The Species of Gut Bacteria Associated with Antitumor Immunity in Cancer Therapy
Abstract
:1. Introduction
2. Bacteria Species Associated with Favorable Modulation in Antitumor Immunity
2.1. Bifidobacterium spp.
2.2. Enterococcus hirae
2.3. Ruminococcaceae (Oscillospiraceae) Family
2.4. Faecalibacterium spp.
2.5. Oscillibacter spp.
2.6. Burkholderia spp.
2.7. Prevotella spp.
3. Bacteria Species Associated with Unfavorable Modulation in Antitumor Immunity
3.1. Fusobacterium nucleatum
3.2. Escherichia coli
3.3. Ruminococcus spp.
3.4. Gammaproteobacteria Class
4. The Bacteria Species Associated with Both Favorable and Unfavorable Modulation in Antitumor Immunity
4.1. Akkermansia muciniphila
4.2. Bacteroides spp.
4.3. Clostridiales (Eubacteriales) Order and Clostridium spp.
4.4. Klebsiella spp.
4.5. Alistipes spp.
5. Prospects
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Dapito, D.H.; Mencin, A.; Gwak, G.-Y.; Pradère, J.-P.; Jang, M.-K.; Mederacke, I.; Caviglia, J.M.; Khiabanian, H.; Adeyemi, A.; Bataller, R.; et al. Promotion of Hepatocellular Carcinoma by the Intestinal Microbiota and TLR4. Cancer Cell 2012, 21, 504–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Daillère, R.; Vétizou, M.; Waldschmitt, N.; Yamazaki, T.; Isnard, C.; Poirier-Colame, V.; Duong, C.P.M.; Flament, C.; Lepage, P.; Roberti, M.P.; et al. Enterococcus hirae and Barnesiella intestinihominis Facilitate Cyclophosphamide-Induced Therapeutic Immunomodulatory Effects. Immunity 2016, 45, 931–943. [Google Scholar] [CrossRef] [Green Version]
- Louis, P.; Flint, H.J. Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol. Lett. 2009, 294, 1–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, G.; Liu, D.; Cooper, T.K.; Kimchi, E.T.; Qi, X.; Avella, D.M.; Li, N.; Yang, Q.X.; Kester, M.; Rountree, C.B.; et al. Successful chemoimmunotherapy against hepatocellular cancer in a novel murine model. J. Hepatol. 2016, 66, 75–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, G.; Liu, D.; Kimchi, E.T.; Kaifi, J.T.; Qi, X.; Manjunath, Y.; Liu, X.; Deering, T.; Avella, D.M.; Fox, T.; et al. Nanoliposome C6-Ceramide Increases the Anti-tumor Immune Response and Slows Growth of Liver Tumors in Mice. Gastroenterology 2018, 154, 1024–1036.e9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qi, X.; Wu, F.; Kim, S.H.; Kaifi, J.T.; Kimchi, E.T.; Snyder, H.; Illendula, A.; Fox, T.; Kester, M.; Staveley-O’Carroll, K.F.; et al. Nanoliposome C6-Ceramide in combination with anti-CTLA4 antibody improves anti-tumor immunity in hepatocellular cancer. FASEB J. 2022, 36, e22250. [Google Scholar] [CrossRef] [PubMed]
- Qi, X.; Yang, M.; Ma, L.; Sauer, M.; Avella, D.; Kaifi, J.T.; Bryan, J.; Cheng, K.; Staveley-O’Carroll, K.F.; Kimchi, E.T.; et al. Synergizing sunitinib and radiofrequency ablation to treat hepatocellular cancer by triggering the antitumor immune response. J. Immunother. Cancer 2020, 8, e001038. [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] [PubMed]
- Baruch, E.N.; Youngster, I.; Ben-Betzalel, G.; Ortenberg, R.; Lahat, A.; Katz, L.; Adler, K.; Dick-Necula, D.; Raskin, S.; Bloch, N.; et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 2020, 371, 602–609. [Google Scholar] [CrossRef] [PubMed]
- Cheng, W.Y.; Wu, C.-Y.; Yu, J. The role of gut microbiota in cancer treatment: Friend or foe? Gut 2020, 69, 1867–1876. [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] [PubMed] [Green Version]
- Shi, Y.; Zheng, W.; Yang, K.; Harris, K.G.; Ni, K.; Xue, L.; Lin, W.; Chang, E.B.; Weichselbaum, R.R.; Fu, Y.-X. Intratumoral accumulation of gut microbiota facilitates CD47-based immunotherapy via STING signaling. J. Exp. Med. 2020, 217, e20192282. [Google Scholar] [CrossRef] [PubMed]
- Sun, S.; Luo, L.; Liang, W.; Yin, Q.; Guo, J.; Rush, A.M.; Lv, Z.; Liang, Q.; Fischbach, M.A.; Sonnenburg, J.L.; et al. Bifidobacterium alters the gut microbiota and modulates the functional metabolism of T regulatory cells in the context of immune checkpoint blockade. Proc. Natl. Acad. Sci. USA 2020, 117, 27509–27515. [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] [Green Version]
- Jin, Y.; Dong, H.; Xia, L.; Yang, Y.; Zhu, Y.; Shen, Y.; Zheng, H.; Yao, C.; Wang, Y.; Lu, S. The Diversity of Gut Microbiome is Associated with Favorable Responses to Anti–Programmed Death 1 Immunotherapy in Chinese Patients With NSCLC. J. Thorac. Oncol. 2019, 14, 1378–1389. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Yin, Q.; Chen, L.; Davis, M.M. Bifidobacterium can mitigate intestinal immunopathology in the context of CTLA-4 blockade. Proc. Natl. Acad. Sci. USA 2017, 115, 157–161. [Google Scholar] [CrossRef] [Green Version]
- Yoon, Y.; Kim, G.; Jeon, B.-N.; Fang, S.; Park, H. Bifidobacterium Strain-Specific Enhances the Efficacy of Cancer Therapeutics in Tumor-Bearing Mice. Cancers 2021, 13, 957. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Routy, B.; le Chatelier, E.; DeRosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, 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] [Green Version]
- Sevcikova, A.; Izoldova, N.; Stevurkova, V.; Kasperova, B.; Chovanec, M.; Ciernikova, S.; Mego, M. The Impact of the Microbiome on Resistance to Cancer Treatment with Chemotherapeutic Agents and Immunotherapy. Int. J. Mol. Sci. 2022, 23, 488. [Google Scholar] [CrossRef]
- Viaud, S.; Saccheri, F.; Mignot, G.; Yamazaki, T.; Daillère, R.; Hannani, D.; Enot, D.P.; Pfirschke, C.; Engblom, C.; Pittet, M.J.; et al. The Intestinal Microbiota Modulates the Anticancer Immune Effects of Cyclophosphamide. Science 2013, 342, 971–976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fluckiger, A.; Daillère, R.; Sassi, M.; Sixt, B.S.; Liu, P.; Loos, F.; Richard, C.; Rabu, C.; Alou, M.T.; Goubet, A.-G.; et al. Cross-reactivity between tumor MHC class I–restricted antigens and an enterococcal bacteriophage. Science 2020, 369, 936–942. [Google Scholar] [CrossRef] [PubMed]
- Goubet, A.-G.; Wheeler, R.; Fluckiger, A.; Qu, B.; Lemaître, F.; Iribarren, K.; Mondragón, L.; Alou, M.T.; Pizzato, E.; Durand, S.; et al. Multifaceted modes of action of the anticancer probiotic Enterococcus hirae. Cell Death Differ. 2021, 28, 2276–2295. [Google Scholar] [CrossRef] [PubMed]
- Rong, Y.; Dong, Z.; Hong, Z.; Jin, Y.; Zhang, W.; Zhang, B.; Mao, W.; Kong, H.; Wang, C.; Yang, B.; et al. Reactivity toward Bifidobacterium longum and Enterococcus hirae demonstrate robust CD8+ T cell response and better prognosis in HBV-related hepatocellular carcinoma. Exp. Cell Res. 2017, 358, 352–359. [Google Scholar] [CrossRef] [PubMed]
- Lee, B.; Lee, J.; Woo, M.-Y.; Lee, M.; Shin, H.-J.; Kim, K.; Park, S. Modulation of the Gut Microbiota Alters the Tumour-Suppressive Efficacy of Tim-3 Pathway Blockade in a Bacterial Species- and Host Factor-Dependent Manner. Microorganisms 2020, 8, 1395. [Google Scholar] [CrossRef] [PubMed]
- De Weirdt, R.; Van De Wiele, T. Micromanagement in the gut: Microenvironmental factors govern colon mucosal biofilm structure and functionality. npj Biofilms Microbiomes 2015, 1, 15026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Panebianco, C.; Adamberg, K.; Jaagura, M.; Copetti, M.; Fontana, A.; Adamberg, S.; Kolk, K.; Vilu, R.; Andriulli, A.; Pazienza, V. Influence of gemcitabine chemotherapy on the microbiota of pancreatic cancer xenografted mice. Cancer Chemother. Pharmacol. 2018, 81, 773–782. [Google Scholar] [CrossRef] [PubMed]
- Fukuoka, S.; Daisuke, M.; Togashi, Y.; Sugiyama, E.; Udagawa, H.; Kirita, K.; Kamada, T.; Kawazoe, A.; Goto, K.; Doi, T.; et al. Association of gut microbiome with immune status and clinical response in solid tumor patients who received on anti-PD-1 therapies. J. Clin. Oncol. 2018, 36, 3011. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Anfossi, S.; Calin, G.A. Gut microbiota: A new player in regulating immune- and chemo-therapy efficacy. Cancer Drug Resist. 2020, 3, 356–370. [Google Scholar] [CrossRef]
- Chaput, N.; Lepage, P.; Coutzac, C.; Soularue, E.; Le Roux, K.; Monot, C.; Boselli, L.; Routier, E.; Cassard, L.; Collins, M.; et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann. Oncol. 2017, 28, 1368–1379. [Google Scholar] [CrossRef]
- Bachem, A.; Makhlouf, C.; Binger, K.J.; de Souza, D.P.; Tull, D.; Hochheiser, K.; Whitney, P.G.; Fernandez-Ruiz, D.; Dähling, S.; Kastenmüller, W.; et al. Microbiota-Derived Short-Chain Fatty Acids Promote the Memory Potential of Antigen-Activated CD8+ T Cells. Immunity 2019, 51, 285–297.e285. [Google Scholar] [CrossRef] [PubMed]
- Peters, B.A.; Wilson, M.; Moran, U.; Pavlick, A.; Izsak, A.; Wechter, T.; Weber, J.; Osman, I.; Ahn, J. Relating the gut metagenome and metatranscriptome to immunotherapy responses in melanoma patients. Genome Med. 2019, 11, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Botticelli, A.; Putignani, L.; Zizzari, I.; Del Chierico, F.; Reddel, S.; Di Pietro, F.; Quagliarello, A.; Onesti, C.E.; Raffaele, G.; Mazzuca, F.; et al. Changes of microbiome profile during nivolumab treatment in NSCLC patients. J. Clin. Oncol. 2018, 36, e15020. [Google Scholar] [CrossRef]
- Tajiri, K.; Shimizu, Y. Gut bacteria may control development of hepatocellular carcinoma. Hepatobiliary Surg. Nutr. 2017, 6, 417–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, J.; Sung, C.Y.J.; Lee, N.; Ni, Y.; Pihlajamäki, J.; Panagiotou, G.; El-Nezami, H. Probiotics modulated gut microbiota suppresses hepatocellular carcinoma growth in mice. Proc. Natl. Acad. Sci. USA 2016, 113, E1306-15. [Google Scholar] [CrossRef] [Green Version]
- Sarhadi, V.; Mathew, B.; Kokkola, A.; Karla, T.; Tikkanen, M.; Rautelin, H.; Lahti, L.; Puolakkainen, P.; Knuutila, S. Gut microbiota of patients with different subtypes of gastric cancer and gastrointestinal stromal tumors. Gut Pathog. 2021, 13, 11. [Google Scholar] [CrossRef]
- Flemer, B.; Lynch, D.B.; Brown, J.M.R.; Jeffery, I.B.; Ryan, F.J.; Claesson, M.J.; O’Riordain, M.; Shanahan, F.; O’Toole, P.W. Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut 2017, 66, 633–643. [Google Scholar] [CrossRef]
- Bhatt, A.P.; Redinbo, M.R.; Bultman, S.J. The role of the microbiome in cancer development and therapy. CA Cancer J. Clin. 2017, 67, 326–344. [Google Scholar] [CrossRef] [Green Version]
- Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.M.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef]
- Huang, F.-Y.; Dai, S.-Z.; Wang, J.-Y.; Lin, Y.-Y.; Wang, C.-C.; Zheng, W.-P.; Tan, G.-H. Engineered porous/hollow Burkholderia pseudomallei loading tumor lysate as a vaccine. Biomaterials 2021, 278, 121141. [Google Scholar] [CrossRef] [PubMed]
- Pernigoni, N.; Zagato, E.; Calcinotto, A.; Troiani, M.; Mestre, R.P.; Calì, B.; Attanasio, G.; Troisi, J.; Minini, M.; Mosole, S.; et al. Commensal bacteria promote endocrine resistance in prostate cancer through androgen biosynthesis. Science 2021, 374, 216–224. [Google Scholar] [CrossRef] [PubMed]
- Castellarin, M.; Warren, R.L.; Freeman, J.D.; Dreolini, L.; Krzywinski, M.; Strauss, J.; Barnes, R.; Watson, P.; Allen-Vercoe, E.; Moore, R.A.; et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012, 22, 299–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kostic, A.D.; Gevers, D.; Pedamallu, C.S.; Michaud, M.; Duke, F.; Earl, A.M.; Ojesina, A.I.; Jung, J.; Bass, A.J.; Tabernero, J.; et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012, 22, 292–298. [Google Scholar] [CrossRef] [Green Version]
- Brennan, C.A.; Garrett, W.S. Fusobacterium nucleatum—Symbiont, opportunist and oncobacterium. Nat. Rev. Microbiol. 2019, 17, 156–166. [Google Scholar] [CrossRef]
- Komiya, Y.; Shimomura, Y.; Higurashi, T.; Sugi, Y.; Arimoto, J.; Umezawa, S.; Uchiyama, S.; Matsumoto, M.; Nakajima, A. Patients with colorectal cancer have identical strains of Fusobacterium nucleatum in their colorectal cancer and oral cavity. Gut 2019, 68, 1335–1337. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Flanagan, L.; Schmid, J.; Ebert, M.; Soucek, P.; Kunicka, T.; Liška, V.; Bruha, J.; Neary, P.; DeZeeuw, N.; Tommasino, M.; et al. Fusobacterium nucleatum associates with stages of colorectal neoplasia development, colorectal cancer and disease outcome. Eur. J. Clin. Microbiol. Infect. Dis. 2014, 33, 1381–1390. [Google Scholar] [CrossRef]
- Mima, K.; Nishihara, R.; Qian, Z.R.; Cao, Y.; Sukawa, Y.; Nowak, J.A.; Yang, J.; Dou, R.; Masugi, Y.; Song, M.; et al. Fusobacterium nucleatumin colorectal carcinoma tissue and patient prognosis. Gut 2016, 65, 1973–1980. [Google Scholar] [CrossRef] [Green Version]
- Wu, J.; Li, Q.; Fu, X. Fusobacterium nucleatum Contributes to the Carcinogenesis of Colorectal Cancer by Inducing Inflammation and Suppressing Host Immunity. Transl. Oncol. 2019, 12, 846–851. [Google Scholar] [CrossRef]
- Rubinstein, M.R.; Wang, X.; Liu, W.; Hao, Y.; Cai, G.; Han, Y.W. Fusobacterium nucleatum Promotes Colorectal Carcinogenesis by Modulating E-Cadherin/β-Catenin Signaling via its FadA Adhesin. Cell Host Microbe 2013, 14, 195–206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sui, X.; Chen, R.; Wang, Z.; Huang, Z.; Kong, N.; Zhang, M.; Han, W.; Lou, F.; Yang, J.; Zhang, Q.; et al. Autophagy and chemotherapy resistance: A promising therapeutic target for cancer treatment. Cell Death Dis. 2013, 4, e838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abed, J.; Emgård, J.E.; Zamir, G.; Faroja, M.; Almogy, G.; Grenov, A.; Sol, A.; Naor, R.; Pikarsky, E.; Atlan, K.A.; et al. Fap2 Mediates Fusobacterium nucleatum Colorectal Adenocarcinoma Enrichment by Binding to Tumor-Expressed Gal-GalNAc. Cell Host Microbe 2016, 20, 215–225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gur, C.; Ibrahim, Y.; Isaacson, B.; Yamin, R.; Abed, J.; Gamliel, M.; Enk, J.; Bar-On, Y.; Stanietsky-Kaynan, N.; Coppenhagen-Glazer, S.; et al. Binding of the Fap2 Protein of Fusobacterium nucleatum to Human Inhibitory Receptor TIGIT Protects Tumors from Immune Cell Attack. Immunity 2015, 42, 344–355. [Google Scholar] [CrossRef] [Green Version]
- Geller, L.T.; Barzily-Rokni, M.; Danino, T.; Jonas, O.H.; Shental, N.; Nejman, D.; Gavert, N.; Zwang, Y.; Cooper, Z.A.; Shee, K.; et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 2017, 357, 1156–1160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cuevas-Ramos, G.; Petit, C.R.; Marcq, I.; Boury, M.; Oswald, E.; Nougayrède, J.-P. Escherichia coli induces DNA damage in vivo and triggers genomic instability in mammalian cells. Proc. Natl. Acad. Sci. USA 2010, 107, 11537–11542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dalmasso, G.; Cougnoux, A.; Delmas, J.; Darfeuille-Michaud, A.; Bonnet, R. The bacterial genotoxin colibactin promotes colon tumor growth by modifying the tumor microenvironment. Gut Microbes 2014, 5, 675–680. [Google Scholar] [CrossRef] [Green Version]
- Wang, F.; Cai, K.; Xiao, Q.; He, L.; Xie, L.; Liu, Z. Akkermansia muciniphila administration exacerbated the development of colitis-associated colorectal cancer in mice. J. Cancer 2022, 13, 124–133. [Google Scholar] [CrossRef]
- Osman, M.A.; Neoh, H.-M.; Ab Mutalib, N.-S.; Chin, S.-F.; Mazlan, L.; Ali, R.A.R.; Zakaria, A.D.; Ngiu, C.S.; Ang, M.Y.; Jamal, R. Parvimonas micra, Peptostreptococcus stomatis, Fusobacterium nucleatum and Akkermansia muciniphila as a four-bacteria biomarker panel of colorectal cancer. Sci. Rep. 2021, 11, 2925. [Google Scholar] [CrossRef]
- Xu, X.; Lv, J.; Guo, F.; Li, J.; Jia, Y.; Jiang, D.; Wang, N.; Zhang, C.; Kong, L.; Liu, Y.; et al. Gut Microbiome Influences the Efficacy of PD-1 Antibody Immunotherapy on MSS-Type Colorectal Cancer via Metabolic Pathway. Front. Microbiol. 2020, 11, 814. [Google Scholar] [CrossRef]
- Botticelli, A.; Vernocchi, P.; Marini, F.; Quagliariello, A.; Cerbelli, B.; Reddel, S.; Del Chierico, F.; Di Pietro, F.; Giusti, R.; Tomassini, A.; et al. Gut metabolomics profiling of non-small cell lung cancer (NSCLC) patients under immunotherapy treatment. J. Transl. Med. 2020, 18, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salgia, N.J.; Bergerot, P.G.; Maia, M.C.; Dizman, N.; Hsu, J.; Gillece, J.D.; Folkerts, M.; Reining, L.; Trent, J.; Highlander, S.K.; et al. Stool Microbiome Profiling of Patients with Metastatic Renal Cell Carcinoma Receiving Anti–PD-1 Immune Checkpoint Inhibitors. Eur. Urol. 2020, 78, 498–502. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Daisley, B.A.; Chanyi, R.M.; Abdur-Rashid, K.; Al, K.F.; Gibbons, S.; Chmiel, J.A.; Wilcox, H.; Reid, G.; Anderson, A.; Dewar, M.; et al. Abiraterone acetate preferentially enriches for the gut commensal Akkermansia muciniphila in castrate-resistant prostate cancer patients. Nat. Commun. 2020, 11, 4822. [Google Scholar] [CrossRef] [PubMed]
- Wexler, H.M. Bacteroides: The Good, the Bad, and the Nitty-Gritty. Clin. Microbiol. Rev. 2007, 20, 593–621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Townsend, G.E.; Han, W.; Schwalm, N.D.; Hong, X.; Bencivenga-Barry, N.A.; Goodman, A.L.; Groisman, E.A. A Master Regulator of Bacteroides thetaiotaomicron Gut Colonization Controls Carbohydrate Utilization and an Alternative Protein Synthesis Factor. mBio 2020, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, J.; Bjursell, M.K.; Himrod, J.; Deng, S.; Carmichael, L.K.; Chiang, H.C.; Hooper, L.V.; Gordon, J.I. A Genomic View of the Human- Bacteroides thetaiotaomicron Symbiosis. Science 2003, 299, 2074–2076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mazmanian, S.; Kasper, D.L. The love–hate relationship between bacterial polysaccharides and the host immune system. Nat. Rev. Immunol. 2006, 6, 849–858. [Google Scholar] [CrossRef]
- Mazmanian, S.K.; Liu, C.H.; Tzianabos, A.O.; Kasper, D.L. An Immunomodulatory Molecule of Symbiotic Bacteria Directs Maturation of the Host Immune System. Cell 2005, 122, 107–118. [Google Scholar] [CrossRef] [Green Version]
- Beck, K.E.; Blansfield, J.A.; Tran, K.Q.; Feldman, A.; Hughes, M.S.; Royal, R.E.; Kammula, U.S.; Topalian, S.L.; Sherry, R.M.; Kleiner, D.; et al. Enterocolitis in Patients with Cancer After Antibody Blockade of Cytotoxic T-Lymphocyte–Associated Antigen 4. J. Clin. Oncol. 2006, 24, 2283–2289. [Google Scholar] [CrossRef]
- Derosa, L.; Routy, B.; Fidelle, M.; Iebba, V.; Alla, L.; Pasolli, E.; Segata, N.; Desnoyer, A.; Pietrantonio, F.; Ferrere, G.; et al. Gut Bacteria Composition Drives Primary Resistance to Cancer Immunotherapy in Renal Cell Carcinoma Patients. Eur. Urol. 2020, 78, 195–206. [Google Scholar] [CrossRef] [PubMed]
- Frankel, A.E.; Coughlin, L.A.; Kim, J.; Froehlich, T.W.; Xie, Y.; Frenkel, E.P.; Koh, A.Y. Metagenomic Shotgun Sequencing and Unbiased Metabolomic Profiling Identify Specific Human Gut Microbiota and Metabolites Associated with Immune Checkpoint Therapy Efficacy in Melanoma Patients. Neoplasia 2017, 19, 848–855. [Google Scholar] [CrossRef] [PubMed]
- Zamani, S.; Taslimi, R.; Sarabi, A.; Jasemi, S.; Sechi, L.A.; Feizabadi, M.M. Enterotoxigenic Bacteroides fragilis: A Possible Etiological Candidate for Bacterially-Induced Colorectal Precancerous and Cancerous Lesions. Front. Cell. Infect. Microbiol. 2020, 9, 449. [Google Scholar] [CrossRef]
- Orberg, E.T.; Fan, H.; Tam, A.J.; Dejea, C.M.; Shields, C.E.D.; Wu, S.; Chung, L.; Finard, B.B.; Wu, X.; Fathi, P.; et al. The myeloid immune signature of enterotoxigenic Bacteroides fragilis-induced murine colon tumorigenesis. Mucosal Immunol. 2016, 10, 421–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shi, W.; Shen, L.; Zou, W.; Wang, J.; Yang, J.; Wang, Y.; Liu, B.; Xie, L.; Zhu, J.; Zhang, Z. The Gut Microbiome Is Associated with Therapeutic Responses and Toxicities of Neoadjuvant Chemoradiotherapy in Rectal Cancer Patients—A Pilot Study. Front. Cell. Infect. Microbiol. 2020, 10, 562463. [Google Scholar] [CrossRef] [PubMed]
- Halley, A.; Leonetti, A.; Gregori, A.; Tiseo, M.; Deng, D.M.; Giovannetti, E.; Peters, G.J. The Role of the Microbiome in Cancer and Therapy Efficacy: Focus on Lung Cancer. Anticancer Res. 2020, 40, 4807–4818. [Google Scholar] [CrossRef] [PubMed]
- Tomita, Y.; Ikeda, T.; Sakata, S.; Saruwatari, K.; Sato, R.; Iyama, S.; Jodai, T.; Akaike, K.; Ishizuka, S.; Saeki, S.; et al. Association of Probiotic Clostridium butyricum Therapy with Survival and Response to Immune Checkpoint Blockade in Patients with Lung Cancer. Cancer Immunol. Res. 2020, 8, 1236–1242. [Google Scholar] [CrossRef] [PubMed]
- Yuan, J.; Chen, C.; Cui, J.; Lu, J.; Yan, C.; Wei, X.; Zhao, X.; Li, N.; Li, S.; Xue, G.; et al. Fatty Liver Disease Caused by High-Alcohol-Producing Klebsiella pneumoniae. Cell Metab. 2019, 30, 675–688.e7. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.-C.; Chuang, C.-H.; Miao, Z.-F.; Yip, K.-L.; Liu, C.-J.; Li, L.-H.; Wu, D.-C.; Cheng, T.; Lin, C.-Y.; Wang, J.-Y. Gut microbiota composition in chemotherapy and targeted therapy of patients with metastatic colorectal cancer. Front. Oncol. 2022, 12, 955313. [Google Scholar] [CrossRef]
- Parker, B.J.; Wearsch, P.A.; Veloo, A.C.M.; Rodriguez-Palacios, A. The Genus Alistipes: Gut Bacteria with Emerging Implications to Inflammation, Cancer, and Mental Health. Front. Immunol. 2020, 11, 906. [Google Scholar] [CrossRef]
- Lee, K.; Oh, H.J.; Kang, M.-S.; Kim, S.; Ahn, S.; Kim, M.J.; Kim, S.-W.; Chang, S. Metagenomic analysis of gut microbiome reveals a dynamic change in Alistipes onderdonkii in the preclinical model of pancreatic cancer, suppressing its proliferation. Appl. Microbiol. Biotechnol. 2021, 105, 8343–8358. [Google Scholar] [CrossRef] [PubMed]
- Heshiki, Y.; Vazquez-Uribe, R.; Li, J.; Ni, Y.; Quainoo, S.; Imamovic, L.; Li, J.; Sørensen, M.; Chow, B.K.C.; Weiss, G.J.; et al. Predictable modulation of cancer treatment outcomes by the gut microbiota. Microbiome 2020, 8, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liang, H.; Jo, J.-H.; Zhang, Z.; MacGibeny, M.A.; Han, J.; Proctor, D.M.; Taylor, M.E.; Che, Y.; Juneau, P.; Apolo, A.B.; et al. Predicting cancer immunotherapy response from gut microbiomes using machine learning models. Oncotarget 2022, 13, 876–889. [Google Scholar] [CrossRef] [PubMed]
- Besançon, M.; Gris, T.; Joncas, F.-H.; Picard, V.; Bergeron, A.; Fradet, Y.; Toren, P. Combining Antiandrogens with Immunotherapy for Bladder Cancer Treatment. Eur. Urol. Open Sci. 2022, 43, 35–44. [Google Scholar] [CrossRef] [PubMed]
- Guallar-Garrido, S.; Julián, E. Bacillus Calmette-Guérin (BCG) Therapy for Bladder Cancer: An Update. ImmunoTargets Ther. 2020, 9, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, M.; Khoukaz, L.; Qi, X.; Kimchi, E.T.; Staveley-O’Carroll, K.F.; Li, G. Diet and Gut Microbiota Interaction-Derived Metabolites and Intrahepatic Immune Response in NAFLD Development and Treatment. Biomedicines 2021, 9, 1893. [Google Scholar] [CrossRef] [PubMed]
- Jiao, N.; Baker, S.S.; Chapa-Rodriguez, A.; Liu, W.; Nugent, C.A.; Tsompana, M.; Mastrandrea, L.; Buck, M.J.; Baker, R.D.; Genco, R.J.; et al. Suppressed hepatic bile acid signalling despite elevated production of primary and secondary bile acids in NAFLD. Gut 2018, 67, 1881–1891. [Google Scholar] [CrossRef]
- Yang, C.; Wan, M.; Xu, D.; Pan, D.; Xia, H.; Yang, L.; Sun, G. Flaxseed Powder Attenuates Non-Alcoholic Steatohepatitis via Modulation of Gut Microbiota and Bile Acid Metabolism through Gut–Liver Axis. Int. J. Mol. Sci. 2021, 22, 10858. [Google Scholar] [CrossRef]
- Yamada, S.; Takashina, Y.; Watanabe, M.; Nagamine, R.; Saito, Y.; Kamada, N.; Saito, H. Bile acid metabolism regulated by the gut microbiota promotes non-alcoholic steatohepatitis-associated hepatocellular carcinoma in mice. Oncotarget 2018, 9, 9925–9939. [Google Scholar] [CrossRef] [Green Version]
- Qi, X.; Yang, M.; Stenberg, J.; Dey, R.; Fogwe, L.; Alam, M.S.; Kimchi, E.T.; Staveley-O’Carroll, K.F.; Li, G. Gut microbiota mediated molecular events and therapy in liver diseases. World J. Gastroenterol. 2020, 26, 7603–7618. [Google Scholar] [CrossRef]
- Ma, C.; Han, M.; Heinrich, B.; Fu, Q.; Zhang, Q.; Sandhu, M.; Agdashian, D.; Terabe, M.; Berzofsky, J.A.; Fako, V.; et al. Gut microbiome–mediated bile acid metabolism regulates liver cancer via NKT cells. Science 2018, 360, eaan5931. [Google Scholar] [CrossRef]
Bacteria | Research Objects | Interventions | Identified Mechanism |
---|---|---|---|
Bifidobacterium spp. | Colon cancer murine model | CD47-based immunotherapy | STING pathway and type I IFN production [12] |
Patients with melanoma | Anti-PD1 Abs | Unknown [14] | |
Patients with NSCLC | Anti-PD1 Abs | CD8+ T cells and NKT cells [15] | |
Enterococcus hirae | Sarcoma murine model | Cyclophosphamide | CD4 Th1 cells [2] |
Ruminococcaceae (family) | PDAC mouse model | Gemcitabine | Unknown [27] |
Patients with metastatic melanoma | Anti-PD1 Abs | Unknown [29] | |
Patients with solid tumors | Nivolumab | Effector CD4+ and CD8+ T cells [30] | |
Faecalibacterium spp. | Patients with metastatic melanoma | Anti-PD1 Abs | Effector CD8+ T cell tumor infiltration [29] |
Patients with melanoma | Anti-CTLA4 Abs | Short chain fatty acid (SCFA) & butyrate [30] | |
Patients with NSCLC | Nivolumab | Unknown [34] | |
Oscillibacter spp. | HCC murine model | N/A | Tumor infiltrating Th17 cells [35,36] |
Patients with gastric cancer | N/A | Unknown [37] | |
Prevotella spp. | Patients with NSCLC | Anti-PD1 Abs | CD8+ T cells and NKT cells [15] |
Alistipes spp. | Patients with NSCLC | Anti-PD1 Abs | CD8+ T cells and NKT cells [15] |
Burkholderia spp. | Metastatic melanoma murine model | Anti-CTLA4 Abs | Unknown [40] |
Bacteria | Research Objects | Interventions | Identified Mechanism |
---|---|---|---|
Ruminococcus spp. | Patients with metastatic melanoma | Anti-PD1 Abs | Unknown [14] |
Patients with NSCLC | Anti-PD1 Abs | Unknown [34,40] | |
Gammaproteobacteria(Glass) | Colon cancer murine model | Gemcitabine | Bacterial enzyme cytidine deaminase [55] |
PDAC murine model | Gemcitabine | Gemcitabine metabolism [55] | |
Fusobacterium nucleatum | Patients with CRC | Oxaliplatin | tumor-associated Neutrophils & M2 macrophages [50] |
Escherichia coli | Patients with melanoma | Anti-PD1 Abs | Treg cells tumor infiltration [29] |
Bacteria | Favorable Role | Unfavorable Role | ||||
---|---|---|---|---|---|---|
Research Objects | Interventions | Identified Mechanism | Research Objects | Interventions | Identified Mechanism | |
Akkermansia muciniphila | Patients with NSCLC and RCC | Anti-PD1 Abs | Interleukin 12-dependent [19] | CRC murine model | N/A | Proliferation-associated gene upregulation [58] |
PDAC murine model | Gemcitabine | Unknown [27] | Patients with CRC | N/A | Unknown [59] | |
CRC murine model | Anti-PD1 Abs | Glycerophospholipid Metabolism [60] | ||||
Patients with NSCLC | Nivolumab | Unknown [61] | ||||
Bacteroides spp. | Patients with NSCLC and murine model | Anti-CTLA4 Abs | ZPS production [68,69] | Patients with melanoma | Anti-CTLA4 Abs | Unknown [31] |
Patients with RCC | Anti-PD1 Abs | Unknown [71] | Patients with melanoma | Anti-PD1 Abs | Unknown [29] | |
Patients with metastatic melanoma | Anti-CTLA4 Abs | Unknown [72] | Patients with CRC | N/A | MDSC [74] | |
Clostridium spp. | Patients with NSCLC | ICB | Unknown [77] | Patients with rectal cancer | nCRT | Unknown [75] |
Klebsiella pneumoniae | Patients with metastatic melanoma | Anti-PD1 Abs | Unknown [40] | Patients with HCC | N/A | Endogenous alcohol production [78] |
Alistipes spp. | Patients with NSCLC | Anti-PD1 Abs | CD8+ T cells & NKT cells [15] | Patients with breast cancer | N/A | Unknown [39] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Qi, X.; Liu, Y.; Hussein, S.; Choi, G.; Kimchi, E.T.; Staveley-O’Carroll, K.F.; Li, G. The Species of Gut Bacteria Associated with Antitumor Immunity in Cancer Therapy. Cells 2022, 11, 3684. https://doi.org/10.3390/cells11223684
Qi X, Liu Y, Hussein S, Choi G, Kimchi ET, Staveley-O’Carroll KF, Li G. The Species of Gut Bacteria Associated with Antitumor Immunity in Cancer Therapy. Cells. 2022; 11(22):3684. https://doi.org/10.3390/cells11223684
Chicago/Turabian StyleQi, Xiaoqiang, Yajun Liu, Samira Hussein, Grace Choi, Eric T. Kimchi, Kevin F. Staveley-O’Carroll, and Guangfu Li. 2022. "The Species of Gut Bacteria Associated with Antitumor Immunity in Cancer Therapy" Cells 11, no. 22: 3684. https://doi.org/10.3390/cells11223684
APA StyleQi, X., Liu, Y., Hussein, S., Choi, G., Kimchi, E. T., Staveley-O’Carroll, K. F., & Li, G. (2022). The Species of Gut Bacteria Associated with Antitumor Immunity in Cancer Therapy. Cells, 11(22), 3684. https://doi.org/10.3390/cells11223684