ecancermedicalscience

Special Issue

Gut microbiota, chemotherapy and the host: the influence of the gut microbiota on cancer treatment

Anna Louise Pouncey, Alasdair James Scott, James Leslie Alexander, Julian Marchesi and James Kinross

Centre for Digestive and Gut Health, Department of Surgery and Cancer, Imperial College London, London SW7 2AZ, UK

Correspondence to: James Kinross. Email: j.kinross@imperial.ac.uk


Abstract

The gut microbiota exists in a dynamic balance between symbiosis and pathogenesis and can influence almost any aspect of host physiology. Growing evidence suggests that the gut microbiota not only plays a key role in carcinogenesis but also influences the efficacy and toxicity of anticancer therapy. The microbiota modulates the host response to chemotherapy via numerous mechanisms, including immunomodulation, xenometabolism and alteration of community structure. Furthermore, exploitation of the microbiota offers opportunities for the personalisation of chemotherapeutic regimens and the development of novel therapies. In this article, we explore the host-chemotherapeutic microbiota axis, from basic science to clinical research, and describe how it may change the face of cancer treatment.

Keywords: gut, microbiome, cancer, oncology, chemotherapy, pharmacobiomics

Copyright: © the authors; licensee ecancermedicalscience. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Published: 05/09/2018; Received: 28/02/2018


Introduction

The microbial commensals of the gastrointestinal system, the gut microbiota, outnumber the cells of the human body, and comprise the largest surface area of microbial interaction with the host immune system [1]. This dynamic interface, consisting of multiple metabolic, immunological and inflammatory pathways, exists in a delicate balance between symbiosis and pathogenesis and can influence many aspects of host physiology. While there is mounting evidence that the gut microbiome plays a key role in carcinogenesis [2], the advancement of high-throughput sequencing, metabolite profiling and bioinformatics algorithms is broadening our understanding of the interactions of the gut microbiota further still. In particular, a relatively novel area of research, ‘pharmacomicrobiomics’ (the study of the effect of microbiota on drug disposition, action and toxicity) is revealing the integral role that microbiota plays in the efficacy and toxicity of cancer treatment [3].

Pharmacomicrobiomics has the potential to enhance therapeutic efficacy and abrogate side effects by manipulating host-chemotherapeutic-microbiota interactions and for personalisation of chemotherapy regimens based on the evaluation of an individual’s microbiome (the genetic composition of their microbiota) [4]. Increased understanding of the complex interplay between gut microbiota and the immune system may also generate novel chemotherapeutic approaches [5, 6].


Host-chemotherapeutic-microbiota interaction

Gut microbiota can modulate the host response to chemotherapy through numerous mechanisms, including immune interactions, xenometabolism and altered community structure [6]. Interaction with the immune system occurs both intra-luminally and within lymphoid organs following chemotherapy-induced bacterial translocation [79]. The microbiota directly metabolises chemotherapeutic medication and may produce toxic secondary metabolites. They also exert an indirect effect on host-chemotherapeutic metabolism through modification of the host microenvironment [10]. As the gut microbiota develops in synchrony and symbiosis with the host, its composition and functionality are highly individualised [11]. During the course of anticancer treatment, the community structure of gut microbiota is affected by multiple factors, such host environment and diet, surgical intervention, use of adjuvant medication (such as antibiotics) and the effect of the chemotherapy administered. Many of these factors generate dysbiosis, a perturbation of the microbial community, which disrupts the symbiotic relationship with the host. Immune interactions, xenometabolism and altered community structure can all cause adverse side effects, and impair chemotherapeutic outcome [12]. Analysis and manipulation of gut microbiota may, therefore, become a vital component for the development of personalised and effective anticancer therapy [6].


Immunomodulation

Modification of the immune response by the microbiota plays a key role in determining chemotherapeutic response. At the mucosal surface, constant interaction between gut microbiota and the innate and adaptive immune systems have been shown to set the immune tone and regulate inflammation [5]. Chemotherapeutic medication can damage the mucosal epithelium, triggering bacterial translocation. While this may cause systemic infection, increased exposure to potential pathogens can also prime the adaptive immune system, augmenting chemotherapeutic response in the host [79]. The mechanisms by which immunomodulation can occur are diverse. Bacterial translocation and T-helper 17 cell activation enhance the action of cyclophosphamide; intraluminal myeloid cell activation enhances the action of oxaliplatin; and microbiota induced T-cell activation is a key in facilitating novel anticancer immunotherapy [9, 13].

The interaction among the immune system, microbiota and cyclophosphamide (CTX) (a commonly used alkylating agent), has been well characterised in both basic and clinical studies. Research by Viaud et al [9] using a murine sarcoma model showed that therapeutic response to CTX was greater in mice with healthy gut microbiota than in mice raised in a germ-free environment. Moreover, depletion of gram-positive gut bacteria with vancomycin pre-treatment resulted in a poor therapeutic response compared to untreated controls [9]. The authors elucidated that transmucosal translocation of specific bacteria (such as Lactobacillus spp., Enterococcus hirae) into mesenteric lymph nodes and the spleen, stimulated T-helper 17 (Th17) cell differentiation resulting in an antitumour adaptive immunological response. Fewer Th17 cells were observed in the spleens of vancomycin pre-treated mice and adoptive transfer of pathogenic Th17 cells from untreated mice could re-establish a therapeutic response [9, 14]. Further investigation demonstrated that administration of E. hirae in antibiotic pre-treated mice could also restore response to CTX [13]. These findings have been corroborated by clinical studies. Among patients with end-stage lung and ovarian cancer, a memory Th1 immune response towards E. hirae was found to be a positive predictor of progression-free survival [6, 10, 11, 14, 17].

The impact of microbiota and myeloid cell interaction on chemotherapeutic efficacy was investigated by Iida et al [8], in a murine lymphoma model. Antibiotic pre-treatment of mice with subcutaneous EL4 lymphomas reduced both DNA damage by oxaliplatin (a DNA cross-linking agent) and the expression of genes responsible for reactive-oxygen species (ROS) generation by myeloid cells. Cybb -/- mice, deficient in NADPH oxidase, an enzyme critical for the production of ROS, also respond poorly to oxaliplatin. It was concluded that a healthy microbiota enhances the antitumour effect of oxaliplatin by priming of myeloid cells for ROS release, enhancing inflammatory cytokine production, and thus tumour eradication [8]. A similar effect was observed with CpG oligodeoxynucleotides (CpG OGN). CpG OGN are synthetic mimics of microbial DNA, which bind to TLR9, and can be injected to stimulate a tumour-eradicating immunological response in mice. In germ-free and antibiotic-treated mice, myeloid cell inflammatory cytokine production and the anticancer adaptive immune response to intra-tumoural administration of CpG OGN were reduced. This could be restored by inoculation with Alistipes shahii [8].

Immune checkpoint inhibitors, which target T-cell regulatory pathways, display promising results in patients with lung cancer or advanced melanoma [15]. It was demonstrated that treatment with a synthetic monoclonal antibody against cytotoxic T-lymphocyte-associated protein 4 (CTLA-4, ipilimumab) could control established MCA205 sarcomas in mice with healthy gut bacteria. However, germ-free or antibiotic-treated mice displayed no response to the CTLA-4 blockade. Melanoma patients responsive to CTLA-4 display T cell reactions specific for Bacteroides species; therapeutic reaction to ipilimumab in germ-free mice could be salvaged with the administration of Bacteriodes fragilis or adoptive transfer B. fragilis-specific T cells. This indicates that the interaction between Bacteriodes species and the immune system plays a significant role in enabling the CTLA-4 induced antitumour response [4].

In another study, Sivan et al [16] observed differential melanoma growth in genetically similar mice sourced from two suppliers—Taconic Farms (Tac) and Jackson Laboratory (Jax). Tumours grew more aggressively in Tac mice compared to Jax mice and this effect could be abrogated by either co-housing or by faecal transfer from Jax mice. Faecal transfer in the opposite direction did not confer increased tumour growth. Furthermore, chemotherapeutic response to antiprogrammed cell death 1 ligand-1 monoclonal antibodies (anti-PD-L1, pembrolizumab) was impaired in Tac mice compared to Jax mice. The authors determined that the attenuated basal tumour growth and enhanced anticancer response to anti-PD-L1 seen in Jax mice were due to the presence of Bifidobacterium spp in their gut flora which are believed to interact with dendritic cells to activate T-cells and stimulate a protective anticancer response. Administration of Bifidobacterium spp. to Tac mice was able to slow basal tumour growth to enhance the antitumour effect of anti-PD-L1 [16].


Metabolism

The gut microbiota has the potential to directly metabolise chemotherapeutic drugs and also to modify the host metabolic milieu, indirectly altering host-chemotherapeutic metabolism [17, 18]. Recent research using the Caenorhabditis elegans nematode worm (C. elegans) as a model for symbiotic host-microbial interactions suggests that the variable clinical response to the first-line chemotherapy for colorectal cancer, fluoropyrimidines, may be due, not only to genetic polymorphisms but also to variations in gut microbiota [10, 19]. Investigations led by Garcia-Gonzalez et al [19] tested the efficacy of camptothecin (CPT), 5-fluorouracil (5-FU) and 5-fluoro-2’-deoxyuridine (FUDR) in C. elegans, which were fed with either E. coli or Comamonas bacteria. As fluoropyrimidines inhibit cell division and impair nematode fertility, the dose of chemotherapeutic needed to prevent live progeny could be assessed. It was found that bacterial species affect the response to chemotherapeutics differently. Nematodes fed with E. coli were two orders of magnitude more sensitive to the sterilising effect of FUDR. Genetic screening and the restorative effect of uracil supplementation were used to elucidate that ribonucleic acid (RNA) metabolism (conversion of 5-FU and FUDR to fluorouridine monophosphate) by bacteria was crucial for the generation of a cytotoxic effect [19]. Further investigation conducted by Scott et al [10] using three-way high-throughput screens verified that bacteria alter the effects of fluoropyrimidines through two distinct pathways. First, bacterial RNA metabolism and vitamins B6 and B9 enable conversion and pro-drug activation. Second, bacteria influence the host metabolic environment, supplying regulatory metabolites that augment 5-FU-induced DNA damage. Concomitant drugs may also affect pro-drug activation. For example, it was also found that metformin inhibits bacterial one-carbon-metabolism, reducing the effect of 5-FU. This highlights the importance of considering all host-microbe-drug interactions when commencing anticancer therapy [10].

Microbial metabolism may cause side effects severe enough to necessitate cessation of chemotherapy. For example, irinotecan-induced mucositis causes severe, dose-limiting diarrhoea in up to 30% of patients [20]. Irinocetan is activated by hydrolysis to form SN-38, an inhibitor of topoisomerase 1. It is later deactivated by hepatic glucuronidation, producing SN-38G, which is excreted into the gut with bile. However, bacterial ß-glucuronidases within the gut lumen can reactivate SN-38G to its active, enterotoxic form, causing mucositis [20]. There are numerous bacterial ß-glucuronidase isoforms, which differ in their substrate pharmacokinetics [21]. Guthrie et al [20] recently demonstrated that differential reactivation of SN-38G by healthy volunteers correlated with the presence of specific bacterial ß-glucuronidases and glucuronide membrane transporters [20]. Microbiome characterisation may, therefore, identify patients at risk of irinotecan-induced mucositis while manipulation of the microbiota may provide novel therapeutic options. Ciprofloxacin and low doses of amoxapine have been shown to be effective in the suppression of bacterial ß-glucuronidase activity and mucositis [22, 23]. Furthermore, bacterial ß-glucuronidases possess unique motifs relative to their human counterparts, which pave the way for the development of bacterial-specific inhibitors [2224].

Inadequate understanding of gut microbiota metabolism can have fatal consequences. This was observed in Japan, when co-administration of the antiviral sorivudine and 5-FU led to toxic circulating levels of 5-FU, causing 16 deaths over a 40 day period [25]. This was caused by inactivation of hepatic dihydropyramidine dehydrogenase (which deactivates 5-FU) by a sorivudine metabolite, BVU [(E)-5-(2-boromovinyl) uracil]. Further investigation revealed that the microbiota, rather than host enzymes, is responsible for producing BVU from sorivudine. Indeed, in a rodent model, circulating levels of BVU and could be abrogated by depletion of gut microbiota with antibiotic administration [18, 26].


Community structure

Bacterial community structure is a key driver of symbiosis between the host and microbiota and the maintenance of intestinal health [27]. Commensal microbiota can prime and modulate host immunity to prevent pathogen overgrowth. Indeed, Bacteroides and Lactobacillus have been shown to induce host production of protective antimicrobial proteins within intestinal crypts [28]. The importance of microbiota to maintain the structure and function of the gut mucosal barrier is revealed by analysis of germ-free mice, which display thin villi, a reduced capillary network, a reduction in surface area and poor peristalsis [29, 30]. Administration of chemotherapeutics can damage the diversity and health of gut microbiota, disrupting this equilibrium [31]. In a rat model, methotrexate-induced mucositis and reduced villous length were associated with a 13-fold decrease in protective anaerobes and a 296-fold decrease in Streptococci [32]. In humans, 1 week of high-dose chemotherapy for nonHodgkin’s lymphoma dramatically reduces the abundance of commensal Faecalibacterium with an increase in the prevalence of potentially pathogenic Escherichia [33]. Chemotherapeutic-induced dysbiosis reduces colonisation resistance to pathogens and a decrease in the abundance of anaerobic bacteria may reduce the production of butyrate—a short chain fatty acid with antiinflammatory properties and an essential trophic factor for enterocytes. The resultant damage to the intestinal barrier increases the risk of colitis, bacterial translocation and infection [31].

Efforts to preserve community structure could, therefore, protect against chemotherapeutic injury. It has been shown that healthy microbiota interact, via Toll-like receptors, with the innate immune system, regulating inflammation and promoting healing. For example, antibiotic-treated mice have been shown to be more susceptible to methotrexate-induced small bowel injury, which can be salvaged with Toll-like receptor 2 ligand administration [34]. An analysis of the intestinal microbiome in patients taking anti-CTLA-4 therapy revealed that abundance of the Bacteriodetes phylum was associated with resistance to colitis. Administration of Bacteriodales has subsequently been shown to reduce CTLA-4 blockade induced colitis in an antibiotic-treated mouse model [4, 35].


Translation toward clinical practice

As our understanding of pharmacomicrobiomics increases, intestinal microbiome analysis holds the potential to predict patient response to chemotherapy prior to treatment and to personalise therapy. A key example of translation from basic research to clinical application follows an investigation into PD-1 inhibitor immunotherapy, which is highly effective in only a small subset of patients. Following the murine research conducted by Zitvogel et al [39] and Sivan et al [16] (described earlier), three clinical studies have recently emerged [4, 16, 3638]. Zitvogel et al [39] found that abnormal gut microbiome composition was associated with failure to respond to anti-PD-1 therapy, while faecal microbial transplantation from responding patients to a germ-free murine model was shown to ameliorate chemotherapeutic response. Metagenomic analysis revealed an abundance of Akkermansia muciniphila associated positively with clinical response and oral supplementation of the bacterium in germ-free mice restored the response to PD-1 blockade [36]. Gopalakrishnan et al [38] examined the microbiomes of melanoma patients undergoing PD-1 immunotherapy. Significantly higher alpha diversity and relative abundance of Ruminococcaceae family were observed in responding patients. Immune profiling of patients revealed enhanced systemic and antitumour immunity, which could be transferred to germ-free mice using faecal transplantation [38]. Finally, Gajewski et al [37] examined baseline stool samples from metastatic melanoma patients. Bifidobacterium longum, Collinsella aerofaciens and Enterococcus faecium were found to be more abundant in responding patients and faecal microbial transplantation from human responders into germ-free mice restored the antitumour effect of PD-1 blockade in the recipient mice [37].

Although different bacteria were identified, this research holds promise for optimisation of chemotherapeutic efficacy, either through intelligent drug selection or manipulation of microbiota. Various measures may be employed prior to, during and after drug administration. These include selective antibiotic therapy (targeting specific bacterial species), probiotic therapy (administration of living micro-organisms), pre-biotic therapy (compounds administered to encourage the functions of certain microbiota) and post-biotic therapy (use of nonviable microbial products to exert immunological or biological activity) [39]. Use of both antibiotic therapies and probiotic VSL#3 administration has been shown to reduce the incidence of irinocetan-induced mucositis in rodent models [23, 40]. Various studies on probiotic administration during chemotherapy have also suggested that they may reduce the incidence of systemic infection [41]. Ecological manipulation with faecal microbiota transplantation may also hold therapeutic potential [42].

As microbiota may have deleterious as well as advantageous effects, the perfect balance of microbial manipulation may not be possible. For example, while the microbiota may enhance chemotherapeutic response to oxaliplatin [8], they may also engender chemotherapy-induced mechanical hyperalgesia [43]. Shen et al [43] demonstrated that the incidence of oxaliplatin-induced mechanical hyperalgesia was lower amongst germ-free mice. This protective effect was lost following gut microbial refaunation. As mechanical hyperalgesia affects more than 30% of patients, and can be severe enough to prevent therapeutic dose administration, a compromise may need to be made in microbial manipulation between enhancement of chemotherapeutic effect and reduction of toxicity [43].

Dysbiosis of the gut microbiota following chemotherapeutic treatment may also be used to predict prognosis. Taur et al [12] divided patients into low-, intermediate- or high-diversity cohorts based on faecal 16S rRNA analysis shortly after allogenic haematopoietic stem cell transplantation (allo-HSCT). Low microbial diversity was associated with a 31% reduction in 3-year survival compared to high diversity, independent of known predictors and co-morbidities [12]. Furthermore, a study of 857 cases of allo-HSCT revealed that broad-spectrum antibiotic administration during treatment was associated with increased graft-versus-host disease (GVHD)-related mortality at 5 years. The analysis in mice demonstrated that antibiotic administration resulted in an increase in Akkermansia mucinophilia and associated loss of the protective mucous lining of the colon, impairment of the intestinal barrier and increased incidence of GVHD [44]. Early studies of faecal microbiota transplantation to tackle steroid-resistant GVHD in four patients have shown promising results [45, 46].


Novel chemotherapeutics

Innovative possibilities for the genetic engineering of bacteria for delivery of chemotherapy, or as vectors for genetic therapy, are emerging. In a study by Pillai et al [47], E.coli was genetically modified to enable excretion of a tumour suppressor, human bone morphogenic protein-2 (BMP-2). When exposed to an in vitro model of colon cancer, the bacterium induced apoptosis of the adenocarcinoma cell line. Din et al [48] used the quorum sensing feedback loops present in E.coli to cause the bacterium to lyse synchronously and release Haemolysin E (an antitumour toxin). This created a pulsatile delivery system with simultaneous control of microbial population density. When tested in vivo on a mouse model of hepatic colorectal metastases, oral administration of the bacterium, in combination with 5-FU, led to a 50% increase in mean survival time [48].


Conclusion

During cancer therapy, the gut microbiota is a dynamic structure, influenced by complex interactions between multiple factors, such as host immunity, chemotherapeutics, concomitant medications, environment and diet [6]. The ability of gut microbiota to augment the chemotherapeutic response has been clearly shown in animal models [8, 9, 13, 14, 3638]. Furthermore, these results have been successfully correlated with clinical research. Specific microbials associated with progression-free survival have been identified amongst patient cohorts and transfer of chemotherapeutic response has been achieved with administration of human faecal samples to a murine model [3638]. These findings have tremendous clinical potential, particularly in the field of novel immunotherapeutics (such anti-PD-L1), which at present are highly effective for only a small subset of patients [3638]. However, gut microbes are not always beneficial. An increased understanding of gut microbial metabolism has revealed contributions to drug toxicity such as mucositis and neuropathy that can even prove fatal [22, 23, 25, 26, 43]. The delicate equilibrium of bacterial community structure and host-microbiota interactions is also disrupted by chemotherapeutic administration, which promotes inflammation and mucosal destruction, and is associated with adverse outcome [3134].

The functional diversity of the microbiota has the capacity for both clinical benefit and harm. Obtaining a comprehensive understanding of the role of gut microbiota and their role in chemotherapy will require a dedicated and innovative systems medicine approach. Manipulation of the gut microbiota to achieve a perfect balance of chemotherapeutic efficacy and reduction of side effects presents a significant challenge [43]. However, with the potential of pharmacomicrobiomics to deliver a new era of cancer treatment, improving efficacy and tolerability, it is surely worth the effort.


Conflicts of interest

The authors declare no conflicts of interest.


References

1. Sender R, Fuchs S, and Milo R (2016) Are we really vastly outnumbered? revisiting the ratio of bacterial to host cells in humans Cell 164 337–340 https://doi.org/10.1016/j.cell.2016.01.013 PMID: 26824647

2. Schwabe RF and Jobin C (2013) The microbiome and cancer Nat Rev Cancer 13 800 https://doi.org/10.1038/nrc3610 PMID: 24132111 PMCID: 3986062

3. Rizkallah MR, Saad R, and Aziz RK (2010) The Human Microbiome Project, personalized medicine and the birth of pharmacomicrobiomics Curr Pharmacogenomics Pers Med 8 182–193 https://doi.org/10.2174/187569210792246326

4. Vétizou M, Pitt JM, and Daillère R, et al (2015) Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota Science 350 1079–1084 https://doi.org/10.1126/science.aad1329 PMID: 26541610 PMCID: 4721659

5. Perez-Chanona E and Trinchieri G (2016) The role of microbiota in cancer therapy Curr Opin Immunol 39 75–81 https://doi.org/10.1016/j.coi.2016.01.003 PMID: 26820225 PMCID: 4801762

6. Alexander JL, Wilson ID, and Teare J, et al (2017) Gut microbiota modulation of chemotherapy efficacy and toxicity Nat Rev Gastroenterol Hepatol 14 356–365 https://doi.org/10.1038/nrgastro.2017.20 PMID: 28270698

7. Erdman SE and Poutahidis T (2017) Gut microbiota modulate host immune cells in cancer development and growth Free Radic Biol Med 105 28–34 https://doi.org/10.1016/j.freeradbiomed.2016.11.013

8. Iida N, Dzutsev A, and Stewart CA, et al (2013) Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment Science 342 967–970 https://doi.org/10.1126/science.1240527 PMID: 24264989

9. Viaud S, Saccheri F, and Mignot G, et al (2013) The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide Science 342(6161) 971–976 https://doi.org/10.1126/science.1240537 PMID: 24264990 PMCID: 4048947

10. Scott TA, Quintaneiro LM, and Norvaisas P, et al (2017) Host-microbe co-metabolism dictates cancer drug efficacy in C. elegans Cell 169 442–456.e18 https://doi.org/10.1016/j.cell.2017.03.040

11. Claesson MJ, Cusack S, and Sullivan O, et al (2011) Composition, variability, and temporal stability of the intestinal microbiota of the elderly Proc Natl Acad Sci 108 4586–4591 https://doi.org/10.1073/pnas.1000097107

12. Taur Y, Jenq RR, and Perales M-A, et al (2014) The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation Blood 124 1174–1182 https://doi.org/10.1182/blood-2014-02-554725 PMID: 24939656 PMCID: 4133489

13. Daillère R, Vétizou M, and Waldschmitt N, et al (2016) Enterococcus hirae and barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects Immunity 45 931–943 https://doi.org/10.1016/j.immuni.2016.09.009 PMID: 27717798

14. Viaud S, Viaud S, and Saccheri F, et al (2016) Two bugs a NOD away from improving cancer therapy efficacy Immunity 45 356–365 https://doi.org/10.1126/science.1240537

15. Sharma P and Allison JP (2015) The future of immune checkpoint therapy Science 348 56–61 https://doi.org/10.1126/science.aaa8172 PMID: 25838373

16. Sivan A, Corrales L, and Hubert N, et al (2015) Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy Science 350 1084–1089 https://doi.org/10.1126/science.aac4255 PMID: 26541606 PMCID: 4873287

17. Kim D (2015) Gut microbiota-mediated drug-antibiotic interactions Drug Metab Dispos 43(10) 1581–1589 https://doi.org/10.1124/dmd.115.063867 PMID: 25926432

18. Klaassen CD and Cui JY (2015) Review: mechanisms of how the intestinal microbiota alters the effects of drugs and bile acids Drug Metab Dispos 43 1505–1521 https://doi.org/10.1124/dmd.115.065698 PMID: 26261286 PMCID: 4576672

19. García-González AP, Ritter AD, and Shrestha S, et al (2017) Bacterial metabolism affects the C. elegans response to cancer chemotherapeutics Cell 169 431–441.e8 https://doi.org/10.1016/j.cell.2017.03.046 PMCID: 5484065

20. Guthrie L, Gupta S, and Daily J, et al (2017) Human microbiome signatures of differential colorectal cancer drug metabolism NPJ Biofilms Microbiomes 3 27 https://doi.org/10.1038/s41522-017-0034-1 PMID: 29104759 PMCID: 5665930

21. Wallace BD, Roberts AB, and Pollet RM, et al (2015) Structure and inhibition of microbiome β-glucuronidases essential to the alleviation of cancer drug toxicity Chem Biol 22 1238–1249 https://doi.org/10.1016/j.chembiol.2015.08.005 PMID: 26364932 PMCID: 4575908

22. Kong R, Liu T, and Zhu X, et al (2014) Old drug new use—amoxapine and its metabolites as potent bacterial β-glucuronidase inhibitors for alleviating cancer drug toxicity Clin Cancer Res 20 3521–3530 https://doi.org/10.1158/1078-0432.CCR-14-0395 PMID: 24780296 PMCID: 4079752

23. Kodawara T, Higashi T, and Negoro Y, et al (2016) The inhibitory effect of ciprofloxacin on the β-glucuronidase-mediated deconjugation of the irinotecan metabolite SN-38-G Basic Clin Pharmacol Toxicol 118 333–337 https://doi.org/10.1111/bcpt.12511

24. Wallace BD, Wang H, and Lane KT, et al (2010) Alleviating cancer drug toxicity by inhibiting a bacterial enzyme Science 330 831–835 https://doi.org/10.1126/science.1191175 PMID: 21051639 PMCID: 3110694

25. Diasio RB (1998) Sorivudine and 5-fluorouracil; A clinically significant drug-drug interaction due to inhibition of dihydropyrimidine dehydrogenase Br J Clin Pharmacol 46 1–4 https://doi.org/10.1046/j.1365-2125.1998.00050.x PMID: 9690942 PMCID: 1873978

26. Nakayama H, Kinouchi T, and Kataoka K, et al (1997) 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 7 35–43 https://doi.org/10.1097/00008571-199702000-00005 PMID: 9110360

27. Chow J, Lee SM, and Shen Y, et al (2010) Host–bacterial symbiosis in health and disease Adv Immunol 107 243–274 https://doi.org/10.1016/B978-0-12-381300-8.00008-3

28. Cash HL, Whitham CV, and Behrendt CL, et al (2006) Symbiotic bacteria direct expression of an intestinal bactericidal lectin Science 313 1126–1130 https://doi.org/10.1126/science.1127119 PMID: 16931762 PMCID: 2716667

29. Jandhyala SM, Talukdar R, and Subramanyam C, et al (2015) Role of the normal gut microbiota World J Gastroenterol 21 8836–8847 https://doi.org/10.3748/wjg.v21.i29.8787

30. Yan F, Cao H, and Cover TL, et al (2011) Colon-specific delivery of a probiotic-derived soluble protein ameliorates intestinal inflammation in mice through an EGFR-dependent mechanism J Clin Invest 121 2242–2253 https://doi.org/10.1172/JCI44031 PMID: 21606592 PMCID: 3104743

31. van Vliet MJ, Tissing WJE, and Dun CAJ, et al (2009) 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 49 262–270 https://doi.org/10.1086/599346 PMID: 19514856

32. Fijlstra M, Ferdous M, and Koning AM, et al (2015) Substantial decreases in the number and diversity of microbiota during chemotherapy-induced gastrointestinal mucositis in a rat model Support Care Cancer 23 1513–1522 https://doi.org/10.1007/s00520-014-2487-6

33. Montassier E, Batard E, and Massart S, et al (2014) 16S rRNA gene pyrosequencing reveals shift in patient faecal microbiota during high-dose chemotherapy as conditioning regimen for bone marrow transplantation Microb Ecol 67 690–699 https://doi.org/10.1007/s00248-013-0355-4 PMID: 24402367

34. Frank M, Hennenberg EM, and Eyking A, et al (2015) Europe PMC Funders Group Europe PMC Funders Author Manuscripts TLR signaling modulates side effects of anticancer therapy in the small intestine J Immunol 194 1983–1995 https://doi.org/10.4049/jimmunol.1402481 PMID: 25589072 PMCID: 4338614

35. Dubin K, Callahan MK, and Ren B, et al (2016) Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis Nat Commun 7 10391 PMID: 26837003 PMCID: 4740747

36. Routy B, Le Chatelier E, and Derosa L, et al (2018) Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors Science 359 91–97 https://doi.org/10.1126/science.aan3706

37. Matson V, Fessler J, and Bao R, et al (2018) The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients Science 359 104–108 https://doi.org/10.1126/science.aao3290 PMID: 29302014

38. Gopalakrishnan V, Spencer CN, and Nezi L, et al (2018) Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients Science 359 97–103 https://doi.org/10.1126/science.aan4236

39. Zitvogel L, Galluzzi L, and Viaud S, et al (2015) Cancer and the gut microbiota: An unexpected link Sci Transl Med 7 1–10 https://doi.org/10.1126/scitranslmed.3010473

40. Bowen J, Stringer A, and Gibson R, et al (2007) VSL#3 Probiotic treatment reduces chemotherapy-induced diarrhoea and weight loss Cancer Biol Ther 6(9) 1449–1454 https://doi.org/10.4161/cbt.6.9.4622 PMID: 17881902

41. Wada M, Nagata S, and Saito M, et al (2010) Effects of the enteral administration of Bifidobacterium breve on patients undergoing chemotherapy for pediatric malignancies Support Care Cancer 18 751–759 https://doi.org/10.1007/s00520-009-0711-6

42. Marchesi JR, Adams DH, and Fava F, et al (2016) The gut microbiota and host health: a new clinical frontier Gut 65 330–339 https://doi.org/10.1136/gutjnl-2015-309990 PMCID: 4752653

43. Shen S, Lim G, and You Z, et al (2017) Gut microbiota is critical for the induction of chemotherapy-induced pain Nat Neurosci 20 1213 https://doi.org/10.1038/nn.4606 PMID: 28714953 PMCID: 5575957

44. Shono Y, Docampo MD, and Peled JU, et al (2016) Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice Sci Transl Med 8 339ra71–339ra71 https://doi.org/10.1126/scitranslmed.aaf2311 PMID: 27194729 PMCID: 4991773

45. Kakihana K, Fujioka Y, and Suda W, et al (2016) Fecal microbiota transplantation for patients with steroid-resistant acute graft-versus-host disease of the gut Blood 128 2083–2088 https://doi.org/10.1182/blood-2016-05-717652 PMID: 27461930 PMCID: 5085256

46. Staffas A, Burgos M, and Brink MRM Van Den (2017) The intestinal microbiota in allogeneic hematopoietic cell transplant and graft-versus-host disease Blood 129 927–934 https://doi.org/10.1182/blood-2016-09-691394 PMCID: 5324712

47. Pillai S, Al-lahham S, and Somasundaram R, et al (2012) E. coli-Produced BMP-2 as a chemopreventive strategy for colon cancer: a proof-of-concept study Gastroenterol Res Pract 2012 895462 https://doi.org/10.1155/2012/895462

48. Din MO, Danino T, and Prindle A, et al (2016) Synchronized cycles of bacterial lysis for in vivo delivery Nature 536 81–85 https://doi.org/10.1038/nature18930 PMID: 27437587 PMCID: 5048415

Related Articles

Margaret Baldwin, Brandon A Niyibizi, Daniella Rangira, Benita Rangira, Madeleine K Kazindu, Daniel Seifu, Cristina Daniela Stefan, Eulade Rugengamanzi, Achille V C Manirakiza
Felipe Carvajal V, Felipe Cardemil, Beatriz P Vásquez, Carolina E Oliva, Tamra A Barría, Maribel A Bruna, Leonor A Moyano, Felipe A Bustos, Paola A Muñoz, Cristóbal F Araya, Jorge E Oyarzún, Eduardo A Villa, Federico A Floriano, Alexis J del Rio, Sebastián R Indo, Enrique A Castellón, Héctor R Contreras
Salah Eddine El Herrag, Soraya Moulessehoul, Douniazad El Mehadji, Djamila Yekrou, Méghit Boumediène Khaled
Anishka Ramadhar, Phoebe N Miller, Mazvita Muchengeti, Juliana Kagura, Kathryn Chu, Cameron Gaskill
Table of Contents
Table of Contents