The Impact of Gut Microbiota-Derived Metabolites on the Tumor Immune Microenvironment
Abstract
Simple Summary
Abstract
1. Introduction
2. The Intestinal Microbiota and Its Relation to Cancer Development and Cancer Immunotherapy
3. The Oncobiome and Cancer
4. Gut-Microbiota-Derived Molecules and Cancer
5. Challenges Associated with Therapeutic Potential of Microbial Metabolites
6. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Belkaid, Y.; Harrison, O.J. Homeostatic Immunity and the Microbiota. Immunity 2017, 46, 562–576. [Google Scholar] [CrossRef] [PubMed]
- Groussin, M.; Mazel, F.; Alm, E.J. Co-evolution and Co-speciation of Host-Gut Bacteria Systems. Cell Host Microbe 2020, 28, 12–22. [Google Scholar] [CrossRef] [PubMed]
- Al Nabhani, Z.; Dulauroy, S.; Marques, R.; Cousu, C.; Al Bounny, S.; Dejardin, F.; Sparwasser, T.; Berard, M.; Cerf-Bensussan, N.; Eberl, G. A Weaning Reaction to Microbiota Is Required for Resistance to Immunopathologies in the Adult. Immunity 2019, 50, 1276–1288.e5. [Google Scholar] [CrossRef] [PubMed]
- Elkrief, A.; Derosa, L.; Zitvogel, L.; Kroemer, G.; Routy, B. The intimate relationship between gut microbiota and cancer immunotherapy. Gut Microbes 2019, 10, 424–428. [Google Scholar] [CrossRef]
- Finlay, B.B.; Goldszmid, R.; Honda, K.; Trinchieri, G.; Wargo, J.; Zitvogel, L. Can we harness the microbiota to enhance the efficacy of cancer immunotherapy? Nat. Rev. Immunol. 2020, 20, 522–528. [Google Scholar] [CrossRef]
- Nejman, D.; Livyatan, I.; Fuks, G.; Gavert, N.; Zwang, Y.; Geller, L.T.; Rotter-Maskowitz, A.; Weiser, R.; Mallel, G.; Gigi, E.; et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 2020, 368, 973–980. [Google Scholar] [CrossRef]
- Dohlman, A.B.; Klug, J.; Mesko, M.; Gao, I.H.; Lipkin, S.M.; Shen, X.; Iliev, I.D. A pan-cancer mycobiome analysis reveals fungal involvement in gastrointestinal and lung tumors. Cell 2022, 185, 3807–3822.e12. [Google Scholar] [CrossRef]
- Fessler, J.; Matson, V.; Gajewski, T.F. Exploring the emerging role of the microbiome in cancer immunotherapy. J. Immunother. Cancer 2019, 7, 108. [Google Scholar] [CrossRef]
- Vitiello, G.A.; Cohen, D.J.; Miller, G. Harnessing the Microbiome for Pancreatic Cancer Immunotherapy. Trends Cancer 2019, 5, 670–676. [Google Scholar] [CrossRef]
- Smith, M.; Dai, A.; Ghilardi, G.; Amelsberg, K.V.; Devlin, S.M.; Pajarillo, R.; Slingerland, J.B.; Beghi, S.; Herrera, P.S.; Giardina, P.; et al. Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat. Med. 2022, 28, 713–723. [Google Scholar] [CrossRef]
- Gopalakrishnan, V.; Helmink, B.A.; Spencer, C.N.; Reuben, A.; Wargo, J.A. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell 2018, 33, 570–580. [Google Scholar] [CrossRef]
- Bejarano, L.; Jordao, M.J.C.; Joyce, J.A. Therapeutic Targeting of the Tumor Microenvironment. Cancer Discov. 2021, 11, 933–959. [Google Scholar] [CrossRef]
- Wherry, E.J.; Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef]
- Chow, A.; Perica, K.; Klebanoff, C.A.; Wolchok, J.D. Clinical implications of T cell exhaustion for cancer immunotherapy. Nat. Rev. Clin. Oncol. 2022, 19, 775–790. [Google Scholar] [CrossRef]
- Chamoto, K.; Al-Habsi, M.; Honjo, T. Role of PD-1 in Immunity and Diseases. Curr. Top. Microbiol. Immunol. 2017, 410, 75–97. [Google Scholar] [CrossRef]
- Sharma, P.; Allison, J.P. Immune checkpoint therapy: Forging ahead. Sci. Transl. Med. 2022, 14, eadf2947. [Google Scholar] [CrossRef]
- Robert, C. A decade of immune-checkpoint inhibitors in cancer therapy. Nat. Commun. 2020, 11, 3801. [Google Scholar] [CrossRef]
- Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef]
- Routy, B.; Gopalakrishnan, V.; Daillere, R.; Zitvogel, L.; Wargo, J.A.; Kroemer, G. The gut microbiota influences anticancer immunosurveillance and general health. Nat. Rev. Clin. Oncol. 2018, 15, 382–396. [Google Scholar] [CrossRef]
- Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillere, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef]
- Derosa, L.; Routy, B.; Thomas, A.M.; Iebba, V.; Zalcman, G.; Friard, S.; Mazieres, J.; Audigier-Valette, C.; Moro-Sibilot, D.; Goldwasser, F.; et al. Intestinal Akkermansia muciniphila predicts clinical response to PD-1 blockade in patients with advanced non-small-cell lung cancer. Nat. Med. 2022, 28, 315–324. [Google Scholar] [CrossRef] [PubMed]
- Elkrief, A.; Derosa, L.; Kroemer, G.; Zitvogel, L.; Routy, B. The negative impact of antibiotics on outcomes in cancer patients treated with immunotherapy: A new independent prognostic factor? Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. ESMO 2019, 30, 1572–1579. [Google Scholar] [CrossRef] [PubMed]
- Elkrief, A.; El Raichani, L.; Richard, C.; Messaoudene, M.; Belkaid, W.; Malo, J.; Belanger, K.; Miller, W.; Jamal, R.; Letarte, N.; et al. Antibiotics are associated with decreased progression-free survival of advanced melanoma patients treated with immune checkpoint inhibitors. Oncoimmunology 2019, 8, e1568812. [Google Scholar] [CrossRef] [PubMed]
- Rosshart, S.P.; Vassallo, B.G.; Angeletti, D.; Hutchinson, D.S.; Morgan, A.P.; Takeda, K.; Hickman, H.D.; McCulloch, J.A.; Badger, J.H.; Ajami, N.J.; et al. Wild Mouse Gut Microbiota Promotes Host Fitness and Improves Disease Resistance. Cell 2017, 171, 1015–1028.e13. [Google Scholar] [CrossRef] [PubMed]
- Rosshart, S.P.; Herz, J.; Vassallo, B.G.; Hunter, A.; Wall, M.K.; Badger, J.H.; McCulloch, J.A.; Anastasakis, D.G.; Sarshad, A.A.; Leonardi, I.; et al. Laboratory mice born to wild mice have natural microbiota and model human immune responses. Science 2019, 365, aaw4361. [Google Scholar] [CrossRef]
- Zambirinis, C.P.; Levie, E.; Nguy, S.; Avanzi, A.; Barilla, R.; Xu, Y.; Seifert, L.; Daley, D.; Greco, S.H.; Deutsch, M.; et al. TLR9 ligation in pancreatic stellate cells promotes tumorigenesis. J. Exp. Med. 2015, 212, 2077–2094. [Google Scholar] [CrossRef]
- Pushalkar, S.; Hundeyin, M.; Daley, D.; Zambirinis, C.P.; Kurz, E.; Mishra, A.; Mohan, N.; Aykut, B.; Usyk, M.; Torres, L.E.; et al. The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression. Cancer Discov. 2018, 8, 403–416. [Google Scholar] [CrossRef]
- Inamura, K. Roles of microbiota in response to cancer immunotherapy. Semin. Cancer Biol. 2020, 65, 164–175. [Google Scholar] [CrossRef]
- Zitvogel, L.; Ma, Y.; Raoult, D.; Kroemer, G.; Gajewski, T.F. The microbiome in cancer immunotherapy: Diagnostic tools and therapeutic strategies. Science 2018, 359, 1366–1370. [Google Scholar] [CrossRef]
- Vetizou, M.; Pitt, J.M.; Daillere, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef]
- Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108. [Google Scholar] [CrossRef] [PubMed]
- Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef] [PubMed]
- Inamura, K. Gut microbiota contributes towards immunomodulation against cancer: New frontiers in precision cancer therapeutics. Semin. Cancer Biol. 2021, 70, 11–23. [Google Scholar] [CrossRef] [PubMed]
- Park, E.M.; Chelvanambi, M.; Bhutiani, N.; Kroemer, G.; Zitvogel, L.; Wargo, J.A. Targeting the gut and tumor microbiota in cancer. Nat. Med. 2022, 28, 690–703. [Google Scholar] [CrossRef]
- Tanoue, T.; Morita, S.; Plichta, D.R.; Skelly, A.N.; Suda, W.; Sugiura, Y.; Narushima, S.; Vlamakis, H.; Motoo, I.; Sugita, K.; et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature 2019, 565, 600–605. [Google Scholar] [CrossRef]
- Skelly, A.N.; Sato, Y.; Kearney, S.; Honda, K. Mining the microbiota for microbial and metabolite-based immunotherapies. Nat. Rev. Immunol. 2019, 19, 305–323. [Google Scholar] [CrossRef]
- Pulendran, B.; Arunachalam, P.S.; O’Hagan, D.T. Emerging concepts in the science of vaccine adjuvants. Nat. Rev. Drug Discov. 2021, 20, 454–475. [Google Scholar] [CrossRef]
- Villemin, C.; Six, A.; Neville, B.A.; Lawley, T.D.; Robinson, M.J.; Bakdash, G. The heightened importance of the microbiome in cancer immunotherapy. Trends Immunol. 2023, 44, 44–59. [Google Scholar] [CrossRef]
- Boesch, M.; Baty, F.; Rothschild, S.I.; Tamm, M.; Joerger, M.; Fruh, M.; Brutsche, M.H. Tumour neoantigen mimicry by microbial species in cancer immunotherapy. Br. J. Cancer 2021, 125, 313–323. [Google Scholar] [CrossRef]
- McCarville, J.L.; Chen, G.Y.; Cuevas, V.D.; Troha, K.; Ayres, J.S. Microbiota Metabolites in Health and Disease. Annu. Rev. Immunol. 2020, 38, 147–170. [Google Scholar] [CrossRef]
- Michaudel, C.; Sokol, H. The Gut Microbiota at the Service of Immunometabolism. Cell Metab. 2020, 32, 514–523. [Google Scholar] [CrossRef]
- Luu, M.; Visekruna, A. Microbial metabolites: Novel therapeutic tools for boosting cancer therapies. Trends Cell Biol. 2021, 31, 873–875. [Google Scholar] [CrossRef]
- Visekruna, A.; Luu, M. The Role of Short-Chain Fatty Acids and Bile Acids in Intestinal and Liver Function, Inflammation, and Carcinogenesis. Front. Cell Dev. Biol. 2021, 9, 703218. [Google Scholar] [CrossRef]
- Kroemer, G.; Zitvogel, L. Inosine: Novel microbiota-derived immunostimulatory metabolite. Cell Res. 2020, 30, 942–943. [Google Scholar] [CrossRef]
- Luu, M.; Riester, Z.; Baldrich, A.; Reichardt, N.; Yuille, S.; Busetti, A.; Klein, M.; Wempe, A.; Leister, H.; Raifer, H.; et al. Microbial short-chain fatty acids modulate CD8(+) T cell responses and improve adoptive immunotherapy for cancer. Nat. Commun. 2021, 12, 4077. [Google Scholar] [CrossRef]
- Fan, Y.; Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 2021, 19, 55–71. [Google Scholar] [CrossRef]
- Sims, T.T.; El Alam, M.B.; Karpinets, T.V.; Dorta-Estremera, S.; Hegde, V.L.; Nookala, S.; Yoshida-Court, K.; Wu, X.; Biegert, G.W.G.; Delgado Medrano, A.Y.; et al. Gut microbiome diversity is an independent predictor of survival in cervical cancer patients receiving chemoradiation. Commun. Biol. 2021, 4, 237. [Google Scholar] [CrossRef]
- Sepich-Poore, G.D.; Zitvogel, L.; Straussman, R.; Hasty, J.; Wargo, J.A.; Knight, R. The microbiome and human cancer. Science 2021, 371, abc4552. [Google Scholar] [CrossRef]
- Galeano Nino, J.L.; Wu, H.; LaCourse, K.D.; Kempchinsky, A.G.; Baryiames, A.; Barber, B.; Futran, N.; Houlton, J.; Sather, C.; Sicinska, E.; et al. Effect of the intratumoral microbiota on spatial and cellular heterogeneity in cancer. Nature 2022, 611, 810–817. [Google Scholar] [CrossRef]
- Thomas, R.M.; Jobin, C. The Microbiome and Cancer: Is the ’Oncobiome’ Mirage Real? Trends Cancer 2015, 1, 24–35. [Google Scholar] [CrossRef]
- Kostic, A.D.; Chun, E.; Robertson, L.; Glickman, J.N.; Gallini, C.A.; Michaud, M.; Clancy, T.E.; Chung, D.C.; Lochhead, P.; Hold, G.L.; et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 2013, 14, 207–215. [Google Scholar] [CrossRef] [PubMed]
- Rubinstein, M.R.; Wang, X.; Liu, W.; Hao, Y.; Cai, G.; Han, Y.W. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/beta-catenin signaling via its FadA adhesin. Cell Host Microbe 2013, 14, 195–206. [Google Scholar] [CrossRef] [PubMed]
- Yu, T.; Guo, F.; Yu, Y.; Sun, T.; Ma, D.; Han, J.; Qian, Y.; Kryczek, I.; Sun, D.; Nagarsheth, N.; et al. Fusobacterium nucleatum Promotes Chemoresistance to Colorectal Cancer by Modulating Autophagy. Cell 2017, 170, 548–563.e16. [Google Scholar] [CrossRef] [PubMed]
- Rubinstein, M.R.; Baik, J.E.; Lagana, S.M.; Han, R.P.; Raab, W.J.; Sahoo, D.; Dalerba, P.; Wang, T.C.; Han, Y.W. Fusobacterium nucleatum promotes colorectal cancer by inducing Wnt/beta-catenin modulator Annexin A1. EMBO Rep. 2019, 20, e47638. [Google Scholar] [CrossRef] [PubMed]
- Parhi, L.; Alon-Maimon, T.; Sol, A.; Nejman, D.; Shhadeh, A.; Fainsod-Levi, T.; Yajuk, O.; Isaacson, B.; Abed, J.; Maalouf, N.; et al. Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression. Nat. Commun. 2020, 11, 3259. [Google Scholar] [CrossRef] [PubMed]
- Udayasuryan, B.; Ahmad, R.N.; Nguyen, T.T.D.; Umana, A.; Monet Roberts, L.; Sobol, P.; Jones, S.D.; Munson, J.M.; Slade, D.J.; Verbridge, S.S. Fusobacterium nucleatum induces proliferation and migration in pancreatic cancer cells through host autocrine and paracrine signaling. Sci. Signal. 2022, 15, eabn4948. [Google Scholar] [CrossRef]
- Thomas, R.M.; Gharaibeh, R.Z.; Gauthier, J.; Beveridge, M.; Pope, J.L.; Guijarro, M.V.; Yu, Q.; He, Z.; Ohland, C.; Newsome, R.; et al. Intestinal microbiota enhances pancreatic carcinogenesis in preclinical models. Carcinogenesis 2018, 39, 1068–1078. [Google Scholar] [CrossRef]
- Yu, D.; Wang, T.; Liang, D.; Mei, Y.; Zou, W.; Guo, S. The Landscape of Microbial Composition and Associated Factors in Pancreatic Ductal Adenocarcinoma Using RNA-Seq Data. Front. Oncol. 2021, 11, 651350. [Google Scholar] [CrossRef]
- Jin, C.; Lagoudas, G.K.; Zhao, C.; Bullman, S.; Bhutkar, A.; Hu, B.; Ameh, S.; Sandel, D.; Liang, X.S.; Mazzilli, S.; et al. Commensal Microbiota Promote Lung Cancer Development via gammadelta T Cells. Cell 2019, 176, 998–1013.e16. [Google Scholar] [CrossRef]
- Leinwand, J.C.; Miller, G. Microbes as biomarkers and targets in pancreatic cancer. Nat. Rev. Clin. Oncol. 2019, 16, 665–666. [Google Scholar] [CrossRef]
- Wei, M.Y.; Shi, S.; Liang, C.; Meng, Q.C.; Hua, J.; Zhang, Y.Y.; Liu, J.; Zhang, B.; Xu, J.; Yu, X.J. The microbiota and microbiome in pancreatic cancer: More influential than expected. Mol. Cancer 2019, 18, 97. [Google Scholar] [CrossRef]
- Aykut, B.; Pushalkar, S.; Chen, R.; Li, Q.; Abengozar, R.; Kim, J.I.; Shadaloey, S.A.; Wu, D.; Preiss, P.; Verma, N.; et al. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature 2019, 574, 264–267. [Google Scholar] [CrossRef]
- Riquelme, E.; Zhang, Y.; Zhang, L.; Montiel, M.; Zoltan, M.; Dong, W.; Quesada, P.; Sahin, I.; Chandra, V.; San Lucas, A.; et al. Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes. Cell 2019, 178, 795–806.e12. [Google Scholar] [CrossRef]
- Agus, A.; Clement, K.; Sokol, H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut 2021, 70, 1174–1182. [Google Scholar] [CrossRef]
- Chen, L.; Zhernakova, D.V.; Kurilshikov, A.; Andreu-Sanchez, S.; Wang, D.; Augustijn, H.E.; Vich Vila, A.; Lifelines Cohort, S.; Weersma, R.K.; Medema, M.H.; et al. Influence of the microbiome, diet and genetics on inter-individual variation in the human plasma metabolome. Nat. Med. 2022, 28, 2333–2343. [Google Scholar] [CrossRef]
- Den Besten, G.; van Eunen, K.; Groen, A.K.; Venema, K.; Reijngoud, D.J.; Bakker, B.M. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 2013, 54, 2325–2340. [Google Scholar] [CrossRef]
- Luu, M.; Visekruna, A. Short-chain fatty acids: Bacterial messengers modulating the immunometabolism of T cells. Eur. J. Immunol. 2019, 49, 842–848. [Google Scholar] [CrossRef]
- Blumberg, R.; Powrie, F. Microbiota, disease, and back to health: A metastable journey. Sci. Transl. Med. 2012, 4, 137rv7. [Google Scholar] [CrossRef]
- Luu, M.; Pautz, S.; Kohl, V.; Singh, R.; Romero, R.; Lucas, S.; Hofmann, J.; Raifer, H.; Vachharajani, N.; Carrascosa, L.C.; et al. The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic-epigenetic crosstalk in lymphocytes. Nat. Commun. 2019, 10, 760. [Google Scholar] [CrossRef]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef]
- Campbell, C.; McKenney, P.T.; Konstantinovsky, D.; Isaeva, O.I.; Schizas, M.; Verter, J.; Mai, C.; Jin, W.B.; Guo, C.J.; Violante, S.; et al. Bacterial metabolism of bile acids promotes generation of peripheral regulatory T cells. Nature 2020, 581, 475–479. [Google Scholar] [CrossRef] [PubMed]
- Coombes, J.L.; Robinson, N.J.; Maloy, K.J.; Uhlig, H.H.; Powrie, F. Regulatory T cells and intestinal homeostasis. Immunol. Rev. 2005, 204, 184–194. [Google Scholar] [CrossRef] [PubMed]
- West, N.R.; McCuaig, S.; Franchini, F.; Powrie, F. Emerging cytokine networks in colorectal cancer. Nat. Rev. Immunol. 2015, 15, 615–629. [Google Scholar] [CrossRef] [PubMed]
- Shine, E.E.; Crawford, J.M. Molecules from the Microbiome. Annu. Rev. Biochem. 2021, 90, 789–815. [Google Scholar] [CrossRef] [PubMed]
- Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Backhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef]
- Kim, M.; Qie, Y.; Park, J.; Kim, C.H. Gut Microbial Metabolites Fuel Host Antibody Responses. Cell Host Microbe 2016, 20, 202–214. [Google Scholar] [CrossRef]
- Kim, C.H.; Park, J.; Kim, M. Gut microbiota-derived short-chain Fatty acids, T cells, and inflammation. Immune Netw. 2014, 14, 277–288. [Google Scholar] [CrossRef]
- Rangan, P.; Mondino, A. Microbial short-chain fatty acids: A strategy to tune adoptive T cell therapy. J. Immunother. Cancer 2022, 10, 4147. [Google Scholar] [CrossRef]
- He, Y.; Fu, L.; Li, Y.; Wang, W.; Gong, M.; Zhang, J.; Dong, X.; Huang, J.; Wang, Q.; Mackay, C.R.; et al. Gut microbial metabolites facilitate anticancer therapy efficacy by modulating cytotoxic CD8(+) T cell immunity. Cell Metab. 2021, 33, 988–1000.e7. [Google Scholar] [CrossRef]
- Round, J.L.; Mazmanian, S.K. Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc. Natl. Acad. Sci. USA 2010, 107, 12204–12209. [Google Scholar] [CrossRef]
- Round, J.L.; Lee, S.M.; Li, J.; Tran, G.; Jabri, B.; Chatila, T.A.; Mazmanian, S.K. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 2011, 332, 974–977. [Google Scholar] [CrossRef]
- Mager, L.F.; Burkhard, R.; Pett, N.; Cooke, N.C.A.; Brown, K.; Ramay, H.; Paik, S.; Stagg, J.; Groves, R.A.; Gallo, M.; et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science 2020, 369, 1481–1489. [Google Scholar] [CrossRef]
- Messaoudene, M.; Pidgeon, R.; Richard, C.; Ponce, M.; Diop, K.; Benlaifaoui, M.; Nolin-Lapalme, A.; Cauchois, F.; Malo, J.; Belkaid, W.; et al. A Natural Polyphenol Exerts Antitumor Activity and Circumvents Anti-PD-1 Resistance through Effects on the Gut Microbiota. Cancer Discov. 2022, 12, 1070–1087. [Google Scholar] [CrossRef]
- Hezaveh, K.; Shinde, R.S.; Klotgen, A.; Halaby, M.J.; Lamorte, S.; Ciudad, M.T.; Quevedo, R.; Neufeld, L.; Liu, Z.Q.; Jin, R.; et al. Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity 2022, 55, 324–340.e8. [Google Scholar] [CrossRef]
- Dziubanska-Kusibab, P.J.; Berger, H.; Battistini, F.; Bouwman, B.A.M.; Iftekhar, A.; Katainen, R.; Cajuso, T.; Crosetto, N.; Orozco, M.; Aaltonen, L.A.; et al. Colibactin DNA-damage signature indicates mutational impact in colorectal cancer. Nat. Med. 2020, 26, 1063–1069. [Google Scholar] [CrossRef]
- Yachida, S.; Mizutani, S.; Shiroma, H.; Shiba, S.; Nakajima, T.; Sakamoto, T.; Watanabe, H.; Masuda, K.; Nishimoto, Y.; Kubo, M.; et al. Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat. Med. 2019, 25, 968–976. [Google Scholar] [CrossRef]
- Ternes, D.; Tsenkova, M.; Pozdeev, V.I.; Meyers, M.; Koncina, E.; Atatri, S.; Schmitz, M.; Karta, J.; Schmoetten, M.; Heinken, A.; et al. The gut microbial metabolite formate exacerbates colorectal cancer progression. Nat. Metab. 2022, 4, 458–475. [Google Scholar] [CrossRef]
- Qiu, J.; Villa, M.; Sanin, D.E.; Buck, M.D.; O’Sullivan, D.; Ching, R.; Matsushita, M.; Grzes, K.M.; Winkler, F.; Chang, C.H.; et al. Acetate promotes T cell effector function during glucose restriction. Cell Rep. 2019, 27, 2063–2074. [Google Scholar] [CrossRef]
- Coutzac, C.; Jouniaux, J.M.; Paci, A.; Schmidt, J.; Mallardo, D.; Seck, A.; Asvatourian, V.; Cassard, L.; Saulnier, P.; Lacroix, L.; et al. Systemic short chain fatty acids limit antitumor effect of CTLA-4 blockade in hosts with cancer. Nat. Commun. 2020, 11, 2168. [Google Scholar] [CrossRef]
- Wang, H.; Rong, X.; Zhao, G.; Zhou, Y.; Xiao, Y.; Ma, D.; Jin, X.; Wu, Y.; Yan, Y.; Yang, H.; et al. The microbial metabolite trimethylamine N-oxide promotes antitumor immunity in triple-negative breast cancer. Cell Metab. 2022, 34, 581–594.e8. [Google Scholar] [CrossRef]
- Iftekhar, A.; Berger, H.; Bouznad, N.; Heuberger, J.; Boccellato, F.; Dobrindt, U.; Hermeking, H.; Sigal, M.; Meyer, T.F. Genomic aberrations after short-term exposure to colibactin-producing E. coli transform primary colon epithelial cells. Nat. Commun. 2021, 12, 1003. [Google Scholar] [CrossRef] [PubMed]
- Rial, N.S.; Lazennec, G.; Prasad, A.R.; Krouse, R.S.; Lance, P.; Gerner, E.W. Regulation of deoxycholate induction of CXCL8 by the adenomatous polyposis coli gene in colorectal cancer. Int. J. Cancer 2009, 124, 2270–2280. [Google Scholar] [CrossRef] [PubMed]
- Marino, E.; Richards, J.L.; McLeod, K.H.; Stanley, D.; Yap, Y.A.; Knight, J.; McKenzie, C.; Kranich, J.; Oliveira, A.C.; Rossello, F.J.; et al. Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes. Nat. Immunol. 2017, 18, 552–562. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Cao, S.; Bashir, M.E.H.; Hesser, L.A.; Su, Y.; Hong, S.M.C.; Thompson, A.; Culleen, E.; Sabados, M.; Dylla, N.P.; et al. Treatment of peanut allergy and colitis in mice via the intestinal release of butyrate from polymeric micelles. Nat. Biomed. Eng. 2023, 7, 38–55. [Google Scholar] [CrossRef]
- Randrianarisoa, E.; Lehn-Stefan, A.; Wang, X.; Hoene, M.; Peter, A.; Heinzmann, S.S.; Zhao, X.; Konigsrainer, I.; Konigsrainer, A.; Balletshofer, B.; et al. Relationship of Serum Trimethylamine N-Oxide (TMAO) Levels with early Atherosclerosis in Humans. Sci. Rep. 2016, 6, 26745. [Google Scholar] [CrossRef]
- Colegio, O.R.; Chu, N.Q.; Szabo, A.L.; Chu, T.; Rhebergen, A.M.; Jairam, V.; Cyrus, N.; Brokowski, C.E.; Eisenbarth, S.C.; Phillips, G.M.; et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 2014, 513, 559–563. [Google Scholar] [CrossRef]
- Feng, Q.; Liu, Z.; Yu, X.; Huang, T.; Chen, J.; Wang, J.; Wilhelm, J.; Li, S.; Song, J.; Li, W.; et al. Lactate increases stemness of CD8+ T cells to augment anti-tumor immunity. Nat. Commun. 2022, 13, 4981. [Google Scholar] [CrossRef]
Microbial Molecules | Cancer Type | References |
---|---|---|
Formate | CRC | Ternes D. et al., Nature Metabolism, 2022 [87] |
Acetate | Melanoma | Qiu J. et al., Cell Reports, 2019 [88] |
Propionate | Metastatic melanoma | Coutzac C. et al., Nature Communications, 2020 [89] |
Butyrate | Adoptive transfer of MC-38 colon adenocarcinoma cells, melanoma, PDAC | He Y. et al., Cell Metabolism, 2021 [79]; Luu M. et al., Nature Communications, 2021 [45] |
Valerate | Melanoma, PDAC | Luu M. et al., Nature Communications, 2021 [45] |
Isovalerate | CRC | Yachida S. et al., Nature Medicine, 2019 [86] |
Insosin | CRC (AOM /DSS model), adoptive transfer of colon adenocarcinoma cells | Mager LF et al., Science, 2020 [82] |
Trimethylamine-N-oxide | Triple-negative breast cancer | Wang H. et al., Cell Metabolism, 2022 [90] |
Colibactin | CRC | Iftekhar A. et al., Nature Communications, 2021 [91] |
Secondary bile acids | CRC | Rial NS et al., International Journal of Cancer, 2009 [92] |
Tryptophan-derived indoles | PDAC | Hezaveh K. et al., Immunity, 2022 [84] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Luu, M.; Schütz, B.; Lauth, M.; Visekruna, A. The Impact of Gut Microbiota-Derived Metabolites on the Tumor Immune Microenvironment. Cancers 2023, 15, 1588. https://doi.org/10.3390/cancers15051588
Luu M, Schütz B, Lauth M, Visekruna A. The Impact of Gut Microbiota-Derived Metabolites on the Tumor Immune Microenvironment. Cancers. 2023; 15(5):1588. https://doi.org/10.3390/cancers15051588
Chicago/Turabian StyleLuu, Maik, Burkhard Schütz, Matthias Lauth, and Alexander Visekruna. 2023. "The Impact of Gut Microbiota-Derived Metabolites on the Tumor Immune Microenvironment" Cancers 15, no. 5: 1588. https://doi.org/10.3390/cancers15051588
APA StyleLuu, M., Schütz, B., Lauth, M., & Visekruna, A. (2023). The Impact of Gut Microbiota-Derived Metabolites on the Tumor Immune Microenvironment. Cancers, 15(5), 1588. https://doi.org/10.3390/cancers15051588