Intestinal Microbiota: A Novel Target to Improve Anti-Tumor Treatment?
Abstract
:1. Introduction
2. Intestinal Microbiota Impacts on Efficacy of Cancer Therapies
2.1. Chemotherapy
2.2. Immunotherapy
2.3. Radiotherapy
2.4. Surgery
3. Modulation of the Gut Microbiome to Enhance Therapy Efficacy: from Predictive Potential to Clinical Assays
3.1. Antibiotics
3.2. Fecal Microbiota Transplantation (FMT)
3.3. Probiotics and Prebiotics
3.3.1. Probiotics
3.3.2. Prebiotics and Synbiotics
3.4. Physical Activity
4. Conclusions & Prospects
Funding
Conflicts of Interest
Abbreviations
AL | Anastomotic leaks |
ATB | Antibiotic |
β-gluc-PB | β-glucuronidase-producing bacteria |
BIRC3 | Baculoviral IAP Repeat Containing 3 |
BVU | 5-(2-bromovinyl)uracil |
CRC | Colorectal cancer |
CTLA-4 | Cytotoxic T-lymphocyte-associated protein 4 |
CTX | Cyclophosphamide |
DCs | Dentritic cells |
FAO/WHO | Food and Agriculture Organization/World Health Organization |
FMT | Fecal Microbiota Transplantation |
GTB | Gemcitabine |
GF | Germ-free |
ICI | Immune checkpoint inhibitor |
IL | Interleukin |
LGG | Lactobacillus rhamnosus GG |
LPS | Lipopolysaccharides |
MMP | Metalloproteinase |
NF-κB | Nuclear factor-kappa B |
ODN | oligodeoxynucleotides |
OXA | Oxaliplatin |
PA | Physical activity |
PD-1 | Programmed cell death 1 |
PD-L1 | Programmed death-ligand 1 |
RNS | Reactive nitrogen species |
ROS | Reactive oxygen species |
SPF | Specific Pathogen Free |
Tc | Cytotoxic T cell |
Th | T helper cell |
TILs | Tumor infiltrating-lymphocytes |
TLR | Toll-like-receptor |
TNF | Tumor Necrosis Factor |
Treg | Regulatory T cell |
5-FU | 5- fluorouracil |
References
- Collins, D.; Hogan, A.M.; Winter, D.C. Microbial and viral pathogens in colorectal cancer. Lancet Oncol. 2011, 12, 504–512. [Google Scholar] [CrossRef]
- Zhang, Y.-J.; Li, S.; Gan, R.-Y.; Zhou, T.; Xu, D.-P.; Li, H.-B. Impacts of Gut Bacteria on Human Health and Diseases. Int. J. Mol. Sci. 2015, 16, 7493–7519. [Google Scholar] [CrossRef] [PubMed]
- Lin, L.; Zhang, J. Role of intestinal microbiota and metabolites on gut homeostasis and human diseases. BMC Immunol. 2017, 18, 2. [Google Scholar] [CrossRef] [PubMed]
- Schwabe, R.F.; Jobin, C. The microbiome and cancer. Nat. Rev. Cancer 2013, 13, 800–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Elinav, E.; Garrett, W.S.; Trinchieri, G.; Wargo, J. The cancer microbiome. Nat. Rev. Cancer 2019, 19, 371–376. [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]
- Zitvogel, L.; Daillère, R.; Roberti, M.P.; Routy, B.; Kroemer, G. Anticancer effects of the microbiome and its products. Nat. Rev. Microbiol. 2017, 15, 465–478. [Google Scholar] [CrossRef] [PubMed]
- Scott, A.J.; Merrifield, C.A.; Younes, J.A.; Pekelharing, E.P. Pre-, pro- and synbiotics in cancer prevention and treatment—A review of basic and clinical research. Ecancermedicalscience 2018, 12. [Google Scholar] [CrossRef] [PubMed]
- Bashiardes, S.; Tuganbaev, T.; Federici, S.; Elinav, E. The microbiome in anti-cancer therapy. Semin. Immunol. 2017, 32, 74–81. [Google Scholar] [CrossRef]
- 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]
- 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] [Green Version]
- Ma, W.; Mao, Q.; Xia, W.; Dong, G.; Yu, C.; Jiang, F. Gut Microbiota Shapes the Efficiency of Cancer Therapy. Front. Microbiol. 2019, 10. [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]
- Guthrie, L.; Gupta, S.; Daily, J.; Kelly, L. Human microbiome signatures of differential colorectal cancer drug metabolism. NPJ Biofilms Microbiomes 2017, 3, 27. [Google Scholar] [CrossRef] [PubMed]
- Nakayama, H.; Kinouchi, T.; Kataoka, K.; Akimoto, S.; Matsuda, Y.; Ohnishi, Y. Intestinal anaerobic bacteria hydrolyse sorivudine, producing the high blood concentration of 5-(E)-(2-bromovinyl)uracil that increases the level and toxicity of 5-fluorouracil. Pharmacogenetics 1997, 7, 35–43. [Google Scholar] [CrossRef] [PubMed]
- Wallace, B.D.; Wang, H.; Lane, K.T.; Scott, J.E.; Orans, J.; Koo, J.S.; Venkatesh, M.; Jobin, C.; Yeh, L.-A.; Mani, S.; et al. Alleviating Cancer Drug Toxicity by Inhibiting a Bacterial Enzyme. Science 2010, 330, 831–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, J.; Feng, W.; Zhang, S.; Chen, L.; Tang, F.; Sheng, Y.; Ao, H.; Peng, C. Gut microbial modulation in the treatment of chemotherapy-induced diarrhea with Shenzhu Capsule. BMC Complement. Altern. Med. 2019, 19, 126. [Google Scholar] [CrossRef] [PubMed]
- Diasio, R.B. Sorivudine and 5-fluorouracil; a clinically significant drug-drug interaction due to inhibition of dihydropyrimidine dehydrogenase. Br. J. Clin. Pharmacol. 1998, 46, 1–4. [Google Scholar] [CrossRef] [PubMed]
- Brandi, G.; Dabard, J.; Raibaud, P.; Di Battista, M.; Bridonneau, C.; Pisi, A.M.; Morselli Labate, A.M.; Pantaleo, M.A.; De Vivo, A.; Biasco, G. Intestinal microflora and digestive toxicity of irinotecan in mice. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2006, 12, 1299–1307. [Google Scholar] [CrossRef]
- Stringer, A.M.; Gibson, R.J.; Logan, R.M.; Bowen, J.M.; Yeoh, A.S.J.; Hamilton, J.; Keefe, D.M.K. Gastrointestinal microflora and mucins may play a critical role in the development of 5-Fluorouracil-induced gastrointestinal mucositis. Exp. Biol. Med. Maywood NJ 2009, 234, 430–441. [Google Scholar] [CrossRef]
- Rigby, R.J.; Carr, J.; Orgel, K.; King, S.L.; Lund, P.K.; Dekaney, C.M. Intestinal bacteria are necessary for doxorubicin-induced intestinal damage but not for doxorubicin-induced apoptosis. Gut Microbes 2016, 7, 414–423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stringer, A.M.; Gibson, R.J.; Bowen, J.M.; Logan, R.M.; Ashton, K.; Yeoh, A.S.J.; Al-Dasooqi, N.; Keefe, D.M.K. Irinotecan-induced mucositis manifesting as diarrhoea corresponds with an amended intestinal flora and mucin profile. Int. J. Exp. Pathol. 2009, 90, 489–499. [Google Scholar] [CrossRef] [PubMed]
- Fijlstra, M.; Ferdous, M.; Koning, A.M.; Rings, E.H.H.M.; Harmsen, H.J.M.; Tissing, W.J.E. Substantial decreases in the number and diversity of microbiota during chemotherapy-induced gastrointestinal mucositis in a rat model. Support. Care Cancer 2015, 23, 1513–1522. [Google Scholar] [CrossRef] [PubMed]
- Hong, B.-Y.; Sobue, T.; Choquette, L.; Dupuy, A.K.; Thompson, A.; Burleson, J.A.; Salner, A.L.; Schauer, P.K.; Joshi, P.; Fox, E.; et al. Chemotherapy-induced oral mucositis is associated with detrimental bacterial dysbiosis. Microbiome 2019, 7, 66. [Google Scholar] [CrossRef] [PubMed]
- Montassier, E.; Gastinne, T.; Vangay, P.; Al-Ghalith, G.A.; Bruley des Varannes, S.; Massart, S.; Moreau, P.; Potel, G.; de La Cochetière, M.F.; Batard, E.; et al. Chemotherapy-driven dysbiosis in the intestinal microbiome. Aliment. Pharmacol. Ther. 2015, 42, 515–528. [Google Scholar] [CrossRef]
- Montassier, E.; Batard, E.; Massart, S.; Gastinne, T.; Carton, T.; Caillon, J.; Le Fresne, S.; Caroff, N.; Hardouin, J.B.; Moreau, P.; et al. 16S rRNA gene pyrosequencing reveals shift in patient faecal microbiota during high-dose chemotherapy as conditioning regimen for bone marrow transplantation. Microb. Ecol. 2014, 67, 690–699. [Google Scholar] [CrossRef] [PubMed]
- Rooks, M.G.; Veiga, P.; Wardwell-Scott, L.H.; Tickle, T.; Segata, N.; Michaud, M.; Gallini, C.A.; Beal, C.; van Hylckama-Vlieg, J.E.T.; Ballal, S.A.; et al. Gut microbiome composition and function in experimental colitis during active disease and treatment-induced remission. ISME J. 2014, 8, 1403–1417. [Google Scholar] [CrossRef]
- Van Vliet, M.J.; Tissing, W.J.E.; Dun, C.A.J.; Meessen, N.E.L.; Kamps, W.A.; de Bont, E.S.J.M.; Harmsen, H.J.M. Chemotherapy treatment in pediatric patients with acute myeloid leukemia receiving antimicrobial prophylaxis leads to a relative increase of colonization with potentially pathogenic bacteria in the gut. Clin. Infect. Dis. 2009, 49, 262–270. [Google Scholar] [CrossRef]
- Wang, Y.; Sun, L.; Chen, S.; Guo, S.; Yue, T.; Hou, Q.; Feng, M.; Xu, H.; Liu, Y.; Wang, P.; et al. The administration of Escherichia coli Nissle 1917 ameliorates irinotecan-induced intestinal barrier dysfunction and gut microbial dysbiosis in mice. Life Sci. 2019, 231, 116529. [Google Scholar] [CrossRef]
- Liu, T.; Wu, Y.; Wang, L.; Pang, X.; Zhao, L.; Yuan, H.; Zhang, C. A More Robust Gut Microbiota in Calorie-Restricted Mice Is Associated with Attenuated Intestinal Injury Caused by the Chemotherapy Drug Cyclophosphamide. mBio 2019, 10, e02903-18. [Google Scholar] [CrossRef] [Green Version]
- Reyna-Figueroa, J.; Barrón-Calvillo, E.; García-Parra, C.; Galindo-Delgado, P.; Contreras-Ochoa, C.; Lagunas-Martínez, A.; Campos-Romero, F.H.; Silva-Estrada, J.A.; Limón-Rojas, A.E. Probiotic Supplementation Decreases Chemotherapy-induced Gastrointestinal Side Effects in Patients With Acute Leukemia. J. Pediatr. Hematol. Oncol. 2019, 41, 468–472. [Google Scholar] [CrossRef]
- Daillère, R.; Vétizou, M.; Waldschmitt, N.; Yamazaki, T.; Isnard, C.; Poirier-Colame, V.; Duong, C.P.M.; Flament, C.; Lepage, P.; Roberti, M.P.; et al. Enterococcus hirae and Barnesiella intestinihominis Facilitate Cyclophosphamide-Induced Therapeutic Immunomodulatory Effects. Immunity 2016, 45, 931–943. [Google Scholar] [CrossRef] [Green Version]
- Kuczma, M.P.; Ding, Z.-C.; Li, T.; Habtetsion, T.; Chen, T.; Hao, Z.; Bryan, L.; Singh, N.; Kochenderfer, J.N.; Zhou, G. The impact of antibiotic usage on the efficacy of chemoimmunotherapy is contingent on the source of tumor-reactive T cells. Oncotarget 2017, 8, 111931–111942. [Google Scholar] [CrossRef] [Green Version]
- Xu, X.; Zhang, X. Effects of cyclophosphamide on immune system and gut microbiota in mice. Microbiol. Res. 2015, 171, 97–106. [Google Scholar] [CrossRef]
- Araya, R.E.; Goldszmid, R.S. Two Bugs a NOD Away from Improving Cancer Therapy Efficacy. Immunity 2016, 45, 714–716. [Google Scholar] [CrossRef] [Green Version]
- Bronckaers, A.; Balzarini, J.; Liekens, S. The cytostatic activity of pyrimidine nucleosides is strongly modulated by Mycoplasma hyorhinis infection: Implications for cancer therapy. Biochem. Pharmacol. 2008, 76, 188–197. [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] [Green Version]
- Geller, L.T.; Straussman, R. Intratumoral bacteria may elicit chemoresistance by metabolizing anticancer agents. Mol. Cell. Oncol. 2018, 5, e1405139. [Google Scholar] [CrossRef]
- Lehouritis, P.; Cummins, J.; Stanton, M.; Murphy, C.T.; McCarthy, F.O.; Reid, G.; Urbaniak, C.; Byrne, W.L.; Tangney, M. Local bacteria affect the efficacy of chemotherapeutic drugs. Sci. Rep. 2015, 5, 14554. [Google Scholar] [CrossRef] [Green Version]
- Vande Voorde, J.; Sabuncuoğlu, S.; Noppen, S.; Hofer, A.; Ranjbarian, F.; Fieuws, S.; Balzarini, J.; Liekens, S. Nucleoside-catabolizing Enzymes in Mycoplasma-infected Tumor Cell Cultures Compromise the Cytostatic Activity of the Anticancer Drug Gemcitabine. J. Biol. Chem. 2014, 289, 13054–13065. [Google Scholar] [CrossRef] [Green Version]
- Yu, T.; Guo, F.; Yu, Y.; Sun, T.; Ma, D.; Han, J.; Qian, Y.; Kryczek, I.; Sun, D.; Nagarsheth, N.; et al. Fusobacterium nucleatum Promotes Chemoresistance to Colorectal Cancer by Modulating Autophagy. Cell 2017, 170, 548–563. [Google Scholar] [CrossRef]
- Zhang, S.; Yang, Y.; Weng, W.; Guo, B.; Cai, G.; Ma, Y.; Cai, S. Fusobacterium nucleatum promotes chemoresistance to 5-fluorouracil by upregulation of BIRC3 expression in colorectal cancer. J. Exp. Clin. Cancer Res. 2019, 38, 14. [Google Scholar] [CrossRef]
- Karasawa, H.; Miura, K.; Fujibuchi, W.; Ishida, K.; Kaneko, N.; Kinouchi, M.; Okabe, M.; Ando, T.; Murata, Y.; Sasaki, H.; et al. Down-regulation of cIAP2 enhances 5-FU sensitivity through the apoptotic pathway in human colon cancer cells. Cancer Sci. 2009, 100, 903–913. [Google Scholar] [CrossRef]
- Paulos, C.M.; Wrzesinski, C.; Kaiser, A.; Hinrichs, C.S.; Chieppa, M.; Cassard, L.; Palmer, D.C.; Boni, A.; Muranski, P.; Yu, Z.; et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J. Clin. Investig. 2007, 117, 2197–2204. [Google Scholar] [CrossRef]
- Uribe-Herranz, M.; Bittinger, K.; Rafail, S.; Guedan, S.; Pierini, S.; Tanes, C.; Ganetsky, A.; Morgan, M.A.; Gill, S.; Tanyi, J.L.; et al. Gut microbiota modulates adoptive cell therapy via CD8α dendritic cells and IL-12. JCI Insight 2018, 3, e94952. [Google Scholar] [CrossRef]
- Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Man Lei, Y.; 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]
- Vetizou, M.; Pitt, J.M.; Daillere, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.M.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef] [Green Version]
- Chaput, N.; Lepage, P.; Coutzac, C.; Soularue, E.; Le Roux, K.; Monot, C.; Boselli, L.; Routier, E.; Cassard, L.; Collins, M.; et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann. Oncol. 2017, 28, 1368–1379. [Google Scholar] [CrossRef]
- 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]
- Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef]
- Tanoue, T.; Morita, S.; Plichta, D.R.; Skelly, A.N.; Suda, W.; Sugiura, Y.; Narushima, S.; Vlamakis, H.; Motoo, I.; Sugita, K.; et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature 2019, 565, 600–605. [Google Scholar] [CrossRef]
- Pitt, J.M.; Vetizou, M.; Waldschmitt, N.; Kroemer, G.; Chamaillard, M.; Boneca, I.G.; Zitvogel, L. Fine-Tuning Cancer Immunotherapy: Optimizing the Gut Microbiome. Cancer Res. 2016, 76, 4602–4607. [Google Scholar] [CrossRef] [Green Version]
- Goldszmid, R.S.; Dzutsev, A.; Viaud, S.; Zitvogel, L.; Restifo, N.P.; Trinchieri, G. Microbiota modulation of myeloid cells in cancer therapy. Cancer Immunol. Res. 2015, 3, 103–109. [Google Scholar] [CrossRef]
- Gorjifard, S.; Goldszmid, R.S. Beating Cancer with a Gut Feeling. Cell Host Microbe 2015, 18, 646–648. [Google Scholar] [CrossRef] [Green Version]
- Dubin, K.; Callahan, M.K.; Ren, B.; Khanin, R.; Viale, A.; Ling, L.; No, D.; Gobourne, A.; Littmann, E.; Huttenhower, C.; et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat. Commun. 2016, 7. [Google Scholar] [CrossRef]
- Frankel, A.E.; Deshmukh, S.; Reddy, A.; Lightcap, J.; Hayes, M.; McClellan, S.; Singh, S.; Rabideau, B.; Glover, T.G.; Roberts, B.; et al. Cancer Immune Checkpoint Inhibitor Therapy and the Gut Microbiota. Integr. Cancer Ther. 2019, 18, 153473541984637. [Google Scholar] [CrossRef]
- Elkrief, A.; El Raichani, L.; Richard, C.; Messaoudene, M.; Belkaid, W.; Malo, J.; Belanger, K.; Miller, W.; Jamal, R.; Letarte, N.; et al. Antibiotics are associated with decreased progression-free survival of advanced melanoma patients treated with immune checkpoint inhibitors. Oncoimmunology 2019, 8, e1568812. [Google Scholar] [CrossRef]
- Roy, S.; Trinchieri, G. Microbiota: A key orchestrator of cancer therapy. Nat. Rev. Cancer 2017, 17, 271–285. [Google Scholar] [CrossRef]
- Baskar, R.; Dai, J.; Wenlong, N.; Yeo, R.; Yeoh, K.-W. Biological response of cancer cells to radiation treatment. Front. Mol. Biosci. 2014, 1, 24. [Google Scholar] [CrossRef]
- Kroemer, G.; Galluzzi, L.; Kepp, O.; Zitvogel, L. Immunogenic cell death in cancer therapy. Annu. Rev. Immunol. 2013, 31, 51–72. [Google Scholar] [CrossRef]
- Barker, H.E.; Paget, J.T.E.; Khan, A.A.; Harrington, K.J. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat. Rev. Cancer 2015, 15, 409–425. [Google Scholar] [CrossRef]
- Cui, M.; Xiao, H.; Luo, D.; Zhang, X.; Zhao, S.; Zheng, Q.; Li, Y.; Zhao, Y.; Dong, J.; Li, H.; et al. Circadian rhythm shapes the gut microbiota affecting host radiosensitivity. Int. J. Mol. Sci. 2016, 17, 1786. [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]
- Kuwahara, Y.; Oikawa, T.; Ochiai, Y.; Roudkenar, M.H.; Fukumoto, M.; Shimura, T.; Ohtake, Y.; Ohkubo, Y.; Mori, S.; Uchiyama, Y.; et al. Enhancement of autophagy is a potential modality for tumors refractory to radiotherapy. Cell Death Dis. 2011, 2, e177. [Google Scholar]
- Digomann, D.; Kurth, I.; Tyutyunnykova, A.; Chen, O.; Löck, S.; Gorodetska, I.; Peitzsch, C.; Skvortsova, I.-I.; Negro, G.; Aschenbrenner, B.; et al. The CD98 Heavy Chain Is a Marker and Regulator of Head and Neck Squamous Cell Carcinoma Radiosensitivity. Clin. Cancer Res. 2019, 25, 3152–3163. [Google Scholar] [CrossRef]
- Digomann, D.; Linge, A.; Dubrovska, A. SLC3A2/CD98hc, autophagy and tumor radioresistance: A link confirmed. Autophagy 2019, 15, 1850–1851. [Google Scholar] [CrossRef]
- Ferreira, M.R.; Muls, A.; Dearnaley, D.P.; Andreyev, H.J.N. Microbiota and radiation-induced bowel toxicity: Lessons from inflammatory bowel disease for the radiation oncologist. Lancet Oncol. 2014, 15, e139–e147. [Google Scholar] [CrossRef]
- Reis Ferreira, M.; Andreyev, J.; Mohammed, K.; Truelove, L.; Gowan, S.M.; Li, J.; Gulliford, S.L.; Marchesi, J.; Dearnaley, D.P. Microbiota and radiotherapy-induced gastrointestinal side-effects (MARS) study: A large pilot study of the microbiome in acute and late radiation enteropathy. Clin. Cancer Res. 2019. [Google Scholar] [CrossRef]
- Gerassy-Vainberg, S.; Blatt, A.; Danin-Poleg, Y.; Gershovich, K.; Sabo, E.; Nevelsky, A.; Daniel, S.; Dahan, A.; Ziv, O.; Dheer, R.; et al. Radiation induces proinflammatory dysbiosis: Transmission of inflammatory susceptibility by host cytokine induction. Gut 2018, 67, 97–107. [Google Scholar] [CrossRef]
- Nam, Y.-D.; Kim, H.J.; Seo, J.-G.; Kang, S.W.; Bae, J.-W. Impact of pelvic radiotherapy on gut microbiota of gynecological cancer patients revealed by massive pyrosequencing. PLoS ONE 2013, 8, e82659. [Google Scholar] [CrossRef]
- Wang, A.; Ling, Z.; Yang, Z.; Kiela, P.R.; Wang, T.; Wang, C.; Cao, L.; Geng, F.; Shen, M.; Ran, X.; et al. Gut Microbial Dysbiosis May Predict Diarrhea and Fatigue in Patients Undergoing Pelvic Cancer Radiotherapy: A Pilot Study. PLoS ONE 2015, 10, e0126312. [Google Scholar] [CrossRef]
- Wang, Z.; Wang, Q.; Wang, X.; Zhu, L.; Chen, J.; Zhang, B.; Chen, Y.; Yuan, Z. Gut microbial dysbiosis is associated with development and progression of radiation enteritis during pelvic radiotherapy. J. Cell. Mol. Med. 2019, 23, 3747–3756. [Google Scholar] [CrossRef]
- Hekmatshoar, Y.; Saadat, Y.R.; Khatibi, S.M.H.; Ozkan, T.; Vahed, F.Z.; Nariman-Saleh-Fam, Z.; Gargari, B.P.; Sunguroglu, A.; Vahed, S.Z. The impact of tumor and gut microbiotas on cancer therapy; Beneficial or detrimental? Life Sci. 2019, 233, 116680. [Google Scholar] [CrossRef]
- Olivas, A.D.; Shogan, B.D.; Valuckaite, V.; Zaborin, A.; Belogortseva, N.; Musch, M.; Meyer, F.; Trimble, W.L.; An, G.; Gilbert, J.; et al. Intestinal Tissues Induce an SNP Mutation in Pseudomonas aeruginosa That Enhances Its Virulence: Possible Role in Anastomotic Leak. PLoS ONE 2012, 7, e44326. [Google Scholar]
- Al-Qadami, G.; Van Sebille, Y.; Le, H.; Bowen, J. Gut microbiota: Implications for radiotherapy response and radiotherapy-induced mucositis. Expert Rev. Gastroenterol. Hepatol. 2019, 13, 485–496. [Google Scholar] [CrossRef]
- Ashraf, S.Q.; Burns, E.M.; Jani, A.; Altman, S.; Young, J.D.; Cunningham, C.; Faiz, O.; Mortensen, N.J. The economic impact of anastomotic leakage after anterior resections in English NHS hospitals: Are we adequately remunerating them? Colorectal Dis. 2013, 15, e190–e198. [Google Scholar] [CrossRef]
- Buchs, N.C.; Gervaz, P.; Secic, M.; Bucher, P.; Mugnier-Konrad, B.; Morel, P. Incidence, consequences, and risk factors for anastomotic dehiscence after colorectal surgery: A prospective monocentric study. Int. J. Colorectal Dis. 2008, 23, 265–270. [Google Scholar] [CrossRef]
- Goto, S.; Hasegawa, S.; Hida, K.; Uozumi, R.; Kanemitsu, Y.; Watanabe, T.; Sugihara, K.; Sakai, Y. Multicenter analysis of impact of anastomotic leakage on long-term oncologic outcomes after curative resection of colon cancer. Surgery 2017, 162, 317–324. [Google Scholar] [CrossRef]
- Hammond, J.; Lim, S.; Wan, Y.; Gao, X.; Patkar, A. The Burden of Gastrointestinal Anastomotic Leaks: An Evaluation of Clinical and Economic Outcomes. J. Gastrointest. Surg. 2014, 18, 1176–1185. [Google Scholar] [CrossRef]
- Mirnezami, A.; Mirnezami, R.; Chandrakumaran, K.; Sasapu, K.; Sagar, P.; Finan, P. Increased Local Recurrence and Reduced Survival from Colorectal Cancer Following Anastomotic Leak: Systematic Review and Meta-Analysis. Ann. Surg. 2011, 253, 890–899. [Google Scholar] [CrossRef]
- Alves, A.; Panis, Y.; Trancar, D.; Regimbeau, J.M.; Pocard, M.; Valleur, P. Factors Associated with Clinically Significant Anastomotic Leakage after Large Bowel Resection: Multivariate Analysis of 707 Patients. World J. Surg. 2002, 26, 499–502. [Google Scholar]
- Borowski, D.W.; Bradburn, D.M.; Mills, S.J.; Bharathan, B.; Wilson, R.G.; Ratcliffe, A.A.; Kelly, S.B. Volume-outcome analysis of colorectal cancer-related outcomes. Br. J. Surg. 2010, 97, 1416–1430. [Google Scholar] [CrossRef]
- Frasson, M.; Granero-Castro, P.; Ramos Rodríguez, J.L.; Flor-Lorente, B.; Braithwaite, M.; Martí Martínez, E.; Álvarez Pérez, J.A.; Codina Cazador, A.; Espí, A.; Garcia-Granero, E.; et al. Risk factors for anastomotic leak and postoperative morbidity and mortality after elective right colectomy for cancer: Results from a prospective, multicentric study of 1102 patients. Int. J. Colorectal Dis. 2016, 31, 105–114. [Google Scholar] [CrossRef]
- Hyman, N.; Manchester, T.L.; Osler, T.; Burns, B.; Cataldo, P.A. Anastomotic Leaks After Intestinal Anastomosis: It’s Later Than You Think. Ann. Surg. 2007, 245, 254–258. [Google Scholar] [CrossRef]
- Chen, E.B.; Cason, C.; Gilbert, J.A.; Ho, K.J. Current State of Knowledge on Implications of Gut Microbiome for Surgical Conditions. J. Gastrointest. Surg. 2018, 22, 1112–1123. [Google Scholar] [CrossRef]
- Alverdy, J.C.; Hyoju, S.K.; Weigerinck, M.; Gilbert, J.A. The gut microbiome and the mechanism of surgical infection: The gut microbiome and the mechanism of surgical infection. Br. J. Surg. 2017, 104, e14–e23. [Google Scholar] [CrossRef]
- Gaines, S.; Shao, C.; Hyman, N.; Alverdy, J.C. Gut microbiome influences on anastomotic leak and recurrence rates following colorectal cancer surgery: Gut microbiome and outcomes after colorectal cancer surgery. Br. J. Surg. 2018, 105, e131–e141. [Google Scholar] [CrossRef]
- Guyton, K.; Alverdy, J.C. The gut microbiota and gastrointestinal surgery. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 43–54. [Google Scholar] [CrossRef]
- Hajjar, R.; Santos, M.M.; Dagbert, F.; Richard, C.S. Current evidence on the relation between gut microbiota and intestinal anastomotic leak in colorectal surgery. Am. J. Surg. 2019. [Google Scholar] [CrossRef]
- Russ, A.; Casillas, M. Gut Microbiota and Colorectal Surgery: Impact on Postoperative Complications. Clin. Colon Rectal Surg. 2016, 29, 253–257. [Google Scholar] [Green Version]
- Cohn, I.; Gelb, A.; Hawthorne, H.R. Strangulation obstruction—the effect of pre- and post-operative antibacterial agents. Ann. Surg. 1953, 138, 748–758. [Google Scholar] [CrossRef]
- Cohn, I.; Rives, J.D. Antibiotic Protection of Colon Anastomoses. Ann. Surg. 1955, 141, 707–717. [Google Scholar] [CrossRef]
- Rollins, K.E.; Javanmard-Emamghissi, H.; Acheson, A.G.; Lobo, D.N. The Role of Oral Antibiotic Preparation in Elective Colorectal Surgery: A Meta-analysis. Ann. Surg. 2019, 270, 43–58. [Google Scholar] [CrossRef]
- Van Praagh, J.B.; de Goffau, M.C.; Bakker, I.S.; van Goor, H.; Harmsen, H.J.M.; Olinga, P.; Havenga, K. Mucus Microbiome of Anastomotic Tissue During Surgery Has Predictive Value for Colorectal Anastomotic Leakage. Ann. Surg. 2019, 269, 911–916. [Google Scholar] [CrossRef]
- Stern, J.R.; Olivas, A.D.; Valuckaite, V.; Zaborina, O.; Alverdy, J.C.; An, G. Agent-based model of epithelial host–pathogen interactions in anastomotic leak. J. Surg. Res. 2013, 184, 730–738. [Google Scholar] [CrossRef]
- Seal, J.B.; Alverdy, J.C.; Zaborina, O.; An, G. Agent-based dynamic knowledge representation of Pseudomonas aeruginosa virulence activation in the stressed gut: Towards characterizing host-pathogen interactions in gut-derived sepsis. Theor. Biol. Med. Model. 2011, 8, 33. [Google Scholar] [CrossRef]
- Wu, L.; Holbrook, C.; Zaborina, O.; Ploplys, E.; Rocha, F.; Pelham, D.; Chang, E.; Musch, M.; Alverdy, J. Pseudomonas aeruginosa Expresses a Lethal Virulence Determinant, the PA-I Lectin/Adhesin, in the Intestinal Tract of a Stressed Host: The Role of Epithelia Cell Contact and Molecules of the Quorum Sensing Signaling System. Ann. Surg. 2003, 238, 754–764. [Google Scholar] [CrossRef]
- Bloemen, J.G.; Schreinemacher, M.H.; de Bruine, A.P.; Buurman, W.A.; Bouvy, N.D.; Dejong, C.H. Butyrate Enemas Improve Intestinal Anastomotic Strength in a Rat Model. Dis. Colon Rectum 2010, 53, 1069–1075. [Google Scholar] [CrossRef]
- Mathew, A.J.; Wann, V.C.; Abraham, D.T.; Jacob, P.M.; Selvan, B.S.; Ramakrishna, B.S.; Nair, A.N. The Effect of Butyrate on the Healing of Colonic Anastomoses in Rats. J. Investig. Surg. 2010, 23, 101–104. [Google Scholar] [CrossRef]
- Levison, M.E. Effect of colon flora and short-chain fatty acids on growth in vitro of Pseudomonas aeruginsoa and Enterobacteriaceae. Infect. Immun. 1973, 8, 30–35. [Google Scholar]
- Shogan, B.D.; Belogortseva, N.; Luong, P.M.; Zaborin, A.; Lax, S.; Bethel, C.; Ward, M.; Muldoon, J.P.; Singer, M.; An, G.; et al. Collagen degradation and MMP9 activation by Enterococcus faecalis contribute to intestinal anastomotic leak. Sci. Transl. Med. 2015, 7, 286ra68. [Google Scholar] [CrossRef]
- Helmink, B.A.; Khan, M.A.W.; Hermann, A.; Gopalakrishnan, V.; Wargo, J.A. The microbiome, cancer, and cancer therapy. Nat. Med. 2019, 25, 377–388. [Google Scholar] [CrossRef]
- Vivarelli, S.; Salemi, R.; Candido, S.; Falzone, L.; Santagati, M.; Stefani, S.; Torino, F.; Banna, G.L.; Tonini, G.; Libra, M. Gut Microbiota and Cancer: From Pathogenesis to Therapy. Cancers 2019, 11, 38. [Google Scholar] [CrossRef]
- Nagano, T.; Otoshi, T.; Hazama, D.; Kiriu, T.; Umezawa, K.; Katsurada, N.; Nishimura, Y. Novel cancer therapy targeting microbiome. OncoTargets Ther. 2019, 12, 3619–3624. [Google Scholar] [CrossRef]
- Huemer, F.; Rinnerthaler, G.; Westphal, T.; Hackl, H.; Hutarew, G.; Gampenrieder, S.P.; Weiss, L.; Greil, R. Impact of antibiotic treatment on immune-checkpoint blockade efficacy in advanced non-squamous non-small cell lung cancer. Oncotarget 2018, 9, 16512–16520. [Google Scholar] [CrossRef] [Green Version]
- Elkrief, A.; Derosa, L.; Kroemer, G.; Zitvogel, L.; Routy, B. The negative impact of antibiotics on outcomes in cancer patients treated with immunotherapy: A new independent prognostic factor? Ann. Oncol. 2019, mdz206. [Google Scholar] [CrossRef]
- Ahmed, J.; Kumar, A.; Parikh, K.; Anwar, A.; Knoll, B.M.; Puccio, C.; Chun, H.; Fanucchi, M.; Lim, S.H. Use of broad-spectrum antibiotics impacts outcome in patients treated with immune checkpoint inhibitors. OncoImmunology 2018, 7, e1507670. [Google Scholar] [CrossRef] [Green Version]
- Hakozaki, T.; Okuma, Y.; Omori, M.; Hosomi, Y. Impact of prior antibiotic use on the efficacy of nivolumab for non-small cell lung cancer. Oncol. Lett. 2019, 17, 2946–2952. [Google Scholar] [CrossRef]
- Derosa, L.; Routy, B.; Enot, D.; Baciarello, G.; Massard, C.; Loriot, Y.; Fizazi, K.; Escudier, B.J.; Zitvogel, L.; Albiges, L. Impact of antibiotics on outcome in patients with metastatic renal cell carcinoma treated with immune checkpoint inhibitors. J. Clin. Oncol. 2017, 35, 462. [Google Scholar] [CrossRef]
- Cougnoux, A.; Delmas, J.; Gibold, L.; Faïs, T.; Romagnoli, C.; Robin, F.; Cuevas-Ramos, G.; Oswald, E.; Darfeuille-Michaud, A.; Prati, F.; et al. Small-molecule inhibitors prevent the genotoxic and protumoural effects induced by colibactin-producing bacteria. Gut 2016, 65, 278–285. [Google Scholar] [CrossRef]
- Gough, E.; Shaikh, H.; Manges, A.R. Systematic Review of Intestinal Microbiota Transplantation (Fecal Bacteriotherapy) for Recurrent Clostridium difficile Infection. Clin. Infect. Dis. 2011, 53, 994–1002. [Google Scholar] [CrossRef] [Green Version]
- Food and Agriculture Organization of the United Nations; World Health Organization; Health Organization. Health and Nutritional Properties of Probiotics in Food including Powder Milk and Live Lactic Acid Bacteria. In Report of a Joint FAO/WHO Expert Consultation on Evaluation of Health and Nutritional Properties of Probiotics in Food Including Powder Milk with Live Lactic Acid Bacteria; FAO/WHO: Córdoba, Argentina, 2001. [Google Scholar]
- Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef]
- Kristensen, N.B.; Bryrup, T.; Allin, K.H.; Nielsen, T.; Hansen, T.H.; Pedersen, O. Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: A systematic review of randomized controlled trials. Genome Med. 2016, 8, 52. [Google Scholar] [CrossRef]
- 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]
- Hendler, R.; Zhang, Y. Probiotics in the Treatment of Colorectal Cancer. Medicines 2018, 5, 101. [Google Scholar] [CrossRef]
- Górska, A.; Przystupski, D.; Niemczura, M.J.; Kulbacka, J. Probiotic Bacteria: A Promising Tool in Cancer Prevention and Therapy. Curr. Microbiol. 2019, 76, 939–949. [Google Scholar] [CrossRef] [Green Version]
- Okawa, T.; Niibe, H.; Arai, T.; Sekiba, K.; Noda, K.; Takeuchi, S.; Hashimoto, S.; Ogawa, N. Effect of lc9018 combined with radiation therapy on carcinoma of the uterine cervix. A phase iii, multicenter, randomized, controlled study. Cancer 1993, 72, 1949–1954. [Google Scholar] [CrossRef]
- Ciorba, M.A.; Riehl, T.E.; Rao, M.S.; Moon, C.; Ee, X.; Nava, G.M.; Walker, M.R.; Marinshaw, J.M.; Stappenbeck, T.S.; Stenson, W.F. Lactobacillus probiotic protects intestinal epithelium from radiation injury in a TLR-2/cyclo-oxygenase-2-dependent manner. Gut 2012, 61, 829–838. [Google Scholar] [CrossRef]
- Delia, P.; Sansotta, G.; Donato, V.; Frosina, P.; Messina, G.; De Renzis, C.; Famularo, G. Use of probiotics for prevention of radiation-induced diarrhea. World J. Gastroenterol. 2007, 13, 912–915. [Google Scholar] [CrossRef]
- Jiang, C.; Wang, H.; Xia, C.; Dong, Q.; Chen, E.; Qiu, Y.; Su, Y.; Xie, H.; Zeng, L.; Kuang, J.; et al. A randomized, double-blind, placebo-controlled trial of probiotics to reduce the severity of oral mucositis induced by chemoradiotherapy for patients with nasopharyngeal carcinoma. Cancer 2019, 125, 1081–1090. [Google Scholar] [CrossRef]
- Consoli, M.L.D.; da Silva, R.S.; Nicoli, J.R.; Bruña-Romero, O.; da Silva, R.G.; de Vasconcelos Generoso, S.; Correia, M.I.T.D. Randomized Clinical Trial: Impact of Oral Administration of Saccharomyces boulardii on Gene Expression of Intestinal Cytokines in Patients Undergoing Colon Resection. J. Parenter. Enter. Nutr. 2016, 40, 1114–1121. [Google Scholar] [CrossRef]
- Louis, P.; Hold, G.L.; Flint, H.J. The gut microbiota, bacterial metabolites and colorectal cancer. Nat. Rev. Microbiol. 2014, 12, 661–672. [Google Scholar] [CrossRef]
- Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef] [Green Version]
- Taur, Y.; Jenq, R.R.; Perales, M.-A.; Littmann, E.R.; Morjaria, S.; Ling, L.; No, D.; Gobourne, A.; Viale, A.; Dahi, P.B.; et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood 2014, 124, 1174–1182. [Google Scholar] [CrossRef]
- Sommacal, H.M.; Bersch, V.P.; Vitola, S.P.; Osvaldt, A.B. Perioperative Synbiotics Decrease Postoperative Complications in Periampullary Neoplasms: A Randomized, Double-Blind Clinical Trial. Nutr. Cancer 2015, 67, 457–462. [Google Scholar] [CrossRef]
- Song, M.; Chan, A.T. The Potential Role of Exercise and Nutrition in Harnessing the Immune System to Improve Colorectal Cancer Survival. Gastroenterology 2018, 155, 596–600. [Google Scholar] [CrossRef] [Green Version]
- Lahiri, S.; Kim, H.; Garcia-Perez, I.; Reza, M.M.; Martin, K.A.; Kundu, P.; Cox, L.M.; Selkrig, J.; Posma, J.M.; Zhang, H.; et al. The gut microbiota influences skeletal muscle mass and function in mice. Sci. Transl. Med. 2019, 11, eaan5662. [Google Scholar] [CrossRef]
- Ticinesi, A.; Nouvenne, A.; Cerundolo, N.; Catania, P.; Prati, B.; Tana, C.; Meschi, T. Gut Microbiota, Muscle Mass and Function in Aging: A Focus on Physical Frailty and Sarcopenia. Nutrients 2019, 11, 1633. [Google Scholar] [CrossRef]
- Nay, K.; Jollet, M.; Goustard, B.; Baati, N.; Vernus, B.; Pontones, M.; Lefeuvre-Orfila, L.; Bendavid, C.; Rué, O.; Mariadassou, M.; et al. Gut bacteria are critical for optimal muscle function: A potential link with glucose homeostasis. Am. J. Physiol. Endocrinol. Metab. 2019, 317, e158–e171. [Google Scholar] [CrossRef]
- Yan, H.; Diao, H.; Xiao, Y.; Li, W.; Yu, B.; He, J.; Yu, J.; Zheng, P.; Mao, X.; Luo, Y.; et al. Gut microbiota can transfer fiber characteristics and lipid metabolic profiles of skeletal muscle from pigs to germ-free mice. Sci. Rep. 2016, 6, 31786. [Google Scholar] [CrossRef] [Green Version]
- Chang, K.-V.; Chen, J.-D.; Wu, W.-T.; Huang, K.-C.; Hsu, C.-T.; Han, D.-S. Association between Loss of Skeletal Muscle Mass and Mortality and Tumor Recurrence in Hepatocellular Carcinoma: A Systematic Review and Meta-Analysis. Liver Cancer 2018, 7, 90–103. [Google Scholar] [CrossRef]
- Fujiwara, N.; Nakagawa, H.; Kudo, Y.; Tateishi, R.; Taguri, M.; Watadani, T.; Nakagomi, R.; Kondo, M.; Nakatsuka, T.; Minami, T.; et al. Sarcopenia, intramuscular fat deposition, and visceral adiposity independently predict the outcomes of hepatocellular carcinoma. J. Hepatol. 2015, 63, 131–140. [Google Scholar] [CrossRef] [Green Version]
- Van Vugt, J.L.A.; Coebergh van den Braak, R.R.J.; Lalmahomed, Z.S.; Vrijland, W.W.; Dekker, J.W.T.; Zimmerman, D.D.E.; Vles, W.J.; Coene, P.-P.L.O.; IJzermans, J.N.M. Impact of low skeletal muscle mass and density on short and long-term outcome after resection of stage I-III colorectal cancer. Eur. J. Surg. Oncol. 2018, 44, 1354–1360. [Google Scholar] [CrossRef]
- Van der Kroft, G.; Bours, D.M.J.L.; Janssen-Heijnen, D.M.; van Berlo, D.C.L.H.; Konsten, D.J.L.M. Value of sarcopenia assessed by computed tomography for the prediction of postoperative morbidity following oncological colorectal resection: A comparison with the malnutrition screening tool. Clin. Nutr. ESPEN 2018, 24, 114–119. [Google Scholar] [CrossRef]
- Reisinger, K.W.; Bosmans, J.W.A.M.; Uittenbogaart, M.; Alsoumali, A.; Poeze, M.; Sosef, M.N.; Derikx, J.P.M. Loss of Skeletal Muscle Mass During Neoadjuvant Chemoradiotherapy Predicts Postoperative Mortality in Esophageal Cancer Surgery. Ann. Surg. Oncol. 2015, 22, 4445–4452. [Google Scholar] [CrossRef] [Green Version]
- Qiu, S.; Jiang, C.; Zhou, L. Physical activity and mortality in patients with colorectal cancer: A meta-analysis of prospective cohort studies. Eur. J. Cancer Prev. 2019. [Google Scholar] [CrossRef]
- Schieber, A.M.P.; Lee, Y.M.; Chang, M.W.; Leblanc, M.; Collins, B.; Downes, M.; Evans, R.M.; Ayres, J.S. Disease tolerance mediated by microbiome E. coli involves inflammasome and IGF-1 signaling. Science 2015, 350, 558–563. [Google Scholar] [CrossRef]
- Bindels, L.B.; Beck, R.; Schakman, O.; Martin, J.C.; De Backer, F.; Sohet, F.M.; Dewulf, E.M.; Pachikian, B.D.; Neyrinck, A.M.; Thissen, J.-P.; et al. Restoring specific lactobacilli levels decreases inflammation and muscle atrophy markers in an acute leukemia mouse model. PLoS ONE 2012, 7, e37971. [Google Scholar] [CrossRef]
- Varian, B.J.; Gourishetti, S.; Poutahidis, T.; Lakritz, J.R.; Levkovich, T.; Kwok, C.; Teliousis, K.; Ibrahim, Y.M.; Mirabal, S.; Erdman, S.E. Beneficial bacteria inhibit cachexia. Oncotarget 2016, 7, 11803–11816. [Google Scholar] [CrossRef] [Green Version]
- Bindels, L.B.; Neyrinck, A.M.; Claus, S.P.; Le Roy, C.I.; Grangette, C.; Pot, B.; Martinez, I.; Walter, J.; Cani, P.D.; Delzenne, N.M. Synbiotic approach restores intestinal homeostasis and prolongs survival in leukaemic mice with cachexia. ISME J. 2016, 10, 1456–1470. [Google Scholar] [CrossRef]
- Bindels, L.B.; Neyrinck, A.M.; Salazar, N.; Taminiau, B.; Druart, C.; Muccioli, G.G.; François, E.; Blecker, C.; Richel, A.; Daube, G.; et al. Non Digestible Oligosaccharides Modulate the Gut Microbiota to Control the Development of Leukemia and Associated Cachexia in Mice. PLoS ONE 2015, 10, e0131009. [Google Scholar] [CrossRef]
- Pötgens, S.A.; Brossel, H.; Sboarina, M.; Catry, E.; Cani, P.D.; Neyrinck, A.M.; Delzenne, N.M.; Bindels, L.B. Klebsiella oxytoca expands in cancer cachexia and acts as a gut pathobiont contributing to intestinal dysfunction. Sci. Rep. 2018, 8, 12321. [Google Scholar] [CrossRef]
- Schmid, D.; Leitzmann, M.F. Association between physical activity and mortality among breast cancer and colorectal cancer survivors: A systematic review and meta-analysis. Ann. Oncol. 2014, 25, 1293–1311. [Google Scholar] [CrossRef]
- Nadler, M.B.; Desnoyers, A.; Langelier, D.M.; Amir, E. The effect of exercise on quality of life, fatigue, physical function and safety in advanced solid tumor cancers: A meta-analysis of randomized control trials. J. Pain Symptom Manag. 2019. [Google Scholar] [CrossRef]
- Liu, L.; He, X.; Feng, L. Exercise on quality of life and cancer-related fatigue for lymphoma survivors: A systematic review and meta-analysis. Support. Care Cancer 2019, 1–14. [Google Scholar] [CrossRef]
- Wang, J.; Huang, L.; Gao, Y.; Wang, Y.; Chen, S.; Huang, J.; Zheng, W.; Bao, P.; Gong, Y.; Zhang, Y.; et al. Physically active individuals have a 23% lower risk of any colorectal neoplasia and a 27% lower risk of advanced colorectal neoplasia than their non-active counterparts: Systematic review and meta-analysis of observational studies. Br. J. Sports Med. 2019. [Google Scholar] [CrossRef]
- Rosero, I.D.; Ramírez-Vélez, R.; Lucia, A.; Martínez-Velilla, N.; Santos-Lozano, A.; Valenzuela, P.L.; Morilla, I.; Izquierdo, M. Systematic Review and Meta-Analysis of Randomized, Controlled Trials on Preoperative Physical Exercise Interventions in Patients with Non-Small-Cell Lung Cancer. Cancers 2019, 11, 944. [Google Scholar] [CrossRef]
- Ashcraft, K.A.; Warner, A.B.; Jones, L.W.; Dewhirst, M.W. Exercise as Adjunct Therapy in Cancer. Semin. Radiat. Oncol. 2019, 29, 16–24. [Google Scholar] [CrossRef]
- Oruç, Z.; Kaplan, M.A. Effect of exercise on colorectal cancer prevention and treatment. World J. Gastrointest. Oncol. 2019, 11, 348–366. [Google Scholar] [CrossRef]
- Courneya, K.S.; Vardy, J.L.; O’Callaghan, C.J.; Friedenreich, C.M.; Campbell, K.L.; Prapavessis, H.; Crawford, J.J.; O’Brien, P.; Dhillon, H.M.; Jonker, D.J.; et al. Effects of a Structured Exercise Program on Physical Activity and Fitness in Colon Cancer Survivors: One Year Feasibility Results from the CHALLENGE Trial. Cancer Epidemiol. Biomark. Prev. 2016, 25, 969–977. [Google Scholar] [CrossRef]
- Zhang, X.; Ashcraft, K.A.; Betof Warner, A.; Nair, S.K.; Dewhirst, M.W. Can Exercise-Induced Modulation of the Tumor Physiologic Microenvironment Improve Antitumor Immunity? Cancer Res. 2019, 79, 2447–2456. [Google Scholar] [CrossRef] [Green Version]
- Pedersen, L.; Idorn, M.; Olofsson, G.H.; Lauenborg, B.; Nookaew, I.; Hansen, R.H.; Johannesen, H.H.; Becker, J.C.; Pedersen, K.S.; Dethlefsen, C.; et al. Voluntary Running Suppresses Tumor Growth through Epinephrine- and IL-6-Dependent NK Cell Mobilization and Redistribution. Cell Metab. 2016, 23, 554–562. [Google Scholar] [CrossRef] [Green Version]
- Hojman, P.; Gehl, J.; Christensen, J.F.; Pedersen, B.K. Molecular Mechanisms Linking Exercise to Cancer Prevention and Treatment. Cell Metab. 2018, 27, 10–21. [Google Scholar] [CrossRef] [Green Version]
- Idorn, M.; Thor Straten, P. Exercise and cancer: From “healthy” to “therapeutic”? Cancer Immunol. Immunother. 2017, 66, 667–671. [Google Scholar] [CrossRef]
- Nilsen, T.I.L.; Romundstad, P.R.; Petersen, H.; Gunnell, D.; Vatten, L.J. Recreational physical activity and cancer risk in subsites of the colon (the Nord-Trøndelag Health Study). Cancer Epidemiol. Biomark. Prev. 2008, 17, 183–188. [Google Scholar] [CrossRef]
- Mailing, L.J.; Allen, J.M.; Buford, T.W.; Fields, C.J.; Woods, J.A. Exercise and the Gut Microbiome: A Review of the Evidence, Potential Mechanisms, and Implications for Human Health. Exerc. Sport Sci. Rev. 2019, 47, 75–85. [Google Scholar] [CrossRef]
- Maillard, F.; Vazeille, E.; Sauvanet, P.; Sirvent, P.; Bonnet, R.; Combaret, L.; Chausse, P.; Chevarin, C.; Otero, Y.F.; Delcros, G.; et al. Preventive Effect of Spontaneous Physical Activity on the Gut-Adipose Tissue in a Mouse Model That Mimics Crohn’s Disease Susceptibility. Cells 2019, 8, 33. [Google Scholar] [CrossRef]
- Allen, J.M.; Mailing, L.J.; Cohrs, J.; Salmonson, C.; Fryer, J.D.; Nehra, V.; Hale, V.L.; Kashyap, P.; White, B.A.; Woods, J.A. Exercise training-induced modification of the gut microbiota persists after microbiota colonization and attenuates the response to chemically-induced colitis in gnotobiotic mice. Gut Microbes 2018, 9, 115–130. [Google Scholar] [CrossRef]
- Lamoureux, E.V.; Grandy, S.A.; Langille, M.G.I. Moderate Exercise Has Limited but Distinguishable Effects on the Mouse Microbiome. mSystems 2017, 2. [Google Scholar] [CrossRef]
- Denou, E.; Marcinko, K.; Surette, M.G.; Steinberg, G.R.; Schertzer, J.D. High-intensity exercise training increases the diversity and metabolic capacity of the mouse distal gut microbiota during diet-induced obesity. Am. J. Physiol. Endocrinol. Metab. 2016, 310, E982–E993. [Google Scholar] [CrossRef]
- Allen, J.M.; Berg Miller, M.E.; Pence, B.D.; Whitlock, K.; Nehra, V.; Gaskins, H.R.; White, B.A.; Fryer, J.D.; Woods, J.A. Voluntary and forced exercise differentially alters the gut microbiome in C57BL/6J mice. J. Appl. Physiol. 2015, 118, 1059–1066. [Google Scholar] [CrossRef] [Green Version]
- Lambert, J.E.; Myslicki, J.P.; Bomhof, M.R.; Belke, D.D.; Shearer, J.; Reimer, R.A. Exercise training modifies gut microbiota in normal and diabetic mice. Appl. Physiol. Nutr. Metab. 2015, 40, 749–752. [Google Scholar] [CrossRef]
- Keohane, D.M.; Woods, T.; O’Connor, P.; Underwood, S.; Cronin, O.; Whiston, R.; O’Sullivan, O.; Cotter, P.; Shanahan, F.; Molloy, M.G.M. Four men in a boat: Ultra-endurance exercise alters the gut microbiome. J. Sci. Med. Sport 2019, 22, 1059–1064. [Google Scholar] [CrossRef]
- Barton, W.; Penney, N.C.; Cronin, O.; Garcia-Perez, I.; Molloy, M.G.; Holmes, E.; Shanahan, F.; Cotter, P.D.; O’Sullivan, O. The microbiome of professional athletes differs from that of more sedentary subjects in composition and particularly at the functional metabolic level. Gut 2018, 67, 625–633. [Google Scholar] [CrossRef]
- Clarke, S.F.; Murphy, E.F.; O’Sullivan, O.; Lucey, A.J.; Humphreys, M.; Hogan, A.; Hayes, P.; O’Reilly, M.; Jeffery, I.B.; Wood-Martin, R.; et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut 2014, 63, 1913–1920. [Google Scholar] [CrossRef] [Green Version]
- Cronin, O.; Barton, W.; Skuse, P.; Penney, N.C.; Garcia-Perez, I.; Murphy, E.F.; Woods, T.; Nugent, H.; Fanning, A.; Melgar, S.; et al. A Prospective Metagenomic and Metabolomic Analysis of the Impact of Exercise and/or Whey Protein Supplementation on the Gut Microbiome of Sedentary Adults. mSystems 2018, 3. [Google Scholar] [CrossRef]
- Newton, R.U.; Christophersen, C.T.; Fairman, C.M.; Hart, N.H.; Taaffe, D.R.; Broadhurst, D.; Devine, A.; Chee, R.; Tang, C.I.; Spry, N.; et al. Does exercise impact gut microbiota composition in men receiving androgen deprivation therapy for prostate cancer? A single-blinded, two-armed, randomised controlled trial. BMJ Open 2019, 9, e024872. [Google Scholar] [CrossRef] [Green Version]
- Zackular, J.P.; Rogers, M.A.M.; Ruffin, M.T.; Schloss, P.D. The Human Gut Microbiome as a Screening Tool for Colorectal Cancer. Cancer Prev. Res. 2014, 7, 1112–1121. [Google Scholar] [CrossRef]
- Bonnet, M.; Buc, E.; Sauvanet, P.; Darcha, C.; Dubois, D.; Pereira, B.; Déchelotte, P.; Bonnet, R.; Pezet, D.; Darfeuille-Michaud, A. Colonization of the human gut by E. coli and colorectal cancer risk. Clin. Cancer Res. 2014, 20, 859–867. [Google Scholar] [CrossRef]
- Villéger, R.; Lopès, A.; Veziant, J.; Gagnière, 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]
- Eklöf, V.; Löfgren-Burström, A.; Zingmark, C.; Edin, S.; Larsson, P.; Karling, P.; Alexeyev, O.; Rutegård, J.; Wikberg, M.L.; Palmqvist, R. Cancer-associated fecal microbial markers in colorectal cancer detection. Int. J. Cancer 2017, 141, 2528–2536. [Google Scholar] [CrossRef]
© 2019 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Villéger, R.; Lopès, A.; Carrier, G.; Veziant, J.; Billard, E.; Barnich, N.; Gagnière, J.; Vazeille, E.; Bonnet, M. Intestinal Microbiota: A Novel Target to Improve Anti-Tumor Treatment? Int. J. Mol. Sci. 2019, 20, 4584. https://doi.org/10.3390/ijms20184584
Villéger R, Lopès A, Carrier G, Veziant J, Billard E, Barnich N, Gagnière J, Vazeille E, Bonnet M. Intestinal Microbiota: A Novel Target to Improve Anti-Tumor Treatment? International Journal of Molecular Sciences. 2019; 20(18):4584. https://doi.org/10.3390/ijms20184584
Chicago/Turabian StyleVilléger, Romain, Amélie Lopès, Guillaume Carrier, Julie Veziant, Elisabeth Billard, Nicolas Barnich, Johan Gagnière, Emilie Vazeille, and Mathilde Bonnet. 2019. "Intestinal Microbiota: A Novel Target to Improve Anti-Tumor Treatment?" International Journal of Molecular Sciences 20, no. 18: 4584. https://doi.org/10.3390/ijms20184584