The Interplay of Autophagy and Tumor Microenvironment in Colorectal Cancer—Ways of Enhancing Immunotherapy Action
Abstract
:1. Introduction
2. The Major Players in the Tumor Microenvironment
2.1. The Heterogeneity of the Tumor Microenvironment
2.2. The Role of T-Lymphocytes
2.3. The Role of Tumor-Associated Myeloid Cells
2.4. Cancer-Associated Fibroblasts (CAFs)
2.5. Angiogenesis and Neo-Vascularization Process in Tumor Stroma
2.6. Other Immune Cell Types in the Tumor Microenvironment
3. The Role of Autophagy in Stroma Development, Inflammation, and the Immunity Response
3.1. The Role of Inflammation in Colorectal Cancer Development
3.2. Hypoxia-Induced Autophagy in the Tumor Microenvironment
3.3. The Cross-Talk between Autophagy and Antigen Presenting Cells
3.4. Autophagy—A Key Regulator of T-Cell Activation
4. The Current State of Immunotherapy in CRC Patients
5. Targeting Autophagy—A Promising Anti-Cancer Strategy
5.1. The Main Autophagy Inhibitors in Cancer Therapy
5.2. Activators of Autophagy for Cancer Therapy
6. Conclusions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
ATGs | Autophagy-related genes |
CAFs | Cancer-associated fibroblasts |
CRC | Colorectal cancer |
CTLs | Cytotoxic T lymphocytes |
CTLA-4 | cytotoxic T-lymphocyte-associated antigen-4 |
CQ | Chloroquine |
PD-1 | Programmed cell death protein 1 |
PD-L1 | Programmed death-ligand 1 |
HCQ | Hydroxyl-chloroquine |
mCRC | metastatic Colorectal cancer |
MDSCs | Myeloid-derived suppressor cell |
MHC I and II | Major histocompatibility complex I and II |
MoAbs | Monoclonal antibodies |
NSCLC | Non-small cell lung cancer |
MMRd | Mismatch repair deficiency |
MSI-H | Microsatellite instability-High |
MSS | Microsatellite stable |
TANs | Tumor-associated neutrophils |
TAMs | Tumor-associated macrophages |
TCR | T-cell receptor |
TILs | Tumor-infiltrating lymphocytes |
Tregs | Regulatory T cells |
3-MA | 3-Methyladenine |
References
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2019. CA Cancer J. Clin. 2019, 69, 7–34. [Google Scholar] [CrossRef] [PubMed]
- Arnold, M.; Sierra, M.S.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global patterns and trends in colorectal cancer incidence and mortality. Gut 2017, 66, 683–691. [Google Scholar] [CrossRef]
- Mizushima, N.; Ohsumi, Y.; Yoshimori, T. Autophagosome Formation in Mammalian Cells. Cell Struct. Funct. 2002, 27, 421–429. [Google Scholar] [CrossRef]
- Koustas, E.; Karamouzis, M.V.; Mihailidou, C.; Schizas, D.; Papavassiliou, A.G. Co-targeting of EGFR and autophagy signaling is an emerging treatment strategy in metastatic colorectal cancer. Cancer Lett. 2017, 396, 94–102. [Google Scholar] [CrossRef] [PubMed]
- Pandurangan, A.K.; Divya, T.; Kumar, K.; Dineshbabu, V.; Velavan, B.; Sudhandiran, G.; AshokKumar, P. Colorectal carcinogenesis: Insights into the cell death and signal transduction pathways: A review. J. Gastrointest. Oncol. 2018, 10, 244–259. [Google Scholar]
- Kroemer, G.; Mariño, G.; Levine, B. Autophagy and the integrated stress response. Mol. Cell 2010, 40, 280–293. [Google Scholar] [CrossRef]
- Burada, F.; Nicoli, E.R.; Ciurea, M.E.; Uscatu, D.C.; Ioana, M.; Gheonea, D.I. Autophagy in colorectal cancer: An important switch from physiology to pathology. World J. Gastrointest. Oncol. 2015, 7, 271–284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Koustas, E.; Sarantis, P.; Papavassiliou, A.G.; Karamouzis, M. V Upgraded role of autophagy in colorectal carcinomas. World J. Gastrointest. Oncol. 2018, 10, 367–369. [Google Scholar] [CrossRef]
- Schaaf, M.B.; Houbaert, D.; Meçe, O.; Agostinis, P. Autophagy in endothelial cells and tumor angiogenesis. Cell Death Differ. 2019, 26, 665–679. [Google Scholar] [CrossRef]
- Colella, B.; Faienza, F.; Di Bartolomeo, S. EMT Regulation by Autophagy: A New Perspective in Glioblastoma Biology. Cancers 2019, 11, 312. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.; Yu, D.-D.; Yan, F.; Jing, Y.-Y.; Han, Z.-P.; Sun, K.; Liang, L.; Hou, J.; Wei, L.-X. The role of autophagy induced by tumor microenvironment in different cells and stages of cancer. Cell Biosci. 2015, 5, 14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- White, E. Deconvoluting the context-dependent role for autophagy in cancer. Nat. Rev. Cancer 2012, 12, 401–410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guo, J.Y.; White, E. Autophagy, metabolism, and cancer. Cold Spring Harb. Symp. Quant. Biol. 2016, 81, 73–78. [Google Scholar] [CrossRef] [PubMed]
- Mellor, H.R.; Harris, A.L. The role of the hypoxia-inducible BH3-only proteins BNIP3 and BNIP3L in cancer. Cancer Metastasis Rev. 2007, 26, 553–566. [Google Scholar] [CrossRef] [PubMed]
- Huijbers, A.; Plantinga, T.S.; Joosten, L.A.B.; Aben, K.K.H.; Gudmundsson, J.; Heijer, M.D.; Kiemeney, L.A.L.M.; Netea, M.G.; Hermus, A.R.M.M.; Netea-Maier, R.T. The effect of the ATG16L1 Thr300Ala polymorphism on susceptibility and outcome of patients with epithelial cell-derived thyroid carcinoma. Endocr. Relat. Cancer 2012, 19, L15–L18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, C.-Y.; Huang, S.-P.; Lin, V.C.; Yu, C.-C.; Chang, T.-Y.; Lu, T.-L.; Chiang, H.-C.; Bao, B.-Y. Genetic variants of the autophagy pathway as prognostic indicators for prostate cancer. Sci. Rep. 2015, 5, 14045. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yun, C.W.; Lee, S.H. The Roles of Autophagy in Cancer. Int. J. Mol. Sci. 2018, 19, 3466. [Google Scholar] [CrossRef]
- Miracco, C.; Cosci, E.; Oliveri, G.; Luzi, P.; Pacenti, L.; Monciatti, I.; Mannucci, S.; De Nisi, M.C.; Toscano, M.; Malagnino, V.; et al. Protein and mRNA expression of autophagy gene Beclin 1 in human brain tumours. Int. J. Oncol. 2007, 30, 429–436. [Google Scholar] [PubMed]
- Pickford, F.; Masliah, E.; Britschgi, M.; Lucin, K.; Narasimhan, R.; Jaeger, P.A.; Small, S.; Spencer, B.; Rockenstein, E.; Levine, B.; et al. The autophagy-related protein beclin 1 shows reduced expression in early Alzheimer disease and regulates amyloid β accumulation in mice. J. Clin. Investig. 2008, 118, 2190–2199. [Google Scholar] [CrossRef] [Green Version]
- Mariño, G.; Salvador-Montoliu, N.; Fueyo, A.; Knecht, E.; Mizushima, N.; López-Otín, C. Tissue-specific Autophagy Alterations and Increased Tumorigenesis in Mice Deficient in Atg4C/Autophagin-3. J. Biol. Chem. 2007, 282, 18573–18583. [Google Scholar] [CrossRef] [Green Version]
- Oikonomou, E.; Koustas, E.; Goulielmaki, M.; Pintzas, A. BRAF vs. RAS oncogenes: Are mutations of the same pathway equal? Differential signalling and therapeutic implications. Oncotarget 2015, 5, 11752–11777. [Google Scholar] [CrossRef]
- Cianfanelli, V.; D’Orazio, M.; Cecconi, F. AMBRA1 and BECLIN 1 interplay in the crosstalk between autophagy and cell proliferation. Cell Cycle 2015, 14, 959–963. [Google Scholar] [CrossRef] [Green Version]
- Yue, Z.; Jin, S.; Yang, C.; Levine, A.J.; Heintz, N. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc. Natl. Acad. Sci. USA 2003, 100, 15077–15082. [Google Scholar] [CrossRef] [Green Version]
- Guo, J.Y.; Chen, H.-Y.; Mathew, R.; Fan, J.; Strohecker, A.M.; Karsli-Uzunbas, G.; Kamphorst, J.J.; Chen, G.; Lemons, J.M.; Karantza, V.; et al. Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis. Genes Dev. 2011, 25, 460–470. [Google Scholar] [CrossRef] [Green Version]
- Zhong, Z.; Sanchez-Lopez, E.; Karin, M. Autophagy, Inflammation and Immunity: A Troika Governing Cancer and Its Treatment. Cell 2016, 166, 288–298. [Google Scholar] [CrossRef]
- Degenhardt, K.; Mathew, R.; Beaudoin, B.; Bray, K.; Anderson, D.; Chen, G.; Mukherjee, C.; Shi, Y.; Gélinas, C.; Fan, Y.; et al. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 2006, 10, 51–64. [Google Scholar] [CrossRef] [Green Version]
- Gerlinger, M.; Rowan, A.J.; Horswell, S.; Math, M.; Larkin, J.; Endesfelder, D.; Gronroos, E.; Martinez, P.; Matthews, N.; Stewart, A.; et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 2012, 366, 883–892. [Google Scholar] [CrossRef]
- Molinari, C.; Marisi, G.; Passardi, A.; Matteucci, L.; De Maio, G.; Ulivi, P. Heterogeneity in Colorectal Cancer: A Challenge for Personalized Medicine? Int. J. Mol. Sci. 2018, 19, 3733. [Google Scholar] [CrossRef]
- Pietras, K.; Östman, A. Hallmarks of cancer: Interactions with the tumor stroma. Exp. Cell Res. 2010, 316, 1324–1331. [Google Scholar] [CrossRef]
- Tape, C.J. The Heterocellular Emergence of Colorectal Cancer. Trends Cancer 2017, 3, 79–88. [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]
- Kather, J.N.; Halama, N.; Jaeger, D. Genomics and emerging biomarkers for immunotherapy of colorectal cancer. Semin. Cancer Biol. 2018, 52, 189–197. [Google Scholar] [CrossRef]
- Bupathi, M.; Wu, C. Biomarkers for immune therapy in colorectal cancer: Mismatch-repair deficiency and others. J. Gastrointest. Oncol. 2016, 7, 713–720. [Google Scholar] [CrossRef]
- Mlecnik, B.; Bindea, G.; Kirilovsky, A.; Angell, H.K.; Obenauf, A.C.; Tosolini, M.; Church, S.E.; Maby, P.; Vasaturo, A.; Angelova, M.; et al. The tumor microenvironment and Immunoscore are critical determinants of dissemination to distant metastasis. Sci. Transl. Med. 2016, 8, 327. [Google Scholar] [CrossRef]
- Nakagawa, K.; Tanaka, K.; Homma, Y.; Nojiri, K.; Kumamoto, T.; Takeda, K.; Endo, I. Low Infiltration of Peritumoral Regulatory T Cells Predicts Worse Outcome Following Resection of Colorectal Liver Metastases. Ann. Surg. Oncol. 2015, 22, 180–186. [Google Scholar] [CrossRef]
- Yu, P.; Fu, Y.X. Tumor-infiltrating T lymphocytes: Friends or foes? Lab. Investig. 2006, 86, 231–245. [Google Scholar] [CrossRef]
- Hu, Z.; Ma, Y.; Shang, Z.; Hu, S.; Liang, K.; Liang, W.; Xing, X.; Wang, Y.; Du, X. Improving immunotherapy for colorectal cancer using dendritic cells combined with anti-programmed death-ligand in vitro. Oncol. Lett. 2018, 15, 5345–5351. [Google Scholar] [CrossRef] [Green Version]
- Pauken, K.E.; Wherry, E.J. Overcoming T cell exhaustion in infection and cancer. Trends Immunol. 2015, 36, 265–276. [Google Scholar] [CrossRef] [Green Version]
- Singh, P.P.; Sharma, P.K.; Krishnan, G.; Lockhart, A.C. Immune checkpoints and immunotherapy for colorectal cancer. Gastroenterol. Rep. 2015, 3, 289–297. [Google Scholar] [CrossRef] [Green Version]
- Derhovanessian, E.; Maier, A.B.; Beck, R.; Jahn, G.; Hähnel, K.; Slagboom, P.; De Craen, A.J.M.; Westendorp, R.G.J.; Pawelec, G. Hallmark Features of Immunosenescence Are Absent in Familial Longevity. J. Immunol. 2010, 185, 4618–4624. [Google Scholar] [CrossRef] [Green Version]
- Koustas, E.; Papavassiliou, A.G.; Karamouzis, M.V. The role of autophagy in the treatment of BRAF mutant colorectal carcinomas differs based on microsatellite instability status. PLoS ONE 2018, 13, e0207227. [Google Scholar] [CrossRef]
- Chen, Y.; Colello, J.; Jarjour, W.; Zheng, S.G. Cellular Metabolic Regulation in the Differentiation and Function of Regulatory T Cells. Cells 2019, 8, 188. [Google Scholar] [CrossRef]
- Morikawa, H.; Sakaguchi, S. Genetic and epigenetic basis of Treg cell development and function: From a FoxP3-centered view to an epigenome-defined view of natural Treg cells. Immunol. Rev. 2014, 259, 192–205. [Google Scholar] [CrossRef]
- Herk, E.H.; Velde, A.A. Treg subsets in inflammatory bowel disease and colorectal carcinoma. Characteristics, role and therapeutic targets. J. Gastroenterol. Hepatol. 2016, 31, 1393–1404. [Google Scholar] [CrossRef] [Green Version]
- Hori, S.; Nomura, T.; Sakaguchi, S. Control of Regulatory T Cell Development by the Transcription Factor Foxp3. Science 2003, 299, 1057–1061. [Google Scholar] [CrossRef]
- Fontenot, J.D.; Gavin, M.A.; Rudensky, A.Y. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat. Immunol. 2003, 4, 330–336. [Google Scholar] [CrossRef]
- Salama, P.; Phillips, M.; Grieu, F.; Morris, M.; Zeps, N.; Joseph, D.; Platell, C.; Iacopetta, B. Tumor-infiltrating FOXP3+ T regulatory cells show strong prognostic significance in colorectal cancer. J. Clin. Oncol. 2009, 27, 186–192. [Google Scholar] [CrossRef]
- Sinicrope, F.A.; Rego, R.L.; Ansell, S.M.; Knutson, K.L.; Foster, N.R.; Sargent, D.J. Intraepithelial Effector (CD3+)/Regulatory (FoxP3+) T-Cell Ratio Predicts a Clinical Outcome of Human Colon Carcinoma. Gastroenterology 2009, 137, 1270–1279. [Google Scholar] [CrossRef]
- Gao, D.; Mittal, V. The role of bone-marrow-derived cells in tumor growth, metastasis initiation and progression. Trends Mol. Med. 2009, 15, 333–343. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Mantovani, A.; Cassatella, M.A.; Costantini, C.; Jaillon, S. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat. Rev. Immunol. 2011, 11, 519–531. [Google Scholar] [CrossRef] [PubMed]
- Szebeni, G.J.; Vizler, C.; Nagy, L.I.; Kitajka, K.; Puskas, L.G.; Tanaka, T.; Shimizu, M. Pro-Tumoral Inflammatory Myeloid Cells as Emerging Therapeutic Targets. Int. J. Mol. Sci. 2016, 17, 1958. [Google Scholar] [CrossRef]
- Shaul, M.E.; Fridlender, Z.G. Neutrophils as active regulators of the immune system in the tumor microenvironment. J. Leukoc. Biol. 2017, 102, 343–349. [Google Scholar] [CrossRef] [PubMed]
- Houghton, A.M.; Rzymkiewicz, D.M.; Ji, H.; Gregory, A.D.; Egea, E.E.; Metz, H.E.; Stolz, D.B.; Land, S.R.; Marconcini, L.A.; Kliment, C.R.; et al. Neutrophil Elastase-Mediated Degradation of IRS-1 Accelerates Lung Tumor Growth. Nat. Med. 2010, 16, 219–223. [Google Scholar] [CrossRef] [Green Version]
- Galdiero, M.R.; Varricchi, G.; Loffredo, S.; Mantovani, A.; Marone, G. Roles ofneutrophils in cancer growth and progression. J. Leukoc. Biol. 2018, 103, 457–464. [Google Scholar] [CrossRef] [PubMed]
- Wislez, M.; Rabbe, N.; Marchal, J.; Milleron, B.; Crestani, B.; Mayaud, C.; Antoine, M.; Soler, P.; Cadranel, J. Hepatocyte growth factor production by neutrophils infiltrating bronchioloalveolar subtype pulmonary adenocarcinoma: Role in tumor progression and death. Cancer Res. 2003, 63, 1405–1412. [Google Scholar] [PubMed]
- Li, Z.; Zhao, R.; Cui, Y.; Zhou, Y.; Wu, X. The dynamic change of neutrophil to lymphocyte ratio can predict clinical outcome in stage I–III colon cancer. Sci. Rep. 2018, 8, 9453. [Google Scholar] [CrossRef] [PubMed]
- Mizuno, R.; Kawada, K.; Itatani, Y.; Ogawa, R.; Kiyasu, Y.; Sakai, Y. The Role of Tumor-Associated Neutrophils in Colorectal Cancer. Int. J. Mol. Sci. 2019, 20, 529. [Google Scholar] [CrossRef] [PubMed]
- Qian, B.Z.; Li, J.; Zhang, H.; Kitamura, T.; Zhang, J.; Campion, L.R.; Kaiser, E.A.; Snyder, L.A.; Pollard, J.W. CCL2 recruits inflammatory monocytes to facilitate breast-tumour metastasis. Nature 2011, 475, 222–225. [Google Scholar] [CrossRef]
- Kumar, V.; Patel, S.; Tcyganov, E.; Gabrilovich, D.I. The nature of myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 2016, 37, 208–220. [Google Scholar] [CrossRef] [Green Version]
- Haverkamp, J.M.; Crist, S.A.; Elzey, B.D.; Cimen, C.; Ratliff, T.L. In vivo suppressive function of myeloid-derived suppressor cells is limited to the inflammatory site. Eur. J. Immunol. 2011, 41, 749–759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Corzo, C.A.; Condamine, T.; Lu, L.; Cotter, M.J.; Youn, J.-I.; Cheng, P.; Cho, H.-I.; Celis, E.; Quiceno, D.G.; Padhya, T.; et al. HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J. Exp. Med. 2010, 207, 2439–2453. [Google Scholar] [CrossRef] [Green Version]
- Jiang, G.M.; Tan, Y.; Wang, H.; Peng, L.; Chen, H.T.; Meng, X.J.; Li, L.L.; Liu, Y.; Li, W.F.; Shan, H. The relationship between autophagy and the immune system and its applications for tumor immunotherapy. Mol. Cancer 2019, 18, 17. [Google Scholar] [CrossRef]
- Li, W.; Tanikawa, T.; Kryczek, I.; Xia, H.; Li, G.; Wu, K.; Wei, S.; Zhao, L.; Vatan, L.; Wen, B.; et al. Aerobic Glycolysis Controls Myeloid-Derived Suppressor Cells and Tumor Immunity via a Specific CEBPB Isoform in Triple-Negative Breast Cancer. Cell Metab. 2018, 28, 87–103.e6. [Google Scholar] [CrossRef] [PubMed]
- De Veirman, K.; Menu, E.; Maes, K.; De Beule, N.; De Smedt, E.; Maes, A.; Vlummens, P.; Fostier, K.; Kassambara, A.; Moreaux, J.; et al. Myeloid-derived suppressor cells induce multiple myeloma cell survival by activating the AMPK pathway. Cancer Lett. 2019, 442, 233–241. [Google Scholar] [CrossRef] [PubMed]
- Xing, F. Cancer associated fibroblasts (CAFs) in tumor microenvironment. Front. Biosci. 2010, 15, 166. [Google Scholar] [CrossRef] [Green Version]
- Koliaraki, V.; Pallangyo, C.K.; Greten, F.R.; Kollias, G. Mesenchymal Cells in Colon Cancer. Gastroenterology 2017, 152, 964–979. [Google Scholar] [CrossRef]
- Grillo, A.R.; Scarpa, M.; D’Inca, R.; Brun, P.; Scarpa, M.; Porzionato, A.; De Caro, R.; Martines, D.; Buda, A.; Angriman, I.; et al. TAK1 is a key modulator of the profibrogenic phenotype of human ileal myofibroblasts in Crohn’s disease. Am. J. Physiol. Gastrointest. Liver Physiol. 2015, 309, 443–454. [Google Scholar] [CrossRef]
- Hawinkels, L.J.A.C.; Paauwe, M.; Verspaget, H.W.; Wiercinska, E.; Van Der Zon, J.M.; Van Der Ploeg, K.; Koelink, P.J.; Lindeman, J.H.N.; Mesker, W.; Ten Dijke, P.; et al. Interaction with colon cancer cells hyperactivates TGF-β signaling in cancer-associated fibroblasts. Oncogene 2014, 33, 97–107. [Google Scholar] [CrossRef]
- Calon, A.; Espinet, E.; Palomo-Ponce, S.; Tauriello, D.V.F.; Iglesias, M.; Céspedes, M.V.; Sevillano, M.; Nadal, C.; Jung, P.; Zhang, X.H.F.; et al. Dependency of Colorectal Cancer on a TGF-β-Driven Program in Stromal Cells for Metastasis Initiation. Cancer Cell 2012, 22, 571–584. [Google Scholar] [CrossRef]
- O’Connell, J.T.; Sugimoto, H.; Cooke, V.G.; MacDonald, B.A.; Mehta, A.I.; LeBleu, V.S.; Dewar, R.; Rocha, R.M.; Brentani, R.R.; Resnick, M.B.; et al. VEGF-A and Tenascin-C produced by S100A4+ stromal cells are important for metastatic colonization. Proc. Natl. Acad. Sci. USA 2011, 108, 16002–16007. [Google Scholar] [CrossRef] [Green Version]
- Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef] [Green Version]
- Charoentong, P.; Finotello, F.; Angelova, M.; Mayer, C.; Efremova, M.; Rieder, D.; Hackl, H.; Trajanoski, Z. Pan-cancer Immunogenomic Analyses Reveal Genotype-Immunophenotype Relationships and Predictors of Response to Checkpoint Blockade. Cell Rep. 2017, 18, 248–262. [Google Scholar] [CrossRef]
- Halama, N.; Braun, M.; Kahlert, C.; Spille, A.; Quack, C.; Rahbari, N.; Koch, M.; Weitz, J.; Kloor, M.; Zoernig, I.; et al. Natural Killer Cells are Scarce in Colorectal Carcinoma Tissue Despite High Levels of Chemokines and Cytokines. Clin. Cancer Res. 2011, 17, 678–689. [Google Scholar] [CrossRef] [Green Version]
- Vaupel, P.; Mayer, A. Hypoxia and anemia: Effects on tumor biology and treatment resistance. Transfus. Clin. Biol. 2005, 12, 5–10. [Google Scholar] [CrossRef]
- Levy, J.M.M.; Towers, C.G.; Thorburn, A. Targeting Autophagy in Cancer Therapy. Nat. Rev. Cancer 2016, 17, 528–542. [Google Scholar] [CrossRef]
- Durães, F.V.; Niven, J.; Dubrot, J.; Hugues, S.; Gannagé, M. Macroautophagy in Endogenous Processing of Self- and Pathogen-Derived Antigens for MHC Class II Presentation. Front. Immunol. 2015, 6, 79. [Google Scholar] [CrossRef] [Green Version]
- Bernstein, C.N.; Blanchard, J.F.; Kliewer, E.; Wajda, A. Cancer risk in patients with inflammatory bowel disease: A population-based study. Cancer 2001, 91, 854–862. [Google Scholar] [CrossRef]
- Fukata, M.; Chen, A.; Vamadevan, A.S.; Cohen, J.; Breglio, K.; Krishnareddy, S.; Hsu, D.; Xu, R.; Harpaz, N.; Dannenberg, A.J.; et al. Toll-like receptor-4 promotes the development of colitis-associated colorectal tumors. Gastroenterology 2007, 133, 1869–1881. [Google Scholar] [CrossRef]
- Chaturvedi, M.M.; Sung, B.; Yadav, V.R.; Kannappan, R.; Aggarwal, B.B. NF-κB addiction and its role in cancer: One size does not fit all. Oncogene 2011, 30, 1615–1630. [Google Scholar] [CrossRef]
- Wu, Y.; Yao, J.; Xie, J.; Liu, Z.; Zhou, Y.; Pan, H.; Han, W. The role of autophagy in colitis-associated colorectal cancer. Signal Transduct. Target. Ther. 2018, 3, 31. [Google Scholar] [CrossRef]
- Ning, C.; Li, Y.-Y.; Wang, Y.; Han, G.-C.; Wang, R.-X.; Xiao, H.; Li, X.-Y.; Hou, C.-M.; Ma, Y.-F.; Sheng, D.-S.; et al. Complement activation promotes colitis-associated carcinogenesis through activating intestinal IL-1β/IL-17A axis. Mucosal Immunol. 2015, 8, 1275–1284. [Google Scholar] [CrossRef]
- Olguín, J.E.; Medina-Andrade, I.; Molina, E.; Vázquez, A.; Pacheco-Fernández, T.; Saavedra, R.; Pérez-Plasencia, C.; Chirino, Y.I.; Vaca-Paniagua, F.; Arias-Romero, L.E.; et al. Early and partial reduction in CD4+Foxp3+ regulatory T cells during colitis-associated colon cancer induces CD4+ and CD8+ T cell activation inhibiting tumorigenesis. J. Cancer 2018, 9, 239–249. [Google Scholar] [CrossRef]
- Ngabire, D.; Kim, G.-D. Autophagy and Inflammatory Response in the Tumor Microenvironment. Int. J. Mol. Sci. 2017, 18, 2016. [Google Scholar] [CrossRef]
- Bellot, G.; Garcia-Medina, R.; Gounon, P.; Chiche, J.; Roux, D.; Pouysségur, J.; Mazure, N.M. Hypoxia-Induced Autophagy Is Mediated through Hypoxia-Inducible Factor Induction of BNIP3 and BNIP3L via Their BH3 Domains. Mol. Cell. Biol. 2009, 29, 2570–2581. [Google Scholar] [CrossRef]
- Lin, A.; Yao, J.; Zhuang, L.; Wang, D.; Han, J.; Lam, E.W.; Network, T.R.; Gan, B. The Foxo-BNIP3 axis exerts a unique regulation of mTORC1 and cell survival under energy stress. Oncogene 2014, 33, 3183–3194. [Google Scholar] [CrossRef]
- Li, Y.-Y.; Feun, L.G.; Thongkum, A.; Tu, C.-H.; Chen, S.-M.; Wangpaichitr, M.; Wu, C.; Kuo, M.T.; Savaraj, N. Autophagic Mechanism in Anti-Cancer Immunity: Its Pros and Cons for Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 1297. [Google Scholar] [CrossRef]
- Çam, H.; Easton, J.B.; High, A.; Houghton, P.J. mTORC1 signaling under hypoxic conditions is controlled by ATM-dependent phosphorylation of HIF-1α. Mol. Cell 2010, 40, 509–520. [Google Scholar] [CrossRef]
- Valečka, J.; Almeida, C.R.; Su, B.; Pierre, P.; Gatti, E. Autophagy and MHC-restricted antigen presentation. Mol. Immunol. 2018, 99, 163–170. [Google Scholar] [CrossRef]
- Keller, C.W.; Loi, M.; Ligeon, L.-A.; Gannage, M.; Lunemann, J.D.; Münz, C. Endocytosis regulation by autophagy proteins in MHC restricted antigen presentation. Curr. Opin. Immunol. 2018, 52, 68–73. [Google Scholar] [CrossRef]
- Folkerts, H.; Hilgendorf, S.; Vellenga, E.; Bremer, E.; Wiersma, V.R. The multifaceted role of autophagy in cancer and the microenvironment. Med. Res. Rev. 2019, 39, 517–560. [Google Scholar] [CrossRef] [PubMed]
- Parekh, V.V.; Wu, L.; Boyd, K.L.; Williams, J.A.; Gaddy, J.A.; Olivares-Villagómez, D.; Cover, T.L.; Zong, W.-X.; Zhang, J.; Van Kaer, L. Impaired autophagy, defective T cell homeostasis and a wasting syndrome in mice with a T cell-specific deletion of Vps34. J. Immunol. 2013, 190, 5086–5101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Loi, M.; Müller, A.; Steinbach, K.; Niven, J.; Barreira da Silva, R.; Paul, P.; Ligeon, L.A.; Caruso, A.; Albrecht, R.A.; Becker, A.C.; et al. Macroautophagy Proteins Control MHC Class I Levels on Dendritic Cells and Shape Anti-viral CD8+ T Cell Responses. Cell Rep. 2016, 15, 1076–1087. [Google Scholar] [CrossRef] [PubMed]
- Germic, N.; Frangez, Z.; Yousefi, S.; Simon, H.-U. Regulation of the innate immune system by autophagy: Monocytes, macrophages, dendritic cells and antigen presentation. Cell Death Differ. 2019, 26, 715–727. [Google Scholar] [CrossRef] [PubMed]
- Münz, C. Autophagy proteins in antigen processing for presentation on MHC molecules. Immunol. Rev. 2016, 272, 17–27. [Google Scholar] [CrossRef]
- Mintern, J.D.; Macri, C.; Chin, W.J.; Panozza, S.E.; Segura, E.; Patterson, N.L.; Zeller, P.; Bourges, D.; Bedoui, S.; McMillan, P.J.; et al. Differential use of autophagy by primary dendritic cells specialized in cross-presentation. Autophagy 2015, 11, 906–917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Loi, M.; Ligeon, L.-A.; Münz, C. MHC Class I Internalization via Autophagy Proteins. Methods Mol. Biol. 2019, 1880, 455–477. [Google Scholar]
- Thiele, F.; Tao, S.; Zhang, Y.; Muschaweckh, A.; Zollmann, T.; Protzer, U.; Abele, R.; Drexler, I. Modified vaccinia virus Ankara-infected dendritic cells present CD4+ T-cell epitopes by endogenous major histocompatibility complex class II presentation pathways. J. Virol. 2015, 89, 2698–2709. [Google Scholar] [CrossRef] [PubMed]
- Bronietzki, A.W.; Schuster, M.; Schmitz, I. Autophagy in T-cell development, activation and differentiation. Immunol. Cell Biol. 2015, 93, 25–34. [Google Scholar] [CrossRef]
- Nedjic, J.; Aichinger, M.; Mizushima, N.; Klein, L. Macroautophagy, endogenous MHC II loading and T cell selection: The benefits of breaking the rules. Curr. Opin. Immunol. 2009, 21, 92–97. [Google Scholar] [CrossRef]
- Khan, N.; Vidyarthi, A.; Pahari, S.; Negi, S.; Aqdas, M.; Nadeem, S.; Agnihotri, T.; Agrewala, J.N. Signaling through NOD-2 and TLR-4 Bolsters the T cell Priming Capability of Dendritic cells by Inducing Autophagy. Sci. Rep. 2016, 6, 19084. [Google Scholar] [CrossRef] [Green Version]
- Tey, S.-K.; Khanna, R. Autophagy mediates transporter associated with antigen processing-independent presentation of viral epitopes through MHC class I pathway. Blood 2012, 120, 994–1004. [Google Scholar] [CrossRef] [Green Version]
- Lee, H.K.; Mattei, L.M.; Steinberg, B.E.; Alberts, P.; Lee, Y.H.; Chervonsky, A.; Mizushima, N.; Grinstein, S.; Iwasaki, A. In Vivo Requirement for Atg5 in Antigen Presentation by Dendritic Cells. Immunity 2010, 32, 227–239. [Google Scholar] [CrossRef] [Green Version]
- Galdiero, M.R.; Bonavita, E.; Barajon, I.; Garlanda, C.; Mantovani, A.; Jaillon, S. Tumor associated macrophages and neutrophils in cancer. Immunobiology 2013, 218, 1402–1410. [Google Scholar] [CrossRef]
- Yang, M.; Liu, J.; Shao, J.; Qin, Y.; Ji, Q.; Zhang, X.; Du, J. Cathepsin S-mediated autophagic flux in tumor-associated macrophages accelerate tumor development by promoting M2 polarization. Mol. Cancer 2014, 13, 43. [Google Scholar] [CrossRef]
- Shao, L.-N.; Xing, C.-G.; Yang, X.-D.; Young, W.; Zhu, B.-S.; Cao, J.-P. Effects of autophagy regulation of tumor-associated macrophages on radiosensitivity of colorectal cancer cells. Mol. Med. Rep. 2016, 13, 2661–2670. [Google Scholar] [CrossRef] [Green Version]
- Viry, E.; Baginska, J.; Berchem, G.; Noman, M.Z.; Medves, S.; Chouaib, S.; Janji, B. Autophagic degradation of GZMB/granzyme B: A new mechanism of hypoxic tumor cell escape from natural killer cell-mediated lysis. Autophagy 2014, 10, 173–175. [Google Scholar] [CrossRef]
- Shibutani, S.T.; Saitoh, T.; Nowag, H.; Münz, C.; Yoshimori, T. Autophagy and autophagy-related proteins in the immune system. Nat. Immunol. 2015, 16, 1014–1024. [Google Scholar] [CrossRef] [Green Version]
- Oral, O.; Yedier, O.; Kilic, S.; Gozuacik, D. Involvement of autophagy in T cellbiology. Histol. Histopathol. 2017, 32, 11–20. [Google Scholar]
- Willinger, T.; Flavell, R.A. Canonical autophagy dependent on the class III phosphoinositide-3 kinase Vps34 is required for naive T-cell homeostasis. Proc. Natl. Acad. Sci. USA 2012, 109, 8670–8675. [Google Scholar] [CrossRef] [Green Version]
- Xu, X.; Araki, K.; Li, S.; Han, J.H.; Ye, L.; Tan, W.G.; Konieczny, B.T.; Bruinsma, M.W.; Martinez, J.; Pearce, E.L.; et al. Autophagy is essential for effector CD8(+) T cell survival and memory formation. Nat. Immunol. 2014, 15, 1152–1161. [Google Scholar] [CrossRef] [PubMed]
- Reed, M.; Morris, S.H.; Jang, S.; Mukherjee, S.; Yue, Z.; Lukacs, N.W. Autophagy-inducing protein beclin-1 in dendritic cells regulates CD4 T cell responses and disease severity during respiratory syncytial virus infection. J. Immunol. 2013, 191, 2526–2537. [Google Scholar] [CrossRef] [PubMed]
- Henson, S.M.; Lanna, A.; Riddel, N.E.; Franzese, O.; Macaulay, R.; Griffiths, S.J.; Puleston, D.J.; Watson, A.S.; Simon, A.K.; Tooze, S.A.; et al. P38 signaling inhibits mTORC1-independent autophagy in senescent human CD8+ T cells. J. Clin. Investig. 2014, 124, 4004–4016. [Google Scholar] [CrossRef]
- Venook, A. Critical Evaluation of Current Treatments in Metastatic Colorectal Cancer. Oncologist 2005, 10, 250–261. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch-repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [Green Version]
- Zhou, C.; Zhang, J. Immunotherapy-based combination strategies for treatment of gastrointestinal cancers: Current status and future prospects. Front. Med. 2019, 13, 12–23. [Google Scholar] [CrossRef]
- Arora, S.P.; Mahalingam, D. Immunotherapy in colorectal cancer: For the select few or all? J. Gastrointest. Oncol. 2018, 9, 170–179. [Google Scholar] [CrossRef]
- Lim, S.-O.; Li, C.-W.; Xia, W.; Cha, J.-H.; Chan, L.-C.; Wu, Y.; Chang, S.-S.; Lin, W.-C.; Hsu, J.-M.; Hsu, Y.-H.; et al. Deubiquitination and Stabilization of PD-L1 by CSN5. Cancer Cell 2016, 30, 925–939. [Google Scholar] [CrossRef] [Green Version]
- Juneja, V.R.; McGuire, K.A.; Manguso, R.T.; LaFleur, M.W.; Collins, N.; Haining, W.N.; Freeman, G.J.; Sharpe, A.H. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J. Exp. Med. 2017, 214, 895–904. [Google Scholar] [CrossRef] [Green Version]
- Yaghoubi, N.; Soltani, A.; Ghazvini, K.; Hassanian, S.M.; Hashemy, S.I. PD-1/ PD-L1 blockade as a novel treatment for colorectal cancer. Biomed. Pharmacother. 2019, 110, 312–318. [Google Scholar] [CrossRef]
- Bin Wang, H.; Yao, H.; Li, C.S.; Liang, L.X.; Zhang, Y.; Chen, Y.X.; Fang, J.-Y.; Xu, J.; Fang, J. Rise of PD-L1 expression during metastasis of colorectal cancer: Implications for immunotherapy. J. Dig. Dis. 2017, 18, 574–581. [Google Scholar] [CrossRef]
- O’Donnell, J.S.; Massi, D.; Teng, M.W.; Mandala, M. PI3K-AKT-mTOR inhibition in cancer immunotherapy, redux. Semin. Biol. 2018, 48, 91–103. [Google Scholar] [CrossRef]
- Battaglin, F.; Naseem, M.; Lenz, H.J.; Salem, M.E. Microsatellite instability in colorectal cancer: Overview of its clinical significance and novel perspectives. Clin. Adv. Hematol. Oncol. 2018, 16, 735–745. [Google Scholar]
- Overman, M.J.; McDermott, R.; Leach, J.L.; Lonardi, S.; Lenz, H.J.; Morse, M.A.; Desai, J.; Hill, A.; Axelson, M.; Moss, R.A.; et al. Nivolumab in patients with metastatic DNA mismatch repair-deficient or microsatellite instability-high colorectal cancer (CheckMate 142): An open-label, multicentre, phase 2 study. Lancet Oncol. 2017, 18, 1182–1191. [Google Scholar] [CrossRef]
- Kang, Y.K.; Boku, N.; Satoh, T.; Ryu, M.H.; Chao, Y.; Kato, K.; Chung, H.C.; Chen, J.S.; Muro, K.; Kang, W.K.; et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017, 390, 2461–2471. [Google Scholar] [CrossRef]
- Fuchs, C.S.; Doi, T.; Jang, R.W.; Muro, K.; Satoh, T.; Machado, M.; Sun, W.; I Jalal, S.; A Shah, M.; Metges, J.-P.; et al. Safety and Efficacy of Pembrolizumab Monotherapy in Patients with Previously Treated Advanced Gastric and Gastroesophageal Junction Cancer: Phase 2 Clinical KEYNOTE-059 Trial. JAMA Oncol. 2018, 4, e180013. [Google Scholar] [CrossRef]
- Chung, K.Y.; Fong, L.; Venook, A.; Beck, S.B.; Dorazio, P.; Criscitiello, P.J.; Healey, D.I.; Huang, B.; Gómez-Navarro, J.; Saltz, L.B.; et al. Phase II Study of the Anti-Cytotoxic T-Lymphocyte–Associated Antigen 4 Monoclonal Antibody, Tremelimumab, in Patients with Refractory Metastatic Colorectal Cancer. J. Clin. Oncol. 2010, 28, 3485–3490. [Google Scholar] [CrossRef]
- Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.; Kemberling, H.; Eyring, A.; Skora, A.; Azad, N.S.; Laheru, D.A.; Donehower, R.C.; et al. PD-1 blockade in tumors with mismatch repair deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef]
- Overman, M.J.; Lonardi, S.; Wong, K.Y.M.; Lenz, H.-J.; Gelsomino, F.; Aglietta, M.; Morse, M.A.; Van Cutsem, E.; McDermott, R.; Hill, A.; et al. Durable Clinical Benefit with Nivolumab Plus Ipilimumab in DNA Mismatch Repair-Deficient/Microsatellite Instability-High Metastatic Colorectal Cancer. J. Clin. Oncol. 2018, 36, 773–779. [Google Scholar] [CrossRef]
- Rico, G.T.; Price, T.J. Atezolizumab for the treatment of colorectal cancer: The latest evidence and clinical potential. Opin. Biol. Ther. 2018, 18, 449–457. [Google Scholar] [CrossRef]
- Calles, A.; Aguado, G.; Sandoval, C.; Álvarez, R. The role of immunotherapy in small cell lung cancer. Clin. Transl. Oncol. 2019, 1–16. [Google Scholar] [CrossRef]
- Herbst, R.S.; Soria, J.-C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515, 563–567. [Google Scholar] [CrossRef] [Green Version]
- Link, J.T.; Overman, M.J. Immunotherapy Progress in Mismatch Repair–Deficient Colorectal Cancer and Future Therapeutic Challenges. Cancer J. 2016, 22, 190–195. [Google Scholar] [CrossRef]
- Emambux, S.; Tachon, G.; Junca, A.; Tougeron, D. Results and challenges of immune checkpoint inhibitors in colorectal cancer. Expert Opin. Biol. Ther. 2018, 18, 561–573. [Google Scholar] [CrossRef]
- Zhong, X.; Tumang, J.R.; Gao, W.; Bai, C.; Rothstein, T.L. PD-L2 expression extends beyond dendritic cells/macrophages to B1 cells enriched for VH11/VH12 and phosphatidylcholine binding. Eur. J. Immunol. 2007, 37, 2405–2410. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Yao, H.; Li, C.; Liang, L.; Zhang, Y.; Shi, H.; Zhou, C.; Chen, Y.; Fang, J.-Y.; Xu, J. PD-L2 expression in colorectal cancer: Independent prognostic effect and targetability by deglycosylation. Oncoimmunology 2017, 6, e1327494. [Google Scholar] [CrossRef] [Green Version]
- Guo, P.-D.; Sun, Z.-W.; Lai, H.-J.; Yang, J.; Wu, P.-P.; Guo, Y.-D.; Sun, J. Clinicopathological analysis of PD-L2 expression in colorectal cancer. OncoTargets Ther. 2018, 11, 7635–7642. [Google Scholar] [CrossRef]
- Taube, J.M.; Klein, A.; Brahmer, J.R.; Xu, H.; Pan, X.; Kim, J.H.; Chen, L.; Pardoll, D.M.; Topalian, S.L.; Anders, R.A. Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin. Cancer Res. 2014, 20, 5064–5074. [Google Scholar] [CrossRef] [Green Version]
- Seto, T.; Sam, D.; Pan, M. Mechanisms of Primary and Secondary Resistanceto Immune Checkpoint Inhibitors in Cancer. Med. Sci. 2019, 7, 14. [Google Scholar]
- Curran, M.A.; Montalvo, W.; Yagita, H.; Allison, J.P. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl. Acad. Sci. USA 2010, 107, 4275–4280. [Google Scholar] [CrossRef] [Green Version]
- Janji, B.; Berchem, G.; Chouaib, S. Targeting Autophagy in the Tumor Microenvironment: New Challenges and Opportunities for Regulating Tumor Immunity. Front. Immunol. 2018, 9, 887. [Google Scholar] [CrossRef] [PubMed]
- Qian, H.-R.; Shi, Z.-Q.; Zhu, H.-P.; Gu, L.-H.; Wang, X.-F.; Yang, Y. Interplay between apoptosis and autophagy in colorectal cancer. Oncotarget 2017, 8, 62759–62768. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rosenfeld, M.R.; Ye, X.; Supko, J.G.; Desideri, S.; A Grossman, S.; Brem, S.; Mikkelson, T.; Wang, D.; Chang, Y.C.; Hu, J.; et al. A phase I/II trial of hydroxychloroquine in conjunction with radiation therapy and concurrent and adjuvant temozolomide in patients with newly diagnosed glioblastoma multiforme. Autophagy 2014, 10, 1359–1368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goulielmaki, M.; Koustas, E.; Moysidou, E.; Vlassi, M.; Sasazuki, T.; Shirasawa, S.; Zografos, G.; Oikonomou, E. BRAF associated autophagy exploitation: BRAF and autophagy inhibitors synergise to efficiently overcome resistance of BRAF mutant colorectal cancer cells. Oncotarget 2015, 7, 9188–9221. [Google Scholar] [CrossRef] [PubMed]
- Boone, B.A.; Bahary, N.; Zureikat, A.H.; Moser, A.J.; Normolle, D.P.; Wu, W.C.; Singhi, A.D.; Bao, P.; Bartlett, D.L.; Liotta, L.A.; et al. Safety and Biologic Response of Pre-operative Autophagy Inhibition in Combination with Gemcitabine in Patients with Pancreatic Adenocarcinoma. Ann. Surg. Oncol. 2015, 22, 4402–4410. [Google Scholar] [CrossRef] [PubMed]
- Pellegrini, P.; Strambi, A.; Zipoli, C.; Hägg-Olofsson, M.; Buoncervello, M.; Linder, S.; De Milito, A. Acidic extracellular pH neutralizes the autophagy-inhibiting activity of chloroquine: Implications for cancer therapies. Autophagy 2014, 10, 562–571. [Google Scholar] [CrossRef] [PubMed]
- Amaravadi, R.K.; Winkler, J.D. Lys05: A new lysosomal autophagy inhibitor. Autophagy 2012, 8, 1383–1384. [Google Scholar] [CrossRef]
- Ronan, B.; Flamand, O.; Vescovi, L.; Dureuil, C.; Durand, L.; Fassy, F.; Bachelot, M.-F.; Lamberton, A.; Mathieu, M.; Bertrand, T.; et al. A highly potent and selective Vps34 inhibitor alters vesicle trafficking and autophagy. Nat. Chem. Biol. 2014, 10, 1013–1019. [Google Scholar] [CrossRef] [PubMed]
- Egan, D.F.; Chun, M.G.; Vamos, M.; Zou, H.; Rong, J.; Miller, C.J.; Lou, H.J.; Raveendra-Panickar, D.; Yang, C.-C.; Sheffler, D.J.; et al. Small Molecule Inhibition of the Autophagy Kinase ULK1 and Identification of ULK1 Substrates. Mol. Cell 2015, 59, 285–297. [Google Scholar] [CrossRef]
- Vakifahmetoglu-Norberg, H.; Xia, H.-G.; Yuan, J. Pharmacologic agents targeting autophagy. J. Clin. Investig. 2015, 125, 5–13. [Google Scholar] [CrossRef] [Green Version]
- Rossi, M.; Munarriz, E.R.; Bartesaghi, S.; Milanese, M.; Dinsdale, D.; Guerra-Martin, M.A.; Bampton, E.T.W.; Glynn, P.; Bonanno, G.; Knight, R.A.; et al. Desmethylclomipramine induces the accumulation of autophagy markers by blocking autophagic flux. J. Cell Sci. 2009, 122, 3330–3339. [Google Scholar] [CrossRef] [Green Version]
- Liu, J.; Xia, H.; Kim, M.; Xu, L.; Li, Y.; Zhang, L.; Cai, Y.; Norberg, H.V.; Zhang, T.; Furuya, T.; et al. Beclin1 Controls the Levels of p53 by Regulating the Deubiquitination Activity of USP10 and USP13. Cell 2011, 147, 223–234. [Google Scholar] [CrossRef] [Green Version]
- Starobinets, H.; Ye, J.; Broz, M.; Barry, K.; Goldsmith, J.; Marsh, T.; Rostker, F.; Krummel, M.; Debnath, J. Antitumor adaptive immunity remains intact following inhibition of autophagy and antimalarial treatment. J. Clin. Investig. 2016, 126, 4417–4429. [Google Scholar] [CrossRef] [Green Version]
- Yang, Z.J.; Chee, C.E.; Huang, S.; Sinicrope, F.A. The Role of Autophagy in Cancer: Therapeutic Implications. Mol. Cancer Ther. 2011, 10, 1533–1541. [Google Scholar] [CrossRef] [Green Version]
- Byun, S.; Lee, E.; Lee, K.W. Therapeutic Implications of Autophagy Inducers in Immunological Disorders, Infection, and Cancer. Int. J. Mol. Sci. 2017, 18, 1959. [Google Scholar] [CrossRef]
- Wang, H.; Li, D.; Li, X.; Ou, X.; Liu, S.; Zhang, Y.; Ding, J.; Xie, B. Mammalian target of rapamycin inhibitor RAD001 sensitizes endometrial cancer cells to paclitaxel-induced apoptosis via the induction of autophagy. Oncol. Lett. 2016, 12, 5029–5035. [Google Scholar] [CrossRef] [Green Version]
- Yeo, S.K.; Paul, R.; Haas, M.; Wang, C.; Guan, J.-L. Improved efficacy of mitochondrial disrupting agents upon inhibition of autophagy in a mouse model of BRCA1-deficient breast cancer. Autophagy 2018, 14, 1214–1225. [Google Scholar] [CrossRef]
- Opydo-Chanek, M.; Gonzalo, O.; Marzo, I. Multifaceted anticancer activity of BH3 mimetics: Current evidence and future prospects. Biochem. Pharmacol. 2017, 136, 12–23. [Google Scholar] [CrossRef]
- Law, B.Y.K.; Chan, W.K.; Xu, S.W.; Wang, J.R.; Bai, L.P.; Liu, L.; Wong, V.K.W. Natural small-molecule enhancers of autophagy induce autophagic cell death in apoptosis-defective cells. Sci. Rep. 2014, 4, 5510. [Google Scholar] [CrossRef]
Number of Study | Immune Target | Agent/Compound | Phase of Study |
---|---|---|---|
NCT01876511 | PD-1 | Pembrolizumab | II |
NCT02981524 | PD-1 | Cyclophosphamide followed by Pembrolizumab | II |
NCT03657641 | PD-1 | Pembrolizumab + Vicriviroc | I/II |
NCT03631407 | PD-1 | Pembrolizumab + Regorafenib | II |
NCT03475004 | PD-1 | Pembrolizumab, Bevacizumab, and Binimetinib | II |
NCT03658772 | PD-1 | Pembrolizumab + grapiprant | I |
NCT03519412 | PD-1 | Pembrolizumab + temozolomide | II |
NCT02713373 | PD-1 | Pembrolizumab + cetuximab | I/II |
NCT02375672 | PD-1 | Pembrolizumab + FOLFOX | II |
NCT03332498 | PD-1 | Pembrolizumab + Ibrutinib | I/II |
NCT02851004 | PD-1 | Pembrolizumab + SBRT | I/II |
NCT02837263 | PD-1 | Pembrolizumab + BBI609 | I |
NCT02992912 | PD-1 | Atezolizumab + stereotactic ablative radiotherapy | II |
NCT03712943 | PD-1 | Nivolumab + Regorafenib | I |
NCT03711058 | PD-1 | Nivolumab + Copanlisib | I/II |
NCT03414983 | PD-1 | Nivolumab, Oxaliplatin, Leucovorin, Fluorouracil, Bevacizumab | II/III |
NCT02860546 | PD-1 | Nivolumab + TAS-102 | II |
NCT03026140 | PD-1 and CTLA-4 | Nivolumab + Ipilimumab +/− celecoxib | I/III |
NCT03693846 | PD-1 and CTLA-4 | Nivolumab + Ipilimumab | II |
NCT03104439 | PD-1 and CTLA-4 | Nivolumab + Ipilimumab + radiotherapy | II |
NCT03377361 | PD-1 and CTLA-4 | Nivolumab + Ipilimumab + Trametinib | I/II |
NCT03832621 | PD-1 and CTLA-4 | Nivolumab, Ipilimumab, Temozolomide | II |
NCT02327078 | PD-1 and IDO | Nivolumab + Epacadostat | VII |
NCT02983578 | PD-L1 | AZD9150 + MEDI4736 | II |
NCT02982694 | PD-L1 | Atezolizumab + Bevacizumab | II |
NCT02777710 | PD-L1 | Durvalumab + Pexidartinib | I |
NCT03827044 | PD-L1 | Avelumab | III |
NCT02669914 | PD-L1 | Durvalumab | II |
NCT02754856 | PD-L1 and CTLA-4 | MEDI4736 + Tremelimumab | I |
NCT03202758 | PD-L1 and CTLA-4 | Durvalumab, Tremelimumab, and FOLFOX | I/II |
Compound | Autophagy Inhibitors |
---|---|
Mechanism of Action | |
Bafilomycin A1 | Inhibitor of v-ATPase, inhibition of lysosomal acidification |
Concanamycin A | Inhibitor of v-ATPase, inhibition of lysosomal acidification |
Azithromycin | Inhibitor of v-ATPase, inhibition of lysosomal acidification |
3-Methyladenine (3-MA) | Inhibitor of class III PI3K |
Chloroquine (CQ) | Neutralizes the acidic pH of intracellular vesicles |
Hydroxy-chloroquine (HCQ) | CQ derivative-Neutralizes the acidic pH of intracellular vesicles |
Lys05 | CQ derivative-alter the acidification of the lysosomes |
SAR405 | Kinase inhibitor of Vps18 and Vps34 |
SBI-0206965 | Inhibitor of ULK1 |
Verteporfin | Inhibit acidification of lysosomes |
Clomipramine | Inhibit acidification of lysosomes |
desmethylclomipramine (DCMI) | Inhibit Autophagosome-Lysosome fusion |
Paclitaxel | Microtubule stabilizer- inhibits phosphorylation of VPS34 at T159 |
SAHA | Interact in autophagosome-lysosome fusion |
Monensin | Inhibit autophagosome-lysosome fusion |
Sputin-1 | Inhibits the activity of ubiquitin-specific peptidases, USP10 and USP13 |
SP600125 | Inhibition of JNK—reduction of Beclin-1 |
U0126 | Inhibitor of MEK1 and MEK2 |
Wortmannin | PI3K inhibitor |
LY294002 | PI3K inhibitor |
SB202190 | Cross-inhibition of the PI3K/mTOR and MAPKs pathway |
SB203580 | Inhibit autophagy by interfering with the trafficking of Atg9 |
MHY1485 | mTOR activator |
Compound/Molecule | Autophagy Inducers |
---|---|
Mechanism of Action | |
Rapamycin | mTORC1 inhibitor |
Temsirolimus | mTORC1 inhibitor |
Deforolimus | mTORC1 inhibitor |
Everolimus | mTORC1 inhibitor |
Metformin | AMPK activator |
Obatoclax | Inhibitor of Bcl-2 family proteins |
isoliensinine | Natural alkaloid |
cepharanthine | Natural alkaloid |
liensinine | Natural alkaloid |
Perifosine | AKT inhibitor |
Tat–Beclin-1 peptide | Releases beclin-1 into cytoplasm-regulate autophagosome formation |
Lithium | Increase the levels of Beclin-1/VPS34 complexes |
GDC-0980 | Dual inhibitor of PI3K and mTORC1 |
GDC-0941 | Inhibitor of class I PI3K |
fluspirilene | Antagonists of L-type Ca2+ channels |
verapamil | Antagonists of L-type Ca2+ channels |
loperamide | Antagonists of L-type Ca2+ channels |
nimodipine | Antagonists of L-type Ca2+ channels |
amiodarone | Antagonists of L-type Ca2+ channels |
© 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
Koustas, E.; Sarantis, P.; Kyriakopoulou, G.; Papavassiliou, A.G.; Karamouzis, M.V. The Interplay of Autophagy and Tumor Microenvironment in Colorectal Cancer—Ways of Enhancing Immunotherapy Action. Cancers 2019, 11, 533. https://doi.org/10.3390/cancers11040533
Koustas E, Sarantis P, Kyriakopoulou G, Papavassiliou AG, Karamouzis MV. The Interplay of Autophagy and Tumor Microenvironment in Colorectal Cancer—Ways of Enhancing Immunotherapy Action. Cancers. 2019; 11(4):533. https://doi.org/10.3390/cancers11040533
Chicago/Turabian StyleKoustas, Evangelos, Panagiotis Sarantis, Georgia Kyriakopoulou, Athanasios G. Papavassiliou, and Michalis V. Karamouzis. 2019. "The Interplay of Autophagy and Tumor Microenvironment in Colorectal Cancer—Ways of Enhancing Immunotherapy Action" Cancers 11, no. 4: 533. https://doi.org/10.3390/cancers11040533
APA StyleKoustas, E., Sarantis, P., Kyriakopoulou, G., Papavassiliou, A. G., & Karamouzis, M. V. (2019). The Interplay of Autophagy and Tumor Microenvironment in Colorectal Cancer—Ways of Enhancing Immunotherapy Action. Cancers, 11(4), 533. https://doi.org/10.3390/cancers11040533