Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Mar 1.
Published in final edited form as: Dig Dis Sci. 2020 Mar;65(3):885–896. doi: 10.1007/s10620-020-06111-x

The gut microbiome modulates response to cancer immunotherapy

Md Abdul Wadud Khan 1, Gabriel Ologun 1, Reetakshi Arora 1, Jennifer L McQuade 2,*, Jennifer A Wargo 1,3,*
PMCID: PMC7678709  NIHMSID: NIHMS1562941  PMID: 32067144

Abstract

With the advent of next generation sequencing approaches, there has been a renaissance in the microbiome field. Microbial taxonomy and function can now be characterized relatively easily and rapidly - no longer mandating complex culturing approaches. With this renaissance, there is now a strong and growing appreciation for the role of the microbiome (referring to microbes and their genomes) in modulating many facets of physiology - including overall immunity. This is particularly true of the gut microbiome, and there is now an evolving body of literature demonstrating a role for gut microbes in modulating responses to cancer treatment - particularly immunotherapy. Gut microbes can modulate immunity and anti-tumor responses via a number of different interactions, and these will be discussed herein. Additionally, data regarding the impact of gut microbes on cancer immunotherapy response will be discussed, as will strategies to manipulate the microbiome to enhance therapeutic responses. These efforts to date are not completely optimized, however there is evidence of efficacy though much additional work is needed in this space. Nonetheless it is clear that the microbiome plays a central role in health and disease, and strategies to manipulate it in cancer and overall precision health are being explored.

Keywords: cancer, immunotherapy, microbiome

Gut microbes and their impact on immunity

Direct molecular and cellular interactions

With the introduction and colonization of the microbial species in the gut ecosystem, the host immune system has evolved to maintain a mutualistic relationship between microbes and host immunity [1]. Gut microbiota themselves have a tremendous impact on host physiology, and play a central role in the education and function of the host immune system. In turn, our immune system may shape the composition and function of the microbiota [2].

Gut microbes may interact with the host via a number of different mechanisms (Figure 1). This includes through pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharide and flagellin, which are recognized by pattern recognition receptors (PRRs; such as Toll-like receptors) located on the surface of intestinal epithelial cells [3]. Commensal bacteria can prime DCs which in turn can signal Toll-like receptors (TLRs) to educate the immune system regarding differential recognition of pathogenic vs non-pathogenic microbes. PAMPs from the gut may also interact directly with immune cells, inducing activation and maturation of antigen-presenting cells (APCs) including dendritic cells (DCs). These APCs may then translocate into mesenteric lymph nodes and mediate priming of B and T cells, which then can disseminate systemically and induce the differentiation of naïve T cells into CD4+ T cells [4] which function at distant sites [5].

Figure 1: Gut microbes and host immunity.

Figure 1:

Bacterial metabolites, anti-microbial peptides and bacteria can prime dendritic cells (DCs) which migrate to the lymph nodes to stimulate T and B cells. PAMPs from the gut may also interact directly with immune cells, inducing activation and maturation of antigen-presenting cells (APCs) including DCs. These APCs may then translocate into mesenteric lymph nodes and mediate priming of lymphocytes, and disseminate systemically and induce the differentiation of naïve T cells. These specific microbes or metabolites can prime DCs that in turn skew the response towards immune suppression or immune activation

Additionally, gut microbes modulate systemic immune responses via soluble immunomodulatory factors, circulating cytokines that can alter the activity of the immune cells. Preclinical data suggests segmented filamentous bacteria (SFB) can induce secretion of IL-22 from ILC3 cells which drive TH17 polarization in lamina propria DCs via production of serum amyloid A [6,7].

Furthermore, commensal bacteria provides a stimuli for the maturation of the Gut-associated lymphoid tissue (GALT), inducing B cell differentiation, maturation and activation [811] and IgA production [12]. The importance of this gut microbiota exposure to mucosal immunity is highlighted by the substantially defective gut mucosal immune systems seen in gnotobiotic mice characterized by a thin gastrointestinal mucus layer and aberrant IgA release in the gut [13,14].

There is also extensive data substantiating the role of gut microbe metabolites in modulating immune responses. Gut microbes produce a complex array of metabolites via utilization of the nutrients derived from the diet and other sources, and these can in turn influence the host immune system by receptor-specific binding to host cells. Examples of metabolites that may impact immunity include short chain fatty acids (SCFAs) and polyamines. SCFAs including acetate, propionate and butyrate and are produced by bacterial fermentation in the gut from dietary fibers that remain undigested or partially digested by the host. As the most abundant microbial metabolites in the intestine, SCFAs plays several crucial roles in host immunity [15]. First, these serve as the major energy sources of intestinal epithelial cells and maintain gut barrier integrity [16]. Prior work has demonstrated that association of acetate producing bacterium Bifidobacterium longum prevents the translocation of toxins from enteropathogenic Escherichia coli O157:H7 into the systemic compartment [17] SCFA may bind to G-coupled protein receptors (GPR43 and GPR41) of intestinal epithelial cells altering host gene expression and inducing autophagy and stimulating production of anti-inflammatory cytokines.. SCFAs can also influence innate immune cells. In the peripheral circulation, SCFAs deactivates nuclear factor-κB (NF-κB) and abrogate the expression of the pro-inflammatory cytokine tumor necrosis factor in mononuclear cells and neutrophils [18,19]. They also mediate immune cell phenotype through epigenetic mechanism, by inhibiting histone deacetylases (HDACs), with recent work demonstrating that this mechanism can promote CD8 memory cell differentiation [20]. Finally, SCFAs also influence antigen-specific adaptive immunity by stimulating the synthesis of IgA by B cells [21,22]. Furthermore, compounds such as tryptophan, indole, and bile acids may interact with aryl hydrocarbon receptor (AhR) and pregnane X receptor (PXR), thus modulating expression of inflammatory genes, upregulainge the expression of tight junctions, and inducing cytokine production by AhR expression lymphoid cells. In contrast, AhR expression on Tregs affects their homing and suppressive activity [23].

Polyamines which can be of dietary, host or microbial sources, are also important players in host immunity. Like SCFAs, polyamines such as putrescine, spermidine and spermine are important in mucosal immunity [16], inducing the secretion of IgA in the gut and maintaining barrier integrity [24,25]. Preclinical data showed that elevated level of polyamine in breastmilk promoted the maturation of gut CD8+ and CD4+ T cells [26]. Oral administration of the polyamine-producing bacterial strain Bifidobacterium animalis subsp. lactis LKM512 along with diet supplemented with the substrate (arginine) has been shown to decrease levels of intestinal TNF and IL-6 [27].

A specific group of gut microbiota, primarily Lactobacillus spp., produces metabolites by metabolizing dietary tryptophan [28]. These metabolites bind to the aryl hydrocarbon receptor (AHR) molecules expressed on the immune, epithelial and some tumor cells, demonstrating important role in regulating colonic inflammation [29]. In addition, the recognition of AHR by microbial metabolites was demonstrated to activate IL-22-producing ILC3 cells [30].

This body of literature demonstrating the profound effects that the gut microbiota exert on both mucosal and systemic immunity thus lays the foundation for investigations into the effects of the gut microbiota on anti-tumor immunity. Dedicated studies assessing metabolic profiles in the gut microbiome and peripheral circulation of patients on immunotherapy are currently underway, and are likely to reveal important insights into functional mechanisms through which gut microbes impact overall immunity and therapeutic response.

Pre-clinical studies substantiating gut microbes in response to immunotherapy

The earliest evidence regarding the role of the gut microbiome on response to immunotherapy was conducted in pre-clinical models. As noted above, there is a vast body of preclinical literature which has established the key role the gut microbiome plays in shaping mucosal as well as systemic immunity. Some of the earliest work indicating that the gut microbiome can also play a role in cancer immunology came from work with the chemotherapeutic cyclophosphamide[31]. Modulating the immune response is a key mechanism of action of cyclophosphamide. One mechanism through which this immune induction occurs is via translation of gram-positive bacteria into secondary lymphoid tissues which then stimulate Th17 and Th1 immune response [31]. Work in preclinical models of adoptive cellular therapy (ACT) has further demonstrated that gut microbiota modulation can help sustain transferred T cells in an IL-12 dependent manner, further supporting a role for the gut microbiota in adaptive anti-tumor immune responses [32].

Two ground-breaking preclinical studies published in Science in 2015, showed that mice with a favorable microbiota had enhanced response to immune checkpoint blockade (ICB) therapy compared to mice with an unfavorable microbiome - and furthermore that the beneficial phenotype was transferable by fecal transplant or cohousing [33,34]. More recent work has demonstrated that fecal microbiota transplant (FMT) from humans who responded versus did not respond to ICB can induce differential response to immunotherapy in mice [35,36]. In these studies, mice who received FMT from complete responder to ICB in turn had tumor response to ICB as well as increased tumor immune infiltrates whereas anti-tumor immunity was not induced in mice who received non-responder donor FMT. The precise mechanisms by which anti-tumor immunity is induced by the gut microbiota remains an area of active investigation. Beyond FMT, other work has focused on trying to define the specific bacterial consortia critical to the induction of anti-tumor immunity, with this work now being translated into clinical trials testing these consortia [37]. Other provocative work has demonstrated that the intratumoral microbiome can also influence response to therapy and can in turn be modulated via the gut microbiome[38,39]. Far from being sterile, pancreatic cancer has a distinct microbiome, apparently as a result of gut translocation[40]. FMT from long-term human survivors of pancreatic cancer into mice was shown to modulate not just the gut microbiome but also the tumor microbiome, as well as shape anti-tumor immunity[41]. Intratumoral bacteria may also induce chemotherapy resistance via in situ drug metabolism[42].

Clinical cohorts and observational studies on gut microbes and response to immunotherapy

These pre-clinical studies laid the foundation for subsequent observational studies in clinical cohorts. The initial evidence that the gut microbiome plays a role in treatment response in cancer were from studies on allogenic hematopoietic stem cell transplant (SCT) where significant association were observed between diversity of the gut microbiome and long-term outcomes in the setting of treatment of acute myelogenous leukemia (AML) with SCT [4345]. Improved survival was demonstrated in patients with a high diversity of the gut microbiome compared to those with an intermediate or low diversity. Subsequent studies focused on the risk of GVHD in the setting of SCT, showing differences in rates of GVHD depending on the composition of the gut microbiome, and also depending on antibiotic regimens used for potential infectious complications in the context of therapy[46].

More recently, several studies demonstrating strong associations between the gut microbiome and response to ICB in human cohorts across a variety of cancer types have been published[35,36,4749], building upon the prior foundational preclinical work.

One of the first of these studies was published by Dubin and colleagues in 2016, and reported results from a cohort study of patients with metastatic melanoma undergoing ICB treatment, and focused on the associations of signatures in the gut microbiome with toxicity to therapy - namely immune checkpoint-associated colitis. In this cohort, the investigators reported that an increased abundance of bacteria belonging to the Bacteriodetes phylum was correlated with a decreased risk of ICB-induced colitis [50] (Table 1). This work was followed by another study authored by Chaput et al in 2017, which demonstrated that the gut microbiome of responders to ICB was distinct from that of non-responders; however, the cohort was somewhat small (n=26). The group found associations of baseline gut microbiota composition with both clinical response as well as colitis; whereby patients whose baseline microbiota was enriched with the Faecalibacterium genus and other Firmicutes, had longer progression free survival (P=0.0039) and overall survival (P=0.051) compared to those dominated by Bacteroides [48].

Table 1.

Observational studies of role of gut microbiome in modulating response to immunotherapy

Reference Population Study Model n Bacteria Influence of Gut Microbiome in ICB Therapy Outcome Mechanism of Immune Modulation Proposed Geography
Gopalakrishnan, V., et al., 2018 Melanoma Patients 112 Ruminococcaceae Enhanced efficacy of PD-1 blockade therapy Higher abundance in responders Increased antigen presentation

Improved effector T cell function
Texas, USA
Elevating levels of effector T cells in peripheral blood and TILs
Increasing densities of CD8+ T cells in tumor microenvironment
Routy, B., et al., 2018 NSCLC and RCC Patients 100 Akkermansia muciniphila Enhanced efficacy of PD-1 blockade Higher abundance in responders Induce dendritic cell secretion of IL-12 Villejuif, France
Prolonging progression free survival
Matson, V., et al., 2018 Melanoma Patients/Mouse -- Bifidobacterium adolescents Enhanced efficacy of PD-1 blockade therapy Higher abundance in responders Enhance the activation of dendritic cells

Increase CD8+ T cells
Chicago, USA
Decreasing peripherally derived Tregs
Bifidobacterium longum Higher abundance in responders
Veillo nella parvula Higher abundance in responders Increased frequency of dendritic cells and greater T helper cell responses

Decreased frequency of regulatory T cells
Klebsiella pneumonia
Enterococcus faecium Decreasing peripherally derived Tregs
Collinsella aerofaciens
Parabacteroides merdae
Roseburia intestinalis Higher abundance in non-responders Increase in CD8+ tumor-infiltrating lymphocytes
Ruminococcus obeum
Chaput, N., et al., 2017 Melanoma Patients 26 Butyrate producing bacterium Enhanced CTLA-4 blockade efficacy therapy Higher abundance in responders -- Villejuif, France
Inducing activation of Treg
Promoting development of tolerogenic macrophages and dendritic cells
Prolonging progression free survival/overall survival
Faecalibacterium prausnitzii Enhanced CTLA-4 blockade efficacy therapy Prolonging overall survival
Elevating colitis risk
Gemmiger formicilis Enhanced efficacy of CTLA-4 blockade therapy -Prolonging progression free survival/overall survival
-Elevating colitis risk
Frankel, A., et al., 2017 Melanoma Patients 39 Bacteroides caccae Enhanced efficacy of ICB blockade therapy Higher abundance in responders -- Texas, USA
Dorea formicogenerans Enhanced efficacy of PD-1 blockade therapy (pembrolizumab)
Bacteroides thetaiotamicron Enhanced efficacy of CTLA-4 plus PD-1 blockade therapy
Faecalibacterium prausnitzii Enhanced efficacy of CTLA-4 plus PD-1 blockade therapy
Holdemania filiformis Enhanced efficacy of CTLA-4 plus PD-1 blockade therapy
Dubin, K., et al., 2016 Melanoma Patients 34 Bacteroidetes -- Decreased colitis secondary to anti- CTLA-4 Stimulate Tregulatory cell differentiation New York, USA
Sivan, A., et al., 2015 Melanoma Mouse -- Bifidobacterium Enhanced efficacy of PD-1 and PDL1 blockade therapy -- Enhance the activation of dendritic cells Increase CD8+ T cells Chicago, USA
Vetizou, M., et al., 2015 Sarcoma, Melanoma and Colon cancer Mouse -- Bacteroides Enhanced efficacy of CTLA-4 blockade therapy -- Induce T helper 1 immune responses in tumor-draining lymph nodes Promote maturation of intratumoral dendritic cells Villejuif, France

MSS=metagenomic shotgun sequencing; rRNA=ribosomal ribonucleic acid; gDNA=genomic deoxyribonucleic acid; PD-1=programmed cell death protein 1;CTLA-4=cytotoxic T-lymphocyte-associated protein 4; NSCLC = non-small cell lung cancer; RCC=renal cell cancer; Treg=regulatory T-cell

Another study published by Frankel and colleague in 2017 demonstrated an association between human gut microbes and metabolites on ICB response in a cohort of patients with metastatic melanoma. In their cohort, responders to treatment with combination anti-CTLA4 and anti-PD1 immune checkpoint blockade had an enrichment of taxa such as Faecalibacterium prausnitznii, Bacteroides thetaiotamicron, and Holdemania filiformis (Table 1) whereas an enrichment in Dorea formicogenerans was found in responders to anti-PD-1 monotherapy [49].

Following this, several groups co-published papers in Science in 2018 reporting a strong association between the diversity and composition of the gut microbiome and response to ICB in patients with melanoma and other cancers[35,36,47]. In these studies, distinct microbial signatures were observed in the gut microbiota of responders vs non-responders to ICB across a range of cancer types, including melanoma, non-small cell lung cancer, and kidney cancer. Interestingly, taxa associated with response differed among the cohorts with modest to little overlap (Table 1) - thus highlighting the complexity of identifying specific taxa to include in a bacterial consortia to enhance responses. However there are some functional redundancies in taxa identified across these cohorts [51] thus supporting the concept that we should be focusing more on bacterial function rather than phylogeny. This notion is strengthened by the observation regarding a higher diversity of gut microbes in responders to ICB, as this might suggest that functional redundancy in the gut microbiome is critically important to fill potential functional “holes” in the repertoire of the gut microbiome.

Further clinical evidence regarding the role of gut microbes in immunotherapy response

An additional piece of evidence substantiating the role of gut microbes in promoting immunotherapy response lies in the growing body of literature suggesting reduced response rates and impaired survival in patients on ICB who receive antibiotics during treatment [52]. One of the first studies to demonstrate this provocative finding was by Routy et al in 2018 [47]. This manuscript showed that if patients received antibiotics before or early after start of treatment with ICB, they had significantly worse survival. However, there were potential confounders in the study limiting conclusions based on these results. Nonetheless, since that time numerous studies have been published demonstrating a negative association between the use of antibiotics and response to immune checkpoint blockade across several cancer types [5355] with a recent meta-analysis summarizing these studies [52].

Together, these studies illuminate the potential deleterious impact of antibiotics on response to immune checkpoint blockade; however, important questions remain. These studies did not include longitudinal microbiome characterization before and during therapy (both ICB and antibiotics) - thus the direct relationship between antibiotic use and impact on the microbiome and associated immunity / anti-tumor immunity is unknown. Nonetheless the data in aggregate is quite compelling, and warrant (and mandate) additional studies to further define the impact of specific classes of anti-microbials on the gut microbiome and therapy response. Such studies will allow us to guide care and will also allow for more targeted approaches and engagement within and outside our respective disciplines regarding antibiotic stewardship.

Strategies targeting gut microbes in patients on immunotherapy

Based on this compelling preclinical and observational data supporting a role for the gut microbiome in cancer therapeutic outcomes, there are now multiple interventional studies examining the ability of gut microbiota modulation to enhance therapeutic efficacy and abrogate treatment-associated toxicity. Several strategies are being employed, building off of prior work on microbiome modulation in other diseases. (Figure 2).

Figure 2: Modulation of gut microbiome.

Figure 2:

Different strategies /approaches that can be employed to modulate the gut microbiome and can be used as interventions to improve immunotherapy treatments and abrogate the treatment associated toxicity i.e. Fecal microbiota transplant (FMT), administration of microbial consortia or probiotics, prebiotics that includes die and supplements, or use of personalized antibiotic therapy or phages.

Fecal microbiota transplant

Fecal microbiota transplant (FMT) is the most well-established strategy to modulate the microbiome. In FMT, the gut microbiome in its entirety is transferred from a donor to a recipient as a stool suspension, administered either endoscopically on in the form of pills. FMT is a proven therapy for recurrent and refractory Clostridium difficile infection (CDI) [56,57] and has also been tested as a treatment modality for many other dysbiotic diseases and is now being investigated in the context of cancer therapy. This includes studies of FMT in SCT patients such as a trial of patients with acute myeloid leukemia, with FMT targeting the prevention of dysbiosis and eradication of multi-drug resistant bacteria (NCT02928523).

There are also a growing number of trials now investigating FMT in cancer patients undergoing immunotherapy (NCT03353402, NCT03341143, NCT03772899, NCT03817125, NCT04130763, NCT04116775, NCT04056026). This field is still at its beginning, and thus these are small pilot Phase I/II studies with primarily safety endpoints, though of course the ultimate strategy is to enhance therapeutic response. These studies vary in selection, delivery method, and FMT duration reflecting that the optimal FMT regimen in this population is still being defined (further discussed below). Very early data from one of these studies being conducted in Israel (NCT03353402) was presented at the 2019 AACR meeting with exciting early safety and efficacy data. This study is enrolling patients with anti-PD1 refractory melanoma who received FMT from a CR donor + anti-PD1, and reported responses in 3 of 10 patients with no adverse safety signals.

FMT is also being studied as a way to abrogate treatment related toxicities. A provocative case study of FMT for steroid-refractory ICB-induced colitis showed remarkable efficacy in two patients, with both histologic and clinical remission [58]. Based on this experience, at least two prospective clinical trials of FMT for ICB-induced toxicity are now underway (NCT03772899 and NCT03819296).

Defined bacterial consortia

Another method of modulating the gut microbiota is through the oral administration of live bacterial consortia or even a single putatively beneficial bacterium as pills (i.e., probiotics). The advantage to this approach is ease and simplicity. One such ongoing trial (NCT03817125) in metastatic melanoma patients undergoing immunotherapy (anti-PD1) employing a defined orally administered microbial formulation (SER-401) is intended to assess safety, clinical response, as well as change in gut microbiome and immunity. Another ongoing clinical trial of a proprietary bacterial strain (MRx0518) in combination with anti-PD-1 therapy is enrolling patients with melanoma, bladder cancer, non-small cell lung cancer and renal cell carcinoma (NCT03637803). While the administration of probiotics along with immunotherapy in cancer is relatively new, it has been employed in combination with other forms of cancer therapies such as chemotherapy and SCT [5962]. However, significant caution should be taken until preclinical and clinical studies fully evaluate the safety of using commercially available probiotics, especially given significant variability and lack of regulation of over the counter probiotics [63].

Diet and prebiotics

Gut microbiota play a major role in harvesting important nutrients for the host by digestion of the food and, as noted above, resultant metabolites play a substantial role in host immunity [6466]. For example, the plant polysaccharide inulin can stimulate an outgrowth of the fiber-fermenting Faecalibacterium and Bifidobacterium species [67], two taxa recently associated with enhanced response to immune checkpoint blockade [36,68] whereas supplementation with inulin and fructo-oligosaccharide may select for such beneficial bacteria as Lactobacillus and Bifidobacterium, [69]. Thus, there is a strong rationale for investigating the effects of dietary interventions on the gut microbiota and immunity in the context of ICB. However, while diet is a convenient and accessible strategy, behavior change is notoriously difficult and defining what diet components or patterns are optimal in this context remains an unmet need.

Fecal transplant and unique issues with regard to patients with cancer

Thus, while gut microbiota can be modulated by a variety of methods, the most established strategy is through the use of FMT. However, there are several important considerations in designing and implementing the FMT trials with cancer patients [63]. These include donor selection, patient population, pre-conditioning, FMT modality, duration of treatment, and endpoints. In regards to donor selection, the primary considerations are healthy donor vs ICB complete responder, and microbiome profiling. Though FMT from healthy donors has proven to be effective in treating non-malignant diseases such as CDI [70], whether FMT from healthy donor is sufficient to induce response to ICB or whether a responder donor is needed has not been established nor has the practice of microbiome profiling to select optimal donors.

Another area of controversy is the use of pre-conditioning antibiotics. Antibiotics are routinely used prior to FMT to ablate the native microbiome and facilitate engraftment. However, as has been noted above, antibiotic use has been associated with worse outcomes with ICB[71,72]. A preclinical study reported that pre-treatment with broad-spectrum antibiotics did not affect the overall engraftment of donor microbiota in the recipients but improved the engraftment of beneficial bacterial taxa such as Bifidobacterium [73]. The effects of the specific antibiotic regimen typically used for FMT preconditioning (oral Vancomcyin) in the context of ICB is also an unanswered question.

The optimal route of administration of FMT in the setting of immunotherapy also needs to be defined. For CDI, a single endoscopically delivered FMT is sufficient for durable engraftment. However, this is not the case for other disease states which are less profoundly dysbiotic and thus may be more resistant to colonization[74]. For example, results of trials of FMT for inflammatory bowel disease have been decidedly mixed, but a much more significant benefit was seen in a recent study that employed ongoing FMT dosing x 8 weeks[75]. Though this study utilize repeated enemas, other studies have demonstrated frozen stool pills to be a convenient and effective alternative to traditional endoscopic approaches and this is an approach also being tested in the setting of immunotherapy[76].

Irrespective of the source for the FMT, the fecal samples may be considered as an imprecise source of immune-potentiating microbial community that may potentially contain harmful bacteria including pathogens, drug resistant bacteria and microbial groups associated with metabolic disorders or even carcinogenesis. A study led by DeFilipp et al. demonstrated that two patients following FMT developed bacteremia with extended-spectrum beta-lactamase (ESBL)–producing Escherichia coli and one of them who underwent hematopoietic cell transplantation died [77]. This warrant updating the donor screening strategy routinely, considering any potential emerging bacteria or viruses that may cause adverse events in the recipients while donors remain asymptomatic. However, overall vast experience with FMT for CDI and other indications support that this is generally a safe and well-tolerated approach.

Conclusions and future directions

Through multiple preclinical and human observational studies, it is now well-established that the gut microbiome is associated with response to cancer immunotherapy. However, there remains considerable work to be done in establishing mechanism as well as the microbial features causally linked to response. Preclinical models further suggest microbiome modulation as a therapeutic strategy to enhance response to immunotherapy and this approach is now being tested in multiple ongoing clinical trials. Modalities being tested include FMT, probiotics and bacterial consortia, and prebiotics and diet. Each of these strategies has important variables to be considered, and these early studies with integrated biospecimen collection will yield important data to help us further optimize these approaches and continue to move this field further.

Key Findings.

  • The gut microbiome plays a key role in shaping mucosal and systemic immunity

  • Work in preclinical models first demonstrated that the gut microbiome can also impact anti-tumor immunity

  • Data from multiple human cohorts now supports that there is a strong association between gut microbiome profile and response to immune checkpoint blockade (ICB)

  • Microbiome modulation can impact response to ICB in mice and various approaches of microbiome modulation are now being tested as a strategy to enhance response and ameliorate toxicity to ICB in multiple human interventional studies

  • The precise mechanisms by which the gut microbiome impacts anti-tumor immunity remains an area of active investigation

Acknowledgements:

J.A.W. is supported by the NIH (1 R01 CA219896-01A1), U.S- Israel Binational Science Foundation (201332), the Melanoma Research Alliance (4022024), American Association for Cancer Research Stand Up To Cancer (SU2C-AACR-IRG-19-17), Department of Defense (W81XWH-16-1-0121), MD Anderson Cancer Center Multidisciplinary Research Program Grant, Andrew Sabin Family Fellows Program, and MD Anderson Cancer Center’s Melanoma Moon Shots Program. J.A.W. is a member of the Parker Institute for Cancer Immunotherapy at MD Anderson Cancer Center. Figures were prepared using Biorender.com.

Biographies

graphic file with name nihms-1562941-b0001.gif

graphic file with name nihms-1562941-b0002.gif

graphic file with name nihms-1562941-b0003.gif

graphic file with name nihms-1562941-b0004.gif

Footnotes

Publisher's Disclaimer: This Author Accepted Manuscript is a PDF file of an unedited peer-reviewed manuscript that has been accepted for publication but has not been copyedited or corrected. The official version of record that is published in the journal is kept up to date and so may therefore differ from this version.

Conflict of Interest

J. Wargo is an inventor on a US patent application (PCT/US17/53.717) submitted by the University of Texas MD Anderson Cancer Center that covers methods to enhance immune checkpoint blockade responses by modulating the microbiome. J. Wargo reports compensation for speaker’s bureau and honoraria from Imedex, Dava Oncology, Omniprex, Illumina, Exelixis, Dhome, Gilead, PeerView, Physician Education Resource, MedImmune and Bristol-Myers Squibb. J. Wargo serves as a consultant / advisory board member for Roche/Genentech, Novartis, AstraZeneca, GlaxoSmithKline, Bristol-Myers Squibb, Merck, Biothera Pharmaceuticals and Microbiome DX. J. Wargo also receives research support from GlaxoSmithKline, Roche/Genentech, Bristol-Myers Squibb, and Novartis.

J. McQuade serves as a consultant for Bristol-Myers Squibb, Merck and Roche.

G. Ologun reports grants from National Institutes of Health T32 CA 009599 and MD Anderson Cancer Center support grant (P30 CA016672).

The other authors declared no conflict of interest.

References

  • 1.Hooper LV, MacPherson AJ Immune adaptations that maintain homeostasis with the intestinal microbiota. Nat Rev Immunol 2010. March;10(3):159–69. [DOI] [PubMed] [Google Scholar]
  • 2.Shi N, Li N, Duan X, Niu H. Interaction between the gut microbiome and mucosal immune system. Mil Med Res 2017. April;4(1). DOI: 10.1186/s40779-017-0122-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Janeway CA. Pillars article: approaching the asymptote? Evolution and revolution in immunology. Cold spring harb symp quant biol. 1989. 54: 1–13. J Immunol 2013. November [cited 2020 Jan 14].; 191(9):4475–87. [PubMed] [Google Scholar]
  • 4.Lathrop SK, Bloom SM, Rao SM, Nutsch K, Lio CW, Santacruz N, et al. Peripheral education of the immune system by colonic commensal microbiota. Nature. 2011. October;478(7368):250–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Stary G, Olive A, Radovic-Moreno AF, Gondek D, Alvarez D, Basto PA, et al. VACCINES. A mucosal vaccine against Chlamydia trachomatis generates two waves of protective memory T cells. Science. 2015. June;348(6241):aaa8205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Sano T, Huang W, Hall JA, Yang Y, Chen A, Gavzy SJ, et al. An IL-23R/IL-22 Circuit Regulates Epithelial Serum Amyloid A to Promote Local Effector Th17 Responses. Cell. 2015. October;163(2):381–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, et al. Induction of Intestinal Th17 Cells by Segmented Filamentous Bacteria. Cell. 2009. October;139(3):485–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Fagarasan S, Muramatsu M, Suzuki K, Nagaoka H, Hiai H, Honjo T. Critical roles of activation-induced cytidine deaminase in the homeostasis of gut flora. Science (80-). 2002. November;298(5597):1424–7. [DOI] [PubMed] [Google Scholar]
  • 9.Lundell A-C, Björnsson V, Ljung A, Ceder M, Johansen S, Lindhagen G, et al. Infant B Cell Memory Differentiation and Early Gut Bacterial Colonization. J Immunol 2012. May;188(9):4315–22. [DOI] [PubMed] [Google Scholar]
  • 10.Ouwehand A, Isolauri E, Salminen S. The role of the intestinal microflora for the development of the immune system in early childhood. Eur J Nutr 2002. November;41(SUPPL. 1):32–7. [DOI] [PubMed] [Google Scholar]
  • 11.Wei B, Su TT, Dalwadi H, Stephan RP, Fujiwara D, Huang TT, et al. Resident enteric microbiota and CD8+ T cells shape the abundance of marginal zone B cells. Eur J Immunol 2008. December;38(12):3411–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Mantis NJ, Rol N, Corthésy B. Secretory IgA’s complex roles in immunity and mucosal homeostasis in the gut. Mucosal Immunol 2011. November;4(6):603–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Johansson MEV, Jakobsson HE, Holmén-Larsson J, Schütte A, Ermund A, Rodríguez-Piñeiro AM, et al. Normalization of Host Intestinal Mucus Layers Requires Long-Term Microbial Colonization. Cell Host Microbe 2015. November;18(5):582–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Spiljar M, Merkler D, Trajkovski M. The Immune System Bridges the Gut Microbiota with Systemic Energy Homeostasis: Focus on TLRs, Mucosal Barrier, and SCFAs. Front Immunol 2017;8:1353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Corrêa-Oliveira R, Fachi JL, Vieira A, Sato FT, Vinolo MAR. Regulation of immune cell function by short-chain fatty acids. Clin Transl Immunol 2016. April;5(4). DOI: 10.1038/cti.2016.17 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol 2016. June;16(6):341–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Fukuda S, Toh H, Hase K, Oshima K, Nakanishi Y, Yoshimura K, et al. Bifidobacteria can protect from enteropathogenic i... [Nature. 2011] - PubMed result. Nature. 2011. January;469(7331):543–7. [DOI] [PubMed] [Google Scholar]
  • 18.Usami M, Kishimoto K, Ohata A, Miyoshi M, Aoyama M, Fueda Y, et al. Butyrate and trichostatin A attenuate nuclear factor kappaB activation and tumor necrosis factor alpha secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells. Nutr Res 2008. May;28(5):321–8. [DOI] [PubMed] [Google Scholar]
  • 19.Vinolo MAR, Rodrigues HG, Hatanaka E, Sato FT, Sampaio SC, Curi R. Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J Nutr Biochem 2011. September;22(9):849–55. [DOI] [PubMed] [Google Scholar]
  • 20.Park J, Kim M, Kang SG, Jannasch AH, Cooper B, Patterson J, et al. Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway. Mucosal Immunol 2015. January;8(1):80–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Pabst O New concepts in the generation and functions of IgA. Nat Rev Immunol 2012. December;12(12):821–32. [DOI] [PubMed] [Google Scholar]
  • 22.Luu M, Weigand K, Wedi F, Breidenbend C, Leister H, Pautz S, et al. Regulation of the effector function of CD8+ T cells by gut microbiota-derived metabolite butyrate. Sci Rep 2018;8(1):14430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Ye J, Qiu J, Bostick JW, Ueda A, Schjerven H, Li S, et al. The Aryl Hydrocarbon Receptor Preferentially Marks and Promotes Gut Regulatory T Cells. Cell Rep 2017. November;21(8):2277–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Dufour C, Dandrifosse G, Forget P, Vermesse F, Romain N, Lepoint P. Spermine and spermidine induce intestinal maturation in the rat. Gastroenterology. 1988;95(1):112–6. [DOI] [PubMed] [Google Scholar]
  • 25.Buts JP, De Keyser N, Kolanowski J, Sokal E, Van Hoof F. Maturation of villus and crypt cell functions in rat small intestine - Role of dietary polyamines. Dig Dis Sci 1993. June;38(6):1091–8. [DOI] [PubMed] [Google Scholar]
  • 26.Pérez-Cano FJ, González-Castro A, Castellote C, Franch A, Castell M. Influence of breast milk polyamines on suckling rat immune system maturation. Dev Comp Immunol 2010. February;34(2):210–8. [DOI] [PubMed] [Google Scholar]
  • 27.Kibe R, Kurihara S, Sakai Y, Suzuki H, Ooga T, Sawaki E, et al. Upregulation of colonic luminal polyamines produced by intestinal microbiota delays senescence in mice. Sci Rep 2014. April;4:4548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hubbard TD, Murray IA, Bisson WH, Lahoti TS, Gowda K, Amin SG, et al. Adaptation of the human aryl hydrocarbon receptor to sense microbiota-derived indoles. Sci Rep 2015. August;5 DOI: 10.1038/srep12689 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Li Y, Innocentin S, Withers DR, Roberts NA, Gallagher AR, Grigorieva EF, et al. Exogenous stimuli maintain intraepithelial lymphocytes via aryl hydrocarbon receptor activation. Cell. 2011. October;147(3):629–40. [DOI] [PubMed] [Google Scholar]
  • 30.Kiss EA, Vonarbourg C, Kopfmann S, Hobeika E, Finke D, Esser C, et al. Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science (80-). 2011. December;334(6062):1561–5. [DOI] [PubMed] [Google Scholar]
  • 31.Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science (80-). 2013;342(6161):971–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Uribe-Herranz M, Rafail S, Beghi S, Gil-de-Gómez L, Verginadis I, Bittinger K, et al. Gut microbiota modulate dendritic cell antigen presentation and radiotherapy-induced antitumor immune response. J Clin Invest 2019. December DOI: 10.1172/JCI124332 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science (80-). 2015. November;350(6264):1084–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015. November;350(6264):1079–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV., et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science (80-). 2018;359(6371):97–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science (80-). 2018. January;359(6371):104–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Tanoue T, Morita S, Plichta DR, Skelly AN, Suda W, Sugiura Y, et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature. 2019. January;565(7741):600–5. [DOI] [PubMed] [Google Scholar]
  • 38.Riquelme E, Zhang Y, Zhang L, Montiel M, Zoltan M, Dong W, et al. Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes. Cell. 2019. August;178(4):795–806.e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, et al. The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression. Cancer Discov 2018;8(4):403–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Riquelme E, Zhang Y, Zhang L, Montiel M, Zoltan M, Dong W, et al. Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes. Cell. 2019. August;178(4):795–806.e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, et al. The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression. Cancer Discov 2018;8(4):403–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Geller LT, Straussman R. Intratumoral bacteria may elicit chemoresistance by metabolizing anticancer agents. Mol Cell Oncol 2018. January;5(1). DOI: 10.1080/23723556.2017.1405139 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Jenq RR, Taur Y, Devlin SM, Ponce DM, Goldberg JD, Ahr KF, et al. Intestinal Blautia Is Associated with Reduced Death from Graft-versus-Host Disease. Biol Blood Marrow Transplant. 2015. August;21(8):1373–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Shono Y, Docampo MD, Peled JU, Perobelli SM, Velardi E, Tsai JJ, et al. Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice. Sci Transl Med 2016;8(339):339ra71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Taur Y, Jenq RR, Perales MA, Littmann ER, Morjaria S, Ling L, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014. August;124(7):1174–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Shono Y, Docampo MD, Peled JU, Perobelli SM, Velardi E, Tsai JJ, et al. Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice. Sci Transl Med 2016;8(339):339ra71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science (80-). 2018. January;359(6371):91–7. [DOI] [PubMed] [Google Scholar]
  • 48.Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol 2017. June;28(6):1368–79. [DOI] [PubMed] [Google Scholar]
  • 49.Frankel AE, Coughlin LA, Kim J, Froehlich TW, Xie Y, Frenkel EP, et al. Metagenomic Shotgun Sequencing and Unbiased Metabolomic Profiling Identify Specific Human Gut Microbiota and Metabolites Associated with Immune Checkpoint Therapy Efficacy in Melanoma Patients. Neoplasia (United States). 2017. October;19(10):848–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Dubin K, Callahan MK, Ren B, Khanin R, Viale A, Ling L, et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun 2016. February;7:10391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gharaibeh RZ, Jobin C. Microbiota and cancer immunotherapy: In search of microbial signals. Gut 2019. March;68(3):385–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Huang XZ, Gao P, Song YX, Xu Y, Sun JX, Chen XW, et al. Antibiotic use and the efficacy of immune checkpoint inhibitors in cancer patients: a pooled analysis of 2740 cancer patients. Oncoimmunology. 2019. December;8(12). DOI: 10.1080/2162402X.2019.1665973 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Derosa L, Hellmann MD, Spaziano M, Halpenny D, Fidelle M, Rizvi H, et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Ann Oncol 2018. June;29(6):1437–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Mielgo-Rubio X, Chara L, Sotelo-Lezama M, Lopez Castro R, Rubio-Martínez J, Velastegui A, et al. MA10.01 Antibiotic Use and PD-1 Inhibitors: Shorter Survival in Lung Cancer, Especially When Given Intravenously. Type of Infection Also Matters. J Thorac Oncol 2018. October;13(10):S389. [Google Scholar]
  • 55.Do TP, Hegde AM, Cherry CR, Stroud CRG, Sharma N, Cherukuri SD, et al. Antibiotic use and overall survival in lung cancer patients receiving nivolumab. J Clin Oncol 2018. May;36(15_suppl):e15109–e15109. [Google Scholar]
  • 56.Kelly CR, Ihunnah C, Fischer M, Khoruts A, Surawicz C, Afzali A, et al. Fecal microbiota transplant for treatment of clostridium difficile infection in immunocompromised patients. Am J Gastroenterol 2014;109(7):1065–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Khoruts A, Rank KM, Newman KM, Viskocil K, Vaughn BP, Hamilton MJ, et al. Inflammatory Bowel Disease Affects the Outcome of Fecal Microbiota Transplantation for Recurrent Clostridium difficile Infection. Clin Gastroenterol Hepatol 2016. October;14(10):1433–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Wang Y, Wiesnoski DH, Helmink BA, Gopalakrishnan V, Choi K, DuPont HL, et al. Fecal microbiota transplantation for refractory immune checkpoint inhibitor-associated colitis. Nat Med 2018;24(12):1804–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Tian Y, Li M, Song W, Jiang R, Li YQ. Effects of probiotics on chemotherapy in patients with lung cancer. Oncol Lett 2019. March;17(3):2836–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Riehl TE, Alvarado D, Ee X, Zuckerman A, Foster L, Kapoor V, et al. Lactobacillus rhamnosus GG protects the intestinal epithelium from radiation injury through release of lipoteichoic acid, macrophage activation and the migration of mesenchymal stem cells. Gut 2019. June;68(6):1003–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Mego M, Holec V, Drgona L, Hainova K, Ciernikova S, Zajac V. Probiotic bacteria in cancer patients undergoing chemotherapy and radiation therapy. Complement Ther Med 2013. December;21(6):712–23. [DOI] [PubMed] [Google Scholar]
  • 62.Panebianco C, Andriulli A, Pazienza V. Pharmacomicrobiomics: exploiting the drug-microbiota interactions in anticancer therapies. Microbiome. 2018;6(1):92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.McQuade JL, Daniel CR, Helmink BA, Wargo JA. Modulating the microbiome to improve therapeutic response in cancer. Lancet Oncol 2019. February;20(2):e77–91. [DOI] [PubMed] [Google Scholar]
  • 64.Bäckhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science (80-) 2005. March;307(5717):1915–20. [DOI] [PubMed] [Google Scholar]
  • 65.Ma N, Guo P, Zhang J, He T, Kim SW, Zhang G, et al. Nutrients mediate intestinal bacteria-mucosal immune crosstalk. Front Immunol 2018;9(JAN):5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Shortt C, Hasselwander O, Meynier A, Nauta A, Fernández EN, Putz P, et al. Systematic review of the effects of the intestinal microbiota on selected nutrients and non-nutrients. Eur J Nutr 2018. February;57(1):25–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Saedisomeolia A, Wood LG, Garg ML, Gibson PG, Wark PAB. Anti-inflammatory effects of long-chain n-3 PUFA in rhinovirus-infected cultured airway epithelial cells. Br J Nutr 2009. February;101(4):533–40. [DOI] [PubMed] [Google Scholar]
  • 68.Gopalakrishnan V, Helmink BA, Spencer CN, Reuben A, Wargo JA. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell. 2018. April;33(4):570–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.García-Peris P, Velasco C, Lozano MA, Moreno Y, Paron L, de la Cuerda C, et al. Effect of a mixture of inulin and fructo-oligosaccharide on Lactobacillus and Bifidobacterium intestinal microbiota of patients receiving radiotherapy: a randomised, double-blind, placebo-controlled trial. Nutr Hosp 27(6):1908–15. [DOI] [PubMed] [Google Scholar]
  • 70.Gough E, Shaikh H, Manges AR. Systematic review of intestinal microbiota transplantation (fecal bacteriotherapy) for recurrent clostridium difficile infection. Clin Infect Dis 2011. November;53(10):994–1002. [DOI] [PubMed] [Google Scholar]
  • 71.Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science (80-). 2018. January;359(6371):91–7. [DOI] [PubMed] [Google Scholar]
  • 72.Huang X-Z, Gao P, Song Y-X, Xu Y, Sun J-X, Chen X-W, et al. Antibiotic use and the efficacy of immune checkpoint inhibitors in cancer patients: a pooled analysis of 2740 cancer patients. Oncoimmunology. 2019;8(12):e1665973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Freitag TL, Hartikainen A, Jouhten H, Sahl C, Meri S, Anttila V-J, et al. Minor effect of antibiotic pre-treatment on the engraftment of donor microbiota in fecal transplantation in mice. Front Microbiol 2019;10:2685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Siegmund B Is intensity the solution for FMT in ulcerative colitis? Lancet (London, England). 2017;389(10075):1170–2. [DOI] [PubMed] [Google Scholar]
  • 75.Paramsothy S, Kamm MA, Kaakoush NO, Walsh AJ, van den Bogaerde J, Samuel D, et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. Lancet. 2017. March;389(10075):1218–28. [DOI] [PubMed] [Google Scholar]
  • 76.Kao D, Roach B, Silva M, Beck P, Rioux K, Kaplan GG, et al. Effect of oral capsule– vs colonoscopy-delivered fecal microbiota transplantation on recurrent Clostridium difficile infection: A randomized clinical trial. JAMA - J Am Med Assoc 2017. November;318(20):1985–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.DeFilipp Z, Bloom PP, Torres Soto M, Mansour MK, Sater MRA, Huntley MH, et al. Drug-Resistant E. coli Bacteremia Transmitted by Fecal Microbiota Transplant. N Engl J Med 2019. October DOI: 10.1056/NEJMoa1910437 [DOI] [PubMed] [Google Scholar]

RESOURCES