Cancer stem cells and epithelial-mesenchymal transition: Concepts and molecular links. Semin. Cancer Biol. 2012, 22, 396–403. [Google Scholar] [CrossRef] [PubMed]
- Peinado, H.; Quintanilla, M.; Cano, A. Transforming growth factor β-1 induces Snail transcription factor in epithelial cell lines. Mechanisms for epithelial mesenchymal transitions. J. Biol. Chem. 2003, 278, 21113–21123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Feng, X.-H.; Derynck, R. Specificity and versatility in Tgf-Β signaling through smads. Annu. Rev. Cell Dev. Biol. 2005, 21, 659–693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kasahara, Y.; Shirota, H.; Umegaki, S.; Ishioka, C. Contribution of Fcγ receptor IIB to creating a suppressive tumor microenvironment in a mouse model. In Proceedings of the Abstracts of the 77th Annual Meeting of the Japanese Cancer Association, Osaka, Japan, 27–29 September 2018. [Google Scholar]
- Fazilaty, H.; Gardaneh, M.; Bahrami, T.; Salmaninejad, A.; Behnam, B. Crosstalk between breast cancer stem cells and metastatic niche: Emerging molecular metastasis pathway? Tumor Biol. 2013, 34, 2019–2030. [Google Scholar] [CrossRef] [PubMed]
- Hiratsuka, S.; Watanabe, A.; Aburatani, H.; Maru, Y. Tumour-mediated upregulation of chemoattractants and recruitment of myeloid cells predetermines lung metastasis. Nat. Cell Biol. 2006, 8, 1369–1375. [Google Scholar] [CrossRef] [PubMed]
- Sceneay, J.; Chow, M.T.; Chen, A.; Halse, H.M.; Wong, C.S.F.; Andrews, D.M.; Sloan, E.K.; Parker, B.S.; Bowtell, D.D.; Smyth, M.J.; et al. Primary tumor hypoxia recruits CD11b+/Ly6Cmed/ Ly6G+ immune suppressor cells and compromises NK cell cytotoxicity in the premetastatic niche. Cancer Res. 2012, 72, 3906–3911. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Cao, X. Characteristics and significance of the pre-metastatic niche. Cancer Cell 2016, 30, 668–681. [Google Scholar] [CrossRef] [Green Version]
- Kitamura, T.; Qian, B.Z.; Pollard, J.W. Immune cell promotion of metastasis. Nat. Rev. Immunol. 2015, 15, 73–86. [Google Scholar] [CrossRef]
- Kaplan, R.N.; Riba, R.D.; Zacharoulis, S.; Bramley, A.H.; Vincent, L.; Costa, C.; MacDonald, D.D.; Jin, D.K.; Shido, K.; Kerns, S.A.; et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 2005, 438, 820–827. [Google Scholar] [CrossRef]
- Zhou, Z.N.; Sharma, V.P.; Beaty, B.T.; Roh-Johnson, M.; Peterson, E.A.; Van Rooijen, N.; Kenny, P.A.; Wiley, H.S.; Condeelis, J.S.; Segall, J.E. Autocrine HBEGF expression promotes breast cancer intravasation, metastasis and macrophage-independent invasion in vivo. Oncogene 2014, 33, 3784–3793. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, J.; Yao, Y.; Gong, C.; Yu, F.; Su, S.; Chen, J.; Liu, B.; Deng, H.; Wang, F.; Lin, L.; et al. CCL18 from tumor-associated macrophages promotes breast cancer metastasis via PITPNM3. Cancer Cell 2011, 19, 541–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sangaletti, S.; Di Carlo, E.; Gariboldi, S.; Miotti, S.; Cappetti, B.; Parenza, M.; Rumio, C.; Brekken, R.A.; Chiodoni, C.; Colombo, M.P. Macrophage-derived SPARC bridges tumor cell-extracellular matrix interactions toward metastasis. Cancer Res. 2008, 68, 9050–9059. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Curiel, T.J.; Coukos, G.; Zou, L.; Alvarez, X.; Cheng, P.; Mottram, P.; Evdemon-Hogan, M.; Conejo-Garcia, J.R.; Zhang, L.; Burow, M.; et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat. Med. 2004, 10, 942–949. [Google Scholar] [CrossRef] [PubMed]
- Kenkel, J.A.; Tseng, W.W.; Davidson, M.G.; Tolentino, L.L.; Choi, O.; Bhattacharya, N.; Seeley, E.S.; Winer, D.A.; Reticker-Flynn, N.E.; Engleman, E.G. An immunosuppressive dendritic cell subset accumulates at secondary sites and promotes metastasis in pancreatic cancer. Cancer Res. 2017, 77, 4158–4170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coffelt, S.B.; Kersten, K.; Doornebal, C.W.; Weiden, J.; Vrijland, K.; Hau, C.S.; Verstegen, N.J.M.; Ciampricotti, M.; Hawinkels, L.J.A.C.; Jonkers, J.; et al. IL-17-producing γδ T cells and neutrophils conspire to promote breast cancer metastasis. Nature 2015, 522, 345–348. [Google Scholar] [CrossRef]
- Wellenstein, M.D.; Coffelt, S.B.; Duits, D.E.M.; van Miltenburg, M.H.; Slagter, M.; de Rink, I.; Henneman, L.; Kas, S.M.; Prekovic, S.; Hau, C.S.; et al. Loss of p53 triggers WNT-dependent systemic inflammation to drive breast cancer metastasis. Nature 2019, 572, 538–542. [Google Scholar] [CrossRef]
- Wculek, S.K.; Malanchi, I. Neutrophils support lung colonization of metastasis-initiating breast cancer cells. Nature 2015, 528, 413–417. [Google Scholar] [CrossRef]
- Nawaz, M.; Fatima, F.; Vallabhaneni, K.C.; Penfornis, P.; Valadi, H.; Ekström, K.; Kholia, S.; Whitt, J.D.; Fernandes, J.D.; Pochampally, R.; et al. Extracellular vesicles: Evolving factors in stem cell biology. Stem Cells Int. 2016. [Google Scholar] [CrossRef] [Green Version]
- Kong, J.; Tian, H.; Zhang, F.; Zhang, Z.; Li, J.; Liu, X.; Li, X.; Liu, J.; Li, X.; Jin, D.; et al. Extracellular vesicles of carcinoma-associated fibroblasts creates a pre-metastatic niche in the lung through activating fibroblasts. Mol. Cancer 2019, 18, 1–16. [Google Scholar] [CrossRef]
- Kalluri, R.; Zeisberg, M. Fibroblasts in cancer. Nat. Rev. Cancer 2006, 6, 392–401. [Google Scholar] [CrossRef] [PubMed]
- Giannoni, E.; Bianchini, F.; Masieri, L.; Serni, S.; Torre, E.; Calorini, L.; Chiarugi, P. Reciprocal activation of prostate cancer cells and cancer-associated fibroblasts stimulates epithelial-mesenchymal transition and cancer stemness. Cancer Res. 2010, 70, 6945–6956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, W.J.; Ho, C.C.; Chang, Y.L.; Chen, H.Y.; Lin, C.A.; Ling, T.Y.; Yu, S.L.; Yuan, S.S.; Louisa Chen, Y.J.; Lin, C.Y.; et al. Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling. Nat. Commun. 2014, 5, 1–17. [Google Scholar] [CrossRef] [PubMed]
- Costa, A.; Kieffer, Y.; Scholer-Dahirel, A.; Pelon, F.; Bourachot, B.; Cardon, M.; Sirven, P.; Magagna, I.; Fuhrmann, L.; Bernard, C.; et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell 2018, 33, 463–479. [Google Scholar] [CrossRef] [Green Version]
- Hu, Y.; Yan, C.; Mu, L.; Huang, K.; Li, X.; Tao, D.; Wu, Y.; Qin, J. Fibroblast-derived exosomes contribute to chemoresistance through priming cancer stem cells in colorectal cancer. PLoS ONE 2015, 10, e0125625. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boelens, M.C.; Wu, T.J.; Nabet, B.Y.; Xu, B.; Qiu, Y.; Yoon, T.; Azzam, D.J.; Twyman-Saint Victor, C.; Wiemann, B.Z.; Ishwaran, H.; et al. Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell 2014, 159, 499–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Su, S.; Chen, J.; Yao, H.; Liu, J.; Yu, S.; Lao, L.; Wang, M.; Luo, M.; Xing, Y.; Chen, F.; et al. CD10+GPR77+ cancer-associated fibroblasts promote cancer formation and chemoresistance by sustaining cancer stemness. Cell 2018, 172, 841–856. [Google Scholar] [CrossRef]
- Donnarumma, E.; Fiore, D.; Nappa, M.; Roscigno, G.; Adamo, A.; Iaboni, M.; Russo, V.; Affinito, A.; Puoti, I.; Quintavalle, C.; et al. Cancer-associated fibroblasts release exosomal microRNAs that dictate an aggressive phenotype in breast cancer. Oncotarget 2017, 8, 19592–19608. [Google Scholar] [CrossRef] [Green Version]
- Luga, V.; Zhang, L.; Viloria-Petit, A.M.; Ogunjimi, A.A.; Inanlou, M.R.; Chiu, E.; Buchanan, M.; Hosein, A.N.; Basik, M.; Wrana, J.L. Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell 2012, 151, 1542–1556. [Google Scholar] [CrossRef] [Green Version]
- Malanchi, I.; Santamaria-Martínez, A.; Susanto, E.; Peng, H.; Lehr, H.A.; Delaloye, J.F.; Huelsken, J. Interactions between cancer stem cells and their niche govern metastatic colonization. Nature 2012, 481, 85–91. [Google Scholar] [CrossRef]
- Fessler, E.; Dijkgraaf, F.E.; De Sousa, E.; Melo, F.; Medema, J.P. Cancer stem cell dynamics in tumor progression and metastasis: Is the microenvironment to blame? Cancer Lett. 2013, 341, 97–104. [Google Scholar] [CrossRef] [PubMed]
- Oskarsson, T.; Acharyya, S.; Zhang, X.H.F.; Vanharanta, S.; Tavazoie, S.F.; Morris, P.G.; Downey, R.J.; Manova-Todorova, K.; Brogi, E.; Massagué, J. Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs. Nat. Med. 2011, 17, 867–874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lytle, N.K.; Barber, A.G.; Reya, T. Stem cell fate in cancer growth, progression and therapy resistance. Nat. Rev. Cancer 2018, 18, 669–680. [Google Scholar] [CrossRef] [PubMed]
- Jiang, W.; Chan, C.K.; Weissman, I.L.; Kim, B.Y.S.; Hahn, S.M. Immune priming of the tumor microenvironment by radiation. Trends Cancer 2016, 2, 638–645. [Google Scholar] [CrossRef]
- Frey, B.; Rückert, M.; Deloch, L.; Rühle, P.F.; Derer, A.; Fietkau, R.; Gaipl, U.S. Immunomodulation by ionizing radiation—Impact for design of radio-immunotherapies and for treatment of inflammatory diseases. Immunol. Rev. 2017, 280, 231–248. [Google Scholar] [CrossRef]
- Jarosz-Biej, M.; Smolarczyk, R.; Cichoń, T.; Kułach, N. Tumor microenvironment as a “game changer” in cancer radiotherapy. Int. J. Mol. Sci. 2019, 20, 3212. [Google Scholar] [CrossRef] [Green Version]
- Holley, A.K.; Miao, L.; St. Clair, D.K.; St. Clair, W.H. Redox-modulated phenomena and radiation therapy: The central role of superoxide dismutases. Antioxidants Redox Signal. 2014, 20, 1567–1589. [Google Scholar] [CrossRef] [Green Version]
- Kuonen, F.; Secondini, C.; Rüegg, C. Molecular pathways: Emerging pathways mediating growth, invasion, and metastasis of tumors progressing in an irradiated microenvironment. Clin. Cancer Res. 2012, 18, 5196–5202. [Google Scholar] [CrossRef] [Green Version]
- Shigdar, S.; Lin, J.; Li, Y.; Yang, C.J.; Wei, M.; Zhu, Y.; Liu, H.; Duan, W. Cancer stem cell targeting: The next generation of cancer therapy and molecular imaging. Ther. Deliv. 2012, 3, 227–244. [Google Scholar] [CrossRef]
- MacDonagh, L.; Gray, S.G.; Breen, E.; Cuffe, S.; Finn, S.P.; O’Byrne, K.J.; Barr, M.P. BBI608 inhibits cancer stemness and reverses cisplatin resistance in NSCLC. Cancer Lett. 2018, 428, 117–126. [Google Scholar] [CrossRef]
- Wong, A.L.; Soo, R.A.; Tan, D.S.; Lee, S.C.; Lim, J.S.; Marban, P.C.; Kong, L.R.; Lee, Y.J.; Wang, L.Z.; Thuya, W.L.; et al. Phase I and biomarker study of OPB-51602, a novel signal transducer and activator of transcription (STAT) 3 inhibitor, in patients with refractory solid malignancies. Ann. Oncol. 2015, 26, 998–1005. [Google Scholar] [CrossRef] [PubMed]
- Han, D.; Yu, T.; Dong, N.; Wang, B.; Sun, F.; Jiang, D. Napabucasin, a novel STAT3 inhibitor suppresses proliferation, invasion and stemness of glioblastoma cells. J. Exp. Clin. Cancer Res. 2019, 38, 289. [Google Scholar] [CrossRef] [PubMed]
- Sikic, B.I.; Lakhani, N.; Patnaik, A.; Shah, S.A.; Chandana, S.R.; Rasco, D.; Colevas, A.D.; O’Rourke, T.; Narayanan, S.; Papadopoulos, K.; et al. First-in-human, first-in-class phase i trial of the anti-CD47 antibody Hu5F9-G4 in patients with advanced cancers. J. Clin. Oncol. 2019, 37, 946–953. [Google Scholar] [CrossRef] [PubMed]
- Zhi, Y.; Mou, Z.; Chen, J.; He, Y.; Dong, H.; Fu, X.; Wu, Y. B7H1 expression and epithelial-to-mesenchymal transition phenotypes on colorectal cancer stem-like cells. PLoS ONE 2015, 10, e0135528. [Google Scholar] [CrossRef]
- Yang, Y.; Wu, K.; Zhao, E.; Li, W.; Shi, L.; Xie, G.; Jiang, B.; Wang, Y.; Li, R.; Zhang, P.; et al. B7-H1 enhances proliferation ability of gastric cancer stem-like cells as a receptor. Oncol. Lett. 2015, 9, 1833–1838. [Google Scholar] [CrossRef] [Green Version]
- Gupta, H.B.; Hurez, V.; Clark, C.A.; Sareddy, G.R.; Vadlamudi, R.; Li, R.; Curiel, T.J. Tumor B7-H1 regulates cancer stem cell generation and virulence. J. Immunol. 2016, 196, 72–73. [Google Scholar]
- Chen, Y.; Di, C.; Zhang, X.; Wang, J.; Wang, F.; Yan, J.F.; Xu, C.; Zhang, J.; Zhang, Q.; Li, H.; et al. Transforming growth factor β signaling pathway: A promising therapeutic target for cancer. J. Cell. Physiol. 2020, 235, 1903–1914. [Google Scholar] [CrossRef]
- Chen, D.S.; Hurwitz, H. Combinations of bevacizumab with cancer immunotherapy. Cancer J. 2018, 24, 193–204. [Google Scholar] [CrossRef]
- Xu, Q.; Liu, G.; Yuan, X.; Xu, M.; Wang, H.; Ji, J.; Konda, B.; Black, K.L.; Yu, J.S. Antigen-specific T-cell response from dendritic cell vaccination using cancer stem-like cell-associated antigens. Stem Cells 2009, 27, 1734–1740. [Google Scholar] [CrossRef] [Green Version]
- Fesnak, A.D.; June, C.H.; Levine, B.L. Engineered T cells: The promise and challenges of cancer immunotherapy. Nat. Rev. Cancer 2016, 16, 566–581. [Google Scholar] [CrossRef]
- Guo, Y.; Feng, K.; Wang, Y.; Han, W. Targeting cancer stem cells by using chimeric antigen receptor-modified T cells: A potential and curable approach for cancer treatment. Protein Cell 2018, 9, 516–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weiner, G.J. Building better monoclonal antibody-based therapeutics. Nat. Rev. Cancer 2015, 15, 361–370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deng, Z.; Wu, Y.; Ma, W.; Zhang, S.; Zhang, Y.Q. Adoptive T-cell therapy of prostate cancer targeting the cancer stem cell antigen EpCAM. BMC Immunol. 2015, 16, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
© 2020 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
Pastò, A.; Consonni, F.M.; Sica, A. Influence of Innate Immunity on Cancer Cell Stemness. Int. J. Mol. Sci. 2020, 21, 3352. https://doi.org/10.3390/ijms21093352
Pastò A, Consonni FM, Sica A. Influence of Innate Immunity on Cancer Cell Stemness. International Journal of Molecular Sciences. 2020; 21(9):3352. https://doi.org/10.3390/ijms21093352
Chicago/Turabian StylePastò, Anna, Francesca Maria Consonni, and Antonio Sica. 2020. "Influence of Innate Immunity on Cancer Cell Stemness" International Journal of Molecular Sciences 21, no. 9: 3352. https://doi.org/10.3390/ijms21093352
APA StylePastò, A., Consonni, F. M., & Sica, A. (2020). Influence of Innate Immunity on Cancer Cell Stemness. International Journal of Molecular Sciences, 21(9), 3352. https://doi.org/10.3390/ijms21093352