2,916
Views
7
CrossRef citations to date
0
Altmetric
Review

Elucidating the gut microbiota composition and the bioactivity of immunostimulatory commensals for the optimization of immune checkpoint inhibitors

, , , , , , , , , , , , , ORCID Icon, , ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon & show all
Article: 1794423 | Received 26 May 2020, Accepted 06 Jul 2020, Published online: 20 Jul 2020

ABSTRACT

Accumulating evidence from preclinical studies and human trials demonstrated the crucial role of the gut microbiota in determining the effectiveness of anticancer therapeutics such as immunogenic chemotherapy or immune checkpoint blockade. In summary, it appears that a diverse intestinal microbiota supports therapeutic anticancer responses, while a dysbiotic microbiota composition that lacks immunostimulatory bacteria or contains overabundant immunosuppressive species causes treatment failure. In this review, we explore preclinical and translational studies highlighting how eubiotic and dysbiotic microbiota composition can affect progression-free survival in cancer patients.

Introduction

The rise of cancer immunotherapy over the past decade has revolutionized the clinical management of a wide array of malignancies that were previously associated with poor prognosis.Citation1 Immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1/PD-L2 and CTLA-4/CD86 axis are at the forefront of current implementations in various indications, alone or in combination, for advanced, metastatic, neoadjuvant, and adjuvant settings. Given the broad bioactivity across multiple histological tumor types, the durability of response, and therapeutic success in second or third line chemo-resistant diseases, ICIs are now positioned as a first-in-class drug and thus constitute major pillar in the oncological armamentarium.Citation2Citation8 As such, ICIs have been approved by multiple regulatory agencies worldwide and are now considered the standard of care in a wide range of solid and hematologic neoplastic diseases including advanced-stage melanoma, non-small-cell lung cancer (NSCLC), head and neck cancer, bladder cancer, or renal cell carcinoma (RCC).Citation9

Figure 1. Key bacteria safely boosting the efficacy of anticancer therapeutics in vivo.

Figure 1. Key bacteria safely boosting the efficacy of anticancer therapeutics in vivo.

Despite the exceptional improvement in objective response rates and overall survival (OS) benefits, ICI responses are currently only observed in a minority (~30%) of patients.Citation5,Citation7 Indeed, most patients manifest primary or secondary resistance to ICIs or even acceleration of the disease called “hyperprogression.”Citation10 Large efforts are being dedicated to identify the “cancer immune set-point,” a notion defined as the point which determines the parameters that govern the strength, timing, and threshold beyond which an effective immune response can occur in a given individual.Citation11,Citation12 Besides tumor intrinsic factors, many non-cell autonomous parameters control primary resistance to ICIs. Recent evidence points to the biological significance of the composition of the gut microbiota in influencing peripheral immune tonus and the effectiveness of immunotherapy in cancer patients.Citation13Citation16

The human gut microbiota modulates many host processes, including metabolism, inflammation, peristalsis, immune functions, and intestinal epithelial barrier fitness.Citation17Citation19 In the last decade, major progress has been made in the comprehension of colon cancer development in interaction with the local microbiota.Citation20 Surprisingly, a ‘deviated’ repertoire of the gut microbiota, called ‘intestinal dysbiosis,’ has been epidemiologically – and sometimes causally – associated with a variety of chronic inflammatory disorders including neoplasia, located at sites distant from the gut. In parallel, discoveries made in preclinical tumor models and in cancer patients have demonstrated that the composition of the intestinal microbiota influences the effectiveness of anticancer agents (such as immunogenic chemotherapies and ICIs) and regulates tumor immunosurveillance.Citation21Citation28 Several lines of evidence have unraveled the link between the gut microbiota composition and ICI-mediated anti-tumor immune responses. This review will summarize arguments supporting the links between the intestinal ecosystem and tumor immunosurveillance, overviewing the deleterious effects of antibiotics on the clinical benefits to be expected from ICIs, the metagenomics-based fingerprints dictating survival, and the key regulatory bacteria associated with tumor control during treatment with immunotherapy.

Antibiotics hinder the efficacy of ICIs

Preclinical studies performed in axenic (gnotobiotic) or broad-spectrum antibiotic (ATB)-treated mice have supported a cause–effect relationship between dysbiosis and the failure of anticancer therapeutics.Citation21Citation23,Citation26,Citation28 Several independent retrospective studies in advanced cancer patients across a diverse range of malignancies (NSCLC, RCC, bladder cancer, melanoma, and geographic locations) revealed that antibiotic treatment taken 1 month before anticancer therapeutics dampens the clinical efficacy of ICIs and immunogenic chemotherapy. These observations specifically highlighted that the disruption of a homeostatic microbiota (i.e., a switch from eubiosis to dysbiosis) and the loss of specific bacterial species may be detrimental for the success of anticancer therapies.Citation28Citation36 Recently, corroborating this notion, Derosa et al. confirmed in a prospective trial investigating the composition of the gut microbiota through shotgun metagenomics that antibiotics prior to second-line PD-1 blockade in advanced RCC patients had a deleterious clinical impact, reducing the microbiota diversity, and increasing Clostridium hathewayi, a species associated with immune tolerance.Citation37 In parallel, microbiota profiling from 70 Japanese NSCLC patients also showed that ATB prior to ICIs decreases bacterial diversity and increases Clostridium hathewayi.Citation38 Intrinsically, identification of key bacteria driving the sensitivity/primary resistance to anticancer treatments is crucial to unravel the role of the gut microbiota in this scenario. Many of the recently published studies in this area highlight the deleterious effect of antibiotics in patients with more advanced disease and with multifariousness incidents that may influence ICI responses.Citation28,Citation37 However, a recent analysis showed that patients with non-metastatic melanoma, a “best-prognosis” subgroup receiving ICIs in the adjuvant setting, also had a survival detriment if exposed to antibiotics. Clearly, harm from antibiotics is not limited to cancer patients with advanced metastatic disease.Citation39 Nevertheless, antibiotic classes should be carefully considered. Some antibiotics can provide a positive ‘eubiotic’ effect on the gut microbiota by reducing the abundance of unfavorable gut bacteria. Vancomycin, mostly targeting gram-positive bacteria, including butyrate-producing bacteria and decreasing short-chain fatty acids (SCFA) concentrations, in combination with radiotherapy was able to potentiate the abscopal antitumor immune effect and tumor growth inhibition in mice. Notably, butyrate, a metabolite produced by the vancomycin-depleted gut bacteria, abrogated the vancomycin effect.Citation40 In fact, high levels of butyrate and propionate in the blood are associated with resistance to CTLA-4 blockade and an increase in the abundance of Treg cells.Citation41 However, these results are in contrast with a small Japanese study (52 patients suffering from a broad range of cancer types) showing that high concentrations of fecal and plasma SCFAs were associated with a response to PD-1 treatment and longer progression-free survival (PFS).Citation42 Additional research is needed to clarify the association between fecal and plasma SCFAs and the efficacy of ICIs. Conversely, there is strong evidence indicating that antibiotics-induced dysbiosis is associated with poor therapeutic efficacy of ICI-based immunotherapy, suggesting a causal link between dysbiosis and poor therapeutic outcome.Citation28Citation33,Citation36,Citation37

Gut oncomicrobiota signatures associated with response to ICIs

Recent advances in next-generation sequencing (NGS) approaches, allowing for the in-depth study of the intestinal microbiota composition, facilitated the discovery of correlations between specific fingerprints of the gut microbiota with the onset and course of certain pathologies.Citation43 Accordingly, the exploration of the composition of the gut microbiota in cancer patients through 16S rRNA gene sequencing or shotgun metagenomics has demonstrated a major impact of the gut microbiota on the clinical activity of ICIs. These analyses led to the hypothesis that the intestinal microbiota can be used to categorize patients receiving ICIs in responders (R) and non-responders (NR) as defined by standardized response evaluation criteria in solid tumors (RECIST 1.1 criteria) regardless of methodologies for DNA extraction and sequencing, geo-distributions of patient populations, and therapies (.

Table 1. Studies highlighting the role of the gut microbiota in the clinical efficacy of anticancer therapeutics.

The first evidence came from a French cohort of metastatic melanoma (MM) patients treated with the anti-CTLA-4 antibody ipilimumab. Twenty-six MM patients were prospectively enrolled to analyze the impact of gut microbiota composition at baseline on clinical response to ipilimumab.Citation44 Interestingly, the authors could segregate cancer patients into clusters driven by specific bacterial fingerprints found using 16S rRNA gene sequencing. Patients belonging to cluster A harbored Faecalibacterium spp. and were associated with longer PFS than Bacteroides spp.-driven cluster B patients. Moreover, patients from cluster A exhibited lower circulating CD4+ Tregs.Citation41,Citation44 An additional study including 39 patients focusing on various ICI regimens (anti-CTLA-4, anti-PD-1, or the combination of both) corroborated the finding that the metagenomics-based analysis of the gut microbiota composition can predict clinical outcome of immune checkpoint blockade in MM patients. It also showed with the bias of a limited number of patients in each immunotherapy arm that the best species predictive for response are different in each regimen.Citation45 Here again, a relative enrichment in Faecalibacterium prausnitzii was strongly associated with responses to a combination of both nivolumab and ipilimumab while Dorea formicigenerans correlated with a favorable clinical outcome during the course of pembrolizumab.Citation45 A study published by Gopalakrishnan et al. revealed that MM patients, from Texas (USA), who responded to anti-PD-1 therapy, had a significantly higher diversity of bacteria in their stool at diagnosis compared to NR. Moreover, a higher relative abundance of Clostridiales, Ruminococcaceae, and Faecalibacterium was observed in individuals with a good prognosis while NR cancer patients had a higher abundance of Bacteroidales.Citation27 The relationship between the dominance of distinct intestinal bacteria and tumor immunosurveillance was discussed when correlating tumor-infiltrating lymphocyte (TIL) phenotyping and 16S rRNA-based bacterial enrichment. The authors showed, in 25 patients, that CD8+, CD3+, FOXP3+, PD1+, and Granzyme B+ TILs were associated with the Faecalibacterium genus, the Ruminococcaceae family, and the Clostridiales order, suggesting the impact of distinct commensals on cytolytic T cells entailing tumor progression.Citation27 Another US report, from Chicago, also demonstrated significant microbiota-related differences in the response to treatment with PD-1 blocking antibodies. 16S rRNA sequencing of gene amplicons in fecal materials of 42 MM patients at baseline demonstrated that R had enrichment in Bifidobacterium longum, Collinsella aerofaciens, and Enterococcus faecium.Citation25 Moreover, a study performed in 25 Dutch MM patients showed that differences in taxa abundance contrasted R and NR, with similar results with previous studies. Indeed, carriers of Streptococcus parasanguinis or Bacteroides massiliensis exhibited prolonged PFS while individuals harboring Peptostreptococcaceae (unclassified species) exhibited a shorter OS and PFS compared to non-carriers.Citation46 Taken together, these epidemiological studies described the association between the composition of the intestinal ecosystem at diagnosis and the clinical outcome of MM patients treated with ICIs.

These particular findings are not restricted to MM. Indeed, the fecal bacteria repertoire has been found to also critically influence the prognosis of advanced NSCLC and RCC cancer patients during the course of ICI-based therapies in France. Quantitative metagenomics analysis performed prior to anti-PD-1 blockade identified a distinct gut metagenomic fingerprint (centered around Akkermansia muciniphila and Alistipes spp.) in stools of 100 patients who benefited from PD-1 inhibition; considering response rates or PFS at 3 months.Citation28 Interestingly, the role of the gut microbiota has also been addressed in an East-Asian NSCLC population.Citation47 In this cohort, 16S rRNA gene sequencing of 37 stools demonstrated that higher diversity of the gut microbiota paved the way to prolonged PFS. Differential gut microbiota signatures contrasted R versus NR cancer patients. Here again, Alistipes putredinis, Prevotella copri, or Bifidobacterium longum were enriched in R patients. The Shannon diversity index of the taxonomic composition was positively correlated with circulating immune antigen-primed–cytotoxic T cells (such as GZMB+CD45RO+CD27+ CD8+ T cells or GZMB+CD45RO+CD27 CD8+ T cells).Citation47 Two additional Japanese studies performed 16S rRNA gene sequencing of fecal materials from NSCLC (n = 70) and NSCLC (n = 14) as well as gastric cancer (n = 24) patients confirmed that higher diversity of the bacterial community and enrichment of the Ruminococcaceae and Clostridiales order predicted benefit to PD-1 blockade.Citation38,Citation48 Furthermore, the relative abundance of members of the Ruminococcaceae familyCitation48 correlated with the density of PD-1+CD8+ T cells among (TILs). Again, another report analyzing the gut microbiota composition from 17 NSCLC patients revealed that Lactobacillus, Clostridium, and Syntrophococcus were overrepresented in R, while Bilophila or SutterellaCitation49 was dominant in NR. Of note, the presence of Bilophila drastically shortened the time to treatment failure.Citation49 A Chinese prospective study including 63 NSCLC cancer patients revealed Parabacteroides and Methanobacteriaceae as species and family members associated with PFS >6 months while stool enriched in Veillonella, Selenomonadales, and NegativicutesCitation50 predicted shorter PFS during PD-1 blockade. In sharp contrast with these findings, ileal enrichment with Veillonella, Selenomonadales, and Negativicutes was found to be associated with increased TIL and favorable prognosis during oxaliplatin-based chemotherapy in proximal colon cancer patients.Citation51

Several teams have confirmed the potential clinical significance of Akkermansia muciniphila in driving a therapeutic benefit to ICIs, more specifically in NSCLC,Citation28 melanoma,Citation46 HCC patients,Citation52 and recently in RCC.Citation37,Citation53 In brief, Derosa et al. reported in 58 RCC cancer patients treated in 2 L with nivolumab that a significant bacterial composition contrasted R versus NR with an overrepresentation of distinct species including Akkermansia muciniphila, Bacteroides salyersiae, or Eubacterium siraeum in patients disposed to becoming R.Citation37

In a parallel study, all RCC cancer patients exhibiting a complete response to ICIs (n = 3) harbored Akkermansia muciniphila although the number of patients was not sufficient to draw definitive conclusions.Citation53 Fecal metagenomics analysis performed in 8 HCC cancer patients identified Akkermansia muciniphila and Ruminococcaceae spp. in the 20 enriched spp. characterizing R patients and B. nordii in a 15 spp-fingerprint associated with NR as already reported.Citation28,Citation52 In Dutch MM patients, A. muciniphila was also listed in the favorable commensals associated with objective responses to ICI therapy.Citation46

Interestingly, focusing on describing also negative species by applying various bioinformatic and clinical subgroup analyses (LEfSe, PLS-DA VIP, networks), Derosa et al. identified a set of species (phylum Firmicutes, family Clostridiaceae, Clostridium clostridioforme, Clostridium hathewayi) as associated with primary resistance to ICIs, enriched by ATB use and metastatic cancer status.Citation37

Although ICIs have revolutionized therapeutic approaches across various malignancies, conventional anticancer regimens such as chemotherapy or radiotherapy still represent the cornerstone of oncological arsenal. Numerous studies have addressed the putative influence of the gut microbiota repertoire in the prediction of clinical responses to these cytotoxic agents. Twenty-six cancer patients diagnosed with miscellaneous malignancies, treated either with cytotoxic compounds or targeted medicine or a combination of the latter drugs with immunotherapy, were enrolled in a prospective study aimed at segregating patients according to their intestinal commensalism. Bacteroides xylanisolvens, Bacteroides ovatus, and Prevotella copri were significantly overrepresented in R compared to NR defined using the RECIST1.1 criterion. In contrast, Clostridium symbiosum and Ruminococcus gnavus were enriched in NR.Citation54 A second study analyzing fecal composition prior to preoperative concurrent chemoradiations in 45 rectal cancer patients concluded that Duodenibacillus massiliensis was linked to complete responses.Citation55

Altogether, the emerging field of oncoimmunomicrobiology is progressively integrating the gut microbiota into the parameters that determine the cancer immune set-point governing the clinical efficacy of immuno-chemo-radio-therapy. First, low alpha diversity of the intestinal ecosystem is associated with dismal prognosis in advanced cancer patients, as also shown in several chronic inflammatory disorders (such as obesity).Citation56 Secondly, some bacteria species arise to be repeatedly associated with favorable clinical outcomes (namely Akkermansia muciniphila, Ruminococcaceae including Faecalibacterium prausnitzi, Bifidobacterium spp.) although variabilities in the main commensal fingerprints associated with a specific pattern of responses appear obvious within analogous patients’ populations and therapies. These variabilities could be explained by many factors such as DNA extraction and sequencing methodologies,Citation55 cohort size, age and gender, geography,Citation57,Citation58 and confounding factors (including diet, lifestyle, exposure to xenobiotics, antibiotic class and window, comedications and comorbidities).Citation59,Citation60 Other important players that affect intestinal barrier integrity are the tumor itself, disease stage, ECOG performance status, medication, peripheral inflammatory tone, and pro-cachexia signs.Citation61,Citation62 Finally, aside from the basal composition of the gut commensalism, dictated by the original network of bacterial co-occurrence, the treatment itself may impact on the relative abundance of microbes, as shown with ipilimumabCitation23 and tyrosine kinase inhibitors (TKIs).Citation37 Overall, TKIs induced a significant and characteristic microbiota shift promoting a higher abundance of immunostimulatory commensals that could be used to improve the efficacy of ICIs in RCC patients such as A. senegalensis and A. muciniphila.

In addition, a different study paved the way in understanding the reciprocal relationship between the intratumoral microbiota and the clinical outcome of resected pancreatic ductal adenocarcinoma (PDAC) cancer patients. Although most of the patients died at an advanced stage with an OS of 9% at 5 y, a minor subset of patients survives longer.Citation63 Interestingly, alpha-diversity of the tumor microbiota was significantly higher in the long-term survivor of PDAC.Citation64 In fact, an enrichment on Proteobacteria (Pseudoxanthomonas) and Actinobacteria (Saccharopolyspora and Streptomyces) was observed in this subset of patients.Citation64 However, intra-tumoral microbes in pancreatic cancer may also be harmful. Pushalkar et al. demonstrated the negative impact of microbes on antitumor immunity with evidence for possible migration of bacteria from the gut to the pancreas.Citation65 Nevertheless, these emerging findings indicate there is a cross-talk between gut microbiota and local microbiota (as exemplified by pancreatic cancer) and that also local microbiota may contribute positively or negatively to carcinogenesis and therapeutic responses.

Identification of key bacteria boosting the antitumoral efficacy of anticancer treatments

Modulating the composition of the gut microbiota and harnessing the immunogenicity of the intestinal microbiota may be a promising strategy with which to circumvent primary resistance to anticancer therapeutics. Several bacterial candidates have been identified, isolated, characterized, and are currently or on the verge to be tested in clinical trials in combination with anticancer treatments or as a standalone therapy.Citation66

Cause-effects relationships between the presence of distinct microbial commensals and antitumor activity have been examined primarily in preclinical models. So far, investigators have performed oral gavages in germ-free or broad ATB-treated mice using a complete human or mouse ecosystem or a complex mixture of several bacteria or “monoclonal” strains, into immunocompetent syngeneic hosts inoculated with ortho-or hetero-topic cancers. The concept of “avatar” mice which consists in colonizing gut-sterilized mice with patients’ stools has proven useful to recapitulate human dysbiosis across various diseases.Citation67,Citation68 In the setting of cancer, avatar mice transferred with feces from patients bearing melanoma, NSCLC, RCC, or colon cancer and transplanted with orthotopic tumors could convey the phenotype of R versus NR following immunotherapy with anti-PD1 and/or anti-CTLA-4 Ab, in 100% cases after oral gavage with R fecal material and in 75% cases when supplementing with NR derived feces.Citation23,Citation25,Citation27,Citation28

Akkermansia muciniphila is a strictly anaerobic Gram-negative bacterium from the phylum Verrucomicrobia displaying a multifaceted mode of action.Citation69,Citation70 Indeed, a Phase I trial conducted in 32 overweight/obese insulin-resistant volunteers demonstrated that supplementation with Akkermansia muciniphila is safe and capable of improving the metabolic fitness.Citation71 Further studies have emphasized its capacity to prolong lifespan in progeroid miceCitation72 while ameliorating the symptoms of amyotrophic lateral sclerosis through nicotinamide accumulation in the central nervous system.Citation73 A recent report showed that A. muciniphila prevents colitis-induced colon cancer by mobilizing TNF producing CTL primed in the mesenteric lymph nodes and expressing low levels of PD1 despite their lytic potential.Citation74 We highlighted the capacity of A. muciniphila to boost immune responses during the course of PD-1 blockade, both in tumor-bearing rodents and humans.Citation28,Citation37 Supplementation of NR-FMT treated avatar mice with A. muciniphila rescued the antitumoral efficacy of PD1-blockade in an IL-12-dependent manner demonstrating that A. muciniphila dictates the clinical outcome of ICIs. In addition, the bacterium dampened the recruitment of immunosuppressive Tregs cells into the tumor microenvironment while eliciting the accumulation of CC-chemokine receptor 9 (CCR9)-expressing Th1 cells in the tumor bed. Accordingly, memory Th1 and Tc1 cell reactivity against A. muciniphila correlated with a clinical benefit of PD-1 blockade in NSCLC and RCC cancer patients.Citation28

Enterococcus hirae has been one of the first bacterial isolates that show antitumoral potential in combination with chemotherapy. This Gram+ bacterium is essential to mediate the antitumoral efficacy of cyclophosphamide (CTX), a prominent alkylating anticancer agent.Citation21,Citation75 CTX promotes the translocation of E. hirae in secondary lymphoid organs (mLN and spleen), inducing FNγ (and IL-17) producing CD4 + T cells and Tc1 cells. Moreover, the combination of E. hirae and CTX reduced Treg numbers in sarcomas, culminating in a significant rise of the CD8/Foxp3 ratio, which in turn anticorrelated with tumor size.Citation21 Hence, oral gavage with E. hirae restored the antitumoral efficacy of CTX lost in ATB-treated mice. In advanced cancer patients, memory CD4+ Th1 cell responses against E. hirae were associated with survival in CTX- or anti-PD-1Citation27 antibody-treated individuals. While the prevalence of E. hirae is minimally detected using shotgun metagenomics-based analyses of patient stool, culturomics allowed for the isolation of E. hirae colonies in 20% cancer patients. Diagnosis of E. hirae in stool culturomics of NSCLC patients at diagnosis before starting second-line PD-1 blockade predicted prolonged survival.Citation28 An independent study revealed that the frequency of circulating T cells recognizing E. hirae correlated with robust CD8+ T cell responses and better prognosis in HBV-related hepatocellular carcinoma,Citation76 suggesting the clinical significance of this particular bacterium across different malignancies.

In addition to E. hirae, other Enterococci spp. have been isolated and characterize for their immunomodulatory potential against cancer cells. A strain of Enterococcus gallinarum, isolated from a healthy human gut, has demonstrated its antiproliferative effects against EMT6 breast, RENCA renal, and LLC1 lung carcinoma.Citation77 This microbial product caused changes in the tumor immune microenvironment and increased the CD8+/FoxP3 ratio. In addition, a TLR5 dependent immuno-stimulatory phenotype of this strain was monitored using reporter cell lines. The authors identified flagellin as the active component of Enterococcus gallinarum.Citation78 Therefore, the antitumoral potential of this strain is currently under investigation in cancer patients amenable to ICI-based therapy in advanced diseases, as well as in neoadjuvant settings to determine its property to modulate the tumor microenvironment before tumor resection (NCT03934827/NCT04193904).

Bifidobacterium is a gram-positive, non-spore-forming, non-motile, non-filamentous polymorphic rod bacterium. Pioneering studies demonstrated that the growth kinetics of B16.SIY melanoma as well as the intratumoral CD8+ T cell accumulation were completely different in mice purchased from different vendors (Jackson Laboratories (JAX) versus Taconic Farms (TAC)) harboring distinct commensal microbiota.Citation24 These differences were ablated when the two mouse colonies were cohoused, demonstrating that the normalization of the gut microbiota could boost anti-cancer immune responses. Further investigations characterizing the composition of the gut microbiota between JAX and TAC highlighted that certain Bifidobacterium species could induce tumor-infiltrating CD8+ T cells. Transfer of Bifidobacterium breve or Bifidobacterium longum or fecal material from JAX mice into TAC mice could all reduce melanoma growth and restore anti-melanoma cytotoxic T lymphocyte (CTL) responses.Citation24 A recent study unveiled that the SIY antigen (TAA) of B1610 displayed antigen mimicry with an epitope belonging to Bifidobacterium breve, accounting for the T cell–mediated antitumor responses achieved by oral supplementation with this probiotic.Citation79 Accordingly, T cells targeting the microbial antigen recognized melanoma tumor cells expressing the SIY tumor-associated antigen. Conversely, tumors expressing the TAA also grew faster in mice lacking Bifidobacterium breve bacterium.Citation79 Of note, memory immune reactivity against B. longum also correlated with robust CD8+ T cell responses and better prognosis in HBV-related hepatocellular carcinoma patients.Citation76

The first bacterial species known to harbor “zwitterionic” peptides capable of engaging CD4+ T cell receptors was Bacteroides fragilis.Citation80Citation82 B. fragilis was very effective in boosting immune responses primed in the setting of sarcoma tumors treated with anti-CTLA4 AbCitation23 as well as colon carcinoma treated with oxaliplatin-based immunogenic chemotherapy.Citation51 Antibiotics blunted the anticancer efficacy of CTLA-4 blockade against various transplantable tumors unless oral supplementation with B. fragilis was performed, which reinstated IL-12-dependent Th1 immune responses. Interestingly, anti-CTLA-4 Abs administered to tumor-bearing avatar mice reconstituted with FMT from melanoma patients foster the overrepresentation of distinct Bacteroides spp. (Bacteroides fragilis or Bacteroides thetaiotaomicron) and recapitulated the phenotype of response observed in patient.Citation23 Interestingly, oral supplementation with B. fragilis (as opposed to Fusobacterium nucleatum or Paraprevotella clara) turned chemotherapy-induced tolerogenic ileal apoptosis into immunogenic cell demise capable of eliciting PD1high follicular helper T cells and B cell responses and of promoting the efficacy of anti-PD1 Abs against established colon cancers.Citation51 Hence, the ileal microbiota enriched in commensals playing the role of adjuvant for ileal apoptosis triggered TFH and the efficacy of PD-1 blockade, even in tumors devoid of neoantigens.

In contrast to the aforementioned approaches, using very common commensals to compensate gut dysbiosis, another study demonstrated that a mixture of several rare strains isolated from fecal materials from healthy Japanese individuals was effective in shaping immunity in the colonic mucosae and tumor microenvironment.Citation83 The authors identified a cocktail of 11 human bacterial strains capable of promoting tent IFNγ producing CD8+ T cells that are not only crucial for combatting infectious pathogens but also for dampening cancer progression.Citation83 Supplementation of germ-free mice with these 11 strains (composed of 7 Bacteroidales spp. and 4 non-Bacteroidales spp.) resulted in the robust induction of IFNγ-CD8+ T cells in the colon through a mechanism requiring Batf3 dependent CD103+ CD11b dendritic cells. Next, they showed the capacity of the cocktail to ameliorate the efficacy of PD-1 blockade in axenic MC38 adenocarcinoma bearing mice. It significantly improved the efficacy of ICIs while increasing the frequency IFNγ+ CD8 TILs phenotypically distinct from the colonic IFNγ+ CD8 T cell subsets. Of note, the bacterial cocktail could reduce tumor growth as a standalone therapy (in the absence of PD-1 blockade).Citation83 This microbial product is currently tested in combination with PD-1 blockade in advanced cancer patients after vancomycin sensitization (NCT04208958).

It is well known that bacteria can sporulate under life-threatening circumstancesCitation84,Citation85 offering an advantage over non-sporulating commensals for the long-lasting colonization of their hosts. This property has been exploited by other investigators in the setting of PD-1 blockade. Firmicutes spores fraction isolated from a healthy donor stool was capable of rescuing the antitumoral efficacy of PD-1 blockade in both conventional mice treated with antibiotics and axenic mice by increasing CD8+ TILs.Citation86

Needless to say that most of the antitumoral efficacy described in all these preclinical studies appear to be strain-specific,Citation22 urging for delineating precise modes of action for each single isolate.

Concluding remarks

Several clinical trials are evaluating the capacity of harnessing the gut microbiota to improve cancer treatments from different angles such as examining the ability to prevent primary resistance to various anticancer treatment modalities, transforming “cold into hot” tumor microenvironment, and mitigating toxicities associated with a single line or combination ICIs.Citation66 Several important issues need to be addressed in the clinical development of live biotherapeutic products or their derivatives (metabolites, antigens, or adjuvants). First, robust preclinical datasets are mandatory to characterize the mechanisms of action of each strain or microbial products to design the most suitable clinical strategy and indications. This will allow for the design of appropriate pharmacodynamic parameters to follow compliance and transient colonization of the patient. Secondly, patient stratification will be necessary to avoid treating patients without overt intestinal dysbiosis, and for whom primary resistance to ICIs may be related to tumor intrinsic factors. Third, compatibility networks between the indigenous microflora and the live biotherapeutic product may be crucial for a long-lasting benefit of repetitive courses of anticancer probiotics. Pre-sensitization with antibiotics or other innovative approaches aimed at eliminating pathobionts associated with ICI failure or precluding colonization or bioactivity of immunogenic commensals may be important to optimize clinical regimen. Regardless of these considerations, this emerging field will benefit from pioneering trials showing the efficacy of FMT from complete responders into patients experiencing primary resistance to PD-1 blockade.

Conflicts of interest

RD is a full-time employee of everImmune, a biotech company dedicated to immunostimulatory bacteria. RD, GK, and LZ are the scientific cofounders of everImmune.

Acknowledgments

LZ and GK are supported by the Ligue contre le Cancer (équipe labellisée); Agence National de la Recherche (ANR) – Projets blancs; ANR under the frame of E-Rare-2, the ERA-Net for Research on Rare Diseases; AMMICa US23/CNRS UMS3655; Association pour la recherche sur le cancer (ARC); Association “Le Cancer du Sein, Parlons-en!”; Cancéropôle Ile-de-France; Chancelerie des universités de Paris (Legs Poix), Fondation pour la Recherche Médicale (FRM); a donation by Elior; European Research Area Network on Cardiovascular Diseases (ERA-CVD, MINOTAUR); Gustave Roussy Odyssea, the European Union Horizon 2020 Project Oncobiome; Fondation Carrefour; High-end Foreign Expert Program in China (GDW20171100085), Institut National du Cancer (INCa); Inserm (HTE); Institut Universitaire de France; LeDucq Foundation; the LabEx Immuno-Oncology (ANR-18-IDEX-0001); the RHU Torino Lumière; the Seerave Foundation; the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); and the SIRIC Cancer Research and Personalized Medicine (CARPEM). A.P.C. is supported by the CPRIT Research Training Program (RP170067). LD is supported by Fondation Philanthropia, Gustave Roussy.

Supplementary

Supplemental data for this article can be accessed on the publisher’s website.

References

  • Pardoll D. Cancer and the immune system: basic concepts and targets for intervention. Semin Oncol. 2015;42(4):523–11. doi:10.1053/j.seminoncol.2015.05.003.
  • Robert C, Thomas L, Bondarenko I, O’Day S, Weber J, Garbe C, Lebbe C, Baurain J-F, Testori A, Grob -J-J, et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N Engl J Med. 2011;364(26):2517–2526. doi:10.1056/NEJMoa1104621.
  • Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, Chow LQ, Vokes EE, Felip E, Holgado E, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373(17):1627–1639. doi:10.1056/NEJMoa1507643.
  • Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, Schuster SJ, Millenson MM, Cattry D, Freeman GJ, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015;372(4):311–319. doi:10.1056/NEJMoa1411087.
  • Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366(26):2443–2454. doi:10.1056/NEJMoa1200690.
  • Ribas A, Hamid O, Daud A, Hodi FS, Wolchok JD, Kefford R, Joshua AM, Patnaik A, Hwu W-J, Weber JS, et al. Association of pembrolizumab with tumor response and survival among patients with advanced melanoma. JAMA. 2016;315(15):1600–1609. doi:10.1001/jama.2016.4059.
  • Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373(19):1803–1813. doi:10.1056/NEJMoa1510665.
  • Hodi FS, O’Day SJ, McDermott DF, Weber RW, Sosman JA, Haanen JB, Gonzalez R, Robert C, Schadendorf D, Hassel JC, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–723. doi:10.1056/NEJMoa1003466.
  • Yu JX, Hubbard-Lucey VM, Tang J. Immuno-oncology drug development goes global. Nat Rev Drug Discov. 2019;18(12):899–900. doi:10.1038/d41573-019-00167-9.
  • Champiat S, Ferrara R, Massard C, Besse B, Marabelle A, Soria J-C, Ferté C. Hyperprogressive disease: recognizing a novel pattern to improve patient management. Nat Rev Clin Oncol. 2018;15(12):748–762. doi:10.1038/s41571-018-0111-2.
  • Chen DS, Mellman I. Elements of cancer immunity and the cancer-immune set point. Nature. 2017;541(7637):321–330. doi:10.1038/nature21349.
  • Sharma P, Hu-Lieskovan S, Wargo JA, Ribas A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168(4):707–723. doi:10.1016/j.cell.2017.01.017.
  • Zitvogel L, Ma Y, Raoult D, Kroemer G, Gajewski TF. The microbiome in cancer immunotherapy: diagnostic tools and therapeutic strategies. Science. 2018;359(6382):1366–1370. doi:10.1126/science.aar6918.
  • Routy B, Gopalakrishnan V, Daillère R, Zitvogel L, Wargo JA, Kroemer G. The gut microbiota influences anticancer immunosurveillance and general health. Nat Rev Clin Oncol. 2018;15(6):382–396. doi:10.1038/s41571-018-0006-2.
  • Kroemer G, Zitvogel L. Cancer immunotherapy in 2017: the breakthrough of the microbiota. Nat Rev Immunol. 2018;18(2):87–88. doi:10.1038/nri.2018.4.
  • Zitvogel L, Daillère R, Roberti MP, Routy B, Kroemer G. Anticancer effects of the microbiome and its products. Nat Rev Microbiol. 2017;15(8):465–478. doi:10.1038/nrmicro.2017.44.
  • Maynard CL, Elson CO, Hatton RD, Weaver CT. Reciprocal interactions of the intestinal microbiota and immune system. Nature. 2012;489(7415):231–241. doi:10.1038/nature11551.
  • Bäckhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science. 2005;307(5717):1915–1920. doi:10.1126/science.1104816.
  • Kamada N, Chen GY, Inohara N, Núñez G. Control of pathogens and pathobionts by the gut microbiota. Nat Immunol. 2013;14(7):685–690. doi:10.1038/ni.2608.
  • de Vos WM, de Vos EAJ. Role of the intestinal microbiome in health and disease: from correlation to causation. Nutr Rev. 2012;70(Suppl 1):S45–56. doi:10.1111/j.1753-4887.2012.00505.x.
  • Viaud S, Saccheri F, Mignot G, Yamazaki T, Daillère R, Hannani D, Enot DP, Pfirschke C, Engblom C, Pittet MJ, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science. 2013;342(6161):971–976. doi:10.1126/science.1240537.
  • Daillère R, Vétizou M, Waldschmitt N, Yamazaki T, Isnard C, Poirier-Colame V, Duong CPM, Flament C, Lepage P, Roberti MP, et al. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity. 2016;45(4):931–943. doi:10.1016/j.immuni.2016.09.009.
  • Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CPM, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–1084. doi:10.1126/science.aad1329.
  • Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Lei YM, Jabri B, Alegre M-L, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350(6264):1084–1089. doi:10.1126/science.aac4255.
  • Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre M-L, Luke JJ, Gajewski TF. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359(6371):104–108. doi:10.1126/science.aao3290.
  • Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342(6161):967–970. doi:10.1126/science.1240527.
  • Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, Prieto PA, Vicente D, Hoffman K, Wei SC, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359(6371):97–103. doi:10.1126/science.aan4236.
  • Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97. doi:10.1126/science.aan3706.
  • Derosa L, Hellmann MD, Spaziano M, Halpenny D, Fidelle M, Rizvi H, Long N, Plodkowski AJ, Arbour KC, Chaft JE, 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;29(6):1437–1444. doi:10.1093/annonc/mdy103.
  • Pinato DJ, Howlett S, Ottaviani D, Urus H, Patel A, Mineo T, Brock C, Power D, Hatcher O, Falconer A, et al. Association of prior antibiotic treatment with survival and response to immune checkpoint inhibitor therapy in patients with cancer. JAMA Oncol. 2019 Sep 12;5(12):1774. doi:10.1001/jamaoncol.2019.2785.
  • Elkrief A, El Raichani L, Richard C, Messaoudene M, Belkaid W, Malo J, Belanger K, Miller W, Jamal R, Letarte N, et al. Antibiotics are associated with decreased progression-free survival of advanced melanoma patients treated with immune checkpoint inhibitors. Oncoimmunology. 2019;8(4):e1568812. doi:10.1080/2162402X.2019.1568812.
  • Zhao S, Gao G, Li W, Li X, Zhao C, Jiang T, Jia Y, He Y, Li A, Su C, et al. Antibiotics are associated with attenuated efficacy of anti-PD-1/PD-L1 therapies in Chinese patients with advanced non-small cell lung cancer. Lung Cancer. 2019;130:10–17. doi:10.1016/j.lungcan.2019.01.017.
  • Lalani A-KA, Xie W, Braun DA, Kaymakcalan M, Bossé D, Steinharter JA, Martini DJ, Simantov R, Lin X, Wei XX, et al. Effect of antibiotic use on outcomes with systemic therapies in metastatic renal cell carcinoma. Eur Urol Oncol. 2019 Sep 24; doi:10.1016/j.euo.2019.09.001.
  • Pflug N, Kluth S, Vehreschild JJ, Bahlo J, Tacke D, Biehl L, Eichhorst B, Fischer K, Cramer P, Fink A-M, et al. Efficacy of antineoplastic treatment is associated with the use of antibiotics that modulate intestinal microbiota. Oncoimmunology. 2016;5(6):e1150399. doi:10.1080/2162402X.2016.1150399.
  • Nenclares P, Bhide SA, Sandoval-Insausti H, Pialat P, Gunn L, Melcher A, Newbold K, Nutting CM, Harrington KJ. Impact of antibiotic use during curative treatment of locally advanced head and neck cancers with chemotherapy and radiotherapy. Eur J Cancer. 2020;131:9–15. doi:10.1016/j.ejca.2020.02.047.
  • Wilson BE, Routy B, Nagrial A, Chin VT. The effect of antibiotics on clinical outcomes in immune-checkpoint blockade: a systematic review and meta-analysis of observational studies. Cancer Immunol Immunother. 2020;69(3):343–354. doi:10.1007/s00262-019-02453-2.
  • Derosa L, Routy B, Fidelle M, Iebba V, Alla L, Pasolli E, Segata N, Desnoyer A, Pietrantonio F, Ferrere G, et al. Gut bacteria composition drives primary resistance to cancer immunotherapy in renal cell carcinoma patients. Eur Urol. 2020 May 3; doi:10.1016/j.eururo.2020.04.044.
  • Meeting Library Gut microbiome to predict efficacy and immune-related toxicities in patients with advanced non-small cell lung cancer treated with anti-PD-1/PD-L1 antibody-based immunotherapy. [accessed May 25, 2020]. https://meetinglibrary.asco.org/record/188511/abstract
  • Mohiuddin JJ, Chu B, Facciabene A, Poirier K, Wang X, Doucette A, Zheng C, Xu W, Anstadt EJ, Amaravadi RK, et al. Association of antibiotic exposure with survival and toxicity in patients with melanoma receiving immunotherapy. J Natl Cancer Inst. 2020 Apr 15; doi:10.1093/jnci/djaa057.
  • Uribe-Herranz M, Rafail S, Beghi S, Gil-de-Gómez L, Verginadis I, Bittinger K, Pustylnikov S, Pierini S, Perales-Linares R, Blair IA, et al. Gut microbiota modulate dendritic cell antigen presentation and radiotherapy-induced antitumor immune response. J Clin Invest. 2020;130(1):466–479. doi:10.1172/JCI124332.
  • Coutzac C, Jouniaux J-M, Paci A, Schmidt J, Mallardo D, Seck A, Asvatourian V, Cassard L, Saulnier P, Lacroix L, et al. Systemic short chain fatty acids limit antitumor effect of CTLA-4 blockade in hosts with cancer. Nat Commun. 2020;11(1):1–13. doi:10.1038/s41467-020-16079-x.
  • Nomura M, Nagatomo R, Doi K, Shimizu J, Baba K, Saito T, Matsumoto S, Inoue K, Muto M. Association of short-chain fatty acids in the gut microbiome with clinical response to treatment with nivolumab or pembrolizumab in patients with solid cancer tumors. JAMA Netw Open. 2020;3(4):e202895. doi:10.1001/jamanetworkopen.2020.2895.
  • Claesson MJ, Clooney AG, O’Toole PW. A clinician’s guide to microbiome analysis. Nat Rev Gastroenterol Hepatol. 2017;14(10):585–595. doi:10.1038/nrgastro.2017.97.
  • Chaput N, Lepage P, Coutzac C, Soularue E, Le Roux K, Monot C, Boselli L, Routier E, Cassard L, Collins M, et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann Oncol. 2017;28(6):1368–1379. doi:10.1093/annonc/mdx108.
  • Frankel AE, Coughlin LA, Kim J, Froehlich TW, Xie Y, Frenkel EP, Koh AY. Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia. 2017;19(10):848–855. doi:10.1016/j.neo.2017.08.004.
  • 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(3):235–246. Publish Ahead of Print. doi:10.1097/CMR.0000000000000656.
  • 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(8):1378–1389. doi:10.1016/j.jtho.2019.04.007.
  • Fukuoka S, Daisuke M, Togashi Y, Sugiyama E, Udagawa H, Kirita K, Kamada T, Kawazoe A, Goto K, Doi T, et al. Association of gut microbiome with immune status and clinical response in solid tumor patients who received on anti-PD-1 therapies. JCO. 2018;36(15_suppl):3011. doi:10.1200/JCO.2018.36.15_suppl.3011.
  • Katayama Y, Yamada T, Shimamoto T, Iwasaku M, Kaneko Y, Uchino J, Takayama K. The role of the gut microbiome on the efficacy of immune checkpoint inhibitors in Japanese responder patients with advanced non-small cell lung cancer. Transl Lung Cancer Res. 2019;8(6):847–853. doi:10.21037/tlcr.2019.10.23.
  • Song P, Yang D, Wang H, Cui X, Si X, Zhang X, Zhang L. Relationship between intestinal flora structure and metabolite analysis and immunotherapy efficacy in Chinese NSCLC patients. Thoracic Cancer. 2020. doi:10.1111/1759-7714.13442
  • Roberti MP, Yonekura S, Duong CPM, Picard M, Ferrere G, Tidjani Alou M, Rauber C, Iebba V, Lehmann CHK, Amon L, et al. Chemotherapy-induced ileal crypt apoptosis and the ileal microbiome shape immunosurveillance and prognosis of proximal colon cancer. Nat Med. 2020 May;25:1–13. doi:10.1038/s41591-020-0882-8.
  • Zheng Y, Wang T, Tu X, Huang Y, Zhang H, Tan D, Jiang W, Cai S, Zhao P, Song R, et al. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J ImmunoTher Cancer. 2019;7(1):193. doi:10.1186/s40425-019-0650-9.
  • Agarwal A, Modliszewski J, Davey L, Reyes-Martinez M, Runyambo D, Corcoran D, Dressman H, George DJ, Valdivia R, Armstrong AJ, et al. Investigating the role of the gastrointestinal microbiome in response to immune checkpoint inhibitors (ICIs) among patients (pts) with metastatic renal cell carcinoma (mRCC). JCO. 2020;38(6_suppl):730. doi:10.1200/JCO.2020.38.6_suppl.730.
  • Heshiki Y, Vazquez-Uribe R, Li J, Ni Y, Quainoo S, Imamovic L, Li J, Sørensen M, Chow BKC, Weiss GJ, et al. Predictable modulation of cancer treatment outcomes by the gut microbiota. Microbiome. 2020;8. doi:10.1186/s40168-020-00811-2.
  • Jang B-S, Chang JH, Chie EK, Kim K, Park JW, Kim MJ, Song E-J, Nam Y-D, Kang SW, Jeong S-Y, et al. Gut microbiome composition is associated with a pathologic response after preoperative chemoradiation in rectal cancer patients. Int J Radiat Oncol Biol Phys. 2020 Apr 18;107(4):736–746. doi:10.1016/j.ijrobp.2020.04.015.
  • Cotillard A, Kennedy SP, Kong LC, Prifti E, Pons N, Le Chatelier E, Almeida M, Quinquis B, Levenez F, Galleron N, et al. Dietary intervention impact on gut microbial gene richness. Nature. 2013;500(7464):585–588. doi:10.1038/nature12480.
  • Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Magris M, Hidalgo G, Baldassano RN, Anokhin AP, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–227. doi:10.1038/nature11053.
  • Senghor B, Sokhna C, Ruimy R, Lagier J-C. Gut microbiota diversity according to dietary habits and geographical provenance. Hum Microbiome J. 2018;7–8:1–9. doi:10.1016/j.humic.2018.01.001.
  • Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, Ze X, Brown D, Stares MD, Scott P, Bergerat A, et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. Isme J. 2011;5(2):220–230. doi:10.1038/ismej.2010.118.
  • Ubeda C, Pamer EG. Antibiotics, microbiota, and immune defense. Trends Immunol. 2012;33(9):459–466. doi:10.1016/j.it.2012.05.003.
  • Bindels LB, Neyrinck AM, Salazar N, Taminiau B, Druart C, Muccioli GG, François E, Blecker C, Richel A, Daube G, et al. Non digestible oligosaccharides modulate the gut microbiota to control the development of leukemia and associated cachexia in mice. PLoS ONE. 2015;10(6):e0131009. doi:10.1371/journal.pone.0131009.
  • Bindels LB, Neyrinck AM, Claus SP, Le Roy CI, Grangette C, Pot B, Martinez I, Walter J, Cani PD, Delzenne NM. Synbiotic approach restores intestinal homeostasis and prolongs survival in leukaemic mice with cachexia. Isme J. 2016;10(6):1456–1470. doi:10.1038/ismej.2015.209.
  • Siegel RL, Miller KD, Jemal A. Cancer statistics, 2018. CA Cancer J Clin. 2018;68(1):7–30. doi:10.3322/caac.21442.
  • Riquelme E, Zhang Y, Zhang L, Montiel M, Zoltan M, Dong W, Quesada P, Sahin I, Chandra V, San Lucas A, et al. Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell. 2019;178(4):795–806.e12. doi:10.1016/j.cell.2019.07.008.
  • Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, Mohan N, Aykut B, Usyk M, Torres LE, et al. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 2018;8(4):403–416. doi:10.1158/2159-8290.CD-17-1134.
  • Daillère R, Derosa L, Bonvalet M, Segata N, Routy B, Gariboldi M, Budinská E, Vries IJMD, Naccarati AG, Zitvogel V, et al. Trial watch: the gut microbiota as a tool to boost the clinical efficacy of anticancer immunotherapy. OncoImmunology. 2020;9(1):1774298. doi:10.1080/2162402X.2020.1774298.
  • Sharon G, Cruz NJ, Kang D-W, Gandal MJ, Wang B, Kim Y-M, Zink EM, Casey CP, Taylor BC, Lane CJ, et al. Human gut microbiota from autism spectrum disorder promote behavioral symptoms in mice. Cell. 2019;177(6):1600–1618.e17. doi:10.1016/j.cell.2019.05.004.
  • Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027–1031. doi:10.1038/nature05414.
  • Derrien M, Belzer C, de Vos WM. Akkermansia muciniphila and its role in regulating host functions. Microb Pathog. 2017;106:171–181. doi:10.1016/j.micpath.2016.02.005.
  • Cani PD, de Vos WM. Next-generation beneficial microbes: the case of akkermansia muciniphila. Front Microbiol. 2017;8:1765. doi:10.3389/fmicb.2017.01765.
  • Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S, Falony G, Raes J, Maiter D, Delzenne NM, et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat Med. 2019;25(7):1096–1103. doi:10.1038/s41591-019-0495-2.
  • Bárcena C, Valdés-Mas R, Mayoral P, Garabaya C, Durand S, Rodríguez F, Fernández-García MT, Salazar N, Nogacka AM, Garatachea N, et al. Healthspan and lifespan extension by fecal microbiota transplantation into progeroid mice. Nat Med. 2019;25(8):1234–1242. doi:10.1038/s41591-019-0504-5.
  • Blacher E, Bashiardes S, Shapiro H, Rothschild D, Mor U, Dori-Bachash M, Kleimeyer C, Moresi C, Harnik Y, Zur M, et al. Potential roles of gut microbiome and metabolites in modulating ALS in mice. Nature. 2019;572(7770):474–480. doi:10.1038/s41586-019-1443-5.
  • Wang L, Tang L, Feng Y, Zhao S, Han M, Zhang C, Yuan G, Zhu J, Cao S, Wu Q, et al. A purified membrane protein from Akkermansia muciniphila or the pasteurised bacterium blunts colitis associated tumourigenesis by modulation of CD8 + T cells in mice. Gut. 2020 Mar 13:gutjnl-2019-320105. doi:10.1136/gutjnl-2019-320105.
  • Sistigu A, Viaud S, Chaput N, Bracci L, Proietti E, Zitvogel L. Immunomodulatory effects of cyclophosphamide and implementations for vaccine design. Semin Immunopathol. 2011;33(4):369–383. doi:10.1007/s00281-011-0245-0.
  • Rong Y, Dong Z, Hong Z, Jin Y, Zhang W, Zhang B, Mao W, Kong H, Wang C, Yang B, et al. Reactivity toward Bifidobacterium longum and Enterococcus hirae demonstrate robust CD8+ T cell response and better prognosis in HBV-related hepatocellular carcinoma. Exp Cell Res. 2017;358(2):352–359. doi:10.1016/j.yexcr.2017.07.009.
  • Stevenson A, Panzica A, Holt A, Laute Caly D, Ettore A, Delday M, Hennessy E, Cowie P, Pradhan M, Jeffery I, et al. Host-microbe interactions mediating antitumorigenic effects of MRX0518, a gut microbiota-derived bacterial strain, in breast, renal and lung carcinoma. JCO. 2018;36(15_suppl):e15006. doi:10.1200/JCO.2018.36.15_suppl.e15006.
  • Lauté-Caly DL, Raftis EJ, Cowie P, Hennessy E, Holt A, Panzica DA, Sparre C, Minter B, Stroobach E, Mulder IE. The flagellin of candidate live biotherapeutic Enterococcus gallinarum MRx0518 is a potent immunostimulant. Sci Rep. 2019;9(1):801. doi:10.1038/s41598-018-36926-8.
  • Bessell CA, Isser A, Havel JJ, Lee S, Bell DR, Hickey JW, Chaisawangwong W, Bieler JG, Srivastava R, Kuo F, et al. Commensal bacteria stimulate antitumor responses via T cell cross-reactivity. JCI Insight. 2020;5(8):8. doi:10.1172/jci.insight.135597.
  • Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell. 2005;122(1):107–118. doi:10.1016/j.cell.2005.05.007.
  • Dasgupta S, Erturk-Hasdemir D, Ochoa-Reparaz J, Reinecker H-C, Kasper DL. Plasmacytoid dendritic cells mediate anti-inflammatory responses to a gut commensal molecule via both innate and adaptive mechanisms. Cell Host Microbe. 2014;15(4):413–423. doi:10.1016/j.chom.2014.03.006.
  • Stingele F, Corthésy B, Kusy N, Porcelli SA, Kasper DL, Tzianabos AO. Zwitterionic polysaccharides stimulate T cells with no preferential V beta usage and promote anergy, resulting in protection against experimental abscess formation. J Immunol. 2004;172(3):1483–1490. doi:10.4049/jimmunol.172.3.1483.
  • Tanoue T, Morita S, Plichta DR, Skelly AN, Suda W, Sugiura Y, Narushima S, Vlamakis H, Motoo I, Sugita K, et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature. 2019;565(7741):600–605. doi:10.1038/s41586-019-0878-z.
  • Cutting SM, Ricca E. Bacterial spore-formers: friends and foes. FEMS Microbiol Lett. 2014;358(2):107–109. doi:10.1111/1574-6968.12572.
  • Hutchison EA, Miller DA, Angert ER. Sporulation in bacteria: beyond the standard model. Microbiol Spectr. 2014;2(5): doi:10.1128/microbiolspec.TBS-0013-2012.
  • Jayaraman L, Sceneay J, Srinivasan S, Halley K, Bist M, Cieciuch K, Marnellos G, Desjardins C, Wortman J, Henn M, et al. Abstract B063: leveraging gut microbiota networks to impact tumor immunotherapy. Cancer Immunol Res. 2019;7(2 Supplement):B063. doi:10.1158/2326-6074.CRICIMTEATIAACR18-B063.