Microbiome in Cancer Development and Treatment
Abstract
:1. Introduction
2. Dominant Bacteria-Driven Mechanisms Associated with Cancer Development
2.1. Helicobacter pylori
2.2. Fusobacterium nucleatum
2.3. Escherichia coli
2.4. Salmonella
2.5. Bacteroides fragilis
2.6. Staphylococcus aureus
2.7. Campylobacter jejuni
2.8. Desulfovibrio
2.9. Porphyromonas
3. Microbiome and Treatment Efficacy
3.1. Microbiome and Chemotherapy
3.2. Microbiome and Immunotherapy
4. Microbiome and Therapy-Induced Late Effects
4.1. Microbiome and Treatment-Induced Cognitive Impairment
4.2. Microbiome and Cardiovascular Toxicity
5. Microbiota Modulation by Probiotics, Prebiotics, and FMT in Cancer Patients
6. Critical Analysis of the Clinical Utility of a Microbiome-Based Approach
7. Conclusions and Future Directions
Author Contributions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Ramirez-Labrada, A.G.; Isla, D.; Artal, A.; Arias, M.; Rezusta, A.; Pardo, J.; Galvez, E.M. The Influence of Lung Microbiota on Lung Carcinogenesis, Immunity, and Immunotherapy. Trends Cancer 2020, 6, 86–97. [Google Scholar] [CrossRef] [PubMed]
- Ciernikova, S.; Sevcikova, A.; Stevurkova, V.; Mego, M. Tumor microbiome—An integral part of the tumor microenvironment. Front. Oncol. 2022, 12, 1063100. [Google Scholar] [CrossRef] [PubMed]
- Chen, X.; Song, M.; Zhang, B.; Zhang, Y. Reactive Oxygen Species Regulate T Cell Immune Response in the Tumor Microenvironment. Oxid. Med. Cell. Longev. 2016, 2016, 1580967. [Google Scholar] [CrossRef] [PubMed]
- Kennel, K.B.; Greten, F.R. Immune cell—Produced ROS and their impact on tumor growth and metastasis. Redox. Biol. 2021, 42, 101891. [Google Scholar] [CrossRef] [PubMed]
- Liu, L.; Jin, R.; Hao, J.; Zeng, J.; Yin, D.; Yi, Y.; Zhu, M.; Mandal, A.; Hua, Y.; Ng, C.K.; et al. Consumption of the Fish Oil High-Fat Diet Uncouples Obesity and Mammary Tumor Growth through Induction of Reactive Oxygen Species in Protumor Macrophages. Cancer Res. 2020, 80, 2564–2574. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
- Ciernikova, S.; Mego, M.; Hainova, K.; Adamcikova, Z.; Stevurkova, V.; Zajac, V. Modification of microflora imbalance: Future directions for prevention and treatment of colorectal cancer? Neoplasma 2015, 62, 345–352. [Google Scholar] [CrossRef]
- Goubet, A.G. Could the tumor-associated microbiota be the new multi-faceted player in the tumor microenvironment? Front. Oncol. 2023, 13, 1185163. [Google Scholar] [CrossRef]
- Nejman, D.; Livyatan, I.; Fuks, G.; Gavert, N.; Zwang, Y.; Geller, L.T.; Rotter-Maskowitz, A.; Weiser, R.; Mallel, G.; Gigi, E.; et al. The human tumor microbiome is composed of tumor type-specific intracellular bacteria. Science 2020, 368, 973–980. [Google Scholar] [CrossRef]
- Wu, S.; Zhu, W.; Thompson, P.; Hannun, Y.A. Evaluating intrinsic and non-intrinsic cancer risk factors. Nat. Commun. 2018, 9, 3490. [Google Scholar] [CrossRef]
- Wu, S.; Powers, S.; Zhu, W.; Hannun, Y.A. Substantial contribution of extrinsic risk factors to cancer development. Nature 2016, 529, 43–47. [Google Scholar] [CrossRef] [PubMed]
- Mbemi, A.; Khanna, S.; Njiki, S.; Yedjou, C.G.; Tchounwou, P.B. Impact of Gene-Environment Interactions on Cancer Development. Int. J. Environ. Res. Public Health 2020, 17, 8089. [Google Scholar] [CrossRef] [PubMed]
- Dorak, M.T.; Karpuzoglu, E. Gender differences in cancer susceptibility: An inadequately addressed issue. Front. Genet. 2012, 3, 268. [Google Scholar] [CrossRef] [PubMed]
- Narimatsu, H.; Yaguchi, Y.T. The Role of Diet and Nutrition in Cancer: Prevention, Treatment, and Survival. Nutrients 2022, 14, 3329. [Google Scholar] [CrossRef] [PubMed]
- Mittelman, S.D. The Role of Diet in Cancer Prevention and Chemotherapy Efficacy. Annu. Rev. Nutr. 2020, 40, 273–297. [Google Scholar] [CrossRef] [PubMed]
- Ciernikova, S.; Sevcikova, A.; Stevurkova, V.; Mego, M. Diet-driven microbiome changes and physical activity in cancer patients. Front. Nutr. 2023, 10, 1285516. [Google Scholar] [CrossRef] [PubMed]
- Shi, B.; Guo, X.; Liu, H.; Jiang, K.; Liu, L.; Yan, N.; Farag, M.A.; Liu, L. Dissecting Maillard reaction production in fried foods: Formation mechanisms, sensory characteristic attribution, control strategy, and gut homeostasis regulation. Food Chem. 2023, 438, 137994. [Google Scholar] [CrossRef] [PubMed]
- Dinu, M.; Abbate, R.; Gensini, G.F.; Casini, A.; Sofi, F. Vegetarian, vegan diets and multiple health outcomes: A systematic review with meta-analysis of observational studies. Crit. Rev. Food Sci. Nutr. 2017, 57, 3640–3649. [Google Scholar] [CrossRef]
- Papadimitriou, N.; Markozannes, G.; Kanellopoulou, A.; Critselis, E.; Alhardan, S.; Karafousia, V.; Kasimis, J.C.; Katsaraki, C.; Papadopoulou, A.; Zografou, M.; et al. An umbrella review of the evidence associating diet and cancer risk at 11 anatomical sites. Nat. Commun. 2021, 12, 4579. [Google Scholar] [CrossRef]
- Fontana, A.; Panebianco, C.; Picchianti-Diamanti, A.; Lagana, B.; Cavalieri, D.; Potenza, A.; Pracella, R.; Binda, E.; Copetti, M.; Pazienza, V. Gut Microbiota Profiles Differ among Individuals Depending on Their Region of Origin: An Italian Pilot Study. Int. J. Environ. Res. Public Health 2019, 16, 4065. [Google Scholar] [CrossRef]
- Sun, S.; Wang, H.; Tsilimigras, M.C.; Howard, A.G.; Sha, W.; Zhang, J.; Su, C.; Wang, Z.; Du, S.; Sioda, M.; et al. Does geographical variation confound the relationship between host factors and the human gut microbiota: A population-based study in China. BMJ Open 2020, 10, e038163. [Google Scholar] [CrossRef] [PubMed]
- Li, J.; Jia, H.; Cai, X.; Zhong, H.; Feng, Q.; Sunagawa, S.; Arumugam, M.; Kultima, J.R.; Prifti, E.; Nielsen, T.; et al. An integrated catalog of reference genes in the human gut microbiome. Nat. Biotechnol. 2014, 32, 834–841. [Google Scholar] [CrossRef] [PubMed]
- Human Microbiome Project, C. A framework for human microbiome research. Nature 2012, 486, 215–221. [Google Scholar] [CrossRef] [PubMed]
- Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef] [PubMed]
- Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [PubMed]
- Whisner, C.M.; Athena Aktipis, C. The Role of the Microbiome in Cancer Initiation and Progression: How Microbes and Cancer Cells Utilize Excess Energy and Promote One Another’s Growth. Curr. Nutr. Rep. 2019, 8, 42–51. [Google Scholar] [CrossRef]
- De Martel, C.; Ferlay, J.; Franceschi, S.; Vignat, J.; Bray, F.; Forman, D.; Plummer, M. Global burden of cancers attributable to infections in 2008: A review and synthetic analysis. Lancet Oncol. 2012, 13, 607–615. [Google Scholar] [CrossRef]
- Vidal, M.C.; Anneberg, T.J.; Cure, A.E.; Althoff, D.M.; Segraves, K.A. The variable effects of global change on insect mutualisms. Curr. Opin. Insect Sci. 2021, 47, 46–52. [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]
- Yang, Q.; Wang, B.; Zheng, Q.; Li, H.; Meng, X.; Zhou, F.; Zhang, L. A Review of Gut Microbiota-Derived Metabolites in Tumor Progression and Cancer Therapy. Adv. Sci. 2023, 10, e2207366. [Google Scholar] [CrossRef]
- Cong, J.; Zhou, P.; Zhang, R. Intestinal Microbiota-Derived Short Chain Fatty Acids in Host Health and Disease. Nutrients 2022, 14, 1977. [Google Scholar] [CrossRef] [PubMed]
- Portincasa, P.; Bonfrate, L.; Vacca, M.; De Angelis, M.; Farella, I.; Lanza, E.; Khalil, M.; Wang, D.Q.; Sperandio, M.; Di Ciaula, A. Gut Microbiota and Short Chain Fatty Acids: Implications in Glucose Homeostasis. Int. J. Mol. Sci. 2022, 23, 1105. [Google Scholar] [CrossRef] [PubMed]
- Kazmierczak-Siedlecka, K.; Marano, L.; Merola, E.; Roviello, F.; Polom, K. Sodium butyrate in both prevention and supportive treatment of colorectal cancer. Front. Cell. Infect. Microbiol. 2022, 12, 1023806. [Google Scholar] [CrossRef] [PubMed]
- Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [Google Scholar] [CrossRef] [PubMed]
- Coutzac, C.; Jouniaux, J.M.; Paci, A.; Schmidt, J.; Mallardo, D.; Seck, A.; Asvatourian, V.; Cassard, L.; Saulnier, P.; Lacroix, L.; et al. Systemic short chain fatty acids limit antitumor effect of CTLA-4 blockade in hosts with cancer. Nat. Commun. 2020, 11, 2168. [Google Scholar] [CrossRef] [PubMed]
- 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.e186. [Google Scholar] [CrossRef] [PubMed]
- Ridlon, J.M.; Kang, D.J.; Hylemon, P.B.; Bajaj, J.S. Bile acids and the gut microbiome. Curr. Opin. Gastroenterol. 2014, 30, 332–338. [Google Scholar] [CrossRef]
- Bernstein, H.; Bernstein, C. Bile acids as carcinogens in the colon and at other sites in the gastrointestinal system. Exp. Biol. Med. 2023, 248, 79–89. [Google Scholar] [CrossRef]
- Marshall, B.J.; Warren, J.R. Unidentified curved bacilli in the stomach of patients with gastritis and peptic ulceration. Lancet 1984, 1, 1311–1315. [Google Scholar] [CrossRef]
- Amieva, M.; Peek, R.M., Jr. Pathobiology of Helicobacter pylori-Induced Gastric Cancer. Gastroenterology 2016, 150, 64–78. [Google Scholar] [CrossRef]
- Brown, L.M. Helicobacter pylori: Epidemiology and routes of transmission. Epidemiol. Rev. 2000, 22, 283–297. [Google Scholar] [CrossRef] [PubMed]
- Chmiela, M.; Kupcinskas, J. Review: Pathogenesis of Helicobacter pylori infection. Helicobacter 2019, 24 (Suppl. S1), e12638. [Google Scholar] [CrossRef] [PubMed]
- Gonciarz, W.; Walencka, M.; Moran, A.P.; Hinc, K.; Obuchowski, M.; Chmiela, M. Upregulation of MUC5AC production and deposition of LEWIS determinants by HELICOBACTER PYLORI facilitate gastric tissue colonization and the maintenance of infection. J. Biomed. Sci. 2019, 26, 23. [Google Scholar] [CrossRef] [PubMed]
- Noto, J.M.; Peek, R.M., Jr. The Helicobacter pylori cag Pathogenicity Island. Methods Mol. Biol. 2012, 921, 41–50. [Google Scholar] [CrossRef]
- Hanada, K.; Uchida, T.; Tsukamoto, Y.; Watada, M.; Yamaguchi, N.; Yamamoto, K.; Shiota, S.; Moriyama, M.; Graham, D.Y.; Yamaoka, Y. Helicobacter pylori infection introduces DNA double-strand breaks in host cells. Infect. Immun. 2014, 82, 4182–4189. [Google Scholar] [CrossRef]
- Backert, S.; Tegtmeyer, N.; Fischer, W. Composition, structure and function of the Helicobacter pylori cag pathogenicity island encoded type IV secretion system. Future Microbiol. 2015, 10, 955–965. [Google Scholar] [CrossRef] [PubMed]
- Saadat, I.; Higashi, H.; Obuse, C.; Umeda, M.; Murata-Kamiya, N.; Saito, Y.; Lu, H.; Ohnishi, N.; Azuma, T.; Suzuki, A.; et al. Helicobacter pylori CagA targets PAR1/MARK kinase to disrupt epithelial cell polarity. Nature 2007, 447, 330–333. [Google Scholar] [CrossRef]
- Graham, D.Y.; Miftahussurur, M. Helicobacter pylori urease for diagnosis of Helicobacter pylori infection: A mini review. J. Adv. Res. 2018, 13, 51–57. [Google Scholar] [CrossRef] [PubMed]
- Palframan, S.L.; Kwok, T.; Gabriel, K. Vacuolating cytotoxin A (VacA), a key toxin for Helicobacter pylori pathogenesis. Front. Cell. Infect. Microbiol. 2012, 2, 92. [Google Scholar] [CrossRef]
- Foegeding, N.J.; Caston, R.R.; McClain, M.S.; Ohi, M.D.; Cover, T.L. An Overview of Helicobacter pylori VacA Toxin Biology. Toxins 2016, 8, 173. [Google Scholar] [CrossRef] [PubMed]
- Fu, H.W.; Lai, Y.C. The Role of Helicobacter pylori Neutrophil-Activating Protein in the Pathogenesis of H. pylori and Beyond: From a Virulence Factor to Therapeutic Targets and Therapeutic Agents. Int. J. Mol. Sci. 2022, 24, 91. [Google Scholar] [CrossRef] [PubMed]
- Amedei, A.; Cappon, A.; Codolo, G.; Cabrelle, A.; Polenghi, A.; Benagiano, M.; Tasca, E.; Azzurri, A.; D’Elios, M.M.; Del Prete, G.; et al. The neutrophil-activating protein of Helicobacter pylori promotes Th1 immune responses. J. Clin. Investig. 2006, 116, 1092–1101. [Google Scholar] [CrossRef] [PubMed]
- Sun, C.H.; Li, B.B.; Wang, B.; Zhao, J.; Zhang, X.Y.; Li, T.T.; Li, W.B.; Tang, D.; Qiu, M.J.; Wang, X.C.; et al. The role of Fusobacterium nucleatum in colorectal cancer: From carcinogenesis to clinical management. Chronic Dis. Transl. Med. 2019, 5, 178–187. [Google Scholar] [CrossRef]
- 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]
- 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] [PubMed]
- Chen, Y.; Huang, Z.; Tang, Z.; Huang, Y.; Huang, M.; Liu, H.; Ziebolz, D.; Schmalz, G.; Jia, B.; Zhao, J. More Than Just a Periodontal Pathogen-the Research Progress on Fusobacterium nucleatum. Front. Cell. Infect. Microbiol. 2022, 12, 815318. [Google Scholar] [CrossRef] [PubMed]
- Liu, H.; Liu, Y.; Liu, W.; Zhang, W.; Xu, J. EZH2-mediated loss of miR-622 determines CXCR4 activation in hepatocellular carcinoma. Nat. Commun. 2015, 6, 8494. [Google Scholar] [CrossRef]
- Gaba, F.I.; Gonzalez, R.C.; Martinez, R.G. The Role of Oral Fusobacterium nucleatum in Female Breast Cancer: A Systematic Review and Meta-Analysis. Int. J. Dent. 2022, 2022, 1876275. [Google Scholar] [CrossRef]
- Han, Y.W.; Ikegami, A.; Rajanna, C.; Kawsar, H.I.; Zhou, Y.; Li, M.; Sojar, H.T.; Genco, R.J.; Kuramitsu, H.K.; Deng, C.X. Identification and characterization of a novel adhesin unique to oral fusobacteria. J. Bacteriol. 2005, 187, 5330–5340. [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]
- Guo, P.; Tian, Z.; Kong, X.; Yang, L.; Shan, X.; Dong, B.; Ding, X.; Jing, X.; Jiang, C.; Jiang, N.; et al. FadA promotes DNA damage and progression of Fusobacterium nucleatum-induced colorectal cancer through up-regulation of chk2. J. Exp. Clin. Cancer Res. 2020, 39, 202. [Google Scholar] [CrossRef] [PubMed]
- Wirbel, J.; Pyl, P.T.; Kartal, E.; Zych, K.; Kashani, A.; Milanese, A.; Fleck, J.S.; Voigt, A.Y.; Palleja, A.; Ponnudurai, R.; et al. Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat. Med. 2019, 25, 679–689. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Huang, J.; Yu, T.; Fang, X.; Lou, L.; Xin, S.; Ji, L.; Jiang, F.; Lou, Y. Fusobacterium nucleatum Promotes the Progression of Colorectal Cancer through Cdk5-Activated Wnt/beta-Catenin Signaling. Front. Microbiol. 2020, 11, 545251. [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] [PubMed]
- Parhi, L.; Alon-Maimon, T.; Sol, A.; Nejman, D.; Shhadeh, A.; Fainsod-Levi, T.; Yajuk, O.; Isaacson, B.; Abed, J.; Maalouf, N.; et al. Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression. Nat. Commun. 2020, 11, 3259. [Google Scholar] [CrossRef] [PubMed]
- Magdy, A.; Elhadidy, M.; Abd Ellatif, M.E.; El Nakeeb, A.; Abdallah, E.; Thabet, W.; Youssef, M.; Khafagy, W.; Morshed, M.; Farid, M. Enteropathogenic Escherichia coli (EPEC): Does it have a role in colorectal tumourigenesis? A Prospective Cohort Study. Int. J. Surg. 2015, 18, 169–173. [Google Scholar] [CrossRef] [PubMed]
- Villeger, R.; Lopes, A.; Veziant, J.; Gagniere, J.; Barnich, N.; Billard, E.; Boucher, D.; Bonnet, M. Microbial markers in colorectal cancer detection and/or prognosis. World J. Gastroenterol. 2018, 24, 2327–2347. [Google Scholar] [CrossRef]
- Scott, D.A.; Kaper, J.B. Cloning and sequencing of the genes encoding Escherichia coli cytolethal distending toxin. Infect. Immun. 1994, 62, 244–251. [Google Scholar] [CrossRef]
- Tremblay, W.; Mompart, F.; Lopez, E.; Quaranta, M.; Bergoglio, V.; Hashim, S.; Bonnet, D.; Alric, L.; Mas, E.; Trouche, D.; et al. Cytolethal Distending Toxin Promotes Replicative Stress Leading to Genetic Instability Transmitted to Daughter Cells. Front. Cell Dev. Biol. 2021, 9, 656795. [Google Scholar] [CrossRef]
- Veziant, J.; Villeger, R.; Barnich, N.; Bonnet, M. Gut Microbiota as Potential Biomarker and/or Therapeutic Target to Improve the Management of Cancer: Focus on Colibactin-Producing Escherichia coli in Colorectal Cancer. Cancers 2021, 13, 2215. [Google Scholar] [CrossRef]
- Nougayrede, J.P.; Homburg, S.; Taieb, F.; Boury, M.; Brzuszkiewicz, E.; Gottschalk, G.; Buchrieser, C.; Hacker, J.; Dobrindt, U.; Oswald, E. Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science 2006, 313, 848–851. [Google Scholar] [CrossRef] [PubMed]
- Arthur, J.C.; Perez-Chanona, E.; Muhlbauer, M.; Tomkovich, S.; Uronis, J.M.; Fan, T.J.; Campbell, B.J.; Abujamel, T.; Dogan, B.; Rogers, A.B.; et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 2012, 338, 120–123. [Google Scholar] [CrossRef] [PubMed]
- Cougnoux, A.; Dalmasso, G.; Martinez, R.; Buc, E.; Delmas, J.; Gibold, L.; Sauvanet, P.; Darcha, C.; Dechelotte, P.; Bonnet, M.; et al. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut 2014, 63, 1932–1942. [Google Scholar] [CrossRef] [PubMed]
- Olier, M.; Marcq, I.; Salvador-Cartier, C.; Secher, T.; Dobrindt, U.; Boury, M.; Bacquie, V.; Penary, M.; Gaultier, E.; Nougayrede, J.P.; et al. Genotoxicity of Escherichia coli Nissle 1917 strain cannot be dissociated from its probiotic activity. Gut Microbes 2012, 3, 501–509. [Google Scholar] [CrossRef] [PubMed]
- Sepe, L.P.; Hartl, K.; Iftekhar, A.; Berger, H.; Kumar, N.; Goosmann, C.; Chopra, S.; Schmidt, S.C.; Gurumurthy, R.K.; Meyer, T.F.; et al. Genotoxic Effect of Salmonella Paratyphi A Infection on Human Primary Gallbladder Cells. mBio 2020, 11, e01911-20. [Google Scholar] [CrossRef] [PubMed]
- Duijster, J.W.; Hansen, J.V.; Franz, E.; Neefjes, J.J.C.; Frisch, M.; Mughini-Gras, L.; Ethelberg, S. Association between Salmonella infection and colon cancer: A nationwide registry-based cohort study. Epidemiol. Infect. 2021, 149, e56. [Google Scholar] [CrossRef] [PubMed]
- Mughini-Gras, L.; Schaapveld, M.; Kramers, J.; Mooij, S.; Neefjes-Borst, E.A.; Pelt, W.V.; Neefjes, J. Increased colon cancer risk after severe Salmonella infection. PLoS ONE 2018, 13, e0189721. [Google Scholar] [CrossRef]
- Lu, R.; Wu, S.; Zhang, Y.G.; Xia, Y.; Zhou, Z.; Kato, I.; Dong, H.; Bissonnette, M.; Sun, J. Salmonella Protein AvrA Activates the STAT3 Signaling Pathway in Colon Cancer. Neoplasia 2016, 18, 307–316. [Google Scholar] [CrossRef]
- Liu, X.; Lu, R.; Xia, Y.; Wu, S.; Sun, J. Eukaryotic signaling pathways targeted by Salmonella effector protein AvrA in intestinal infection in vivo. BMC Microbiol. 2010, 10, 326. [Google Scholar] [CrossRef]
- Wu, H.; Jones, R.M.; Neish, A.S. The Salmonella effector AvrA mediates bacterial intracellular survival during infection in vivo. Cell. Microbiol. 2012, 14, 28–39. [Google Scholar] [CrossRef]
- Van Elsland, D.M.; Duijster, J.W.; Zhang, J.; Stevenin, V.; Zhang, Y.; Zha, L.; Xia, Y.; Franz, E.; Sun, J.; Mughini-Gras, L.; et al. Repetitive non-typhoidal Salmonella exposure is an environmental risk factor for colon cancer and tumor growth. Cell Rep. Med. 2022, 3, 100852. [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]
- Pierce, J.V.; Bernstein, H.D. Genomic Diversity of Enterotoxigenic Strains of Bacteroides fragilis. PLoS ONE 2016, 11, e0158171. [Google Scholar] [CrossRef] [PubMed]
- Yu, L.C.; Wei, S.C.; Ni, Y.H. Impact of microbiota in colorectal carcinogenesis: Lessons from experimental models. Intest. Res. 2018, 16, 346–357. [Google Scholar] [CrossRef] [PubMed]
- Bundgaard-Nielsen, C.; Baandrup, U.T.; Nielsen, L.P.; Sorensen, S. The presence of bacteria varies between colorectal adenocarcinomas, precursor lesions and non-malignant tissue. BMC Cancer 2019, 19, 399. [Google Scholar] [CrossRef] [PubMed]
- Geis, A.L.; Fan, H.; Wu, X.; Wu, S.; Huso, D.L.; Wolfe, J.L.; Sears, C.L.; Pardoll, D.M.; Housseau, F. Regulatory T-cell Response to Enterotoxigenic Bacteroides fragilis Colonization Triggers IL17-Dependent Colon Carcinogenesis. Cancer Discov. 2015, 5, 1098–1109. [Google Scholar] [CrossRef] [PubMed]
- Chung, L.; Thiele Orberg, E.; Geis, A.L.; Chan, J.L.; Fu, K.; DeStefano Shields, C.E.; Dejea, C.M.; Fathi, P.; Chen, J.; Finard, B.B.; et al. Bacteroides fragilis Toxin Coordinates a Pro-carcinogenic Inflammatory Cascade via Targeting of Colonic Epithelial Cells. Cell Host Microbe 2018, 23, 203–214.e205. [Google Scholar] [CrossRef]
- Boleij, A.; Hechenbleikner, E.M.; Goodwin, A.C.; Badani, R.; Stein, E.M.; Lazarev, M.G.; Ellis, B.; Carroll, K.C.; Albesiano, E.; Wick, E.C.; et al. The Bacteroides fragilis toxin gene is prevalent in the colon mucosa of colorectal cancer patients. Clin. Infect. Dis. 2015, 60, 208–215. [Google Scholar] [CrossRef]
- Pinchuk, I.V.; Beswick, E.J.; Reyes, V.E. Staphylococcal enterotoxins. Toxins 2010, 2, 2177–2197. [Google Scholar] [CrossRef]
- Espersen, F.; Frimodt-Moller, N.; Rosdahl, V.T.; Jessen, O.; Faber, V.; Rosendal, K. Staphylococcus aureus bacteremia in patients with hematological malignancies and/or agranulocytosis. Acta Medica Scand 1987, 222, 465–470. [Google Scholar] [CrossRef]
- Velasco, E.; Thuler, L.C.; Martins, C.A.; Nucci, M.; Dias, L.M.; Goncalves, V.M. Epidemiology of bloodstream infections at a cancer center. Sao Paulo Med. J. 2000, 118, 131–138. [Google Scholar] [CrossRef] [PubMed]
- Turk, S.; Yanpar, H.; Baesmat, A.S.; Canli, S.D.; Cinar, O.E.; Malkan, U.Y.; Turk, C.; Haznedaroglu, I.C.; Ucar, G. Enterotoxins A and B produced by Staphylococcus aureus increase cell proliferation, invasion and cytarabine resistance in acute myeloid leukemia cell lines. Heliyon 2023, 9, e19743. [Google Scholar] [CrossRef] [PubMed]
- Akbari, A.; Farahnejad, Z.; Akhtari, J.; Abastabar, M.; Mobini, G.R.; Mehbod, A.S. Staphylococcus aureus Enterotoxin B Down-Regulates the Expression of Transforming Growth Factor-Beta (TGF-beta) Signaling Transducers in Human Glioblastoma. Jundishapur J. Microbiol. 2016, 9, e27297. [Google Scholar] [CrossRef] [PubMed]
- Ejtehadifar, M.; Halabian, R.; Fooladi, A.A.I.; Ghazavi, A.; Mosayebi, G. Anti-cancer effects of Staphylococcal Enterotoxin type B on U266 cells co-cultured with Mesenchymal Stem Cells. Microb. Pathog. 2017, 113, 438–444. [Google Scholar] [CrossRef] [PubMed]
- Krejsgaard, T.; Willerslev-Olsen, A.; Lindahl, L.M.; Bonefeld, C.M.; Koralov, S.B.; Geisler, C.; Wasik, M.A.; Gniadecki, R.; Kilian, M.; Iversen, L.; et al. Staphylococcal enterotoxins stimulate lymphoma-associated immune dysregulation. Blood 2014, 124, 761–770. [Google Scholar] [CrossRef] [PubMed]
- Sun, X.; Threadgill, D.; Jobin, C. Campylobacter jejuni induces colitis through activation of mammalian target of rapamycin signaling. Gastroenterology 2012, 142, 86–95.e5. [Google Scholar] [CrossRef] [PubMed]
- Konkel, M.E.; Talukdar, P.K.; Negretti, N.M.; Klappenbach, C.M. Taking Control: Campylobacter jejuni Binding to Fibronectin Sets the Stage for Cellular Adherence and Invasion. Front. Microbiol. 2020, 11, 564. [Google Scholar] [CrossRef]
- He, Z.; Gharaibeh, R.Z.; Newsome, R.C.; Pope, J.L.; Dougherty, M.W.; Tomkovich, S.; Pons, B.; Mirey, G.; Vignard, J.; Hendrixson, D.R.; et al. Campylobacter jejuni promotes colorectal tumorigenesis through the action of cytolethal distending toxin. Gut 2019, 68, 289–300. [Google Scholar] [CrossRef]
- Attene-Ramos, M.S.; Wagner, E.D.; Plewa, M.J.; Gaskins, H.R. Evidence that hydrogen sulfide is a genotoxic agent. Mol. Cancer Res. 2006, 4, 9–14. [Google Scholar] [CrossRef]
- Dahmus, J.D.; Kotler, D.L.; Kastenberg, D.M.; Kistler, C.A. The gut microbiome and colorectal cancer: A review of bacterial pathogenesis. J. Gastrointest. Oncol. 2018, 9, 769–777. [Google Scholar] [CrossRef]
- Kapral, M.; Weglarz, L.; Parfiniewicz, B.; Lodowska, J.; Jaworska-Kik, M. Quantitative evaluation of transcriptional activation of NF-kappaB p65 and p50 subunits and IkappaBalpha encoding genes in colon cancer cells by Desulfovibrio desulfuricans endotoxin. Folia Microbiol. 2010, 55, 657–661. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Dai, J.; Lan, X.; Fan, B.; Dong, T.; Zhang, Y.; Han, M. Intestinal bacteria are potential biomarkers and therapeutic targets for gastric cancer. Microb. Pathog. 2021, 151, 104747. [Google Scholar] [CrossRef] [PubMed]
- Fan, X.; Alekseyenko, A.V.; Wu, J.; Peters, B.A.; Jacobs, E.J.; Gapstur, S.M.; Purdue, M.P.; Abnet, C.C.; Stolzenberg-Solomon, R.; Miller, G.; et al. Human oral microbiome and prospective risk for pancreatic cancer: A population-based nested case-control study. Gut 2018, 67, 120–127. [Google Scholar] [CrossRef] [PubMed]
- Olsen, I.; Yilmaz, O. Possible role of Porphyromonas gingivalis in orodigestive cancers. J. Oral Microbiol. 2019, 11, 1563410. [Google Scholar] [CrossRef] [PubMed]
- Zhou, L.; Dorfer, C.E.; Chen, L.; Fawzy El-Sayed, K.M. Porphyromonas gingivalis lipopolysaccharides affect gingival stem/progenitor cells attributes through NF-kappaB, but not Wnt/beta-catenin, pathway. J. Clin. Periodontol. 2017, 44, 1112–1122. [Google Scholar] [CrossRef] [PubMed]
- Al-Attar, A.; Alimova, Y.; Kirakodu, S.; Kozal, A.; Novak, M.J.; Stromberg, A.J.; Orraca, L.; Gonzalez-Martinez, J.; Martinez, M.; Ebersole, J.L.; et al. Activation of Notch-1 in oral epithelial cells by P. gingivalis triggers the expression of the antimicrobial protein PLA(2)-IIA. Mucosal Immunol. 2018, 11, 1047–1059. [Google Scholar] [CrossRef] [PubMed]
- Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013, 342, 967–970. [Google Scholar] [CrossRef] [PubMed]
- Vetizou, M.; Pitt, J.M.; Daillere, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef]
- 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]
- Burotto, M.; Wilkerson, J.; Stein, W.D.; Bates, S.E.; Fojo, T. Adjuvant and neoadjuvant cancer therapies: A historical review and a rational approach to understand outcomes. Semin. Oncol. 2019, 46, 83–99. [Google Scholar] [CrossRef]
- Liedtke, C.; Mazouni, C.; Hess, K.R.; Andre, F.; Tordai, A.; Mejia, J.A.; Symmans, W.F.; Gonzalez-Angulo, A.M.; Hennessy, B.; Green, M.; et al. Response to neoadjuvant therapy and long-term survival in patients with triple-negative breast cancer. J. Clin. Oncol. 2008, 26, 1275–1281. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez-Angulo, A.M.; Morales-Vasquez, F.; Hortobagyi, G.N. Overview of resistance to systemic therapy in patients with breast cancer. Adv. Exp. Med. Biol. 2007, 608, 1–22. [Google Scholar] [CrossRef] [PubMed]
- Einhorn, L.H. Treatment of testicular cancer: A new and improved model. J. Clin. Oncol. 1990, 8, 1777–1781. [Google Scholar] [CrossRef] [PubMed]
- Poletto, S.; Novo, M.; Paruzzo, L.; Frascione, P.M.M.; Vitolo, U. Treatment strategies for patients with diffuse large B-cell lymphoma. Cancer Treat. Rev. 2022, 110, 102443. [Google Scholar] [CrossRef] [PubMed]
- Anand, U.; Dey, A.; Chandel, A.K.S.; Sanyal, R.; Mishra, A.; Pandey, D.K.; De Falco, V.; Upadhyay, A.; Kandimalla, R.; Chaudhary, A.; et al. Cancer chemotherapy and beyond: Current status, drug candidates, associated risks and progress in targeted therapeutics. Genes Dis. 2023, 10, 1367–1401. [Google Scholar] [CrossRef] [PubMed]
- Alexander, J.L.; Wilson, I.D.; Teare, J.; Marchesi, J.R.; Nicholson, J.K.; Kinross, J.M. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 356–365. [Google Scholar] [CrossRef] [PubMed]
- Li, N.; Bai, C.; Zhao, L.; Sun, Z.; Ge, Y.; Li, X. The Relationship Between Gut Microbiome Features and Chemotherapy Response in Gastrointestinal Cancer. Front. Oncol. 2021, 11, 781697. [Google Scholar] [CrossRef]
- 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]
- Shuwen, H.; Xi, Y.; Yuefen, P.; Jiamin, X.; Quan, Q.; Haihong, L.; Yizhen, J.; Wei, W. Effects of postoperative adjuvant chemotherapy and palliative chemotherapy on the gut microbiome in colorectal cancer. Microb. Pathog. 2020, 149, 104343. [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]
- Viaud, S.; Saccheri, F.; Mignot, G.; Yamazaki, T.; Daillere, 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]
- Dizman, N.; Hsu, J.; Bergerot, P.G.; Gillece, J.D.; Folkerts, M.; Reining, L.; Trent, J.; Highlander, S.K.; Pal, S.K. Randomized trial assessing impact of probiotic supplementation on gut microbiome and clinical outcome from targeted therapy in metastatic renal cell carcinoma. Cancer Med. 2021, 10, 79–86. [Google Scholar] [CrossRef] [PubMed]
- Daillere, R.; Vetizou, 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] [PubMed]
- 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]
- Yamamura, K.; Izumi, D.; Kandimalla, R.; Sonohara, F.; Baba, Y.; Yoshida, N.; Kodera, Y.; Baba, H.; Goel, A. Intratumoral Fusobacterium nucleatum Levels Predict Therapeutic Response to Neoadjuvant Chemotherapy in Esophageal Squamous Cell Carcinoma. Clin. Cancer Res. 2019, 25, 6170–6179. [Google Scholar] [CrossRef] [PubMed]
- Sims, T.T.; El Alam, M.B.; Karpinets, T.V.; Dorta-Estremera, S.; Hegde, V.L.; Nookala, S.; Yoshida-Court, K.; Wu, X.; Biegert, G.W.G.; Delgado Medrano, A.Y.; et al. Gut microbiome diversity is an independent predictor of survival in cervical cancer patients receiving chemoradiation. Commun. Biol. 2021, 4, 237. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Teng, H.; Wang, Y.; Sui, X.; Fan, J.; Li, S.; Lei, X.; Shi, C.; Sun, W.; Song, M.; Wang, H.; et al. Gut microbiota-mediated nucleotide synthesis attenuates the response to neoadjuvant chemoradiotherapy in rectal cancer. Cancer Cell 2023, 41, 124–138.e6. [Google Scholar] [CrossRef]
- Bukavina, L.; Ginwala, R.; Sindhani, M.; Prunty, M.; Geynisman, D.; Pooja, G.; Valentine, H.; Calaway, A.; Brown, J.R.; Correa, A.; et al. Role of Gut Microbiome in Neoadjuvant Chemotherapy Response in Urothelial Carcinoma: A Multi-Institutional Prospective Cohort Evaluation. bioRxiv 2023. [Google Scholar] [CrossRef]
- Yi, Y.; Shen, L.; Shi, W.; Xia, F.; Zhang, H.; Wang, Y.; Zhang, J.; Wang, Y.; Sun, X.; Zhang, Z.; et al. Gut Microbiome Components Predict Response to Neoadjuvant Chemoradiotherapy in Patients with Locally Advanced Rectal Cancer: A Prospective, Longitudinal Study. Clin. Cancer Res. 2021, 27, 1329–1340. [Google Scholar] [CrossRef]
- Tsai, H.F.; Hsu, P.N. Cancer immunotherapy by targeting immune checkpoints: Mechanism of T cell dysfunction in cancer immunity and new therapeutic targets. J. Biomed. Sci. 2017, 24, 35. [Google Scholar] [CrossRef] [PubMed]
- Johnson, D.B.; Nebhan, C.A.; Moslehi, J.J.; Balko, J.M. Immune-checkpoint inhibitors: Long-term implications of toxicity. Nat. Rev. Clin. Oncol. 2022, 19, 254–267. [Google Scholar] [CrossRef] [PubMed]
- Reckova, M.; Mladosievicova, B. An ongoing evolution of cardio-oncology with the rapid development of modern immunotherapy. Int. J. Cardiol. 2022, 347, 60–61. [Google Scholar] [CrossRef]
- Zomborska, E.; Kasperova, S.; Slopovsky, J.; Pazderova, N.; Kasperova, B.; Penz, P.; Nyitrayova, O.; Salek, T.; Porsok, S.; Mladosievicova, B.; et al. Fatal myocarditis after the first dose of nivolumab. Klin. Onkol. 2022, 35, 486–492. [Google Scholar] [CrossRef] [PubMed]
- Brahmer, J.R.; Abu-Sbeih, H.; Ascierto, P.A.; Brufsky, J.; Cappelli, L.C.; Cortazar, F.B.; Gerber, D.E.; Hamad, L.; Hansen, E.; Johnson, D.B.; et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune checkpoint inhibitor-related adverse events. J. Immunother. Cancer 2021, 9, e002435. [Google Scholar] [CrossRef] [PubMed]
- Lu, Y.; Yuan, X.; Wang, M.; He, Z.; Li, H.; Wang, J.; Li, Q. Gut microbiota influence immunotherapy responses: Mechanisms and therapeutic strategies. J. Hematol. Oncol. 2022, 15, 47. [Google Scholar] [CrossRef] [PubMed]
- Bagchi, S.; Yuan, R.; Engleman, E.G. Immune Checkpoint Inhibitors for the Treatment of Cancer: Clinical Impact and Mechanisms of Response and Resistance. Annu. Rev. Pathol. 2021, 16, 223–249. [Google Scholar] [CrossRef]
- He, Y.; Fu, L.; Li, Y.; Wang, W.; Gong, M.; Zhang, J.; Dong, X.; Huang, J.; Wang, Q.; Mackay, C.R.; et al. Gut microbial metabolites facilitate anticancer therapy efficacy by modulating cytotoxic CD8(+) T cell immunity. Cell Metab. 2021, 33, 988–1000.e7. [Google Scholar] [CrossRef] [PubMed]
- Davar, D.; Dzutsev, A.K.; McCulloch, J.A.; Rodrigues, R.R.; Chauvin, J.M.; Morrison, R.M.; Deblasio, R.N.; Menna, C.; Ding, Q.; Pagliano, O.; et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science 2021, 371, 595–602. [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]
- Peng, Z.; Cheng, S.; Kou, Y.; Wang, Z.; Jin, R.; Hu, H.; Zhang, X.; Gong, J.F.; Li, J.; Lu, M.; et al. The Gut Microbiome Is Associated with Clinical Response to Anti-PD-1/PD-L1 Immunotherapy in Gastrointestinal Cancer. Cancer Immunol. Res. 2020, 8, 1251–1261. [Google Scholar] [CrossRef] [PubMed]
- Di Modica, M.; Gargari, G.; Regondi, V.; Bonizzi, A.; Arioli, S.; Belmonte, B.; De Cecco, L.; Fasano, E.; Bianchi, F.; Bertolotti, A.; et al. Gut Microbiota Condition the Therapeutic Efficacy of Trastuzumab in HER2-Positive Breast Cancer. Cancer Res. 2021, 81, 2195–2206. [Google Scholar] [CrossRef]
- Vernaci, G.; Savarino, E.V.; Patuzzi, I.; Facchin, S.; Zingone, F.; Massa, D.; Faggioni, G.; Giarratano, T.; Miglietta, F.; Griguolo, G.; et al. Characterization of Gut Microbiome Composition in Patients with Triple-Negative Breast Cancer Treated with Neoadjuvant Chemotherapy. Oncologist 2023, 28, e703–e711. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.H.; Cho, S.Y.; Yoon, Y.; Park, C.; Sohn, J.; Jeong, J.J.; Jeon, B.N.; Jang, M.; An, C.; Lee, S.; et al. Bifidobacterium bifidum strains synergize with immune checkpoint inhibitors to reduce tumour burden in mice. Nat. Microbiol. 2021, 6, 277–288. [Google Scholar] [CrossRef] [PubMed]
- Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillere, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [PubMed]
- Vernocchi, P.; Gili, T.; Conte, F.; Del Chierico, F.; Conta, G.; Miccheli, A.; Botticelli, A.; Paci, P.; Caldarelli, G.; Nuti, M.; et al. Network Analysis of Gut Microbiome and Metabolome to Discover Microbiota-Linked Biomarkers in Patients Affected by Non-Small Cell Lung Cancer. Int. J. Mol. Sci. 2020, 21, 8730. [Google Scholar] [CrossRef] [PubMed]
- Hakozaki, T.; Richard, C.; Elkrief, A.; Hosomi, Y.; Benlaifaoui, M.; Mimpen, I.; Terrisse, S.; Derosa, L.; Zitvogel, L.; Routy, B.; et al. The Gut Microbiome Associates with Immune Checkpoint Inhibition Outcomes in Patients with Advanced Non-Small Cell Lung Cancer. Cancer Immunol. Res. 2020, 8, 1243–1250. [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]
- 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]
- 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]
- Ahmad, S.S.; Reinius, M.A.; Hatcher, H.M.; Ajithkumar, T.V. Anticancer chemotherapy in teenagers and young adults: Managing long term side effects. BMJ 2016, 354, i4567. [Google Scholar] [CrossRef] [PubMed]
- Kunika; Frey, N.; Rangrez, A.Y. Exploring the Involvement of Gut Microbiota in Cancer Therapy-Induced Cardiotoxicity. Int. J. Mol. Sci. 2023, 24, 7261. [Google Scholar] [CrossRef] [PubMed]
- Bajic, J.E.; Johnston, I.N.; Howarth, G.S.; Hutchinson, M.R. From the Bottom-Up: Chemotherapy and Gut-Brain Axis Dysregulation. Front. Behav. Neurosci. 2018, 12, 104. [Google Scholar] [CrossRef] [PubMed]
- Erny, D.; Hrabe de Angelis, A.L.; Jaitin, D.; Wieghofer, P.; Staszewski, O.; David, E.; Keren-Shaul, H.; Mahlakoiv, T.; Jakobshagen, K.; Buch, T.; et al. Host microbiota constantly control maturation and function of microglia in the CNS. Nat. Neurosci. 2015, 18, 965–977. [Google Scholar] [CrossRef] [PubMed]
- Ciernikova, S.; Mego, M.; Chovanec, M. Exploring the Potential Role of the Gut Microbiome in Chemotherapy-Induced Neurocognitive Disorders and Cardiovascular Toxicity. Cancers 2021, 13, 782. [Google Scholar] [CrossRef]
- Parker, A.; Fonseca, S.; Carding, S.R. Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health. Gut Microbes 2020, 11, 135–157. [Google Scholar] [CrossRef] [PubMed]
- Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef] [PubMed]
- Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 2018, 1693, 128–133. [Google Scholar] [CrossRef]
- Kong, C.; Liang, L.; Liu, G.; Du, L.; Yang, Y.; Liu, J.; Shi, D.; Li, X.; Ma, Y. Integrated metagenomic and metabolomic analysis reveals distinct gut-microbiome-derived phenotypes in early-onset colorectal cancer. Gut 2023, 72, 1129–1142. [Google Scholar] [CrossRef]
- Gershon, M.D. 5-Hydroxytryptamine (serotonin) in the gastrointestinal tract. Curr. Opin. Endocrinol. Diabetes Obes. 2013, 20, 14–21. [Google Scholar] [CrossRef]
- Shah, P.A.; Park, C.J.; Shaughnessy, M.P.; Cowles, R.A. Serotonin as a Mitogen in the Gastrointestinal Tract: Revisiting a Familiar Molecule in a New Role. Cell. Mol. Gastroenterol. Hepatol. 2021, 12, 1093–1104. [Google Scholar] [CrossRef] [PubMed]
- Perez-Pardo, P.; Dodiya, H.B.; Engen, P.A.; Forsyth, C.B.; Huschens, A.M.; Shaikh, M.; Voigt, R.M.; Naqib, A.; Green, S.J.; Kordower, J.H.; et al. Role of TLR4 in the gut-brain axis in Parkinson’s disease: A translational study from men to mice. Gut 2019, 68, 829–843. [Google Scholar] [CrossRef] [PubMed]
- Janelsins, M.C.; Heckler, C.E.; Peppone, L.J.; Kamen, C.; Mustian, K.M.; Mohile, S.G.; Magnuson, A.; Kleckner, I.R.; Guido, J.J.; Young, K.L.; et al. Cognitive Complaints in Survivors of Breast Cancer After Chemotherapy Compared with Age-Matched Controls: An Analysis from a Nationwide, Multicenter, Prospective Longitudinal Study. J. Clin. Oncol. 2017, 35, 506–514. [Google Scholar] [CrossRef] [PubMed]
- Chovanec, M.; Vasilkova, L.; Setteyova, L.; Obertova, J.; Palacka, P.; Rejlekova, K.; Sycova-Mila, Z.; Kalavska, K.; Svetlovska, D.; Cingelova, S.; et al. Long-Term Cognitive Functioning in Testicular Germ-Cell Tumor Survivors. Oncologist 2018, 23, 617–623. [Google Scholar] [CrossRef]
- Loman, B.R.; Jordan, K.R.; Haynes, B.; Bailey, M.T.; Pyter, L.M. Chemotherapy-induced neuroinflammation is associated with disrupted colonic and bacterial homeostasis in female mice. Sci. Rep. 2019, 9, 16490. [Google Scholar] [CrossRef]
- Shabbir, U.; Tyagi, A.; Elahi, F.; Aloo, S.O.; Oh, D.H. The Potential Role of Polyphenols in Oxidative Stress and Inflammation Induced by Gut Microbiota in Alzheimer’s Disease. Antioxidants 2021, 10, 1370. [Google Scholar] [CrossRef]
- Li, Y.; Peng, Y.; Shen, Y.; Zhang, Y.; Liu, L.; Yang, X. Dietary polyphenols: Regulate the advanced glycation end products-RAGE axis and the microbiota-gut-brain axis to prevent neurodegenerative diseases. Crit. Rev. Food Sci. Nutr. 2023, 63, 9816–9842. [Google Scholar] [CrossRef]
- Filosa, S.; Di Meo, F.; Crispi, S. Polyphenols-gut microbiota interplay and brain neuromodulation. Neural Regen. Res. 2018, 13, 2055–2059. [Google Scholar] [CrossRef]
- Figueira, I.; Garcia, G.; Pimpao, R.C.; Terrasso, A.P.; Costa, I.; Almeida, A.F.; Tavares, L.; Pais, T.F.; Pinto, P.; Ventura, M.R.; et al. Polyphenols journey through blood-brain barrier towards neuronal protection. Sci. Rep. 2017, 7, 11456. [Google Scholar] [CrossRef]
- Duenas, M.; Munoz-Gonzalez, I.; Cueva, C.; Jimenez-Giron, A.; Sanchez-Patan, F.; Santos-Buelga, C.; Moreno-Arribas, M.V.; Bartolome, B. A survey of modulation of gut microbiota by dietary polyphenols. Biomed. Res. Int. 2015, 2015, 850902. [Google Scholar] [CrossRef]
- Cross, C.; Davies, M.; Bateman, E.; Crame, E.; Joyce, P.; Wignall, A.; Ariaee, A.; Gladman, M.; Wardill, H.; Bowen, J. Fibre-rich diet attenuates chemotherapy-related neuroinflammation in mice. Brain Behav. Immun. 2023, 115, 13–25. [Google Scholar] [CrossRef] [PubMed]
- Venkidesh, B.S.; Narasimhamurthy, R.K.; Jnana, A.; Reghunathan, D.; Sharan, K.; Chandraguthi, S.G.; Saigal, M.; Murali, T.S.; Mumbrekar, K.D. Pelvic irradiation induces behavioural and neuronal damage through gut dysbiosis in a rat model. Chem. Biol. Interact. 2023, 386, 110775. [Google Scholar] [CrossRef] [PubMed]
- Smith, M.; Dai, A.; Ghilardi, G.; Amelsberg, K.V.; Devlin, S.M.; Pajarillo, R.; Slingerland, J.B.; Beghi, S.; Herrera, P.S.; Giardina, P.; et al. Author Correction: Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat. Med. 2023, 29, 2954. [Google Scholar] [CrossRef] [PubMed]
- Chovanec, M.; Kalavska, K.; Obertova, J.; Palacka, P.; Rejlekova, K.; Sycova-Mila, Z.; Orszaghova, Z.; Lesko, P.; De Angelis, V.; Vasilkova, L.; et al. Cognitive impairment and biomarkers of gut microbial translocation in testicular germ cell tumor survivors. Front. Oncol. 2023, 13, 1146032. [Google Scholar] [CrossRef] [PubMed]
- Lyon, A.R.; Lopez-Fernandez, T.; Couch, L.S.; Asteggiano, R.; Aznar, M.C.; Bergler-Klein, J.; Boriani, G.; Cardinale, D.; Cordoba, R.; Cosyns, B.; et al. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur. Heart J. 2022, 43, 4229–4361. [Google Scholar] [CrossRef]
- Couch, L.S.; Lyon, A.R.; Lopez-Fernandez, T. Cardio-oncology: A new field requiring guidance. Eur. Heart J. Cardiovasc. Imaging 2023, 24, e47. [Google Scholar] [CrossRef]
- Drobni, Z.D.; Alvi, R.M.; Taron, J.; Zafar, A.; Murphy, S.P.; Rambarat, P.K.; Mosarla, R.C.; Lee, C.; Zlotoff, D.A.; Raghu, V.K.; et al. Association Between Immune Checkpoint Inhibitors with Cardiovascular Events and Atherosclerotic Plaque. Circulation 2020, 142, 2299–2311. [Google Scholar] [CrossRef]
- Mladosievicova, B.; Petrikova, L.; Valaskova, Z.; Bernadic, M., Jr.; Chovanec, M.; Mego, M.; Bernadic, M., Sr. Atherosclerosis in cancer patients. Bratisl. Lek. Listy 2019, 120, 636–640. [Google Scholar] [CrossRef]
- Mertens, A.C.; Yasui, Y.; Neglia, J.P.; Potter, J.D.; Nesbit, M.E., Jr.; Ruccione, K.; Smithson, W.A.; Robison, L.L. Late mortality experience in five-year survivors of childhood and adolescent cancer: The Childhood Cancer Survivor Study. J. Clin. Oncol. 2001, 19, 3163–3172. [Google Scholar] [CrossRef]
- Cespedes Feliciano, E.M.; Chen, W.Y.; Bradshaw, P.T.; Prado, C.M.; Alexeeff, S.; Albers, K.B.; Castillo, A.L.; Caan, B.J. Adipose Tissue Distribution and Cardiovascular Disease Risk among Breast Cancer Survivors. J. Clin. Oncol. 2019, 37, 2528–2536. [Google Scholar] [CrossRef]
- Sturgeon, K.M.; Deng, L.; Bluethmann, S.M.; Zhou, S.; Trifiletti, D.M.; Jiang, C.; Kelly, S.P.; Zaorsky, N.G. A population-based study of cardiovascular disease mortality risk in US cancer patients. Eur. Heart J. 2019, 40, 3889–3897. [Google Scholar] [CrossRef] [PubMed]
- Grilz, E.; Posch, F.; Nopp, S.; Konigsbrugge, O.; Lang, I.M.; Klimek, P.; Thurner, S.; Pabinger, I.; Ay, C. Relative risk of arterial and venous thromboembolism in persons with cancer vs. persons without cancer-a nationwide analysis. Eur. Heart J. 2021, 42, 2299–2307. [Google Scholar] [CrossRef] [PubMed]
- Florido, R.; Daya, N.R.; Ndumele, C.E.; Koton, S.; Russell, S.D.; Prizment, A.; Blumenthal, R.S.; Matsushita, K.; Mok, Y.; Felix, A.S.; et al. Cardiovascular Disease Risk among Cancer Survivors: The Atherosclerosis Risk in Communities (ARIC) Study. J. Am. Coll. Cardiol. 2022, 80, 22–32. [Google Scholar] [CrossRef] [PubMed]
- Yamashita, T.; Kasahara, K.; Emoto, T.; Matsumoto, T.; Mizoguchi, T.; Kitano, N.; Sasaki, N.; Hirata, K. Intestinal Immunity and Gut Microbiota as Therapeutic Targets for Preventing Atherosclerotic Cardiovascular Diseases. Circ. J. 2015, 79, 1882–1890. [Google Scholar] [CrossRef] [PubMed]
- Zollner, J.; Howe, L.G.; Edey, L.F.; O’Dea, K.P.; Takata, M.; Gordon, F.; Leiper, J.; Johnson, M.R. The response of the innate immune and cardiovascular systems to LPS in pregnant and nonpregnant mice. Biol. Reprod. 2017, 97, 258–272. [Google Scholar] [CrossRef]
- Sun, W.; Du, D.; Fu, T.; Han, Y.; Li, P.; Ju, H. Alterations of the Gut Microbiota in Patients with Severe Chronic Heart Failure. Front. Microbiol. 2021, 12, 813289. [Google Scholar] [CrossRef] [PubMed]
- Halaweish, H.F.; Boatman, S.; Staley, C. Encapsulated Fecal Microbiota Transplantation: Development, Efficacy, and Clinical Application. Front. Cell. Infect. Microbiol. 2022, 12, 826114. [Google Scholar] [CrossRef] [PubMed]
- Peri, F.; Granucci, F.; Weiss, J. Endotoxin, TLR4 signaling and beyond. Mol. Immunol. 2015, 63, 125–126. [Google Scholar] [CrossRef]
- Wang, L.; Chen, Q.; Qi, H.; Wang, C.; Wang, C.; Zhang, J.; Dong, L. Doxorubicin-Induced Systemic Inflammation Is Driven by Upregulation of Toll-like Receptor TLR4 and Endotoxin Leakage. Cancer Res. 2016, 76, 6631–6642. [Google Scholar] [CrossRef]
- Herrema, H.; Nieuwdorp, M.; Groen, A.K. Microbiome and Cardiovascular Disease. In Prevention and Treatment of Atherosclerosis: Improving State-of-the-Art Management and Search for Novel Targets; von Eckardstein, A., Binder, C.J., Eds.; Springer: Cham, Switzerland, 2022; pp. 311–334. [Google Scholar]
- Zhu, W.; Wang, Z.; Tang, W.H.W.; Hazen, S.L. Gut Microbe-Generated Trimethylamine N-Oxide From Dietary Choline Is Prothrombotic in Subjects. Circulation 2017, 135, 1671–1673. [Google Scholar] [CrossRef]
- Tang, W.H.; Hazen, S.L. The contributory role of gut microbiota in cardiovascular disease. J. Clin. Investig. 2014, 124, 4204–4211. [Google Scholar] [CrossRef] [PubMed]
- Heianza, Y.; Ma, W.; Manson, J.E.; Rexrode, K.M.; Qi, L. Gut Microbiota Metabolites and Risk of Major Adverse Cardiovascular Disease Events and Death: A Systematic Review and Meta-Analysis of Prospective Studies. J. Am. Heart Assoc. 2017, 6, e004947. [Google Scholar] [CrossRef] [PubMed]
- Velasquez, M.T.; Ramezani, A.; Manal, A.; Raj, D.S. Trimethylamine N-Oxide: The Good, the Bad and the Unknown. Toxins 2016, 8, 326. [Google Scholar] [CrossRef] [PubMed]
- Washburn, R.L.; Cox, J.E.; Muhlestein, J.B.; May, H.T.; Carlquist, J.F.; Le, V.T.; Anderson, J.L.; Horne, B.D. Pilot Study of Novel Intermittent Fasting Effects on Metabolomic and Trimethylamine N-oxide Changes During 24-hour Water-Only Fasting in the FEELGOOD Trial. Nutrients 2019, 11, 246. [Google Scholar] [CrossRef] [PubMed]
- Mueller, D.M.; Allenspach, M.; Othman, A.; Saely, C.H.; Muendlein, A.; Vonbank, A.; Drexel, H.; von Eckardstein, A. Plasma levels of trimethylamine-N-oxide are confounded by impaired kidney function and poor metabolic control. Atherosclerosis 2015, 243, 638–644. [Google Scholar] [CrossRef] [PubMed]
- Aldana-Hernandez, P.; Leonard, K.A.; Zhao, Y.Y.; Curtis, J.M.; Field, C.J.; Jacobs, R.L. Dietary Choline or Trimethylamine N-oxide Supplementation Does Not Influence Atherosclerosis Development in Ldlr−/− and Apoe−/− Male Mice. J. Nutr. 2020, 150, 249–255. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Ma, Y.; Zhong, Y.; Liu, Q.; Chen, C.; Qiang, L.; Wang, X. KALRN mutations promote antitumor immunity and immunotherapy response in cancer. J. Immunother. Cancer 2020, 8, e000293. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.; Liu, Y.; Wang, Y.; Chen, X.; Wang, C.; Chen, X.; Yuan, X.; Liu, L.; Yang, J.; Zhou, X. Prevotellaceae produces butyrate to alleviate PD-1/PD-L1 inhibitor-related cardiotoxicity via PPARalpha-CYP4X1 axis in colonic macrophages. J. Exp. Clin. Cancer Res. 2022, 41, 1. [Google Scholar] [CrossRef]
- Kalyanaraman, B. Teaching the basics of the mechanism of doxorubicin-induced cardiotoxicity: Have we been barking up the wrong tree? Redox. Biol. 2020, 29, 101394. [Google Scholar] [CrossRef]
- Lin, H.; Meng, L.; Sun, Z.; Sun, S.; Huang, X.; Lin, N.; Zhang, J.; Lu, W.; Yang, Q.; Chi, J.; et al. Yellow Wine Polyphenolic Compound Protects against Doxorubicin-Induced Cardiotoxicity by Modulating the Composition and Metabolic Function of the Gut Microbiota. Circ. Heart Fail. 2021, 14, e008220. [Google Scholar] [CrossRef]
- Ciernikova, S.; Sevcikova, A.; Drgona, L.; Mego, M. Modulating the gut microbiota by probiotics, prebiotics, postbiotics, and fecal microbiota transplantation: An emerging trend in cancer patient care. Biochim. Biophys. Acta Rev. Cancer 2023, 1878, 188990. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Xia, Y.; Chen, H.; Hong, L.; Feng, J.; Yang, J.; Yang, Z.; Shi, C.; Wu, W.; Gao, R.; et al. The effect of perioperative probiotics treatment for colorectal cancer: Short-term outcomes of a randomized controlled trial. Oncotarget 2016, 7, 8432–8440. [Google Scholar] [CrossRef] [PubMed]
- Hassan, H.; Rompola, M.; Glaser, A.W.; Kinsey, S.E.; Phillips, R.S. Systematic review and meta-analysis investigating the efficacy and safety of probiotics in people with cancer. Support. Care Cancer 2018, 26, 2503–2509. [Google Scholar] [CrossRef] [PubMed]
- Redman, M.G.; Ward, E.J.; Phillips, R.S. The efficacy and safety of probiotics in people with cancer: A systematic review. Ann. Oncol. 2014, 25, 1919–1929. [Google Scholar] [CrossRef]
- Wardill, H.R.; Van Sebille, Y.Z.A.; Ciorba, M.A.; Bowen, J.M. Prophylactic probiotics for cancer therapy-induced diarrhoea: A meta-analysis. Curr. Opin. Support. Palliat. Care 2018, 12, 187–197. [Google Scholar] [CrossRef]
- Danis, R.; Mego, M.; Antonova, M.; Stepanova, R.; Svobodnik, A.; Hejnova, R.; Wawruch, M. Orally Administered Probiotics in the Prevention of Chemotherapy (±Radiotherapy)-Induced Gastrointestinal Toxicity: A Systematic Review with Meta-Analysis of Randomized Trials. Integr. Cancer Ther. 2022, 21, 15347354221144309. [Google Scholar] [CrossRef]
- Rodriguez-Arrastia, M.; Martinez-Ortigosa, A.; Rueda-Ruzafa, L.; Folch Ayora, A.; Ropero-Padilla, C. Probiotic Supplements on Oncology Patients’ Treatment-Related Side Effects: A Systematic Review of Randomized Controlled Trials. Int. J. Environ. Res. Public Health 2021, 18, 4265. [Google Scholar] [CrossRef]
- Huang, F.; Li, S.; Chen, W.; Han, Y.; Yao, Y.; Yang, L.; Li, Q.; Xiao, Q.; Wei, J.; Liu, Z.; et al. Postoperative Probiotics Administration Attenuates Gastrointestinal Complications and Gut Microbiota Dysbiosis Caused by Chemotherapy in Colorectal Cancer Patients. Nutrients 2023, 15, 356. [Google Scholar] [CrossRef]
- Mego, M.; Chovanec, J.; Vochyanova-Andrezalova, I.; Konkolovsky, P.; Mikulova, M.; Reckova, M.; Miskovska, V.; Bystricky, B.; Beniak, J.; Medvecova, L.; et al. Prevention of irinotecan induced diarrhea by probiotics: A randomized double blind, placebo controlled pilot study. Complement. Ther. Med. 2015, 23, 356–362. [Google Scholar] [CrossRef]
- Mego, M.; Danis, R.; Chovanec, J.; Jurisova, S.; Bystricky, B.; Porsok, S.; Konkolovsky, P.; Vaclav, V.; Wagnerova, M.; Stresko, M.; et al. Randomized double-blind, placebo-controlled multicenter phase III study of prevention of irinotecan-induced diarrhea by a probiotic mixture containing Bifidobacterium BB-12((R))Lactobacillus rhamnosus LGG((R)) in colorectal cancer patients. Front. Oncol. 2023, 13, 1168654. [Google Scholar] [CrossRef]
- Lee, J.Y.; Chu, S.H.; Jeon, J.Y.; Lee, M.K.; Park, J.H.; Lee, D.C.; Lee, J.W.; Kim, N.K. Effects of 12 weeks of probiotic supplementation on quality of life in colorectal cancer survivors: A double-blind, randomized, placebo-controlled trial. Dig. Liver Dis. 2014, 46, 1126–1132. [Google Scholar] [CrossRef] [PubMed]
- Venkidesh, B.S.; Shankar, S.R.; Narasimhamurthy, R.K.; Rao, S.B.S.; Mumbrekar, K.D. Radioprotective potential of probiotics against gastrointestinal and neuronal toxicity: A preclinical study. Clin. Transl. Oncol. 2023, 25, 3165–3173. [Google Scholar] [CrossRef] [PubMed]
- Xia, C.; Jiang, C.; Li, W.; Wei, J.; Hong, H.; Li, J.; Feng, L.; Wei, H.; Xin, H.; Chen, T. A Phase II Randomized Clinical Trial and Mechanistic Studies Using Improved Probiotics to Prevent Oral Mucositis Induced by Concurrent Radiotherapy and Chemotherapy in Nasopharyngeal Carcinoma. Front. Immunol. 2021, 12, 618150. [Google Scholar] [CrossRef] [PubMed]
- Mirza, M.A.; Aruna, D.; Irukulla, M. Efficacy of Bacillus clausii UBBC—07 spores in the amelioration of oral mucositis in head and neck cancer patients undergoing radiation therapy. Cancer Treat. Res. Commun. 2022, 31, 100523. [Google Scholar] [CrossRef] [PubMed]
- Lages, P.C.; Generoso, S.V.; Correia, M. Postoperative symbiotic in patients with head and neck cancer: A double-blind randomised trial. Br. J. Nutr. 2018, 119, 190–195. [Google Scholar] [CrossRef]
- DE Sanctis, V.; Belgioia, L.; Cante, D.; MR, L.A.P.; Caspiani, O.; Guarnaccia, R.; Argenone, A.; Muto, P.; Musio, D.; Felice, D.E.F.; et al. Lactobacillus brevis CD2 for Prevention of Oral Mucositis in Patients with Head and Neck Tumors: A Multicentric Randomized Study. Anticancer Res. 2019, 39, 1935–1942. [Google Scholar] [CrossRef] [PubMed]
- Tian, Y.; Li, M.; Song, W.; Jiang, R.; Li, Y.Q. Effects of probiotics on chemotherapy in patients with lung cancer. Oncol. Lett. 2019, 17, 2836–2848. [Google Scholar] [CrossRef]
- Cui, M.; Xiao, H.; Li, Y.; Zhou, L.; Zhao, S.; Luo, D.; Zheng, Q.; Dong, J.; Zhao, Y.; Zhang, X.; et al. Faecal microbiota transplantation protects against radiation-induced toxicity. EMBO Mol. Med. 2017, 9, 448–461. [Google Scholar] [CrossRef]
- 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 2021, 371, 602–609. [Google Scholar] [CrossRef]
ClinicalTrials.gov Identifier: | Study Design | Malignancy | Purpose | Patients (n) | Intervention Model Description | Study Status |
---|---|---|---|---|---|---|
NCT05155618 | An interventional randomized study, crossover assignment | Prostate cancer | To evaluate the shifts in the microbiome and correlation with changes in cytokines and adipokines, particularly in the context of late-onset toxicity | 300 adults | Participants in the intervention group will meet a dietitian and physiotherapist to obtain personalized diet and exercise recommendations. Patients undergoing radiotherapy will receive a 6-month intervention followed by a 6-month follow-up. | Recruiting |
NCT04775355 | An observational prospective study | Prostate cancer | To analyze gut microbiome during androgen deprivation therapy and radiotherapy and reveal changes in the gut microbial community related to late toxicity | 30 adults | Participants will complete questionnaires prior to radiotherapy, mid-way through therapy, and after completion of radiotherapy. | Recruiting |
NCT03294122 | An observational prospective study | Head and neck cancer/prostate cancer | To examine how intestinal/salivary microbiomes affect toxicity | 400 adults | Anti-cancer therapies will include radiotherapy with possible adjuvant hormone therapies/concomitant chemotherapies. CTCAE will define a grading system for late toxicities. | Unknown |
NCT05349227 | An interventional randomized, open-label study, crossover assignment | Ovarian cancer/ breast cancer/ lung cancer/ gastric cancer | To monitor changes in depression, cognitive function or impairment, sleep-related impairment and analyze the gut microbiome in fecal samples at baseline of study enrollment and month 6 following enrollment | 660 adults | Patients will be divided into wrist-worn devices monitored groups where they will receive either 6 months of digital coaching immediately followed by 6 months of monitoring or 6 months of monitoring followed by 6 months of digital health coaching. | Recruiting |
NCT06098404 | An observational prospective study | Cancer | To study the composition of gut microbiome and correlate fatigue incidence with gut microbiome composition | 250 adults | The cancer-related cognitive impairment will be monitored using MoCA consisting of 9 questions (focused on memory, attention, language, abstraction, delayed recall, and orientation), while fatigue and cognition with FACS (20-question assessment). | Not yet recruiting |
NCT06050733 | An observational cross-sectional study | Solid cancer | To observe the associations between cognition and fatigue with gut microbiome composition | 16 adults | Fatigue and cognition will be evaluated by FACS (20-question assessment), MFSI-SF (30-question assessment), and MoCA consisting of 9 questions in patients receiving standard therapy with PD-1/PD-L1 blockade. | Recruiting |
NCT04691284 | An interventional prospective, single-center, non-randomized, open-label study, single-group assignment | Hematologic malignancies | To compare microbiome alternations with quality of life, spirituality, and cognitive functions | 100 adults | Enrolled patients will receive high-dose chemotherapy and hematopoietic cell transplantation or CAR-T cell therapy. | Recruiting |
NCT06088940 | An interventional double-blinded, placebo-controlled, randomized study, parallel assignment | Cancer | To investigate probiotics vs. placebo effect on gut taxons and correlate bacterial operational taxonomic units with gastrointestinal and psychosocial functions | 66 adults | Cancer patient survivors will receive one probiotic (Lactobacillus and Bifidobacterium strains)/placebo (maltodextrin) capsule daily for 12 weeks. The effect of probiotics on diarrhea/gas/bloating/anxiety/fatigue symptoms and cognitive function will be measured. | Not yet recruiting |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ciernikova, S.; Sevcikova, A.; Mladosievicova, B.; Mego, M. Microbiome in Cancer Development and Treatment. Microorganisms 2024, 12, 24. https://doi.org/10.3390/microorganisms12010024
Ciernikova S, Sevcikova A, Mladosievicova B, Mego M. Microbiome in Cancer Development and Treatment. Microorganisms. 2024; 12(1):24. https://doi.org/10.3390/microorganisms12010024
Chicago/Turabian StyleCiernikova, Sona, Aneta Sevcikova, Beata Mladosievicova, and Michal Mego. 2024. "Microbiome in Cancer Development and Treatment" Microorganisms 12, no. 1: 24. https://doi.org/10.3390/microorganisms12010024