Systematic Reviews Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Oncol. Nov 24, 2022; 13(11): 929-942
Published online Nov 24, 2022. doi: 10.5306/wjco.v13.i11.929
Gut microbiota diversity and composition in predicting immunotherapy response and immunotherapy-related colitis in melanoma patients: A systematic review
Oliver Oey, Department of Medical Oncology, St John of God Midland Public and Private Hospital, Midland, Perth 6004, WA, Australia
Oliver Oey, School of Medicine, University of Western Australia, Perth 6009, WA, Australia
Yu-Yang Liu, School of Medicine, University of Queensland, Brisbane 4072, QLD, Australia
Angela Felicia Sunjaya, Faculty of Medicine, Tarumanagara University, Jakarta 11440, Indonesia
Daniel Martin Simadibrata, School of Medicine, University of Indonesia, Jakarta 10430, Indonesia
Muhammad Adnan Khattak, Department of Medical Oncology, Fiona Stanley Hospital, Perth 6150, WA, Australia
Muhammad Adnan Khattak, Elin Gray, School of Medical Sciences, Edith Cowan University, Perth 6027, WA, Australia
Muhammad Adnan Khattak, Elin Gray, Centre for Precision Health, Edith Cowan University, Perth 6027, WA, Australia
ORCID number: Oliver Oey (0000-0003-0673-6804); Yu-Yang Liu (0000-0002-7438-7258); Angela Felicia Sunjaya (0000-0001-8831-0449); Daniel Martin Simadibrata (0000-0002-7512-2112); Elin Gray (0000-0002-8613-3570).
Author contributions: Oey O, Simadibrata DM, Gray E and Khattak MA contributed to the study conception and design; Oey O and Liu Y performed data extraction; Oey O and Simadibrata DM performed risk of bias assessment; Oey O, Liu Y, Sunjaya AF, Simadibrata DM, Khattak MA and Gray E performed data analysis; Oey O written the first draft of the manuscript; all authors commented on previous versions of the manuscript, read and approved the final manuscript.
Conflict-of-interest statement: Khattak MA reports receiving travel support from Merck Sharp and Dohme (MSD), Bristol-Myers Squibb and Merck Serono. Gray E reports receiving travel sponsorship from MSD. Oey O, Liu Y, Sunjaya AF, and Simadibrata DM report no competing interests.
PRISMA 2009 Checklist statement: All authors have read the PRISMA 2009 Checklist, and the manuscript was prepared and revised according to the PRISMA 2009 Checklist.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Oliver Oey, MD, Doctor, Researcher, Department of Internal Medicine, St John of God Midland Public and Private Hospital, Midland, No. 1 Clayton Street, Perth 6004, WA, Australia. oliver.oey@sjog.org.au
Received: October 19, 2022
Peer-review started: October 19, 2022
First decision: October 28, 2022
Revised: October 30, 2022
Accepted: November 6, 2022
Article in press: November 6, 2022
Published online: November 24, 2022

Abstract
BACKGROUND

Gut microbiome (GM) composition and diversity have recently been studied as a biomarker of response to immune checkpoint blockade therapy (ICB) and of ICB-related colitis.

AIM

To conduct a systematic review on the role of GM composition and diversity in predicting response and colitis in patients with melanoma treated with ICB.

METHODS

The review protocol was registered in PROSPERO: CRD42021228018. From a total of 300 studies, nine studies met inclusion criteria. Two studies were phase I clinical trials, while the remainder were prospective observational studies. All but one study has moderate risk of bias. In addition, we conducted a relevant search by Reference Citation Analysis (RCA) (https://www.referencecitationanalysis.com).

RESULTS

Fecal samples enriched in Firmicutes phylum were associated with good response to ICB, whereas the Bacteroidales family was associated with poor response to ICB. Samples with greater GM diversity were associated with more favorable response to ICB [hazard ratio (HR) = 3.57, 95% confidence interval = 1.02-12.52, P < 0.05]. Fecal samples with a higher abundance in Firmicutes were more susceptible to ICB-related colitis (P < 0.01) whereas samples enriched in Bacteroidetes were more resistant to ICB-related colitis (P < 0.05). Overall, there was limited concordance in the organisms in the GM identified to be associated with response to ICB, and studies evaluating GM diversity showed conflicting results.

CONCLUSION

This highlights the need for further prospective studies to confirm whether the GM could be used as a biomarker and potential intervention to modulate ICB response in melanoma patients.

Key Words: Melanoma, Gut microbiome, Microbiota, Immunotherapy, Biomarker, Immune checkpoint blockade therapy

Core Tip: Since the introduction of immune checkpoint inhibitors as part of standard of care for melanoma patients, there has been a growing interest in identifying biomarkers of response and immune related adverse events. Amongst these biomarkers, the composition of the gut microbiome has been one of the most intriguing discoveries. Our aim was to ascertain the current published evidence on the gut microbiome diversity and composition as a biomarker of response to immunotherapy. We demonstrated high variability in the results and limited concordance on the organisms identified. We highlight the conflicting aspects of these reports as well as their few commonalities.



INTRODUCTION

Melanoma is the most lethal form of skin cancer accounting for 73% of skin-cancer related mortality and over 50000 deaths worldwide annually[1,2]. Survival for metastatic melanoma has significantly improved since the introduction of immunotherapy and targeted therapy with a 5-year survival rate of up to 50%[3-5]. Currently, the standard first-line therapy for metastatic melanoma include BRAF-targeted therapies and immune checkpoint blockade (ICB) consisting either anti-programmed death (PD)-1 monotherapy or combination of anti-PD-1 as well as anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) therapy[6]. Despite the considerable benefit of ICB, 40%-60% of melanoma patients do not experience objective responses to the therapy[7-9]. Thus, tremendous efforts are now focused on identifying novel biomarkers which could accurately predict the subset of patients who would benefit from ICB[10-14]. These biomarkers include tumor mutational burden, cytokines, circulating tumor DNA, human leukocyte antigen, gut microbiota (GM) diversity and composition, among many others[15].

The GM is a community of 100 trillion microorganisms of more than 1000 species mainly bacteria but also, archaea, viruses and fungi which colonize the human intestines[16]. The relationship which exists between GM and the host is a mutualistic relationship where one benefits the other[16]. In return for the nutrients derived from the host, the GM performs numerous critical functions such as fermentation of dietary fiber into short-chain fatty acids; synthesis of vitamins; protection against pathologic gut microbes; and induction and regulation of the immune system[17,18]. The gut microbial balance is pivotal in the optimal functioning of all of these roles and thus any discrepancy in this delicate equilibrium could produce a state of dysbiosis which has been associated with many pathologies including cancer[19]. In the context of cancer, preclinical studies have demonstrated that some GM subpopulations have pro-tumorigenic effects, whereas others have tumor-suppressive effects[20-22]. Additionally, the GM has also been shown to modulate response to chemotherapy and immunotherapy[23-25]. This could be linked to the role of GM in metabolizing anti-cancer compounds and regulating the host’s immune response[16]. Thus, GM has been studied intensely as a potential biomarker of response to ICB[12,26-31]. This is particularly relevant for melanoma, where ICB has become standard of care given its demonstrated pronounced effectiveness.

Studies investigating GM composition and/or diversity in patients with melanoma have identified distinct GM composition in responders to ICB compared to non-responders, offering hope of a novel biomarker for predicting response to ICB[12,26-32]. Additionally, studies exploring whether certain GM composition and diversity could be predictive of ICB-related colitis - one of the major factors of ICB treatment cessation and thus failure to derive full benefit of ICB - have also been conducted[27,33]. This systematic review will be the first to compile the existing data regarding the role of GM composition and diversity in predicting response to ICB and ICB-related colitis specifically in patients with melanoma. Notably previous reviews have combined multiple cancers.

MATERIALS AND METHODS
Literature search strategies

This review was conducted following the preferred reporting Items for systematic reviews and meta-analyses guidelines[34]. The review protocol was submitted to the international prospective register of systemic reviews (PROSPERO Registration number: CRD42021228018).

In this comprehensive literature search, original studies exploring the variation in GM community in fecal samples of melanoma patients who responded and did not respond to immunotherapy, experienced colitis and did not experience colitis were identified. Medline and Embase were searched for eligible papers published prior to December 2021 using the following search terms: (fecal OR gut) AND (microbiota OR microbiome) AND (melanoma) AND (immunotherapy OR checkpoint OR nivolumab OR ipilimumab OR pembrolizumab). OpenGrey and the Grey Literature Report were also searched for eligible unpublished papers and grey literature. The following keywords and its synonyms will be used for our search strategy: “fecal microbiota”, “melanoma”, “immunotherapy”.

Duplicate and irrelevant publication types such as symposium agendas were removed from the initial search results. Titles and abstracts of relevant publications were screened independently by Oliver O and Simadibrata DM based on inclusion and exclusion criteria stated below. Subsequently, reference lists within each relevant publication were examined for further pertinent studies. The full texts of these publications were then reviewed.

Inclusion criteria

Inclusion criteria for the systematic review included randomized controlled trials (RCTs), original cohort, case-control studies published in a peer-reviewed journal exploring GM diversity and composition in fecal samples from melanoma patients treated with ICB which can be anti-PD-1 and/or anti-PD-L1 and/or anti-CTLA-4. Studies included should assess treatment outcome and/or ICB-related colitis incidence following treatment with ICB. Treatment outcomes should be determined by RECIST criteria and/or progression free survival (PFS) and/or overall survival (OS) and ICB-related colitis confirmed by colonoscopy.

Only studies which utilized fecal samples obtained from human subjects receiving ICB were included. Studies which assessed treatment response to immunotherapy in animal models were excluded. Two reviewers (Oliver O, Liu YY) independently screened and read the full text of the included articles for eligibility.

Data extraction

Two investigators (Oliver O, Liu YY) independently reviewed the eligible studies and extracted data from each study. Extracted variables included title, first author, year of publication, number of participants, type of immunotherapy received, GM analysis method, and study outcomes (GM composition and diversity in responders/non-responders and ICB-related colitis/non-ICB-related colitis patients). Extracted GM composition data included a list of the GM at the level of phyla, class, order, family, genus and species, whereas extracted GM diversity extracted included alpha diversity or the Shannon index. Any discrepancies found by the investigators on data extraction were resolved by consensus. In addition, we conducted a relevant search by Reference Citation Analysis (RCA) (https://www.referencecitationanalysis.com).

Quality assessment

Non-randomized studies, including cohort studies, case-control studies and single-arm clinical trial that were included in this systematic review were independently evaluated by Oliver O and Simadibrata DM for any risk of bias using the Risk of Bias in Non-randomized Studies of interventions (ROBINS-I) assessment tool, a tool which assesses seven items: confounding, selection, intervention classification, deviation from intervention, missing data, measurement of outcome and selection of reported result. Each item was assessed according to the ROBINS-I guideline, where each bias domain can be classified as either low, moderate, serious or critical risk of bias, or no information mentioned.

RESULTS
Study selection and risk of bias assessment

The initial search from Medline, Embase, OpenGrey and Grey Literature retrieved 300 studies. After deduplication, the studies were screened by reviewing their abstracts and 10 articles selected for full assessment (Figure 1). One study by Vétizou et al[35] was excluded because while the fecal samples were obtained from patients treated with anti-CTLA-4, treatment response to ICB was assessed in an in-vivo mice model of melanoma following fecal transplantation rather than humans.

Figure 1
Figure 1 Prisma flow diagram of study selection.

From the nine included studies, two studies were phase I clinical trials, while the remainder were prospective observational studies[26,28]. Unfortunately, no RCTs were available to date. According to the ROBINS-I assessment tool, all but one study was shown to have moderate risk of bias (Table 1). The study by Matson et al[29] had a serious risk of bias as there was a lack of clarity regarding the definition of intervention used.

Table 1 Risk of bias assessment with Risk of Bias In Non-randomised Studies - of Interventions.
Ref.
Confounding
Selection
Intervention classification
Deviation from intervention
Missing data
Measurement of outcome
Selection of reported result
Overall
Dubin et al[33], 2016ModerateLowLowLowLowLowLowModerate
Chaput et al[27], 2017ModerateLowLowModerateLowLowModerateModerate
Gopalakrishnan et al[12], 2018ModerateLowNo informationLowLowLowLowModerate
Matson et al[29], 2018ModerateNo informationSeriousNo informationNo informationLowModerateSerious
Peters et al[30], 2019ModerateLowNo informationNo informationLowLowLowModerate
Baruch et al[26], 2020ModerateLowLowLowModerateLowLowModerate
Wind et al[31], 2020ModerateNo informationNo informationNo informationNo informationLowLowModerate
Davar et al[28], 2021ModerateModerateLowLowNo informationLowLowModerate
Andrews et al[36], 2021ModerateLowLowLowLowLowLowModerate
GM composition and diversity in predicting immunotherapy response

Eight studies assessed the role of GM composition and/or diversity and response to ICB in melanoma patients (Table 2). Seven studies compared the GM between responders and non-responders to ICB, and two studies analyzed the GM in patients undergoing fecal microbiota transplant (FMT).

Table 2 Characteristics of included studies exploring link between gut microbiome composition and diversity and response to immune-checkpoint blockade therapy in metastatic melanoma patients treated with immune-checkpoint blockade therapy.
Ref.
Year
Therapy
Method
Sample size/ time point
Dominant microbes
Microbial diversity
Chaput et al[27], 2017 2017Anti-CTLA-416S rRNA gene sequencing of fecal samples 26 before txResponders: Faecalibacterium and FirmicutesN/A
Matson et al[29], 2018 2018Anti-PD-1 or anti-CTLA-416S rRNA gene and shotgun metagenome sequencing of fecal samples; qPCR on selected bacteria42 before txResponders: Bifidobacterium longum, Collinsella aerofaciens, and Enterococcus faecium Non-responders: Ruminococcus obeum and Roseburia intestinalisN/A
Gopalakrishnan et al[12], 20182018Anti-PD-116S rRNA gene and shotgun metagenome sequencing of fecal samples43 before txResponders: Clostridiales, in particular Faecalibacterium Non-responders: Bacteroidales, in particular Bacteroides thetaiotaomicron; as well as Escherichia coli, and Anaerotruncus colihominisHigher alpha diversity in patients with longer PFS
Peters et al[30], 20192019Anti-PD-1 or anti-CTLA-416S rRNA gene and shotgun metagenome sequencing of fecal samples 27 before txResponders: Faecalibacterium, Parabacteroides, and Faecalibacterium prausnitzii Non-responders: Bacteroides and Biophilia Higher microbial community richness and diversity was associated with longer PFS
Wind et al[31], 2020 2020Anti-PD-1 or anti-CTLA-4Shotgun metagenome sequencing of fecal samples25 before txResponders: Ruminococcus gnavus, Streptococcus parasanguinis, and Bacteroides massiliensis. Non-responders: Bifidobacterium longum and Peptostreptococcaceae No significant difference in alpha-diversity between responder and non responders
Baruch et al[26], 20202020Anti-PD-1 refractory16S rRNA gene and shotgun metagenome sequencing of fecal samples 10 anti-PD-1 refractory patientsFMT donors (responders): Lachnospiraceae, Veillonellaceae, and Ruminococcaceae Post FMT Responders: Enterococcaceae, Enterococcus, and Streptococcus australis Non-responders: Veillonella atypicaNo significant difference in GM composition prior to FMT, but significant difference post-FMT between responders and non-responders Lower microbial richness in the donor of responding recipients
Davar et al[28].2021 2021Anti-PD-1 refractoryShotgun metagenomic sequencing of fecal samples15 anti-PD-1 refractory patients, before FMTResponders: Firmicutes (Lachnospiraceae and Ruminococcaceae families) and Actinobacteria (Bifidobacteriaceae and Coriobacteriaceae families)Higher GM diversity of donors who were complete responders compared to donors who were partial responders No significant difference in GM diversity between donors and recipients prior to FMT
Andrews et al[36], 20212021Combined ICB - Anti-PD-1 and anti-CTLA-416S rRNA gene and shotgun metagenome sequencing of fecal samples 38Responders: Bacteroides stercoris, Parabacteroides distasonis, Fournierella massiliensis. Non-responders: Klebsiella aerogenes and Lactobacillus rogosaeNo significant difference in GM diversity between responders and non-responders

The study by Chaput et al[27] assessing fecal GM composition of 26 metastatic melanoma patients prior to and post commencing anti-CTLA-4 therapy revealed that GM composition varied according to response. Patients showing long term response to therapy (nine out of 26 patients) were found with fecal samples with significantly higher Faecalibacterium percentages (P = 0.0092) while patients with poor clinical benefit had higher proportions of Bacteroides (P = 0.034). When patients were grouped based on their microbiota composition, those with high prevalence of Faecalibacterium and other Firmicutes had a longer PFS (P = 0.0039) and to a lesser extent longer OS (P = 0.051) relative to patients whose fecal samples were abundant with Bacteroides. Additionally, these patient groups were noted to derive long-term clinical benefit compared to the latter (67% vs 0%; P = 0.0017)[28].

In an analysis of stool samples from 42 metastatic melanoma patients prior to treatment with anti-PD-1 (n = 38) and anti-CTLA-4 (n = 4) therapy, Matson et al[29] showed a significant difference in GM composition between responders (16 patients) and non-responders (26 patients) (P < 0.01). In responders, eight microbial species namely, Enterococcus faecium, Collinsella aerofaciens, Bifidobacterium adolescentis, Klebsiella pneumoniae, Veillonella parvula, Parabacteroides merdae, Lactobacillus sp. and Bifidobacterium longum were found to be more abundant in responders than in non-responders[29]. In non-responders, two microbial species, specifically, Ruminococcus obeum and Roseburia intestinalis were more abundant[29]. To further assess the applicability of GM composition as a biomarker of response to ICB, they explored the correlation between the ratio of total numbers of potentially “beneficial” and “nonbeneficial” operational taxonomic units (OTUs), and change in tumor size, as assessed by the RECIST[29]. Patients with an OTU ratio of greater than 1.5 demonstrated clinical response to ICB[29].

In another study by Gopalakrishnan et al[12], fecal samples of 43 metastatic melanoma patients prior to treatment with anti-PD-1 therapy were analyzed. In responders (30 patients), analysis of fecal samples revealed abundance of GM from Ruminococcaceae family of the Clostridiales order, whereas in non-responders (13 patients), abundance of GM from the Bacteroidales order was noted[12]. Further analyses demonstrated that Faecalibacterium genus was notably enriched in fecal samples from responders and Bacteroides thetaiotaomicron, Escherichia coli, and Anaerotruncus colihominis were enriched in non-responders[12]. In addition, to investigate durability of response, patients were stratified based on their fecal composition of Faecalibacterium genus and Bacteroidales order and correlated to their PFS[12]. Results demonstrated that patients with Faecalibacterium-enriched fecal samples have longer PFS than those with low abundance (P = 0.03) and patients with Bacteroidales-enriched fecal samples have shorter PFS than those with low abundance (P = 0.05). Beyond specific microbial taxa, GM diversity, as assessed by Simpson's reciprocal index, was higher in responders compared to non-responders (P < 0.01)[12]. Moreover, high GM diversity was significantly associated with anti-PD-1 therapy response, when compared to patient groups of intermediate diversity [hazard ratio (HR) = 3.60, 95% confidence interval (CI): 1.02-12.74, P < 0.05) and low diversity (HR = 3.57, 95%CI: 1.02-12.52, P < 0.05). Other important predictors of therapy response include abundance of Faecalibacterium (HR = 2.92, 95%CI: 1.08-7.89) and Bacteroidales (HR = 0.39, 95%CI: 0.15-1.03) in the fecal microbiome[12].

Peters et al[30] examined the correlation between GM taxa and PFS in pre-treatment fecal samples of 27 metastatic melanoma patients receiving anti-PD-L1 and/or anti-CTLA-4. GM which was associated with shorter PFS included genera Bacteroides and Bilophila, and species Bacteroides ovatus, Blautia producta, and Ruminococcus gnavus, whereas those which correlated with longer PFS included genera Faecalibacterium and Parabacteroides and species Faecalibacterium prausnitzii[12]. With regards to GM richness the authors compared the β-diversity or between-sample microbiome diversity relative to survival. Multivariate analysis adjusting for age, sex, BMI, stage, number of sites of metastases, and antibiotic use in the last 6 mo revealed that higher GM richness was correlated with longer PFS (number of 16S sub - OTUs: HR [95%CI] = 0.97 [0.95, 1.00], P = 0.02; number of shotgun subspecies: HR [95%CI] = 0.89 [0.79, 0.99], P = 0.03)[30]. Furthermore, analysis of the 16S but not shotgun dataset showed that higher diversity of GM, as assessed by the Shannon index, was associated with longer PFS (P = 0.02)[12].

Similarly, Wind et al[31] analyzed fecal samples from 25 metastatic melanoma patients - 12 responders, 13 non-responders - prior to start of treatment with anti-PD-1 or anti-CTLA-4. Analysis revealed that the fecal samples of responders were mainly enriched in Ruminococcus gnavus, Escherichia coli, Eubacterium biforme, Phascolarctobacterium succinatutens and Streptococcus salivarius, whereas samples from non-responders were abundant in Bifidobacterium longum, Prevotella copri, Coprococus sp, Eggerthella unclassified and Eubacterium ramulus[31]. When correlated with survival, fecal samples of participants enriched in Bacteroides massiliensis and Streptococcus parasanguinis were associated with longer PFS (HR: 3.79, 95%CI: 1.06-13.52 P = 0.04) and OS (HR: 5.05, 95%CI: 1.33-19.21, P = 0.017) respectively, whereas those who were carriers of Peptostreptococcaceae were associated with shorter PFS (HR: 0.18, 95%CI: 0.05-0.62, P = 0.007) and OS (HR: 0.12, 95%CI: 0.01-0.96, P = 0.046)[31]. In terms of GM diversity, as assessed by Shannon index, no significant difference between responders and non-responders was noted[31].

The study by Andrews et al[36] analyzed gut microbiome samples from a subset of 77 metastatic melanoma patients - 27 responders, 11 non-responders - who underwent combined ICB. There was no significant association in Firmicutes phyla and Clostridiales order and response to ICB (P = 0.39 and P = 0.38, respectively) and no significant difference in alpha diversity between responders and non-responders to ICB[36]. Fecal samples from responders were mainly enriched with Bacteroides stercoris, Parabacteroides distasonis and Fournierella massiliensis (P = 0.03, P = 0.04 and P = 0.008, respectively) while fecal samples from non-responders were abundant in Klebsiella aerogenes and Lactobacillus rogosae (P = 0.04 and P = 0.02, respectively)[36].

In a first clinical trial of its kind (phase 1), Baruch et al[26] demonstrated that FMT from anti-PD-1 treated metastatic melanoma patients who were complete responders (2 donors), triggered response to anti-PD-1 therapy in metastatic melanoma patients who were refractory to at least one line of anti-PD-1 therapy. Out of 10 patients included in the trial, 3 patients demonstrated objective responses with 1 achieving complete response and 2 patients achieving partial response[26]. Notably, the PFS milestone of 6 mo was reached in all responders[26]. Upon analysis of pre-treatment fecal samples of donors, donor of the responding recipients had a lower microbial richness than the other donor of the non-responding patients[26]. There was no significant difference on the GM composition prior to FMT of recipients who responded compared to those who did not respond[26]. Metagenome sequencing found that recipients post FMT have higher proportions of Veillonellaceae family and a lower relative abundance of Bifidobacterium bifidum. Donors were found with high amounts of Lachnospiraceae, Veillonellaceae, and Ruminococcaceae. Comparison of a small subset of non-responders with responders, found statistically significant higher abundance of Enterococcaceae, Enterococcus, and Streptococcus australis, and a lower relative abundance of Veillonella atypica. However clear deductions on specific GM taxa cannot be made, as there were non-responders and pre-treatment fecal samples with similar dynamics. it is crucial to note that this trial was primarily designed to assess safety of FMT and not statistically powered to assess efficacy[26].

In a separate trial, Davar et al[28] showed that fecal microbial transplant (FMT) from metastatic melanoma patients (7 donors) who had complete (4 donors) or partial response (3 donors) to anti-PD-1 therapy helped overcome resistance in anti-PD-1 treatment-refractory metastatic melanoma patients (15 patients). Following FMT and anti-PD-1 therapy, 6 out of 15 patients achieved clinical benefit, with 3 patients achieving objective responses and 3 patients experiencing stable disease lasting more than 12 mo[28]. Analysis of stools after FMT revealed that samples from responders were abundant in the phyla, Firmicutes (Lachnospiraceae and Ruminococcaceae) and Actinobacteria (Bifidobacteriaceae and Coriobacteriaceae) and had decreased proportions in phylum Bacteroidetes[28]. In terms of GM diversity assessed with inverse Simpson index, GM diversity of donors who were complete responders were more diverse than donors who were partial responders. There was no significant difference in GM diversity between donors and recipients prior to FMT[28].

Gut microbiota composition and diversity in predicting ICB-related colitis

To date only three studies have reported on the correlation between pre-treatment GM composition and/or diversity and ICB-related colitis (Table 3).

Table 3 Characteristics studies exploring link between gut microbiome composition and diversity and immune-checkpoint blockade therapy-related colitis in metastatic melanoma patients treated with immune-checkpoint blockade therapy.
Ref.
Year
Therapy
Method
Sample size/ timepoint
Dominant microbes
Microbial diversity
Dubin et al[33], 2015 2015Anti-CTLA-4 immunotherapy 16S rRNA gene and shotgun metagenome sequencing of fecal samples34Colitis-resistant: Bacteroidetes (Bacteroidaceae, Rikenellaceae and Barnesiellaceae)No significant difference in microbial richness and diversity
Chaput et al[27], 2017 2017 Anti-CTLA-4 immunotherapy 16S rRNA gene sequencing of fecal samples26Colitis-resistant: Bacteroidetes; Colitis-prone: Firmicutes Decreased bacterial diversity was associated with colitis
Andrews et al[36], 2021Combined ICB - Anti-PD-1 and anti-CTLA-416S rRNA gene and shotgun metagenome sequencing of fecal samples38Colitis resistant: Firmicutes; Colitis prone: BacteriodetesNo significant difference in alpha diversity

Firstly, a prospective study by Dubin et al[33] explored the link between GM composition, and subsequent colitis development in 34 metastatic melanoma patients treated with ipilimumab, showed that the Bacteroidetes phylum was more abundant (P = < 0.05) in fecal samples of the 24 patients who did not develop ipilimumab-induced colitis compared to those who did. Further analysis revealed that within the Bacteroidetes phylum, the population of Bacteroidaceae, Rikenellaceae and Barnesiellaceae was significantly more abundant in the former than the latter (P < 0.01, P < 0.05 and P < 0.05 respectively)[33]. However, there was no significant difference in microbial richness and diversity, as assessed by Shannon and inverse Simpson indices, between those who developed ipilimumab-induced colitis relative to those who did not[33].

In a similar study by Chaput et al[27], analysis of fecal samples of metastatic melanoma patients receiving ipilimumab demonstrated high proportions of Firmicutes in patients who developed ipilimumab-induced colitis (P = 0.009). In contrast, fecal samples of those that did not develop colitis were mainly enriched with Bacteroidetes (P = 0.011)[27]. Accordingly, patients with the former GM composition also tend to have a shorter colitis-free cumulative incidence compared with patients with the latter composition[27]. Several OTUs known to be predictive to colitis such as F. prausnitzii L2-6, butyrate producing bacterium L2-21 and G. formicilis ATCC 27749 were associated with longer OS[27].

Finally, Andrews et al[36], analyzed gut microbiome samples in metastatic melanoma patients undergoing combined ICB and their link to ileitis and colitis events. No significant difference in alpha diversity was observed between those that did and did not develop colitis[36]. Fecal samples of patients developing colitis were enriched in Bacteroides intestinalis and Intestinibacter bartlettii (P = 0.009 and P = 0.009, respectively) while those that did not were abundant in Anaerotignum lactatifermentans and Dorea formicigenerans (P = 0.016 and P = 0.06, respectively)[36]. For both B. intestinalis and D. formicigenerans, associations with their risk of colitis were still maintained after adjustment using a logistic regression model [OR = 4.54 (95%CI = 1.06-24.7) and OR = 0.35 (95%CI = 0.082-1.35), respectively][36].

DISCUSSION

Our review of current reports assessing the GM composition relative to response to ICB, indicated high variability in the results and limited concordance on the organisms identified (Figure 2). Amongst the few commonalities, we found that fecal samples enriched in organisms from the Firmicutes phylum (Lachnospiraceae and Ruminococcaceae family) especially the Faecalibacterium genus were associated with ICB responders in 4 of 9 studies[12,27,28,31], while Bacteroidetes phylum was found in higher proportions in non-responders in 2 of the studies[12,30]. However, other than these two findings, there was no clear correlation between specific GM composition and response to ICB.

Figure 2
Figure 2 Phylogenetic tree showing family and species of gut microbiome abundant in responders and non-responders to immune-checkpoint blockade therapy in all included studies. Gut microbiome species highlighted in red: Abundant in non-responders to immune-checkpoint blockade therapy; blue: Abundant in responders to immune-checkpoint blockade therapy; purple: Abundant in both responders and non-responders.

In fact, our analysis mainly identified inconsistencies in the GM composition reported to be associated with response to ICB. For instance, Bifidobacterium longum was found to be abundant in responders in the study by Wind et al[31], but found to be enriched in non-responders in the study by Matson et al[29]. Some species from the Firmicutes family were found in both responders and non-responders such as Roseburia intestinalis[29]. Similarly, species from the Bacteroidales order were found in both responders and non-responders, such as Bacteroides massiliensis[31]. The overlap in GM composition in responders and non-responders may suggest that the functional capacity of the GM may be more important than individual GM family/order/species in determining response to ICB[30].

In contrast to individual species or taxas, GM diversity have been heralded to a marker of good health[37]. Here four studies - Gopalakrishnan et al[12], Peters et al[30], Wind et al[31] and, Andrews et al[36] - assessed its potential to predict ICB responsiveness. The two first studies demonstrated that higher GM diversity in the responder group compared to non-responder arm[12,30]. However, the other two studies, Wind et al[31] and Andrews et al[36], found no differences in GM diversity between both groups. Nevertheless, in other cancer types such as renal cell carcinoma and non-small cell lung cancer, greater GM diversity has also been associated with improved responses to anti-PD-1 therapy[37-39].

Study results showing associations with GM diversity were consistent with previous studies which showed that greater GM diversity is prevalent in healthy state across multiple diseases, plausibly suggesting that a greater GM diversity produces the optimal immune environment needed for normal physiological functioning[40-42]. One major reason is the promotion of a favorable immune phenotype, as evidenced by the positive correlation between Shannon diversity index and several CD8+ T cell and NK cell signatures, required to produce a robust anti-tumoral response[38].

Previous studies have demonstrated that GM from Firmicutes family and Bacteroidales order play a significant role in mediating the response to immunotherapy in melanoma patients[12,27,29]. For instance, abundance of Firmicutes was associated with increased frequencies of CD4+, CD8+ T cells, CD 45+ myeloid and lymphoid tumor-infiltrating cells and preserved cytokine response to anti-PD-1 therapy[12]. Additionally, abundance of Firmicutes was linked with decreased frequency of intestinal and systemic regulatory T cells (Tregs) and B7+ T cells, cells responsible for limiting immune response robustness[27]. This resulted in increased antigen presentation and effector T cell function in both the periphery and tumor microenvironment[12,27,29]. However, other GM such as Bacteroidales were unfavorable in terms of anti-tumoral response in that its abundance was associated with higher frequencies of Tregs and myeloid-derived suppressor cells and a blunted cytokine response[12]. These findings combined demonstrated that certain GM play a crucial role in mediating systemic and antitumor immune responses which have clear implications on efficacy on ICB therapy in metastatic melanoma patients.

Notably, GM has also been shown to potentially serve as not just a predictor of ICB therapy response, but also for boosting response to ICB therapy. FMT on anti-PD-1 treatment-refractory metastatic melanoma patients produced a complete response to anti-PD-1 therapy in one-third (9 out of 25 patients) of the otherwise therapy refractory patients[26,28].

Another aspect of the GM analyzed here, was its association with ICB-related colitis. The three studies included in this review demonstrated that GM which was abundant in ICB-related colitis-prone patients was enriched in responders to ICB (Firmicutes) while GM which was abundant in ICB-related colitis-resistant patients was enriched in non-responders to ICB (Bacteroidetes). This is consistent with the understanding that a more effective anti-tumoral response will produce greater off-target effects. The Bacteroidetes phyla has been linked with low-grade systemic inflammation, which could explain the observation that Bacteroidetes phyla was abundant in ICB-related colitis-resistant patients[27]. In line with this observation is the finding that level of Bacteroidetes is lower in inflammatory bowel disease - an autoimmune condition which produces chronic inflammation of the digestive tract - patients relative to healthy patients[43]. Conversely, Firmicutes phyla, especially F. prausnitzii has been associated with induction of Tregs which express high levels of CTLA-4, fueling speculation that it may cause sequestration of Tregs within the intestine[44]. Since Tregs express high levels of CTLA-4, their actions are inhibited, thereby limiting self-tolerance and promoting the development of colitis. These findings reiterate that GM has an immunomodulatory role, giving them the potential to be utilized as biomarkers of ICB-related colitis, in addition to response to ICB.

Our systematic review has several strengths. Firstly, unlike previous reviews which combined studies in various cancer types, this review focused solely on the effect of GM composition and diversity only in patients with melanoma. Secondly, we conducted a comprehensive search for RCTs and observational studies, performed a risk-of-bas assessment and studied clinically important outcomes - clinical response and ICB-related colitis - an adverse event reported in up to 25% of patients treated with ICB[45]. Thirdly, we only included studies which assessed response to immunotherapy in humans, not animals. However, several limitations exist in our systematic review. Studies which we included used distinct approaches when segregating patients into the responder and non-responder groups, using different response criteria to evaluate treatment response in patients. Additionally, there were differences in methods of stool collection and analysis of GM composition and diversity. For example, Chaput et al[46] collected multiple stool samples every 3 wk of ICB, while other studies such as Dubin et al[33] and Matson et al[29] collected stool samples only prior to initiation of ICB. Furthermore, only 4 studies considered confounding factors such as variation in diet and antibiotic use[27,29-31]. Therefore, inter-study comparison of the GM composition and diversity in responders vs non-responders and those who experienced colitis vs non-colitis should be addressed with caution. Furthermore, included studies only enrolled a small number of patients, which could explain inconsistent results between studies.

CONCLUSION

In conclusion, GM composition and diversity holds some potential as a biomarker of response and toxicity to ICB in melanoma. Larger prospective studies with standardized experimental protocol ought to be conducted to elucidate whether distinct GM signatures are required for robust response to different ICB regimens. Additionally, more studies correlating metagenomic and metatranscriptomic data of GM to outcomes of melanoma patients on immunotherapy ought to be performed as the functional capacity may be more important rather than individual GM family/order/species. In addition, we eagerly await the outcome of multiple large-scale RCTs involving FMT in the context of ICB-refractory melanoma such as NCT04577729 and NCT04988841 (PICASSO) (ClinicalTrails.gov).We foresee that together with other promising biomarkers, GM composition and diversity will be integrated into a multiparameter model to accurately predict which subset of melanoma patients are likely to respond to ICB[10,11,47].

ARTICLE HIGHLIGHTS
Research background

Survival for metastatic melanoma has significantly improved since the introduction of immune checkpoint blockade (ICB) therapy. However, despite their considerable efficacy, 40%-60% of melanoma patients do not experience objective responses to the therapy. Additionally, some patients experience ICB-related colitis as a consequence of ICB therapy, preventing them from deriving the full benefit of ICB therapy. Recent studies have demonstrated that the gut microbiome (GM) may affect tumor immunity by regulating the host immune system and tumor micro-environment, thus suggesting that GM may affect response to ICB therapy and susceptibility of ICB-related colitis.

Research motivation

The GM has shown great potential as a biomarker of response to ICB therapy in melanoma patients. Previous studies investigating GM composition and/or diversity in patients with melanoma have identified distinct GM composition and diversity in responders to ICB compared to non-responders, as well as those more susceptible to ICB-related colitis than those who are not.

Research objectives

To be the first to compile the existing data regarding the role of GM composition and diversity in predicting response to ICB and ICB-related colitis specifically in patients with melanoma.

Research methods

Comprehensive literature search was done in various platforms using the following search terms: (fecal OR gut) AND (microbiota OR microbiome) AND (melanoma) AND (immunotherapy OR checkpoint OR nivolumab OR ipilimumab OR pembrolizumab). From a total of 300 studies, nine studies met inclusion criteria. Two studies were phase I clinical trials, while the remainder were prospective observational studies. All but one study has moderate risk of bias. Data from these studies including but not limited to, number of participants, type of immunotherapy received, GM analysis method, and GM composition and diversity were collected and interpreted.

Research results

Fecal samples enriched in Firmicutes phylum were associated with good response to ICB therapy, however they were associated with increased susceptibility to ICB-related colitis. Fecal samples enriched in Bacteroidales family were associated with poor response to ICB. Samples with greater GM diversity were associated with more favorable response to ICB. Fecal samples enriched in Bacteroidetes were associated with decreased incidence of ICB-related colitis. Overall, there was limited concordance in the organisms in the GM identified to be associated with response to ICB, and studies evaluating GM diversity showed conflicting results.

Research conclusions

GM composition and diversity holds some potential as a biomarker of response and toxicity to ICB in melanoma. Further prospective studies, including several RCTs that are underway, are needed to confirm whether the GM could be used as a biomarker and potential intervention to modulate ICB response in melanoma patients.

Research perspectives

With other promising biomarkers, GM composition and diversity holds potential to be integrated into a multiparameter model to accurately predict which subset of melanoma patients are likely to respond to ICB.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Corresponding Author's Membership in Professional Societies: American Society of Clinical Oncology; American Association for Cancer Research.

Specialty type: Oncology

Country/Territory of origin: Australia

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: MI SC, China; Wen XL, China S-Editor: Wang LL L-Editor: A P-Editor: Wang LL

References
1.  Gershenwald JE, Guy GP Jr. Stemming the Rising Incidence of Melanoma: Calling Prevention to Action. J Natl Cancer Inst. 2016;108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 50]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
2.  Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin. 2021;71:209-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50630]  [Cited by in F6Publishing: 42895]  [Article Influence: 14298.3]  [Reference Citation Analysis (47)]
3.  Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, Wolchok JD, Hersey P, Joseph R, Weber JS, Dronca R, Mitchell TC, Patnaik A, Zarour HM, Joshua AM, Zhao Q, Jensen E, Ahsan S, Ibrahim N, Ribas A. Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann Oncol. 2019;30:582-588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 579]  [Cited by in F6Publishing: 547]  [Article Influence: 109.4]  [Reference Citation Analysis (0)]
4.  Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Rutkowski P, Lao CD, Cowey CL, Schadendorf D, Wagstaff J, Dummer R, Ferrucci PF, Smylie M, Hogg D, Hill A, Márquez-Rodas I, Haanen J, Guidoboni M, Maio M, Schöffski P, Carlino MS, Lebbé C, McArthur G, Ascierto PA, Daniels GA, Long GV, Bastholt L, Rizzo JI, Balogh A, Moshyk A, Hodi FS, Wolchok JD. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med. 2019;381:1535-1546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1826]  [Cited by in F6Publishing: 2116]  [Article Influence: 423.2]  [Reference Citation Analysis (0)]
5.  Robert C, Grob JJ, Stroyakovskiy D, Karaszewska B, Hauschild A, Levchenko E, Chiarion Sileni V, Schachter J, Garbe C, Bondarenko I, Gogas H, Mandalá M, Haanen JBAG, Lebbé C, Mackiewicz A, Rutkowski P, Nathan PD, Ribas A, Davies MA, Flaherty KT, Burgess P, Tan M, Gasal E, Voi M, Schadendorf D, Long GV. Five-Year Outcomes with Dabrafenib plus Trametinib in Metastatic Melanoma. N Engl J Med. 2019;381:626-636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 676]  [Cited by in F6Publishing: 760]  [Article Influence: 152.0]  [Reference Citation Analysis (0)]
6.  Domingues B, Lopes JM, Soares P, Pópulo H. Melanoma treatment in review. Immunotargets Ther. 2018;7:35-49.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 309]  [Cited by in F6Publishing: 390]  [Article Influence: 65.0]  [Reference Citation Analysis (0)]
7.  Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, Akerley W, van den Eertwegh AJ, Lutzky J, Lorigan P, Vaubel JM, Linette GP, Hogg D, Ottensmeier CH, Lebbé C, Peschel C, Quirt I, Clark JI, Wolchok JD, Weber JS, Tian J, Yellin MJ, Nichol GM, Hoos A, Urba WJ. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363:711-723.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10799]  [Cited by in F6Publishing: 11097]  [Article Influence: 792.6]  [Reference Citation Analysis (0)]
8.  Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, Ferrucci PF, Hill A, Wagstaff J, Carlino MS, Haanen JB, Maio M, Marquez-Rodas I, McArthur GA, Ascierto PA, Long GV, Callahan MK, Postow MA, Grossmann K, Sznol M, Dreno B, Bastholt L, Yang A, Rollin LM, Horak C, Hodi FS, Wolchok JD. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med. 2015;373:23-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5972]  [Cited by in F6Publishing: 5702]  [Article Influence: 633.6]  [Reference Citation Analysis (0)]
9.  Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, Daud A, Carlino MS, McNeil C, Lotem M, Larkin J, Lorigan P, Neyns B, Blank CU, Hamid O, Mateus C, Shapira-Frommer R, Kosh M, Zhou H, Ibrahim N, Ebbinghaus S, Ribas A; KEYNOTE-006 investigators. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med. 2015;372:2521-2532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4026]  [Cited by in F6Publishing: 4121]  [Article Influence: 457.9]  [Reference Citation Analysis (1)]
10.  Chan TA, Yarchoan M, Jaffee E, Swanton C, Quezada SA, Stenzinger A, Peters S. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann Oncol. 2019;30:44-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1568]  [Cited by in F6Publishing: 1517]  [Article Influence: 303.4]  [Reference Citation Analysis (0)]
11.  Chowell D, Morris LGT, Grigg CM, Weber JK, Samstein RM, Makarov V, Kuo F, Kendall SM, Requena D, Riaz N, Greenbaum B, Carroll J, Garon E, Hyman DM, Zehir A, Solit D, Berger M, Zhou R, Rizvi NA, Chan TA. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science. 2018;359:582-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 768]  [Cited by in F6Publishing: 688]  [Article Influence: 114.7]  [Reference Citation Analysis (0)]
12.  Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, Prieto PA, Vicente D, Hoffman K, Wei SC, Cogdill AP, Zhao L, Hudgens CW, Hutchinson DS, Manzo T, Petaccia de Macedo M, Cotechini T, Kumar T, Chen WS, Reddy SM, Szczepaniak Sloane R, Galloway-Pena J, Jiang H, Chen PL, Shpall EJ, Rezvani K, Alousi AM, Chemaly RF, Shelburne S, Vence LM, Okhuysen PC, Jensen VB, Swennes AG, McAllister F, Marcelo Riquelme Sanchez E, Zhang Y, Le Chatelier E, Zitvogel L, Pons N, Austin-Breneman JL, Haydu LE, Burton EM, Gardner JM, Sirmans E, Hu J, Lazar AJ, Tsujikawa T, Diab A, Tawbi H, Glitza IC, Hwu WJ, Patel SP, Woodman SE, Amaria RN, Davies MA, Gershenwald JE, Hwu P, Lee JE, Zhang J, Coussens LM, Cooper ZA, Futreal PA, Daniel CR, Ajami NJ, Petrosino JF, Tetzlaff MT, Sharma P, Allison JP, Jenq RR, Wargo JA. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359:97-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2846]  [Cited by in F6Publishing: 2679]  [Article Influence: 446.5]  [Reference Citation Analysis (0)]
13.  Pavlick AC, Fecher L, Ascierto PA, Sullivan RJ. Frontline Therapy for BRAF-Mutated Metastatic Melanoma: How Do You Choose, and Is There One Correct Answer? Am Soc Clin Oncol Educ Book. 2019;39:564-571.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 31]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
14.  Marsavela G, Lee J, Calapre L, Wong SQ, Pereira MR, McEvoy AC, Reid AL, Robinson C, Warburton L, Abed A, Khattak MA, Meniawy TM, Dawson SJ, Sandhu S, Carlino MS, Menzies AM, Scolyer RA, Long GV, Amanuel B, Millward M, Ziman MR, Rizos H, Gray ES. Circulating Tumor DNA Predicts Outcome from First-, but not Second-line Treatment and Identifies Melanoma Patients Who May Benefit from Combination Immunotherapy. Clin Cancer Res. 2020;26:5926-5933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 38]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
15.  Morad G, Helmink BA, Sharma P, Wargo JA. Hallmarks of response, resistance, and toxicity to immune checkpoint blockade. Cell. 2021;184:5309-5337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 512]  [Article Influence: 170.7]  [Reference Citation Analysis (0)]
16.  Lepage P, Leclerc MC, Joossens M, Mondot S, Blottière HM, Raes J, Ehrlich D, Doré J. A metagenomic insight into our gut's microbiome. Gut. 2013;62:146-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 239]  [Cited by in F6Publishing: 229]  [Article Influence: 20.8]  [Reference Citation Analysis (0)]
17.  Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell. 2014;157:121-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2488]  [Cited by in F6Publishing: 2823]  [Article Influence: 282.3]  [Reference Citation Analysis (0)]
18.  Morowitz MJ, Carlisle EM, Alverdy JC. Contributions of intestinal bacteria to nutrition and metabolism in the critically ill. Surg Clin North Am. 2011;91:771-785, viii.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 107]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
19.  Sheflin AM, Whitney AK, Weir TL. Cancer-promoting effects of microbial dysbiosis. Curr Oncol Rep. 2014;16:406.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 158]  [Article Influence: 17.6]  [Reference Citation Analysis (0)]
20.  Jan G, Belzacq AS, Haouzi D, Rouault A, Métivier D, Kroemer G, Brenner C. Propionibacteria induce apoptosis of colorectal carcinoma cells via short-chain fatty acids acting on mitochondria. Cell Death Differ. 2002;9:179-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 245]  [Cited by in F6Publishing: 232]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
21.  Lara-Tejero M, Galán JE. A bacterial toxin that controls cell cycle progression as a deoxyribonuclease I-like protein. Science. 2000;290:354-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 397]  [Cited by in F6Publishing: 377]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
22.  Paulos CM, Wrzesinski C, Kaiser A, Hinrichs CS, Chieppa M, Cassard L, Palmer DC, Boni A, Muranski P, Yu Z, Gattinoni L, Antony PA, Rosenberg SA, Restifo NP. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J Clin Invest. 2007;117:2197-2204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 365]  [Cited by in F6Publishing: 400]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
23.  Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ, Schlitzer A, Ginhoux F, Apetoh L, Chachaty E, Woerther PL, Eberl G, Bérard M, Ecobichon C, Clermont D, Bizet C, Gaboriau-Routhiau V, Cerf-Bensussan N, Opolon P, Yessaad N, Vivier E, Ryffel B, Elson CO, Doré J, Kroemer G, Lepage P, Boneca IG, Ghiringhelli F, Zitvogel L. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342:971-976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1147]  [Cited by in F6Publishing: 1357]  [Article Influence: 123.4]  [Reference Citation Analysis (0)]
24.  Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth N, Chen Y, Chen H, Hong J, Zou W, Fang JY. Fusobacterium nucleatum Promotes Chemoresistance to Colorectal Cancer by Modulating Autophagy. Cell. 2017;170:548-563.e16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 814]  [Cited by in F6Publishing: 1154]  [Article Influence: 164.9]  [Reference Citation Analysis (0)]
25.  Zheng Y, Wang T, Tu X, Huang Y, Zhang H, Tan D, Jiang W, Cai S, Zhao P, Song R, Li P, Qin N, Fang W. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J Immunother Cancer. 2019;7:193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 280]  [Article Influence: 56.0]  [Reference Citation Analysis (0)]
26.  Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N, Rotin D, Anafi L, Avivi C, Melnichenko J, Steinberg-Silman Y, Mamtani R, Harati H, Asher N, Shapira-Frommer R, Brosh-Nissimov T, Eshet Y, Ben-Simon S, Ziv O, Khan MAW, Amit M, Ajami NJ, Barshack I, Schachter J, Wargo JA, Koren O, Markel G, Boursi B. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371:602-609.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 357]  [Cited by in F6Publishing: 680]  [Article Influence: 170.0]  [Reference Citation Analysis (0)]
27.  Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, Boselli L, Routier E, Cassard L, Collins M, Vaysse T, Marthey L, Eggermont A, Asvatourian V, Lanoy E, Mateus C, Robert C, Carbonnel F. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol. 2017;28:1368-1379.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 585]  [Cited by in F6Publishing: 775]  [Article Influence: 129.2]  [Reference Citation Analysis (0)]
28.  Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin JM, Morrison RM, Deblasio RN, Menna C, Ding Q, Pagliano O, Zidi B, Zhang S, Badger JH, Vetizou M, Cole AM, Fernandes MR, Prescott S, Costa RGF, Balaji AK, Morgun A, Vujkovic-Cvijin I, Wang H, Borhani AA, Schwartz MB, Dubner HM, Ernst SJ, Rose A, Najjar YG, Belkaid Y, Kirkwood JM, Trinchieri G, Zarour HM. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science. 2021;371:595-602.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 327]  [Cited by in F6Publishing: 663]  [Article Influence: 221.0]  [Reference Citation Analysis (0)]
29.  Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre ML, Luke JJ, Gajewski TF. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359:104-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1390]  [Cited by in F6Publishing: 1748]  [Article Influence: 291.3]  [Reference Citation Analysis (0)]
30.  Peters BA, Wilson M, Moran U, Pavlick A, Izsak A, Wechter T, Weber JS, Osman I, Ahn J. Relating the gut metagenome and metatranscriptome to immunotherapy responses in melanoma patients. Genome Med. 2019;11:61.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 120]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
31.  Wind TT, Gacesa R, Vich Vila A, de Haan JJ, Jalving M, Weersma RK, Hospers GAP. Gut microbial species and metabolic pathways associated with response to treatment with immune checkpoint inhibitors in metastatic melanoma. Melanoma Res. 2020;30:235-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 35]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
32.  Carlino MS, Larkin J, Long GV. Immune checkpoint inhibitors in melanoma. Lancet. 2021;398:1002-1014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 414]  [Article Influence: 138.0]  [Reference Citation Analysis (0)]
33.  Dubin K, Callahan MK, Ren B, Khanin R, Viale A, Ling L, No D, Gobourne A, Littmann E, Huttenhower C, Pamer EG, Wolchok JD. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun. 2016;7:10391.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 560]  [Cited by in F6Publishing: 675]  [Article Influence: 84.4]  [Reference Citation Analysis (0)]
34.  Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JP, Clarke M, Devereaux PJ, Kleijnen J, Moher D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. J Clin Epidemiol. 2009;62:e1-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6253]  [Cited by in F6Publishing: 7022]  [Article Influence: 468.1]  [Reference Citation Analysis (0)]
35.  Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CP, Poirier-Colame V, Roux A, Becharef S, Formenti S, Golden E, Cording S, Eberl G, Schlitzer A, Ginhoux F, Mani S, Yamazaki T, Jacquelot N, Enot DP, Bérard M, Nigou J, Opolon P, Eggermont A, Woerther PL, Chachaty E, Chaput N, Robert C, Mateus C, Kroemer G, Raoult D, Boneca IG, Carbonnel F, Chamaillard M, Zitvogel L. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350:1079-1084.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1834]  [Cited by in F6Publishing: 2167]  [Article Influence: 240.8]  [Reference Citation Analysis (0)]
36.  Andrews MC, Duong CPM, Gopalakrishnan V, Iebba V, Chen WS, Derosa L, Khan MAW, Cogdill AP, White MG, Wong MC, Ferrere G, Fluckiger A, Roberti MP, Opolon P, Alou MT, Yonekura S, Roh W, Spencer CN, Curbelo IF, Vence L, Reuben A, Johnson S, Arora R, Morad G, Lastrapes M, Baruch EN, Little L, Gumbs C, Cooper ZA, Prieto PA, Wani K, Lazar AJ, Tetzlaff MT, Hudgens CW, Callahan MK, Adamow M, Postow MA, Ariyan CE, Gaudreau PO, Nezi L, Raoult D, Mihalcioiu C, Elkrief A, Pezo RC, Haydu LE, Simon JM, Tawbi HA, McQuade J, Hwu P, Hwu WJ, Amaria RN, Burton EM, Woodman SE, Watowich S, Diab A, Patel SP, Glitza IC, Wong MK, Zhao L, Zhang J, Ajami NJ, Petrosino J, Jenq RR, Davies MA, Gershenwald JE, Futreal PA, Sharma P, Allison JP, Routy B, Zitvogel L, Wargo JA. Gut microbiota signatures are associated with toxicity to combined CTLA-4 and PD-1 blockade. Nat Med. 2021;27:1432-1441.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 195]  [Article Influence: 65.0]  [Reference Citation Analysis (0)]
37.  Valdes AM, Walter J, Segal E, Spector TD. Role of the gut microbiota in nutrition and health. BMJ. 2018;361:k2179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 839]  [Cited by in F6Publishing: 949]  [Article Influence: 158.2]  [Reference Citation Analysis (0)]
38.  Jin Y, Dong H, Xia L, Yang Y, Zhu Y, Shen Y, Zheng H, Yao C, Wang Y, Lu S. The Diversity of Gut Microbiome is Associated With Favorable Responses to Anti-Programmed Death 1 Immunotherapy in Chinese Patients With NSCLC. J Thorac Oncol. 2019;14:1378-1389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 275]  [Article Influence: 55.0]  [Reference Citation Analysis (0)]
39.  Salgia NJ, Bergerot PG, Maia MC, Dizman N, Hsu J, Gillece JD, Folkerts M, Reining L, Trent J, Highlander SK, Pal SK. Stool Microbiome Profiling of Patients with Metastatic Renal Cell Carcinoma Receiving Anti-PD-1 Immune Checkpoint Inhibitors. Eur Urol. 2020;78:498-502.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 102]  [Article Influence: 25.5]  [Reference Citation Analysis (0)]
40.  Mosca A, Leclerc M, Hugot JP. Gut Microbiota Diversity and Human Diseases: Should We Reintroduce Key Predators in Our Ecosystem? Front Microbiol. 2016;7:455.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 289]  [Cited by in F6Publishing: 333]  [Article Influence: 41.6]  [Reference Citation Analysis (0)]
41.  Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML, Jones WJ, Roe BA, Affourtit JP, Egholm M, Henrissat B, Heath AC, Knight R, Gordon JI. A core gut microbiome in obese and lean twins. Nature. 2009;457:480-484.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5397]  [Cited by in F6Publishing: 5338]  [Article Influence: 333.6]  [Reference Citation Analysis (0)]
42.  Vincent C, Stephens DA, Loo VG, Edens TJ, Behr MA, Dewar K, Manges AR. Reductions in intestinal Clostridiales precede the development of nosocomial Clostridium difficile infection. Microbiome. 2013;1:18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 102]  [Cited by in F6Publishing: 91]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
43.  Verma R, Verma AK, Ahuja V, Paul J. Real-time analysis of mucosal flora in patients with inflammatory bowel disease in India. J Clin Microbiol. 2010;48:4279-4282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 82]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
44.  Romano E, Kusio-Kobialka M, Foukas PG, Baumgaertner P, Meyer C, Ballabeni P, Michielin O, Weide B, Romero P, Speiser DE. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc Natl Acad Sci U S A. 2015;112:6140-6145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 392]  [Cited by in F6Publishing: 443]  [Article Influence: 49.2]  [Reference Citation Analysis (0)]
45.  Som A, Mandaliya R, Alsaadi D, Farshidpour M, Charabaty A, Malhotra N, Mattar MC. Immune checkpoint inhibitor-induced colitis: A comprehensive review. World J Clin Cases. 2019;7:405-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 145]  [Cited by in F6Publishing: 150]  [Article Influence: 30.0]  [Reference Citation Analysis (2)]
46.  Nel Van Zyl K, Whitelaw AC, Newton-Foot M. The effect of storage conditions on microbial communities in stool. PLoS One. 2020;15:e0227486.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
47.  Pedersen JG, Madsen AT, Gammelgaard KR, Aggerholm-Pedersen N, Sørensen BS, Øllegaard TH, Jakobsen MR. Inflammatory Cytokines and ctDNA Are Biomarkers for Progression in Advanced-Stage Melanoma Patients Receiving Checkpoint Inhibitors. Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]