Intratumoral Microbiome: Foe or Friend in Reshaping the Tumor Microenvironment Landscape?
Abstract
:1. Introduction
2. Types of Specialized Microenvironments in the TME
2.1. Immune Microenvironment
2.2. Metabolic Microenvironment
2.3. Hypoxic Microenvironment
2.4. Acidic Microenvironment
2.5. Mechanical Microenvironment
2.6. Innervated Microenvironment
3. Human Microbiome: Origin, Composition, Localization and General Functions in Cancer
3.1. Gut Microbiome
3.2. Skin and Oral Microbiome
3.3. Blood Microbiome
3.4. Intratumoral Microbiome
3.4.1. Intratumoral Fungi and Viruses
3.4.2. Determinants of Intratumoral Microbiome Abundance
3.4.3. Impact of Intratumoral Microbiome-Produced Metabolites on Cancer Properties
4. Crosstalk between the Intratumoral Microbiome and the Tumor Immune Microenvironment (TIME)
4.1. Microbiome as a Driver of TME Immunostimulation
4.1.1. TME Infiltration by Immune Cells
4.1.2. Antitumor Immune Cell Activities
4.1.3. Phenotypic Alterations of Immune Cells
4.2. Microbiome as a Driver of TME Immunosuppression
4.2.1. TME Infiltration by Immune Cells
4.2.2. Tumor-Promoting Function of Immune Cells
4.2.3. Phenotypic Alterations of Immune Cells
5. Crosstalk between the Intratumoral Microbiome and the Acidic TME
6. Crosstalk between the Intratumoral Microbiome and the Hypoxic TME
7. Crosstalk between the Intratumoral Microbiome and the Metabolic TME
8. Crosstalk between the Intratumoral Microbiome and the Mechanical TME
9. Crosstalk between the Intratumoral Microbiome and the Innervated TME
10. Perspectives on Microbiome-Mediated, Multifaceted and Multileveled Effects on the TME Landscape Remodeling—Therapeutic Interventions
11. Challenges and Limitations in Studying Microbiome–TME Interactions
12. Conclusions
Supplementary Materials
Author Contributions
Funding
Data Availability Statement
Conflicts of Interest
References
- Paget, S. The distribution of secondary growths in cancer of the breast. Cancer Metastasis Rev. 1989, 8, 98–101. [Google Scholar] [CrossRef] [PubMed]
- Maman, S.; Witz, I.P. A history of exploring cancer in context. Nat. Rev. Cancer 2018, 18, 359–376. [Google Scholar] [CrossRef] [PubMed]
- de Visser, K.E.; Joyce, J.A. The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth. Cancer Cell 2023, 41, 374–403. [Google Scholar] [CrossRef] [PubMed]
- Reavis, H.D.; Chen, H.I.; Drapkin, R. Tumor innervation: Cancer has some nerve. Trends Cancer 2020, 6, 1059–1067. [Google Scholar] [CrossRef] [PubMed]
- Wagner, M.; Wiig, H. Tumor interstitial fluid formation, characterization, and clinical implications. Front. Oncol. 2015, 5, 115. [Google Scholar] [CrossRef] [PubMed]
- Heldin, C.H.; Rubin, K.; Pietras, K.; Ostman, A. High interstitial fluid pressure—An obstacle in cancer therapy. Nat. Rev. Cancer 2004, 4, 806–813. [Google Scholar] [CrossRef] [PubMed]
- Sun, X.X.; Yu, Q. Intra-tumor heterogeneity of cancer cells and its implications for cancer treatment. Acta Pharmacol. Sin. 2015, 36, 1219–1227. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
- Laplane, L.; Duluc, D.; Larmonier, N.; Pradeu, T.; Bikfalvi, A. The multiple layers of the tumor environment. Trends Cancer 2018, 4, 802–809. [Google Scholar] [CrossRef]
- Jin, M.Z.; Jin, W.L. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct. Target. Ther. 2020, 5, 166. [Google Scholar] [CrossRef]
- Zhu, B.; Wang, X.; Li, L. Human gut microbiome: The second genome of human body. Protein Cell 2010, 1, 718–725. [Google Scholar] [CrossRef] [PubMed]
- Lloyd-Price, J.; Abu-Ali, G.; Huttenhower, C. The healthy human microbiome. Genome Med. 2016, 8, 51. [Google Scholar] [CrossRef] [PubMed]
- Hou, K.; Wu, Z.X.; Chen, X.Y.; Wang, J.Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in health and diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Yang, S.; Dai, H.; Lu, Y.; Li, R.; Gao, C.; Pan, S. Trimethylamine n-oxide promotes cell proliferation and angiogenesis in colorectal cancer. J. Immunol. Res. 2022, 2022, 7043856. [Google Scholar] [CrossRef] [PubMed]
- Xu, Y.; Wang, F.; Mi, K.; Wang, X.; Wang, D.; Zhao, Q.; Wang, J.; Liu, Z.; Zhang, Q.; Liu, Y.; et al. Biglycan regulated colorectal cancer progress by modulating enteric neuron-derived il-10 and abundance of bacteroides thetaiotaomicron. iScience 2023, 26, 107515. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Galeano Niño, 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]
- 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]
- Narunsky-Haziza, L.; Sepich-Poore, G.D.; Livyatan, I.; Asraf, O.; Martino, C.; Nejman, D.; Gavert, N.; Stajich, J.E.; Amit, G.; Gonzalez, A.; et al. Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell 2022, 185, 3789–3806.e17. [Google Scholar] [CrossRef]
- Chuah, S.; Chew, V. High-dimensional immune-profiling in cancer: Implications for immunotherapy. J. Immunother. Cancer 2020, 8, e000363. [Google Scholar] [CrossRef] [PubMed]
- Propper, D.J.; Balkwill, F.R. Harnessing cytokines and chemokines for cancer therapy. Nat. Rev. Clin. Oncol. 2022, 19, 237–253. [Google Scholar] [CrossRef] [PubMed]
- Yang, P.; Peng, Y.; Feng, Y.; Xu, Z.; Feng, P.; Cao, J.; Chen, Y.; Chen, X.; Cao, X.; Yang, Y.; et al. Immune cell-derived extracellular vesicles—New strategies in cancer immunotherapy. Front. Immunol. 2021, 12, 771551. [Google Scholar] [CrossRef] [PubMed]
- Warburg, O.; Wind, F.; Negelein, E. The Metabolism of Tumors in the Body. J. Gen. Physiol. 1927, 8, 519–530. [Google Scholar] [CrossRef] [PubMed]
- Gong, T.; Zheng, C.; Ou, X.; Zheng, J.; Yu, J.; Chen, S.; Duan, Y.; Liu, W. Glutamine metabolism in cancers: Targeting the oxidative homeostasis. Front. Oncol. 2022, 12, 994672. [Google Scholar] [CrossRef] [PubMed]
- Mao, X.Y.; Jin, M.Z.; Chen, J.F.; Zhou, H.H.; Jin, W.L. Live or let die: Neuroprotective and anti-cancer effects of nutraceutical antioxidants. Pharmacol. Ther. 2018, 183, 137–151. [Google Scholar] [CrossRef] [PubMed]
- Cairns, R.A.; Harris, I.S.; Mak, T.W. Regulation of cancer cell metabolism. Nat. Rev. Cancer 2011, 11, 85–95. [Google Scholar] [CrossRef] [PubMed]
- Schafer, F.Q.; Buettner, G.R. Redox environment of the cell as viewed through the redox state of the glutathione disulfide/glutathione couple. Free Radic. Biol. Med. 2001, 30, 1191–1212. [Google Scholar] [CrossRef] [PubMed]
- Trachootham, D.; Alexandre, J.; Huang, P. Targeting cancer cells by ros-mediated mechanisms: A radical therapeutic approach? Nat. Rev. Drug Discov. 2009, 8, 579–591. [Google Scholar] [CrossRef]
- Sena, L.A.; Chandel, N.S. Physiological roles of mitochondrial reactive oxygen species. Mol. Cell 2012, 48, 158–167. [Google Scholar] [CrossRef]
- Sies, H. Hydrogen peroxide as a central redox signaling molecule in physiological oxidative stress: Oxidative eustress. Redox Biol. 2017, 11, 613–619. [Google Scholar] [CrossRef] [PubMed]
- Harris, I.S.; DeNicola, G.M. The complex interplay between antioxidants and ros in cancer. Trends Cell Biol. 2020, 30, 440–451. [Google Scholar] [CrossRef] [PubMed]
- Harris, I.S.; Treloar, A.E.; Inoue, S.; Sasaki, M.; Gorrini, C.; Lee, K.C.; Yung, K.Y.; Brenner, D.; Knobbe-Thomsen, C.B.; Cox, M.A.; et al. Glutathione and thioredoxin antioxidant pathways synergize to drive cancer initiation and progression. Cancer Cell 2015, 27, 211–222. [Google Scholar] [CrossRef] [PubMed]
- Schaaf, M.B.; Garg, A.D.; Agostinis, P. Defining the role of the tumor vasculature in antitumor immunity and immunotherapy. Cell Death Dis. 2018, 9, 115. [Google Scholar] [CrossRef] [PubMed]
- Schito, L.; Semenza, G.L. Hypoxia-Inducible Factors: Master Regulators of Cancer Progression. Trends Cancer 2016, 2, 758–770. [Google Scholar] [CrossRef] [PubMed]
- Qiu, G.Z.; Jin, M.Z.; Dai, J.X.; Sun, W.; Feng, J.H.; Jin, W.L. Reprogramming of the tumor in the hypoxic niche: The emerging concept and associated therapeutic strategies. Trends Pharmacol. Sci. 2017, 38, 669–686. [Google Scholar] [CrossRef] [PubMed]
- Webb, B.A.; Chimenti, M.; Jacobson, M.P.; Barber, D.L. Dysregulated pH: A perfect storm for cancer progression. Nat. Rev. Cancer 2011, 11, 671–677. [Google Scholar] [CrossRef] [PubMed]
- Corbet, C.; Feron, O. Tumour acidosis: From the passenger to the driver’s seat. Nat. Rev. Cancer 2017, 17, 577–593. [Google Scholar] [CrossRef]
- Sonveaux, P.; Vegran, F.; Schroeder, T.; Wergin, M.C.; Verrax, J.; Rabbani, Z.N.; De Saedeleer, C.J.; Kennedy, K.M.; Diepart, C.; Jordan, B.F.; et al. Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J. Clin. Investig. 2008, 118, 3930–3942. [Google Scholar] [CrossRef]
- Dimmer, K.S.; Friedrich, B.; Lang, F.; Deitmer, J.W.; Broer, S. The low-affinity monocarboxylate transporter mct4 is adapted to the export of lactate in highly glycolytic cells. Biochem. J. 2000, 350 Pt 1, 219–227. [Google Scholar] [CrossRef]
- Reshkin, S.J.; Cardone, R.A.; Harguindey, S. Na+-H+ exchanger, pH regulation and cancer. Recent Pat. Anti-Cancer Drug Discov. 2013, 8, 85–99. [Google Scholar] [CrossRef]
- Mboge, M.Y.; McKenna, R.; Frost, S.C. Advances in Anti-Cancer Drug Development Targeting Carbonic Anhydrase IX and XII. Top. Anticancer Res. 2015, 5, 3–42. [Google Scholar]
- Chiche, J.; Brahimi-Horn, M.C.; Pouyssegur, J. Tumour hypoxia induces a metabolic shift causing acidosis: A common feature in cancer. J. Cell. Mol. Med. 2010, 14, 771–794. [Google Scholar] [CrossRef]
- Valkenburg, K.C.; de Groot, A.E.; Pienta, K.J. Targeting the tumour stroma to improve cancer therapy. Nat. Rev. Clin. Oncol. 2018, 15, 366–381. [Google Scholar] [CrossRef]
- Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 2020, 11, 5120. [Google Scholar] [CrossRef] [PubMed]
- Henke, E.; Nandigama, R.; Ergun, S. Extracellular matrix in the tumor microenvironment and its impact on cancer therapy. Front. Mol. Biosci. 2019, 6, 160. [Google Scholar] [CrossRef]
- Cooper, J.; Giancotti, F.G. Integrin signaling in cancer: Mechanotransduction, stemness, epithelial plasticity, and therapeutic resistance. Cancer Cell 2019, 35, 347–367. [Google Scholar] [CrossRef]
- Canel, M.; Serrels, A.; Frame, M.C.; Brunton, V.G. E-cadherin-integrin crosstalk in cancer invasion and metastasis. J. Cell Sci. 2013, 126, 393–401. [Google Scholar] [CrossRef]
- Pundavela, J.; Roselli, S.; Faulkner, S.; Attia, J.; Scott, R.J.; Thorne, R.F.; Forbes, J.F.; Bradshaw, R.A.; Walker, M.M.; Jobling, P.; et al. Nerve fibers infiltrate the tumor microenvironment and are associated with nerve growth factor production and lymph node invasion in breast cancer. Mol. Oncol. 2015, 9, 1626–1635. [Google Scholar] [CrossRef] [PubMed]
- Ferdoushi, A.; Griffin, N.; Marsland, M.; Xu, X.; Faulkner, S.; Gao, F.; Liu, H.; King, S.J.; Denham, J.W.; van Helden, D.F.; et al. Tumor innervation and clinical outcome in pancreatic cancer. Sci. Rep. 2021, 11, 7390. [Google Scholar] [CrossRef]
- Wang, W.; Li, L.; Chen, N.; Niu, C.; Li, Z.; Hu, J.; Cui, J. Nerves in the tumor microenvironment: Origin and effects. Front. Cell Dev. Biol. 2020, 8, 601738. [Google Scholar] [CrossRef] [PubMed]
- Hernandez, S.; Serrano, A.G.; Solis Soto, L.M. The role of nerve fibers IN the tumor immune microenvironment of solid tumors. Adv. Biol. 2022, 6, e2200046. [Google Scholar] [CrossRef]
- Li, X.; Ma, G.; Ma, Q.; Li, W.; Liu, J.; Han, L.; Duan, W.; Xu, Q.; Liu, H.; Wang, Z.; et al. Neurotransmitter substance p mediates pancreatic cancer perineural invasion via nk-1r in cancer cells. Mol. Cancer Res. MCR 2013, 11, 294–302. [Google Scholar] [CrossRef] [PubMed]
- Yu, A.I.; Zhao, L.; Eaton, K.A.; Ho, S.; Chen, J.; Poe, S.; Becker, J.; Gonzalez, A.; McKinstry, D.; Hasso, M.; et al. Gut microbiota Modulate cd8 t cell responses to influence colitis-associated tumorigenesis. Cell Rep. 2020, 31, 107471. [Google Scholar] [CrossRef] [PubMed]
- Zegarra Ruiz, D.F.; Kim, D.V.; Norwood, K.; Saldana-Morales, F.B.; Kim, M.; Ng, C.; Callaghan, R.; Uddin, M.; Chang, L.C.; Longman, R.S.; et al. Microbiota manipulation to increase macrophage IL-10 improves colitis and limits colitis-associated colorectal cancer. Gut Microbes 2022, 14, 2119054. [Google Scholar] [CrossRef] [PubMed]
- Overacre-Delgoffe, A.E.; Bumgarner, H.J.; Cillo, A.R.; Burr, A.H.P.; Tometich, J.T.; Bhattacharjee, A.; Bruno, T.C.; Vignali, D.A.A.; Hand, T.W. Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity 2021, 54, 2812–2824.e4. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Tang, W.; Putluri, V.; Ambati, C.R.; Dorsey, T.H.; Putluri, N.; Ambs, S. Liver- and microbiome-derived bile acids accumulate in human breast tumors and inhibit growth and improve patient survival. Clin. Cancer Res. 2019, 25, 5972–5983. [Google Scholar] [CrossRef] [PubMed]
- Muller, P.A.; Matheis, F.; Schneeberger, M.; Kerner, Z.; Jove, V.; Mucida, D. Microbiota-modulated cart(+) enteric neurons autonomously regulate blood glucose. Science 2020, 370, 314–321. [Google Scholar] [CrossRef]
- Chen, Y.E.; Bousbaine, D.; Veinbachs, A.; Atabakhsh, K.; Dimas, A.; Yu, V.K.; Zhao, A.; Enright, N.J.; Nagashima, K.; Belkaid, Y.; et al. Engineered skin bacteria induce antitumor t cell responses against melanoma. Science 2023, 380, 203–210. [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]
- Tan, Q.; Ma, X.; Yang, B.; Liu, Y.; Xie, Y.; Wang, X.; Yuan, W.; Ma, J. Periodontitis pathogen Porphyromonas gingivalis promotes pancreatic tumorigenesis via neutrophil elastase from tumor-associated neutrophils. Gut Microbes 2022, 14, 2073785. [Google Scholar] [CrossRef]
- Castillo, D.J.; Rifkin, R.F.; Cowan, D.A.; Potgieter, M. The Healthy Human Blood Microbiome: Fact or Fiction? Front. Cell. Infect. Microbiol. 2019, 9, 148. [Google Scholar] [CrossRef] [PubMed]
- Tan, C.C.S.; Ko, K.K.K.; Chen, H.; Liu, J.; Loh, M.; Consortium, S.G.K.H.; Chia, M.; Nagarajan, N. No evidence for a common blood microbiome based on a population study of 9770 healthy humans. Nat. Microbiol. 2023, 8, 973–985. [Google Scholar] [CrossRef] [PubMed]
- Woerner, J.; Huang, Y.; Hutter, S.; Gurnari, C.; Sanchez, J.M.H.; Wang, J.; Huang, Y.; Schnabel, D.; Aaby, M.; Xu, W.; et al. Circulating microbial content in myeloid malignancy patients is associated with disease subtypes and patient outcomes. Nat. Commun. 2022, 13, 1038. [Google Scholar] [CrossRef] [PubMed]
- Yang, D.; Wang, X.; Zhou, X.; Zhao, J.; Yang, H.; Wang, S.; Morse, M.A.; Wu, J.; Yuan, Y.; Li, S.; et al. Blood microbiota diversity determines response of advanced colorectal cancer to chemotherapy combined with adoptive T cell immunotherapy. Oncoimmunology 2021, 10, 1976953. [Google Scholar] [CrossRef]
- Gye, W.E. The Ætiology of Malignant New Growths. Lancet 1925, 206, 109–117. [Google Scholar] [CrossRef]
- Coley, W.B. The treatment of malignant tumors by repeated inoculations of erysipelas: With a report of ten original cases. Clin. Orthop. Relat Res. 1893, 105, 487. [Google Scholar]
- Zhu, Z.; Cai, J.; Hou, W.; Xu, K.; Wu, X.; Song, Y.; Bai, C.; Mo, Y.Y.; Zhang, Z. Microbiome and spatially resolved metabolomics analysis reveal the anticancer role of gut akkermansia muciniphila by crosstalk with intratumoral microbiota and reprogramming tumoral metabolism in mice. Gut Microbes 2023, 15, 2166700. [Google Scholar] [CrossRef]
- Bertocchi, A.; Carloni, S.; Ravenda, P.S.; Bertalot, G.; Spadoni, I.; Lo Cascio, A.; Gandini, S.; Lizier, M.; Braga, D.; Asnicar, F.; et al. Gut vascular barrier impairment leads to intestinal bacteria dissemination and colorectal cancer metastasis to liver. Cancer Cell 2021, 39, 708–724.e11. [Google Scholar] [CrossRef]
- Abed, J.; Emgard, 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]
- 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]
- Shirai, H.; Ito, C.; Tsukada, K. pH-taxis drives aerobic bacteria in duodenum to migrate into the pancreas with tumors. Sci. Rep. 2022, 12, 1783. [Google Scholar] [CrossRef]
- Wong-Rolle, A.; Dong, Q.; Zhu, Y.; Divakar, P.; Hor, J.L.; Kedei, N.; Wong, M.; Tillo, D.; Conner, E.A.; Rajan, A.; et al. Spatial meta-transcriptomics reveal associations of intratumor bacteria burden with lung cancer cells showing a distinct oncogenic signature. J. Immunother. Cancer 2022, 10, e004698. [Google Scholar] [CrossRef] [PubMed]
- Fu, A.; Yao, B.; Dong, T.; Chen, Y.; Yao, J.; Liu, Y.; Li, H.; Bai, H.; Liu, X.; Zhang, Y.; et al. Tumor-resident intracellular microbiota promotes metastatic colonization in breast cancer. Cell 2022, 185, 1356–1372.e26. [Google Scholar] [CrossRef]
- Wang, J.; Li, X.; Wu, X.; Wang, Z.; Zhang, C.; Cao, G.; Liu, K.; Yan, T. Uncovering the microbiota in renal cell carcinoma tissue using 16s rrna gene sequencing. J. Cancer Res. Clin. Oncol. 2021, 147, 481–491. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Wang, Y.; Wang, J. A comprehensive analysis of intratumor microbiome in head and neck squamous cell carcinoma. Eur. Arch. Oto Rhino Laryngol. 2022, 279, 4127–4136. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Guo, H.; Gao, X.; Wang, J. The intratumor microbiota signatures associate with subtype, tumor stage, and survival status of esophageal carcinoma. Front. Oncol. 2021, 11, 754788. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Wang, Y.; Li, Z.; Gao, X.; Huang, D. Global analysis of microbiota signatures in four major types of gastrointestinal cancer. Front. Oncol. 2021, 11, 685641. [Google Scholar] [CrossRef]
- Liu, W.; Zhang, X.; Xu, H.; Li, S.; Lau, H.C.; Chen, Q.; Zhang, B.; Zhao, L.; Chen, H.; Sung, J.J.; et al. Microbial Community Heterogeneity within Colorectal Neoplasia and its Correlation with Colorectal Carcinogenesis. Gastroenterology 2021, 160, 2395–2408. [Google Scholar] [CrossRef]
- Stritzker, J.; Weibel, S.; Seubert, C.; Gotz, A.; Tresch, A.; van Rooijen, N.; Oelschlaeger, T.A.; Hill, P.J.; Gentschev, I.; Szalay, A.A. Enterobacterial tumor colonization in mice depends on bacterial metabolism and macrophages but is independent of chemotaxis and motility. Int. J. Med. Microbiol. 2010, 300, 449–456. [Google Scholar] [CrossRef] [PubMed]
- Chai, X.; Wang, J.; Li, H.; Gao, C.; Li, S.; Wei, C.; Huang, J.; Tian, Y.; Yuan, J.; Lu, J.; et al. Intratumor microbiome features reveal antitumor potentials of intrahepatic cholangiocarcinoma. Gut Microbes 2023, 15, 2156255. [Google Scholar] [CrossRef]
- Neuzillet, C.; Marchais, M.; Vacher, S.; Hilmi, M.; Schnitzler, A.; Meseure, D.; Leclere, R.; Lecerf, C.; Dubot, C.; Jeannot, E.; et al. Prognostic value of intratumoral fusobacterium nucleatum and association with immune-related gene expression in oral squamous cell carcinoma patients. Sci. Rep. 2021, 11, 7870. [Google Scholar] [CrossRef] [PubMed]
- Kalaora, S.; Nagler, A.; Nejman, D.; Alon, M.; Barbolin, C.; Barnea, E.; Ketelaars, S.L.C.; Cheng, K.; Vervier, K.; Shental, N.; et al. Identification of bacteria-derived hla-bound peptides in melanoma. Nature 2021, 592, 138–143. [Google Scholar] [CrossRef] [PubMed]
- Huang, J.H.; Wang, J.; Chai, X.Q.; Li, Z.C.; Jiang, Y.H.; Li, J.; Liu, X.; Fan, J.; Cai, J.B.; Liu, F. The intratumoral bacterial metataxonomic signature of hepatocellular carcinoma. Microbiol. Spectr. 2022, 10, e0098322. [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] [PubMed]
- Bullman, S.; Pedamallu, C.S.; Sicinska, E.; Clancy, T.E.; Zhang, X.; Cai, D.; Neuberg, D.; Huang, K.; Guevara, F.; Nelson, T.; et al. Analysis of fusobacterium persistence and antibiotic response in colorectal cancer. Science 2017, 358, 1443–1448. [Google Scholar] [CrossRef] [PubMed]
- Shiao, S.L.; Kershaw, K.M.; Limon, J.J.; You, S.; Yoon, J.; Ko, E.Y.; Guarnerio, J.; Potdar, A.A.; McGovern, D.P.B.; Bose, S.; et al. Commensal bacteria and fungi differentially regulate tumor responses to radiation therapy. Cancer Cell 2021, 39, 1202–1213.e6. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Bose, D.; Banerjee, S.; Singh, R.K.; Wise, L.M.; Robertson, E.S. Vascular endothelial growth factor encoded by parapoxviruses can regulate metabolism and survival of triple negative breast cancer cells. Cell Death Dis. 2020, 11, 996. [Google Scholar] [CrossRef]
- Dhakal, A.; Upadhyay, R.; Wheeler, C.; Hoyd, R.; Karivedu, V.; Gamez, M.E.; Valentin, S.; Vanputten, M.; Bhateja, P.; Bonomi, M.; et al. Association between tumor microbiome and hypoxia across anatomic subsites of head and neck cancers. Int. J. Mol. Sci. 2022, 23, 15531. [Google Scholar] [CrossRef] [PubMed]
- Perry, L.M.; Cruz, S.M.; Kleber, K.T.; Judge, S.J.; Darrow, M.A.; Jones, L.B.; Basmaci, U.N.; Joshi, N.; Settles, M.L.; Durbin-Johnson, B.P.; et al. Human soft tissue sarcomas harbor an intratumoral viral microbiome which is linked with natural killer cell infiltrate and prognosis. J. Immunother. Cancer 2023, 11, e004285. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.P.; Jiang, J.K.; Chen, C.Y.; Chou, T.Y.; Chen, Y.C.; Chang, Y.T.; Lin, S.F.; Chan, C.H.; Yang, C.Y.; Lin, C.H.; et al. Human cytomegalovirus preferentially infects the neoplastic epithelium of colorectal cancer: A quantitative and histological analysis. J. Clin. Virol. 2012, 54, 240–244. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Qi, M.; He, K.; Liu, H.; Yan, W.; Zhao, L.; Jia, Y.; He, L.; Lv, C.; Zhang, M.; et al. Neospora caninum inhibits tumor development by activating the immune response and destroying tumor cells in a b16f10 melanoma model. Parasites Vectors 2022, 15, 332. [Google Scholar] [CrossRef] [PubMed]
- Luo, M.; Liu, Y.; Hermida, L.C.; Gertz, E.M.; Zhang, Z.; Li, Q.; Diao, L.; Ruppin, E.; Han, L. Race is a key determinant of the human intratumor microbiome. Cancer Cell 2022, 40, 901–902. [Google Scholar] [CrossRef] [PubMed]
- Chiba, A.; Bawaneh, A.; Velazquez, C.; Clear, K.Y.J.; Wilson, A.S.; Howard-McNatt, M.; Levine, E.A.; Levi-Polyachenko, N.; Yates-Alston, S.A.; Diggle, S.P.; et al. Neoadjuvant chemotherapy shifts breast tumor microbiota populations to regulate drug responsiveness and the development of metastasis. Mol. Cancer Res. 2020, 18, 130–139. [Google Scholar] [CrossRef] [PubMed]
- Guo, W.; Zhang, Y.; Guo, S.; Mei, Z.; Liao, H.; Dong, H.; Wu, K.; Ye, H.; Zhang, Y.; Zhu, Y.; et al. Tumor microbiome contributes to an aggressive phenotype in the basal-like subtype of pancreatic cancer. Commun. Biol. 2021, 4, 1019. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.; An, H.; He, Y.; Fu, G.; Jiang, Z. Comprehensive analysis of microbiota signature across 32 cancer types. Front. Oncol. 2023, 13, 1127225. [Google Scholar] [CrossRef] [PubMed]
- Gottschalk, G. Bacterial Metabolism, 2nd ed.; Springer: New York, NY, USA, 1986. [Google Scholar]
- Huang, Y.; Zhu, N.; Zheng, X.; Liu, Y.; Lu, H.; Yin, X.; Hao, H.; Tan, Y.; Wang, D.; Hu, H.; et al. Intratumor microbiome analysis identifies positive association between megasphaera and survival of chinese patients with pancreatic ductal adenocarcinomas. Front. Immunol. 2022, 13, 785422. [Google Scholar] [CrossRef] [PubMed]
- Tsai, C.K.; Lin, C.Y.; Kang, C.J.; Liao, C.T.; Wang, W.L.; Chiang, M.H.; Yen, T.C.; Lin, G. Nuclear magnetic resonance metabolomics biomarkers for identifying high risk patients with extranodal extension in oral squamous cell carcinoma. J. Clin. Med. 2020, 9, 951. [Google Scholar] [CrossRef]
- Yu, T.; Ji, L.; Lou, L.; Ye, S.; Fang, X.; Li, C.; Jiang, F.; Gao, H.; Lou, Y.; Li, X. Fusobacterium nucleatum affects cell apoptosis by regulating intestinal flora and metabolites to promote the development of colorectal cancer. Front. Microbiol. 2022, 13, 841157. [Google Scholar] [CrossRef] [PubMed]
- Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-chain fatty-acid-producing bacteria: Key components of the human gut microbiota. Nutrients 2023, 15, 2211. [Google Scholar] [CrossRef] [PubMed]
- Archer, S.Y.; Johnson, J.; Kim, H.J.; Ma, Q.; Mou, H.; Daesety, V.; Meng, S.; Hodin, R.A. The histone deacetylase inhibitor butyrate downregulates cyclin b1 gene expression via a p21/waf-1-dependent mechanism in human colon cancer cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2005, 289, G696–G703. [Google Scholar] [CrossRef] [PubMed]
- Filippone, A.; Casili, G.; Scuderi, S.A.; Mannino, D.; Lanza, M.; Campolo, M.; Paterniti, I.; Capra, A.P.; Colarossi, C.; Bonasera, A.; et al. Sodium propionate contributes to tumor cell growth inhibition through ppar-gamma signaling. Cancers 2022, 15, 217. [Google Scholar] [CrossRef]
- Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [Google Scholar] [CrossRef]
- Yang, X.; Zhang, Z.; Shen, X.; Xu, J.; Weng, Y.; Wang, W.; Xue, J. Clostridium butyricum and its metabolite butyrate promote ferroptosis susceptibility in pancreatic ductal adenocarcinoma. Cell. Oncol. 2023, 46, 1645–1658. [Google Scholar] [CrossRef] [PubMed]
- Huang, Q.; Wei, X.; Li, W.; Ma, Y.; Chen, G.; Zhao, L.; Jiang, Y.; Xie, S.; Chen, Q.; Chen, T. Endogenous propionibacterium acnes promotes ovarian cancer progression via regulating hedgehog signalling pathway. Cancers 2022, 14, 5178. [Google Scholar] [CrossRef]
- Shu, M.; Wang, Y.; Yu, J.; Kuo, S.; Coda, A.; Jiang, Y.; Gallo, R.L.; Huang, C.M. Fermentation of Propionibacterium acnes, a commensal bacterium in the human skin microbiome, as skin probiotics against methicillin-resistant Staphylococcus aureus. PLoS ONE 2013, 8, e55380. [Google Scholar] [CrossRef]
- Bender, M.J.; McPherson, A.C.; Phelps, C.M.; Pandey, S.P.; Laughlin, C.R.; Shapira, J.H.; Medina Sanchez, L.; Rana, M.; Richie, T.G.; Mims, T.S.; et al. Dietary tryptophan metabolite released by intratumoral Lactobacillus reuteri facilitates immune checkpoint inhibitor treatment. Cell 2023, 186, 846–1862. [Google Scholar] [CrossRef]
- Tintelnot, J.; Xu, Y.; Lesker, T.R.; Schonlein, M.; Konczalla, L.; Giannou, A.D.; Pelczar, P.; Kylies, D.; Puelles, V.G.; Bielecka, A.A.; et al. Microbiota-derived 3-iaa influences chemotherapy efficacy in pancreatic cancer. Nature 2023, 615, 168–174. [Google Scholar] [CrossRef]
- Kitada, Y.; Muramatsu, K.; Toju, H.; Kibe, R.; Benno, Y.; Kurihara, S.; Matsumoto, M. Bioactive polyamine production by a novel hybrid system comprising multiple indigenous gut bacterial strategies. Sci. Adv. 2018, 4, eaat0062. [Google Scholar] [CrossRef]
- Luu, K.; Ye, J.Y.; Lagishetty, V.; Liang, F.; Hauer, M.; Sedighian, F.; Kwaan, M.R.; Kazanjian, K.K.; Hecht, J.R.; Lin, A.Y.; et al. Fecal and tissue microbiota are associated with tumor t-cell infiltration and mesenteric lymph node involvement in colorectal cancer. Nutrients 2023, 15, 316. [Google Scholar] [CrossRef]
- Zhu, G.; Su, H.; Johnson, C.H.; Khan, S.A.; Kluger, H.; Lu, L. Intratumour microbiome associated with the infiltration of cytotoxic cd8+ t cells and patient survival in cutaneous melanoma. Eur. J. Cancer 2021, 151, 25–34. [Google Scholar] [CrossRef]
- Wu, H.; Leng, X.; Liu, Q.; Mao, T.; Jiang, T.; Liu, Y.; Li, F.; Cao, C.; Fan, J.; Chen, L.; et al. Intratumoral microbiota composition regulates chemoimmunotherapy response in esophageal squamous cell carcinoma. Cancer Res. 2023, 83, 3131–3144. [Google Scholar] [CrossRef]
- Janku, F.; Zhang, H.H.; Pezeshki, A.; Goel, S.; Murthy, R.; Wang-Gillam, A.; Shepard, D.R.; Helgason, T.; Masters, T.; Hong, D.S.; et al. Intratumoral Injection of Clostridium novyi-nt spores in patients with treatment-refractory advanced solid tumors. Clin. Cancer Res. 2021, 27, 96–106. [Google Scholar] [CrossRef]
- Brown, M.C.; Mosaheb, M.M.; Mohme, M.; McKay, Z.P.; Holl, E.K.; Kastan, J.P.; Yang, Y.; Beasley, G.M.; Hwang, E.S.; Ashley, D.M.; et al. Viral infection of cells within the tumor microenvironment mediates antitumor immunotherapy via selective tbk1-irf3 signaling. Nat. Commun. 2021, 12, 1858. [Google Scholar] [CrossRef]
- Naghavian, R.; Faigle, W.; Oldrati, P.; Wang, J.; Toussaint, N.C.; Qiu, Y.; Medici, G.; Wacker, M.; Freudenmann, L.K.; Bonte, P.E.; et al. Microbial peptides activate tumour-infiltrating lymphocytes in glioblastoma. Nature 2023, 617, 807–817. [Google Scholar] [CrossRef]
- 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]
- Lam, K.C.; Araya, R.E.; Huang, A.; Chen, Q.; Di Modica, M.; Rodrigues, R.R.; Lopes, A.; Johnson, S.B.; Schwarz, B.; Bohrnsen, E.; et al. Microbiota triggers sting-type I ifn-dependent monocyte reprogramming of the tumor microenvironment. Cell 2021, 184, 5338–5356.e21. [Google Scholar] [CrossRef] [PubMed]
- Hu, C.; Xu, B.; Wang, X.; Wan, W.H.; Lu, J.; Kong, D.; Jin, Y.; You, W.; Sun, H.; Mu, X.; et al. Gut microbiota-derived short-chain fatty acids regulate group 3 innate lymphoid cells in hcc. Hepatology 2023, 77, 48–64. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Wang, W.; Xu, H.; Ye, Q.; Tao, F.; Wheeldon, I.; Yuan, A.; Hu, Y.; Wu, J. Systemic immune responses to irradiated tumours via the transport of antigens to the tumour periphery by injected flagellate bacteria. Nat. Biomed. Eng. 2022, 6, 44–53. [Google Scholar] [CrossRef] [PubMed]
- Geng, Z.; Cao, Z.; Liu, R.; Liu, K.; Liu, J.; Tan, W. Aptamer-assisted tumor localization of bacteria for enhanced biotherapy. Nat. Commun. 2021, 12, 6584. [Google Scholar] [CrossRef] [PubMed]
- Wang, L.; Cao, Z.; Zhang, M.; Lin, S.; Liu, J. Spatiotemporally controllable distribution of combination therapeutics in solid tumors by dually modified bacteria. Adv. Mater. 2022, 34, e2106669. [Google Scholar] [CrossRef] [PubMed]
- Murphy, C.; Rettedal, E.; Lehouritis, P.; Devoy, C.; Tangney, M. Intratumoural production of tnfalpha by bacteria mediates cancer therapy. PLoS ONE 2017, 12, e0180034. [Google Scholar] [CrossRef]
- Savage, T.M.; Vincent, R.L.; Rae, S.S.; Huang, L.H.; Ahn, A.; Pu, K.; Li, F.; de los Santos-Alexis, K.; Coker, C.; Danino, T.; et al. Chemokines expressed by engineered bacteria recruit and orchestrate antitumor immunity. Sci. Adv. 2023, 9, eadc9436. [Google Scholar] [CrossRef] [PubMed]
- Choi, Y.; Lichterman, J.N.; Coughlin, L.A.; Poulides, N.; Li, W.; Del Valle, P.; Palmer, S.N.; Gan, S.; Kim, J.; Zhan, X.; et al. Immune checkpoint blockade induces gut microbiota translocation that augments extraintestinal antitumor immunity. Sci. Immunol. 2023, 8, eabo2003. [Google Scholar] [CrossRef] [PubMed]
- Burkhardt, F.; Denise, B.L.; Jillian, M.R.B.; Ian, B.J.; Feargal, J.R.; Marcus, J.C.; Micheal, O.; Fergus, S.; Paul, W.O. Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut 2017, 66, 633. [Google Scholar] [CrossRef]
- Sheng, D.; Yue, K.; Li, H.; Zhao, L.; Zhao, G.; Jin, C.; Zhang, L. The interaction between intratumoral microbiome and immunity is related to the prognosis of ovarian cancer. Microbiol. Spectr. 2023, 11, e0354922. [Google Scholar] [CrossRef]
- Mirji, G.; Worth, A.; Bhat, S.A.; El Sayed, M.; Kannan, T.; Goldman, A.R.; Tang, H.Y.; Liu, Q.; Auslander, N.; Dang, C.V.; et al. The microbiome-derived metabolite tmao drives immune activation and boosts responses to immune checkpoint blockade in pancreatic cancer. Sci. Immunol. 2022, 7, eabn0704. [Google Scholar] [CrossRef]
- Le Tourneau, C.; Delord, J.P.; Goncalves, A.; Gavoille, C.; Dubot, C.; Isambert, N.; Campone, M.; Tredan, O.; Massiani, M.A.; Mauborgne, C.; et al. Molecularly targeted therapy based on tumour molecular profiling versus conventional therapy for advanced cancer (SHIVA): A multicentre, open-label, proof-of-concept, randomised, controlled phase 2 trial. Lancet Oncol. 2015, 16, 1324–1334. [Google Scholar] [CrossRef]
- Hilmi, M.; Kamal, M.; Vacher, S.; Dupain, C.; Ibadioune, S.; Halladjian, M.; Sablin, M.P.; Marret, G.; Ajgal, Z.C.; Nijnikoff, M.; et al. Intratumoral microbiome is driven by metastatic site and associated with immune histopathological parameters: An ancillary study of the shiva clinical trial. Eur. J. Cancer 2023, 183, 152–161. [Google Scholar] [CrossRef]
- Liu, Z.; Zhang, X.; Zhang, H.; Zhang, H.; Yi, Z.; Zhang, Q.; Liu, Q.; Liu, X. Multi-omics analysis reveals intratumor microbes as immunomodulators in colorectal cancer. Microbiol. Spectr. 2023, 11, e0503822. [Google Scholar] [CrossRef]
- Mima, K.; Sukawa, Y.; Nishihara, R.; Qian, Z.R.; Yamauchi, M.; Inamura, K.; Kim, S.A.; Masuda, A.; Nowak, J.A.; Nosho, K.; et al. Fusobacterium nucleatum and t cells in colorectal carcinoma. JAMA Oncol. 2015, 1, 653–661. [Google Scholar] [CrossRef]
- Lee, J.A.; Yoo, S.Y.; Oh, H.J.; Jeong, S.; Cho, N.Y.; Kang, G.H.; Kim, J.H. Differential immune microenvironmental features of microsatellite-unstable colorectal cancers according to fusobacterium nucleatum status. Cancer Immunol. Immunother. CII 2021, 70, 47–59. [Google Scholar] [CrossRef]
- Triner, D.; Devenport, S.N.; Ramakrishnan, S.K.; Ma, X.; Frieler, R.A.; Greenson, J.K.; Inohara, N.; Nunez, G.; Colacino, J.A.; Mortensen, R.M.; et al. Neutrophils restrict tumor-associated microbiota to reduce growth and invasion of colon tumors in mice. Gastroenterology 2019, 156, 1467–1482. [Google Scholar] [CrossRef]
- Noguti, J.; Chan, A.A.; Bandera, B.; Brislawn, C.J.; Protic, M.; Sim, M.S.; Jansson, J.K.; Bilchik, A.J.; Lee, D.J. Both the intratumoral immune and microbial microenvironment are linked to recurrence in human colon cancer: Results from a prospective, multicenter nodal ultrastaging trial. Oncotarget 2018, 9, 23564–23576. [Google Scholar] [CrossRef]
- Wang, X.; Jia, Y.; Wen, L.; Mu, W.; Wu, X.; Liu, T.; Liu, X.; Fang, J.; Luan, Y.; Chen, P.; et al. Porphyromonas gingivalis promotes colorectal carcinoma by activating the hematopoietic nlrp3 inflammasome. Cancer Res. 2021, 81, 2745–2759. [Google Scholar] [CrossRef]
- Chakladar, J.; Kuo, S.Z.; Castaneda, G.; Li, W.T.; Gnanasekar, A.; Yu, M.A.; Chang, E.Y.; Wang, X.Q.; Ongkeko, W.M. The pancreatic microbiome is associated with carcinogenesis and worse prognosis in males and smokers. Cancers 2020, 12, 2672. [Google Scholar] [CrossRef] [PubMed]
- Gnanasekar, A.; Castaneda, G.; Iyangar, A.; Magesh, S.; Perez, D.; Chakladar, J.; Li, W.T.; Bouvet, M.; Chang, E.Y.; Ongkeko, W.M. The intratumor microbiome predicts prognosis across gender and subtypes in papillary thyroid carcinoma. Comput. Struct. Biotechnol. J. 2021, 19, 1986–1997. [Google Scholar] [CrossRef] [PubMed]
- Ma, J.; Gnanasekar, A.; Lee, A.; Li, W.T.; Haas, M.; Wang-Rodriguez, J.; Chang, E.Y.; Rajasekaran, M.; Ongkeko, W.M. Influence of intratumor microbiome on clinical outcome and immune processes in prostate cancer. Cancers 2020, 12, 2524. [Google Scholar] [CrossRef]
- Wong, L.M.; Shende, N.; Li, W.T.; Castaneda, G.; Apostol, L.; Chang, E.Y.; Ongkeko, W.M. Comparative analysis of age- and gender-associated microbiome in lung adenocarcinoma and lung squamous cell carcinoma. Cancers 2020, 12, 1447. [Google Scholar] [CrossRef]
- Tzeng, A.; Sangwan, N.; Jia, M.; Liu, C.C.; Keslar, K.S.; Downs-Kelly, E.; Fairchild, R.L.; Al-Hilli, Z.; Grobmyer, S.R.; Eng, C. Human breast microbiome correlates with prognostic features and immunological signatures in breast cancer. Genome Med. 2021, 13, 60. [Google Scholar] [CrossRef]
- Soto-Pantoja, D.R.; Gaber, M.; Arnone, A.A.; Bronson, S.M.; Cruz-Diaz, N.; Wilson, A.S.; Clear, K.Y.J.; Ramirez, M.U.; Kucera, G.L.; Levine, E.A.; et al. Diet alters entero-mammary signaling to regulate the breast microbiome and tumorigenesis. Cancer Res. 2021, 81, 3890–3904. [Google Scholar] [CrossRef]
- Ling, Z.; Shao, L.; Liu, X.; Cheng, Y.; Yan, C.; Mei, Y.; Ji, F.; Liu, X. Regulatory t cells and plasmacytoid dendritic cells within the tumor microenvironment in gastric cancer are correlated with gastric microbiota dysbiosis: A Preliminary Study. Front. Immunol. 2019, 10, 533. [Google Scholar] [CrossRef]
- Zhang, S.; Zhang, S.; Ma, X.; Zhan, J.; Pan, C.; Zhang, H.; Xie, X.; Wen, J.; Xie, X. Intratumoral microbiome impacts immune infiltrates in tumor microenvironment and predicts prognosis in esophageal squamous cell carcinoma patients. Front. Cell. Infect. Microbiol. 2023, 13, 1165790. [Google Scholar] [CrossRef]
- Kosumi, K.; Baba, Y.; Yamamura, K.; Nomoto, D.; Okadome, K.; Yagi, T.; Toihata, T.; Kiyozumi, Y.; Harada, K.; Eto, K.; et al. Intratumour Fusobacterium nucleatum and immune response to oesophageal cancer. Br. J. Cancer 2023, 128, 1155–1165. [Google Scholar] [CrossRef]
- Ghaddar, B.; Biswas, A.; Harris, C.; Omary, M.B.; Carpizo, D.R.; Blaser, M.J.; De, S. Tumor microbiome links cellular programs and immunity in pancreatic cancer. Cancer Cell 2022, 40, 1240–1253.e5. [Google Scholar] [CrossRef]
- Alam, A.; Levanduski, E.; Denz, P.; Villavicencio, H.S.; Bhatta, M.; Alhorebi, L.; Zhang, Y.; Gomez, E.C.; Morreale, B.; Senchanthisai, S.; et al. Fungal mycobiome drives il-33 secretion and type 2 immunity in pancreatic cancer. Cancer Cell 2022, 40, 153–167.e11. [Google Scholar] [CrossRef] [PubMed]
- Zhu, Y.; Shi, T.; Lu, X.; Xu, Z.; Qu, J.; Zhang, Z.; Shi, G.; Shen, S.; Hou, Y.; Chen, Y.; et al. Fungal-induced glycolysis in macrophages promotes colon cancer by enhancing innate lymphoid cell secretion of il-22. EMBO J. 2021, 40, e105320. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- 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] [PubMed]
- Peng, R.; Liu, S.; You, W.; Huang, Y.; Hu, C.; Gao, Y.; Jia, X.; Li, G.; Xu, Z.; Chen, Y. Gastric Microbiome Alterations Are Associated with Decreased CD8+ Tissue-Resident Memory T Cells in the Tumor Microenvironment of Gastric Cancer. Cancer Immunol. Res. 2022, 10, 1224–1240. [Google Scholar] [CrossRef] [PubMed]
- 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] [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]
- Wang, S.P.; Rubio, L.A.; Duncan, S.H.; Donachie, G.E.; Holtrop, G.; Lo, G.; Farquharson, F.M.; Wagner, J.; Parkhill, J.; Louis, P.; et al. Pivotal roles for pH, lactate, and lactate-utilizing bacteria in the stability of a human colonic microbial ecosystem. mSystems 2020, 5, 10–1128. [Google Scholar] [CrossRef] [PubMed]
- Lamaudiere, M.T.F.; Arasaradnam, R.; Weedall, G.D.; Morozov, I.Y. The colorectal cancer microbiota alter their transcriptome to adapt to the acidity, reactive oxygen species, and metabolite availability of gut microenvironments. mSphere 2023, 8, e0062722. [Google Scholar] [CrossRef]
- Sun, J.; Tang, Q.; Yu, S.; Xie, M.; Zheng, W.; Chen, G.; Yin, Y.; Huang, X.; Wo, K.; Lei, H.; et al. F. nucleatum facilitates oral squamous cell carcinoma progression via glut1-driven lactate production. EBioMedicine 2023, 88, 104444. [Google Scholar] [CrossRef]
- Sun, C.; Li, T.; Song, X.; Huang, L.; Zang, Q.; Xu, J.; Bi, N.; Jiao, G.; Hao, Y.; Chen, Y.; et al. Spatially resolved metabolomics to discover tumor-associated metabolic alterations. Proc. Natl. Acad. Sci. USA 2019, 116, 52–57. [Google Scholar] [CrossRef]
- Miska, J.; Rashidi, A.; Lee-Chang, C.; Gao, P.; Lopez-Rosas, A.; Zhang, P.; Burga, R.; Castro, B.; Xiao, T.; Han, Y.; et al. Polyamines drive myeloid cell survival by buffering intracellular pH to promote immunosuppression in glioblastoma. Sci. Adv. 2021, 7, eabc8929. [Google Scholar] [CrossRef]
- Lan, A.; Lagadic-Gossmann, D.; Lemaire, C.; Brenner, C.; Jan, G. Acidic extracellular pH shifts colorectal cancer cell death from apoptosis to necrosis upon exposure to propionate and acetate, major end-products of the human probiotic propionibacteria. Apoptosis 2007, 12, 573–591. [Google Scholar] [CrossRef]
- Queirós, O.; Preto, A.; Pacheco, A.; Pinheiro, C.; Azevedo-Silva, J.; Moreira, R.; Pedro, M.; Ko, Y.H.; Pedersen, P.L.; Baltazar, F.; et al. Butyrate activates the monocarboxylate transporter MCT4 expression in breast cancer cells and enhances the antitumor activity of 3-bromopyruvate. J. Bioenerg. Biomembr. 2012, 44, 141–153. [Google Scholar] [CrossRef]
- Borthakur, A.; Saksena, S.; Gill, R.K.; Alrefai, W.A.; Ramaswamy, K.; Dudeja, P.K. Regulation of monocarboxylate transporter 1 (MCT1) promoter by butyrate in human intestinal epithelial cells: Involvement of NF-kappaB pathway. J. Cell. Biochem. 2008, 103, 1452–1463. [Google Scholar] [CrossRef]
- Rogers, A.H.; Zilm, P.S.; Gully, N.J.; Pfennig, A.L.; Marsh, P.D. Aspects of the growth and metabolism of Fusobacterium nucleatum atcc 10953 in continuous culture. Oral Microbiol. Immunol. 1991, 6, 250–255. [Google Scholar] [CrossRef]
- Kimura, N.T.; Taniguchi, S.; Aoki, K.; Baba, T. Selective localization and growth of Bifidobacterium bifidum in mouse tumors following intravenous administration. Cancer Res. 1980, 40, 2061–2068. [Google Scholar]
- Malmgren, R.A.; Flanigan, C.C. Localization of the vegetative form of clostridium tetani in mouse tumors following intravenous spore administration. Cancer Res. 1955, 15, 473–478. [Google Scholar] [PubMed]
- Li, X.; Wang, X.; Huang, R.; Stucky, A.; Chen, X.; Sun, L.; Wen, Q.; Zeng, Y.; Fletcher, H.; Wang, C.; et al. The machine-learning-mediated interface of microbiome and genetic risk stratification in neuroblastoma reveals molecular pathways related to patient survival. Cancers 2022, 14, 2874. [Google Scholar] [CrossRef]
- Fachi, J.L.; Felipe, J.S.; Pral, L.P.; da Silva, B.K.; Correa, R.O.; de Andrade, M.C.P.; da Fonseca, D.M.; Basso, P.J.; Camara, N.O.S.; de Sales, E.S.E.L.; et al. Butyrate protects mice from clostridium difficile-induced colitis through an hif-1-dependent mechanism. Cell Rep. 2019, 27, 750–761.e7. [Google Scholar] [CrossRef]
- Kelly, C.J.; Zheng, L.; Campbell, E.L.; Saeedi, B.; Scholz, C.C.; Bayless, A.J.; Wilson, K.E.; Glover, L.E.; Kominsky, D.J.; Magnuson, A.; et al. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell Host Microbe 2015, 17, 662–671. [Google Scholar] [CrossRef]
- Zgouras, D.; Wachtershauser, A.; Frings, D.; Stein, J. Butyrate impairs intestinal tumor cell-induced angiogenesis by inhibiting HIF-1alpha nuclear translocation. Biochem. Biophys. Res. Commun. 2003, 300, 832–838. [Google Scholar] [CrossRef]
- Bell, H.N.; Rebernick, R.J.; Goyert, J.; Singhal, R.; Kuljanin, M.; Kerk, S.A.; Huang, W.; Das, N.K.; Andren, A.; Solanki, S.; et al. Reuterin in the healthy gut microbiome suppresses colorectal cancer growth through altering redox balance. Cancer Cell 2022, 40, 185–200.e6. [Google Scholar] [CrossRef] [PubMed]
- Das, N.K.; Schwartz, A.J.; Barthel, G.; Inohara, N.; Liu, Q.; Sankar, A.; Hill, D.R.; Ma, X.; Lamberg, O.; Schnizlein, M.K.; et al. Microbial metabolite signaling is required for systemic iron homeostasis. Cell Metab. 2020, 31, 115–130.e6. [Google Scholar] [CrossRef] [PubMed]
- Viljoen, K.S.; Dakshinamurthy, A.; Goldberg, P.; Blackburn, J.M. Quantitative profiling of colorectal cancer-associated bacteria reveals associations between fusobacterium spp., enterotoxigenic bacteroides fragilis (ETBF) and clinicopathological features of colorectal cancer. PLoS ONE 2015, 10, e0119462. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Liu, J.; Deng, M.; Chen, X.; Jiang, L.; Zhang, J.; Tao, L.; Yu, W.; Qiu, Y. Enterococcus faecalis promotes the progression of colorectal cancer via its metabolite: Biliverdin. J. Transl. Med. 2023, 21, 72. [Google Scholar] [CrossRef] [PubMed]
- Sonveaux, P.; Copetti, T.; De Saedeleer, C.J.; Végran, F.; Verrax, J.; Kennedy, K.M.; Moon, E.J.; Dhup, S.; Danhier, P.; Frérart, F.; et al. Targeting the lactate transporter MCT1 in endothelial cells inhibits lactate-induced HIF-1 activation and tumor angiogenesis. PLoS ONE 2012, 7, e33418. [Google Scholar] [CrossRef] [PubMed]
- Dai, D.; Yang, Y.; Yu, J.; Dang, T.; Qin, W.; Teng, L.; Ye, J.; Jiang, H. Interactions between gastric microbiota and metabolites in gastric cancer. Cell Death Dis. 2021, 12, 1104. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Dai, D.; Jin, W.; Huang, Y.; Zhang, Y.; Chen, Y.; Wang, W.; Lin, W.; Chen, X.; Zhang, J.; et al. Microbiota and metabolites alterations in proximal and distal gastric cancer patients. J. Transl. Med. 2022, 20, 439. [Google Scholar] [CrossRef]
- Giallourou, N.; Urbaniak, C.; Puebla-Barragan, S.; Vorkas, P.A.; Swann, J.R.; Reid, G. Characterizing the breast cancer lipidome and its interaction with the tissue microbiota. Commun. Biol. 2021, 4, 1229. [Google Scholar] [CrossRef] [PubMed]
- AlMalki, R.H.; Sebaa, R.; Al-Ansari, M.M.; Al-Alwan, M.; Alwehaibi, M.A.; Rahman, A.M.A. E. coli Secretome metabolically modulates mda-mb-231 breast cancer cells’ energy metabolism. Int. J. Mol. Sci. 2023, 24, 4219. [Google Scholar] [CrossRef] [PubMed]
- Urbaniak, C.; Gloor, G.B.; Brackstone, M.; Scott, L.; Tangney, M.; Reid, G. The microbiota of breast tissue and its association with breast cancer. Appl. Environ. Microbiol. 2016, 82, 5039–5048. [Google Scholar] [CrossRef]
- Smith, A.; Cao, X.; Gu, Q.; Kubi Amos-Abanyie, E.; Tolley, E.A.; Vidal, G.; Lyn-Cook, B.; Starlard-Davenport, A. Characterization of the Metabolome of Breast Tissues from Non-Hispanic Black and Non-Hispanic White Women Reveals Correlations between Microbial Dysbiosis and Enhanced Lipid Metabolism Pathways in Triple-Negative Breast Tumors. Cancers 2022, 14, 4075. [Google Scholar] [CrossRef] [PubMed]
- Canale, F.P.; Basso, C.; Antonini, G.; Perotti, M.; Li, N.; Sokolovska, A.; Neumann, J.; James, M.J.; Geiger, S.; Jin, W.; et al. Metabolic modulation of tumours with engineered bacteria for immunotherapy. Nature 2021, 598, 662–666. [Google Scholar] [CrossRef] [PubMed]
- Choi, S.Y.; Collins, C.C.; Gout, P.W.; Wang, Y. Cancer-generated lactic acid: A regulatory, immunosuppressive metabolite? J. Pathol. 2013, 230, 350–355. [Google Scholar] [CrossRef] [PubMed]
- LaCourse, K.D.; Zepeda-Rivera, M.; Kempchinsky, A.G.; Baryiames, A.; Minot, S.S.; Johnston, C.D.; Bullman, S. The cancer chemotherapeutic 5-fluorouracil is a potent fusobacterium nucleatum inhibitor and its activity is modified by intratumoral microbiota. Cell Rep. 2022, 41, 111625. [Google Scholar] [CrossRef] [PubMed]
- Colbert, L.E.; El Alam, M.B.; Wang, R.; Karpinets, T.; Lo, D.; Lynn, E.J.; Harris, T.A.; Elnaggar, J.H.; Yoshida-Court, K.; Tomasic, K.; et al. Tumor-resident Lactobacillus iners confer chemoradiation resistance through lactate-induced metabolic rewiring. Cancer Cell 2023, 41, 1945–1962.e11. [Google Scholar] [CrossRef] [PubMed]
- Sitthirak, S.; Suksawat, M.; Phetcharaburanin, J.; Wangwiwatsin, A.; Klanrit, P.; Namwat, N.; Khuntikeo, N.; Titapun, A.; Jarearnrat, A.; Sangkhamanon, S.; et al. Chemotherapeutic resistant cholangiocarcinoma displayed distinct intratumoral microbial composition and metabolic profiles. PeerJ 2022, 10, e13876. [Google Scholar] [CrossRef] [PubMed]
- Byndloss, M.X.; Olsan, E.E.; Rivera-Chavez, F.; Tiffany, C.R.; Cevallos, S.A.; Lokken, K.L.; Torres, T.P.; Byndloss, A.J.; Faber, F.; Gao, Y.; et al. Microbiota-activated ppar-gamma signaling inhibits dysbiotic Enterobacteriaceae expansion. Science 2017, 357, 570–575. [Google Scholar] [CrossRef] [PubMed]
- Alfano, M.; Canducci, F.; Nebuloni, M.; Clementi, M.; Montorsi, F.; Salonia, A. The interplay of extracellular matrix and microbiome in urothelial bladder cancer. Nat. Rev. Urol. 2016, 13, 77–90. [Google Scholar] [CrossRef]
- Thomas, A.M.; Jesus, E.C.; Lopes, A.; Aguiar, S., Jr.; Begnami, M.D.; Rocha, R.M.; Carpinetti, P.A.; Camargo, A.A.; Hoffmann, C.; Freitas, H.C.; et al. Tissue-associated bacterial alterations in rectal carcinoma patients revealed by 16S rrna community profiling. Front. Cell. Infect. Microbiol. 2016, 6, 179. [Google Scholar] [CrossRef]
- Li, W.T.; Iyangar, A.S.; Reddy, R.; Chakladar, J.; Bhargava, V.; Sakamoto, K.; Ongkeko, W.M.; Rajasekaran, M. The bladder microbiome is associated with epithelial-mesenchymal transition in muscle invasive urothelial bladder carcinoma. Cancers 2021, 13, 3649. [Google Scholar] [CrossRef]
- Chen, S.; Fan, L.; Lin, Y.; Qi, Y.; Xu, C.; Ge, Q.; Zhang, Y.; Wang, Q.; Jia, D.; Wang, L.; et al. Bifidobacterium adolescentis orchestrates cd143(+) cancer-associated fibroblasts to suppress colorectal tumorigenesis by Wnt signaling-regulated GAS1. Cancer Commun. 2023, 43, 1027–1047. [Google Scholar] [CrossRef]
- Xu, Z.; Lv, Z.; Chen, F.; Zhang, Y.; Xu, Z.; Huo, J.; Liu, W.; Yu, S.; Tuersun, A.; Zhao, J.; et al. Dysbiosis of human tumor microbiome and aberrant residence of Actinomyces in tumor-associated fibroblasts in young-onset colorectal cancer. Front. Immunol. 2022, 13, 1008975. [Google Scholar] [CrossRef] [PubMed]
- Buda, A.; Qualtrough, D.; Jepson, M.A.; Martines, D.; Paraskeva, C.; Pignatelli, M. Butyrate downregulates alpha2beta1 integrin: A possible role in the induction of apoptosis in colorectal cancer cell lines. Gut 2003, 52, 729–734. [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/beta-catenin signaling via its fada adhesin. Cell Host Microbe 2013, 14, 195–206. [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]
- Chen, M.F.; Lu, M.S.; Hsieh, C.C.; Chen, W.C. Porphyromonas gingivalis promotes tumor progression in esophageal squamous cell carcinoma. Cell. Oncol. 2021, 44, 373–384. [Google Scholar] [CrossRef] [PubMed]
- Shen, J.; Zhai, J.; You, Q.; Zhang, G.; He, M.; Yao, X.; Shen, L. Cancer-associated fibroblasts-derived VCAM1 induced by H. pylori infection facilitates tumor invasion in gastric cancer. Oncogene 2020, 39, 2961–2974. [Google Scholar] [CrossRef]
- Krzysiek-Maczka, G.; Targosz, A.; Szczyrk, U.; Strzalka, M.; Sliwowski, Z.; Brzozowski, T.; Czyz, J.; Ptak-Belowska, A. Role of Helicobacter pylori infection in cancer-associated fibroblast-induced epithelial-mesenchymal transition in vitro. Helicobacter 2018, 23, e12538. [Google Scholar] [CrossRef]
- Chen, Y.; Yang, S.; Tavormina, J.; Tampe, D.; Zeisberg, M.; Wang, H.; Mahadevan, K.K.; Wu, C.J.; Sugimoto, H.; Chang, C.C.; et al. Oncogenic collagen I homotrimers from cancer cells bind to alpha3beta1 integrin and impact tumor microbiome and immunity to promote pancreatic cancer. Cancer Cell 2022, 40, 818–834.e9. [Google Scholar] [CrossRef]
- Culbert, A.; Chow, A.; Gharaibah, R.; Sangwan, N.; Dwidar, M.; Newsome, R.; Dai, J.; Ma, S.; Fredenburg, K.; Yilmaz, E.; et al. 1309 Differential microbial enrichment is associated with oral cancer and perineural invasion. J. Immunother. Cancer 2022, 10, A1360. [Google Scholar] [CrossRef]
- Chakladar, J.; John, D.; Magesh, S.; Uzelac, M.; Li, W.T.; Dereschuk, K.; Apostol, L.; Brumund, K.T.; Rodriguez, J.W.; Ongkeko, W.M. The intratumor bacterial and fungal microbiome is characterized by HPV, smoking, and alcohol consumption in head and neck squamous cell carcinoma. Int. J. Mol. Sci. 2022, 23, 13250. [Google Scholar] [CrossRef]
- Zhang, Z.; Feng, H.; Qiu, Y.; Xu, Z.; Xie, Q.; Ding, W.; Liu, H.; Li, G. Dysbiosis of gastric mucosal fungal microbiota in the gastric cancer microenvironment. J. Immunol. Res. 2022, 2022, 6011632. [Google Scholar] [CrossRef]
- Jiang, S.H.; Hu, L.P.; Wang, X.; Li, J.; Zhang, Z.G. Neurotransmitters: Emerging targets in cancer. Oncogene 2020, 39, 503–515. [Google Scholar] [CrossRef]
- Barrett, E.; Ross, R.P.; O‘Toole, P.W.; Fitzgerald, G.F.; Stanton, C. gamma-Aminobutyric acid production by culturable bacteria from the human intestine. J. Appl. Microbiol. 2012, 113, 411–417. [Google Scholar] [CrossRef]
- Chen, X.H.; Wang, A.; Chu, A.N.; Gong, Y.H.; Yuan, Y. Mucosa-associated microbiota in gastric cancer tissues compared with non-cancer tissues. Front. Microbiol. 2019, 10, 1261. [Google Scholar] [CrossRef] [PubMed]
- Lin, Y.; Fan, L.; Qi, Y.; Xu, C.; Jia, D.; Jiang, Y.; Chen, S.; Wang, L. Bifidobacterium adolescentis induces Decorin(+) macrophages via TLR2 to suppress colorectal carcinogenesis. J. Exp. Clin. Cancer Res. 2023, 42, 172. [Google Scholar] [CrossRef]
- Özoğul, F. Production of biogenic amines by Morganella morganii, Klebsiella pneumoniae and hafnia alvei using a rapid hplc method. Eur. Food Res. Technol. 2004, 219, 465–469. [Google Scholar] [CrossRef]
- Shishov, V.A.; Kirovskaia, T.A.; Kudrin, V.S.; Oleskin, A.V. Amine neuromediators, their precursors, and oxidation products in the culture of Escherichia coli K-12. Prikl. Biokhim Mikrobiol. 2009, 45, 550–554. [Google Scholar]
- Li, S.; Xia, H.; Wang, Z.; Zhang, X.; Song, T.; Li, J.; Xu, L.; Zhang, N.; Fan, S.; Li, Q.; et al. Intratumoral microbial heterogeneity affected tumor immune microenvironment and determined clinical outcome of hbv-related HCC. Hepatology 2023, 78, 1079–1091. [Google Scholar] [CrossRef] [PubMed]
- Younginger, B.S.; Mayba, O.; Reeder, J.; Nagarkar, D.R.; Modrusan, Z.; Albert, M.L.; Byrd, A.L. Enrichment of oral-derived bacteria in inflamed colorectal tumors and distinct associations of Fusobacterium in the mesenchymal subtype. Cell Rep. Med. 2023, 4, 100920. [Google Scholar] [CrossRef]
- Yam, A.O.; Bailey, J.; Lin, F.; Jakovija, A.; Youlten, S.E.; Counoupas, C.; Gunzer, M.; Bald, T.; Woodruff, T.M.; Triccas, J.A.; et al. Neutrophil conversion to a tumor-killing phenotype underpins effective microbial therapy. Cancer Res. 2023, 83, 1315–1328. [Google Scholar] [CrossRef]
- Jiang, L.; Duan, B.; Jia, P.; Zhang, Y.; Yan, X. The Role of Intratumor Microbiomes in Cervical Cancer Metastasis. Cancers 2023, 15, 509. [Google Scholar] [CrossRef]
- Fidelle, M.; Rauber, C.; Alves Costa Silva, C.; Tian, A.L.; Lahmar, I.; de La Varende, A.M.; Zhao, L.; Thelemaque, C.; Lebhar, I.; Messaoudene, M.; et al. A microbiota-modulated checkpoint directs immunosuppressive intestinal T cells into cancers. Science 2023, 380, eabo2296. [Google Scholar] [CrossRef] [PubMed]
- Zhou, J.; Huang, G.; Wong, W.C.; Hu, D.H.; Zhu, J.W.; Li, R.; Zhou, H. The impact of antibiotic use on clinical features and survival outcomes of cancer patients treated with immune checkpoint inhibitors. Front. Immunol. 2022, 13, 968729. [Google Scholar] [CrossRef] [PubMed]
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. |
© 2024 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
Kyriazi, A.A.; Karaglani, M.; Agelaki, S.; Baritaki, S. Intratumoral Microbiome: Foe or Friend in Reshaping the Tumor Microenvironment Landscape? Cells 2024, 13, 1279. https://doi.org/10.3390/cells13151279
Kyriazi AA, Karaglani M, Agelaki S, Baritaki S. Intratumoral Microbiome: Foe or Friend in Reshaping the Tumor Microenvironment Landscape? Cells. 2024; 13(15):1279. https://doi.org/10.3390/cells13151279
Chicago/Turabian StyleKyriazi, Athina A., Makrina Karaglani, Sofia Agelaki, and Stavroula Baritaki. 2024. "Intratumoral Microbiome: Foe or Friend in Reshaping the Tumor Microenvironment Landscape?" Cells 13, no. 15: 1279. https://doi.org/10.3390/cells13151279
APA StyleKyriazi, A. A., Karaglani, M., Agelaki, S., & Baritaki, S. (2024). Intratumoral Microbiome: Foe or Friend in Reshaping the Tumor Microenvironment Landscape? Cells, 13(15), 1279. https://doi.org/10.3390/cells13151279