Microbial metabolites and immunotherapy: Basic rationale and clinical indications
Tài liệu tham khảo
Belkaid, 2017, Homeostatic Immunity and the Microbiota, Immunity, 46, 562, 10.1016/j.immuni.2017.04.008
Cullin, 2021, Microbiome and cancer, Cancer Cell, 39, 1317, 10.1016/j.ccell.2021.08.006
Gopalakrishnan, 2018, Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients, Science, 359, 97, 10.1126/science.aan4236
Matson, 2018, The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients, Science, 359, 104, 10.1126/science.aao3290
Routy, 2018, Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors, Science, 359, 91, 10.1126/science.aan3706
Baruch, 2021, Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients, Science, 371, 602, 10.1126/science.abb5920
Davar, 2021, Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients, Science, 371, 595, 10.1126/science.abf3363
Helmink, 2019, The microbiome, cancer, and cancer therapy, Nat. Med, 25, 377, 10.1038/s41591-019-0377-7
Capietto, 2017, Characterizing neoantigens for personalized cancer immunotherapy, Curr. Opin. Immunol., 46, 58, 10.1016/j.coi.2017.04.007
Yi, 2018, The role of neoantigen in immune checkpoint blockade therapy, Exp. Hematol. Oncol., 7, 28, 10.1186/s40164-018-0120-y
Kim, 2016, Immune escape to PD-L1/PD-1 blockade: seven steps to success (or failure), Ann. Oncol., 27, 1492, 10.1093/annonc/mdw217
Schreiber, 2011, Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion, Science, 331, 1565, 10.1126/science.1203486
Mariathasan, 2018, TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells, Nature, 554, 544, 10.1038/nature25501
Sivan, 2015, Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy, Science, 350, 1084, 10.1126/science.aac4255
Vetizou, 2015, Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota, Science, 350, 1079, 10.1126/science.aad1329
Andrews, 2021, Gut microbiota signatures are associated with toxicity to combined CTLA-4 and PD-1 blockade, Nat. Med, 27, 1432, 10.1038/s41591-021-01406-6
Masetti, 2021, Microbiome-derived metabolites in allogeneic hematopoietic stem cell transplantation, Int J. Mol. Sci., 22, 10.3390/ijms22031197
Shono, 2018, Gut microbiota injury in allogeneic haematopoietic stem cell transplantation, Nat. Rev. Cancer, 18, 283, 10.1038/nrc.2018.10
Iida, 2013, Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment, Science, 342, 967, 10.1126/science.1240527
Pflug, 2016, Efficacy of antineoplastic treatment is associated with the use of antibiotics that modulate intestinal microbiota, Oncoimmunology, 5, 10.1080/2162402X.2016.1150399
Smith, 2022, Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy, Nat. Med, 28, 713, 10.1038/s41591-022-01702-9
Mager, 2020, Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy, Science, 369, 1481, 10.1126/science.abc3421
Renga, 2022, Optimizing therapeutic outcomes of immune checkpoint blockade by a microbial tryptophan metabolite, J. Immunother. Cancer, 10, 10.1136/jitc-2021-003725
He, 2021, Gut microbial metabolites facilitate anticancer therapy efficacy by modulating cytotoxic CD8(+) T cell immunity, Cell Metab., 33, 988, 10.1016/j.cmet.2021.03.002
Lam, 2021, Microbiota triggers STING-type I IFN-dependent monocyte reprogramming of the tumor microenvironment, Cell, 184, 5338, 10.1016/j.cell.2021.09.019
Luu, 2021, Microbial short-chain fatty acids modulate CD8(+) T cell responses and improve adoptive immunotherapy for cancer, Nat. Commun., 12, 4077, 10.1038/s41467-021-24331-1
Bishai, 2021, Small molecule metabolites at the host-microbiota interface, J. Immunol., 207, 1725, 10.4049/jimmunol.2100528
Twomey, 2021, Cancer immunotherapy update: FDA-Approved checkpoint inhibitors and companion diagnostics, AAPS J., 23, 39, 10.1208/s12248-021-00574-0
Welihinda, 2018, Enhancement of inosine-mediated A2AR signaling through positive allosteric modulation, Cell Signal, 42, 227, 10.1016/j.cellsig.2017.11.002
Hasko, 2000, Inosine inhibits inflammatory cytokine production by a posttranscriptional mechanism and protects against endotoxin-induced shock, J. Immunol., 164, 1013, 10.4049/jimmunol.164.2.1013
Liaudet, 2002, Inosine exerts a broad range of antiinflammatory effects in a murine model of acute lung injury, Ann. Surg., 235, 568, 10.1097/00000658-200204000-00016
Mabley, 2003, Inosine reduces inflammation and improves survival in a murine model of colitis, Am. J. Physiol. Gastrointest. Liver Physiol., 284, G138, 10.1152/ajpgi.00060.2002
Azambuja, 2019, Inhibition of the adenosinergic pathway in cancer rejuvenates innate and adaptive immunity, Int J. Mol. Sci., 20, 10.3390/ijms20225698
Leone, 2015, A2aR antagonists: Next generation checkpoint blockade for cancer immunotherapy, Comput. Struct. Biotechnol. J., 13, 265, 10.1016/j.csbj.2015.03.008
Vigano, 2019, Targeting adenosine in cancer immunotherapy to enhance T-cell function, Front Immunol., 10, 925, 10.3389/fimmu.2019.00925
Fiorucci, 2018, Bile acids activated receptors regulate innate immunity, Front Immunol., 9, 1853, 10.3389/fimmu.2018.01853
Jones, 2008, Functional and comparative metagenomic analysis of bile salt hydrolase activity in the human gut microbiome, Proc. Natl. Acad. Sci. USA, 105, 13580, 10.1073/pnas.0804437105
Vavassori, 2009, The bile acid receptor FXR is a modulator of intestinal innate immunity, J. Immunol., 183, 6251, 10.4049/jimmunol.0803978
Mencarelli, 2009, The bile acid sensor farnesoid X receptor is a modulator of liver immunity in a rodent model of acute hepatitis, J. Immunol., 183, 6657, 10.4049/jimmunol.0901347
Cipriani, 2011, The bile acid receptor GPBAR-1 (TGR5) modulates integrity of intestinal barrier and immune response to experimental colitis, PLoS One, 6, 10.1371/journal.pone.0025637
Skelly, 2019, Mining the microbiota for microbial and metabolite-based immunotherapies, Nat. Rev. Immunol., 19, 305, 10.1038/s41577-019-0144-5
Guo, 2016, Bile acids control inflammation and metabolic disorder through inhibition of NLRP3 inflammasome, Immunity, 45, 802, 10.1016/j.immuni.2016.09.008
Hao, 2017, Farnesoid X receptor regulation of the NLRP3 inflammasome underlies cholestasis-associated sepsis, Cell Metab., 25, 856, 10.1016/j.cmet.2017.03.007
Ma, 2018, Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells, Science, 360, 10.1126/science.aan5931
Mirji, 2022, The microbiome-derived metabolite TMAO drives immune activation and boosts responses to immune checkpoint blockade in pancreatic cancer, Sci. Immunol., 7, eabn0704, 10.1126/sciimmunol.abn0704
Wang, 2022, The microbial metabolite trimethylamine N-oxide promotes antitumor immunity in triple-negative breast cancer, Cell Metab., 34, 581, 10.1016/j.cmet.2022.02.010
Met, 2019, Principles of adoptive T cell therapy in cancer, Semin Immunopathol., 41, 49, 10.1007/s00281-018-0703-z
Chong, 2021, P. Lymphoma program investigators at the University of, five-year outcomes for refractory B-Cell lymphomas with CAR T-cell therapy, New Engl. J. Med, 384, 673, 10.1056/NEJMc2030164
Santomasso, 2019, The other side of CAR T-cell therapy: cytokine release syndrome neurologic toxicity, and financial burden, Am. Soc. Clin. Oncol. Educ. Book, 39, 433, 10.1200/EDBK_238691
Rooks, 2016, Gut microbiota, metabolites and host immunity, Nat. Rev. Immunol., 16, 341, 10.1038/nri.2016.42
Cummings, 1987, Short chain fatty acids in human large intestine, portal, hepatic and venous blood, Gut, 28, 1221, 10.1136/gut.28.10.1221
Arpaia, 2013, Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation, Nature, 504, 451, 10.1038/nature12726
Furusawa, 2013, Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells, Nature, 504, 446, 10.1038/nature12721
Kim, 2016, Gut microbial metabolites fuel host antibody responses, Cell Host Microbe, 20, 202, 10.1016/j.chom.2016.07.001
Chun, 2019, Metabolite-sensing receptor Ffar2 regulates colonic group 3 innate lymphoid cells and gut immunity, Immunity, 51, 871, 10.1016/j.immuni.2019.09.014
Usami, 2008, Butyrate and trichostatin A attenuate nuclear factor kappaB activation and tumor necrosis factor alpha secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells, Nutr. Res, 28, 321, 10.1016/j.nutres.2008.02.012
Chang, 2014, The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition, Proc. Natl. Acad. Sci. USA, 111, 2247, 10.1073/pnas.1322269111
Singh, 2010, Blockade of dendritic cell development by bacterial fermentation products butyrate and propionate through a transporter (Slc5a8)-dependent inhibition of histone deacetylases, J. Biol. Chem., 285, 27601, 10.1074/jbc.M110.102947
Luu, 2018, Regulation of the effector function of CD8(+) T cells by gut microbiota-derived metabolite butyrate, Sci. Rep., 8, 14430, 10.1038/s41598-018-32860-x
Bachem, 2019, Microbiota-derived short-chain fatty acids promote the memory potential of antigen-activated CD8(+) T Cells, Immunity, 51, 285, 10.1016/j.immuni.2019.06.002
Trompette, 2014, Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis, Nat. Med, 20, 159, 10.1038/nm.3444
Hagihara, 2020, Clostridium butyricum modulates the microbiome to protect intestinal barrier function in mice with antibiotic-induced dysbiosis, Iscience, 23, 10.1016/j.isci.2019.100772
Tomita, 2020, Association of Probiotic Clostridium butyricum Therapy with Survival and Response to Immune Checkpoint Blockade in Patients with Lung Cancer, Cancer Immunol. Res, 8, 1236, 10.1158/2326-6066.CIR-20-0051
Dizman, 2022, Nivolumab plus ipilimumab with or without live bacterial supplementation in metastatic renal cell carcinoma: a randomized phase 1 trial, Nat. Med, 28, 704, 10.1038/s41591-022-01694-6
Docampo, 2022, Alloreactive T cells deficient of the short-chain fatty acid receptor GPR109A induce less graft-versus-host disease, Blood, 139, 2392, 10.1182/blood.2021010719
Wang, 2020, Inosine is an alternative carbon source for CD8(+)-T-cell function under glucose restriction, Nat. Metab., 2, 635, 10.1038/s42255-020-0219-4
Apetoh, 2007, Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy, Nat. Med, 13, 1050, 10.1038/nm1622
Ghiringhelli, 2009, Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors, Nat. Med, 15, 1170, 10.1038/nm.2028
Corrales, 2016, Endogenous and pharmacologic targeting of the STING pathway in cancer immunotherapy, Cytokine, 77, 245, 10.1016/j.cyto.2015.08.258
Woo, 2014, STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors, Immunity, 41, 830, 10.1016/j.immuni.2014.10.017
Tanaka, 2012, STING specifies IRF3 phosphorylation by TBK1 in the cytosolic DNA signaling pathway, Sci. Signal, 5, ra20, 10.1126/scisignal.2002521
Fitzgerald, 2003, IKKepsilon and TBK1 are essential components of the IRF3 signaling pathway, Nat. Immunol., 4, 491, 10.1038/ni921
Ishikawa, 2008, STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling, Nature, 455, 674, 10.1038/nature07317
Dunn, 2006, Interferons, immunity and cancer immunoediting, Nat. Rev. Immunol., 6, 836, 10.1038/nri1961
Fuertes, 2013, Type I interferon response and innate immune sensing of cancer, Trends Immunol., 34, 67, 10.1016/j.it.2012.10.004
Atarashi, 2008, ATP drives lamina propria T(H)17 cell differentiation, Nature, 455, 808, 10.1038/nature07240
Ganal, 2012, Priming of natural killer cells by nonmucosal mononuclear phagocytes requires instructive signals from commensal microbiota, Immunity, 37, 171, 10.1016/j.immuni.2012.05.020
Erttmann, 2022, The gut microbiota prime systemic antiviral immunity via the cGAS-STING-IFN-I axis, Immunity, 55, 847, 10.1016/j.immuni.2022.04.006
Li, 2020, Prebiotic-induced anti-tumor immunity attenuates tumor growth, Cell Rep., 30, 1753, 10.1016/j.celrep.2020.01.035
Leventhal, 2020, Immunotherapy with engineered bacteria by targeting the STING pathway for anti-tumor immunity, Nat. Commun., 11, 2739, 10.1038/s41467-020-16602-0
Riese, 2021, 500 SYNB1891, a bacterium engineered to produce a STING agonist, demonstrates target engagement in humans following intratumoral injection, J. Immunother. Cancer, 9, 10.1136/jitc-2021-SITC2021.500
Chao, 2019, Therapeutic targeting of the macrophage immune checkpoint CD47 in myeloid malignancies, Front Oncol., 9, 1380, 10.3389/fonc.2019.01380
Chao, 2012, The CD47-SIRPalpha pathway in cancer immune evasion and potential therapeutic implications, Curr. Opin. Immunol., 24, 225, 10.1016/j.coi.2012.01.010
Liu, 2015, CD47 blockade triggers T cell-mediated destruction of immunogenic tumors, Nat. Med., 21, 1209, 10.1038/nm.3931
Deng, 2014, STING-dependent cytosolic DNA sensing promotes radiation-induced type I interferon-dependent antitumor immunity in immunogenic tumors, Immunity, 41, 843, 10.1016/j.immuni.2014.10.019
Chen, 2016, Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing, Nat. Immunol., 17, 1142, 10.1038/ni.3558
Shi, 2020, Intratumoral accumulation of gut microbiota facilitates CD47-based immunotherapy via STING signaling, J. Exp. Med, 217, 10.1084/jem.20192282
Conroy, 2022, Immune-related adverse events and the balancing act of immunotherapy, Nat. Commun., 13, 392, 10.1038/s41467-022-27960-2
Wang, 2018, Fecal microbiota transplantation for refractory immune checkpoint inhibitor-associated colitis, Nat. Med, 24, 1804, 10.1038/s41591-018-0238-9
Krautkramer, 2021, Gut microbial metabolites as multi-kingdom intermediates, Nat. Rev. Microbiol, 19, 77, 10.1038/s41579-020-0438-4
Roager, 2018, Microbial tryptophan catabolites in health and disease, Nat. Commun., 9, 3294, 10.1038/s41467-018-05470-4
Zelante, 2013, Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22, Immunity, 39, 372, 10.1016/j.immuni.2013.08.003
Stockinger, 2014, The aryl hydrocarbon receptor: multitasking in the immune system, Annu Rev. Immunol., 32, 403, 10.1146/annurev-immunol-032713-120245
Quintana, 2008, Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor, Nature, 453, 65, 10.1038/nature06880
Bessede, 2014, Aryl hydrocarbon receptor control of a disease tolerance defence pathway, Nature, 511, 184, 10.1038/nature13323
Gandhi, 2010, Activation of the aryl hydrocarbon receptor induces human type 1 regulatory T cell-like and Foxp3(+) regulatory T cells, Nat. Immunol., 11, 846, 10.1038/ni.1915
Cervantes-Barragan, 2017, Lactobacillus reuteri induces gut intraepithelial CD4(+)CD8alphaalpha(+) T cells, Science, 357, 806, 10.1126/science.aah5825
Wilck, 2017, Salt-responsive gut commensal modulates TH17 axis and disease, Nature, 551, 585, 10.1038/nature24628
Sugimura, 2021, Lactobacillus gallinarum modulates the gut microbiota and produces anti-cancer metabolites to protect against colorectal tumourigenesis, Gut, 71, 2011, 10.1136/gutjnl-2020-323951
Agus, 2018, Gut microbiota regulation of tryptophan metabolism in health and disease, Cell Host Microbe, 23, 716, 10.1016/j.chom.2018.05.003
Zou, 2016, PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations, Sci. Transl. Med, 8, 328rv4, 10.1126/scitranslmed.aad7118
Hatae, 2020, Combination of host immune metabolic biomarkers for the PD-1 blockade cancer immunotherapy, JCI Insight, 5, 10.1172/jci.insight.133501
Wikoff, 2009, Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites, Proc. Natl. Acad. Sci. USA, 106, 3698, 10.1073/pnas.0812874106