The Gut Microbiota and Stem Cell Transplantation
Perhaps one of the earliest demonstrations of the role of the gut microbiota in response and toxicity to cancer therapy was in the setting of allogeneic stem cell transplant (allo-HSCT) for hematologic malignancies. Dysbiosis and impaired systemic immunity is fairly common in these patients, as they are often treated with concurrent therapies that significantly alter immunity and composition of the gut microbiota—including immunosuppressants, broad-spectrum antibiotics, and even total body irradiation (
,
). Several investigators hypothesized that dysbiosis could be associated with altered responses and potentially with toxicity to therapy. Analyses of longitudinal fecal samples demonstrated a disruption of the existing state of equilibrium of the gut microbiota post-HSCT, with a loss of bacterial diversity and stability, and dominance of
Enterococcus,
Streptococcus, and various
Proteobacteria (
,
). Importantly, health-promoting bacteria such as
Faecalibacteriumand
Ruminococcus were reduced (
).
Dysbiosis in the setting of HSCT has also been associated with differences in long-term survival, with patients having a lower diversity of microbiota in their gut at the time of HSCT having shortened overall survival and higher mortality rates (specifically transplant-related mortality), compared with those with a high diversity of gut microbiota (
). Further confirmation of this finding was reported when low levels of 3-indoxyl sulfate in the urine, a by-product of L-tryptophan metabolism by commensal microbiota and a marker for bacterial dysbiosis, was found to be associated with worse overall survival following allo-HSCT (
). In addition to diversity, compositional differences in the gut microbiota have also been studied in response and survival after HSCT, with a higher abundance of bacteria within the genus
Blautiaassociated with improved overall survival (
) and a higher abundance of
Eubacterium limosum associated with a reduced risk of relapse (
).
In addition to the relationship to response and survival, the influence of the gut microbiota has also been studied in the context of toxicity to HSCT therapies—particularly with regard to graft-versus-host disease (GVHD). GVHD is characterized by the vigorous activation of immune-competent donor immune cells (mostly T cells) and causes significant damage to a variety of organs including the skin, liver, and gut, as well as sites of hematopoiesis (
). GVHD susceptibility varies with the type and extent of conditioning regimen, the degree of HLA mismatch, and the activation status of donor cells. Severe acute GVHD has a long-term survival probability of less than 5% (
) and chronic GVHD is also associated with significant morbidity and mortality.
The onset of acute GVHD is associated with significant shifts in the composition of the microbiota, with a loss of overall diversity and reduction of health-promoting obligate anaerobes such as
Faecalibacterium, Ruminococcus, Lactobacillus, and
Blautia,and an enrichment of
Enterococcus and
Clostridiales (
,
,
,
). A high abundance of the genus
Blautia of the Clostridia class was found to be associated with reduced GVHD lethality in two independent cohorts of patients undergoing treatment with allo-HSCT (
). The gut microbiota may also be affected by antibiotic treatment for infectious complications during HSCT, with differences in GVHD-associated mortality seen with different antibiotic regimens (
).
Modulation of the gut microbiota to abrogate toxicity has been studied in pre-clinical models with mixed results. The administration of the probiotic
Lactobacillus rhamnosus GG alone or in combination with Ciprofloxacin before and during transplantation in mice was associated with reduced rates of GVHD and improved overall survival (
). In this work, the authors hypothesized that probiotic supplementation contributed to the preservation of gut mucosal integrity, as surveying the mLNs revealed an absence of enteric pathogens (
). However, results in human cohorts have been more heterogeneous, with early studies demonstrating reduced rates of GVHD in patients treated with broad-spectrum antibiotics prior to HSCT (
) and more recent studies demonstrating detrimental effects of antibiotic use with higher rates of GVHD (
,
). More refined strategies to modulate the gut microbiota to reduce the risk of GVHD are now being tested in clinical trials including dietary modifications and fecal microbiome transplant (FMT) (
NCT03359980,
NCT03148743,
NCT03214289, and
NCT02763033). These trials are based on the central hypothesis that re-shaping the intestinal microbiota to its pre-treatment eubiotic state would lessen the risk of subsequent GVHD development, and are primarily exploratory in nature, seeking to assess the safety and feasibility of such modalities. Nevertheless, these trials will serve as a foundation, and additional trials will be implemented based on insights gained.
Gut Microbiota and Immunotherapy
Although stem cell transplant may be considered one of the earliest effective forms of cancer immunotherapy, there are now a host of novel immunotherapeutic approaches, and, not unsurprisingly, these are similarly affected by the gut microbiota. Important initial insights came from murine models (
,
), and many of these findings have now been validated in patient cohorts treated with immunotherapy, specifically immune checkpoint blockade.
Over a decade ago, investigators from the National Cancer Institute demonstrated that administration of antibiotics significantly abrogated anti-tumor activity in a murine model of adoptive cell therapy for melanoma. The authors posed that the proliferation of transferred T cells in the tumor was augmented by the translocation of the gut microbiota to the mLNs associated with total body irradiation, which was used as a preparative regimen. They surmised that the translocation of gut bacteria helped to prime an immune response via TLR4 signaling (
). This notion is supported by studies demonstrating impaired responses to intra-tumoral injection of TLR agonists in GF or antibiotic-treated mice. In this setting, tumor-associated myeloid cells are primed by commensal gut bacteria (via TLR4 signaling) for the production of tumor necrosis factor and other inflammatory cytokines that mediate the anti-tumor effect of these agents (
).
Although not traditionally considered immunotherapy, effective treatment with conventional chemotherapy is also dependent on intact immune responses, thus substantiating the notion that the gut microbiota could shape responses to these forms of therapy as well. This has certainly been demonstrated in platinum-based chemotherapies and cyclophosphamide therapy. During treatment with cyclophosphamide, translocation of commensal bacteria (specifically Gram-positive organisms such as
Lactobacillus johnsonii and
Enterococcus hirae) into mLNs can potentially facilitate robust Th17 responses in the spleen and the induction of memory Th1 responses. Immune responses to cyclophosphamide have also been shown to be dependent on MyD88 and TLR signaling—suggesting that commensal microorganisms may play a role. Indeed, the effects of cyclophosphamide and other chemotherapy regimens were abrogated in GF or antibiotic-treated mice and were differentially affected by the presence of particular bacterial species (
,
).
Importantly, the impact of the gut microbiota has also been studied in the setting of treatment with immune checkpoint inhibitors, which target immunomodulatory molecules on the surface of T cells (or their ligands) to enhance anti-tumor immune responses. Despite the enthusiasm for treatment with these agents, a significant proportion of patients do not experience objective responses and, when responses do occur, may not be durable. Tremendous efforts have focused on identifying predictors of the response to immune checkpoint blockade as well as strategies to overcome therapeutic resistance (
,
). Emerging evidence suggests that the gut microbiota may play a significant role in modulating responses to these therapies.
The impact of the gut microbiota on response to immune checkpoint blockade was first studied in mouse models, with landmark publications in
Science in 2015 demonstrating that the composition of the gut microbiota could influence the response to immune checkpoint inhibitors targeting the cytotoxic T lymphocyte antigen 4 (CTLA-4) and the programmed death receptor 1 (PD-1) (
,
). In the case of CTLA-4 blockade, notable changes in the abundance of gut microbiota in mice were seen following anti-CTLA-4 therapy, with a relative increase in
Bacteroidales and
Burkholderialesand a decrease in
Clostridiales. The efficacy of anti-CTLA-4 therapy was markedly reduced in GF mice and SPF mice with broad-spectrum antibiotics. Furthermore, oral feeding with
Bacteroides fragilis in combination with either
Bacteroides thetaiotaomicron or
Burkholderia cepacia augmented the action of anti-CTLA-4 therapy by eliciting a Th1 response in the lymph nodes and facilitating the maturation of intra-tumoral DCs. The translational impact of these findings was demonstrated when FMT from patients having dominant
Bacteroides species in their gut resulted in improved tumor control compared with FMT from patients with distinct
Bacteroides or
Prevotella species (
). These findings were complemented by parallel studies in the context of treatment with PD-1 blockade, demonstrating significant differences in tumor outgrowth in genetically similar mice with differing gut microbiomes purchased from two separate vendors. Therapeutic responses also differed in these mice, and beneficial effects from mice with a more “favorable” microbiota could be transplanted to other mice using FMT or co-housing. Profiling of the gut microbiome revealed an over-representation of
Bifidobacterium species in mice with delayed tumor outgrowth and favorable responses to PD-1-based therapy. Furthermore, supplementation with an oral probiotic containing
Bifidobacterium restored anti-tumor efficacy of PD-L1-blockade in mice with an “unfavorable” gut microbiota, which primarily occurred through enhanced DC maturation resulting in increased tumor-specific CD8
+ T cell activity (
).
These studies were further supplemented by multiple studies published in the past several months demonstrating a role for the gut microbiota in patients on immune checkpoint blockade (
,
,
,
,
). Several provocative findings were reported substantiating the role of the gut microbiota in shaping responses to therapy. First, the impact of antibiotic use on response to immune checkpoint blockade was shown in a large cohort of patients with non-small-cell lung cancer, renal cell carcinoma, or urothelial cancer. Patients treated with antibiotics for routine indications shortly before, during, or shortly after treatment with anti-PD-1/PD-L1 mAB had significantly lower progression-free survival and overall survival rates compared with patients who had not received antibiotics. This suggests that disrupting the gut microbiota (via antibiotic use) could potentially impair anti-tumor immune responses as well as response to immune checkpoint blockade (
). The group also studied the gut microbiota directly by performing whole metagenomic sequencing in fecal samples from these patients, demonstrating that responders to PD-1 blockade had differential composition of gut bacteria, including specific genera highlighted by the group as being enriched in responding patients (
Akkermansia and
Alistipes). FMT was performed in GF and SPF mice using a stool sample from either responder (R) or non-responder (NR) patients prior to treatment with PD-1 blockade, demonstrating enhanced responses in the setting of R-FMT. In these studies, the efficacy of anti-PD-1 in GF mice receiving NR-FMT could be restored by administration of
Akkermansia muciniphila alone or in combination with
E. hirae, where administration of
A. muciniphilia was associated with increased intra-tumoral immune infiltrates, mediated by the recruitment of CCR9
+CXCR3
+CD4
+ T cells into the tumor bed and an increased ratio of CD4
+ T cells to CD4
+FoxP3
+ T cells (Tregs) in response to PD-1 blockade (
). These findings were corroborated in two additional papers published in the same issue of
Science describing the impact of gut microbiota on responses to anti-PD-1 therapy in patients with metastatic melanoma.
The study by Gopalakrishnan et al. revealed that patients who responded to anti-PD-1 therapy had a significantly higher diversity of bacteria in their gut microbiota as well as a higher relative abundance of
Clostridiales,
Ruminococcaceae, and
Faecalibacterium (
,
). In contrast, NR had significantly lower diversity of gut bacteria and higher abundance of Bacteroidales. Importantly, comparing the composition of bacteria in the gut with immune profiling in the tumor microenvironment revealed that patients with a favorable gut microbiota had increased expression of cytolytic T cell markers and antigen processing and presentation compared with patients with unfavorable gut microbiota. Mechanistic studies were performed in GF mice with FMT from R versus NR, recapitulating findings in parallel published studies that mice receiving FMT from R had significantly delayed tumor outgrowth and enhanced responses to treatment with immune checkpoint blockade (
). Another cohort of patients with metastatic melanoma studied by
also demonstrated significant differences in response to treatment with immune checkpoint blockade based on profiles within the gut microbiota. Specifically, the group found that patients who responded to anti-PD-1 therapy had enrichment of
Bifidobacterium longum, Collinsella aerofaciens, and
Enterococcus faecium in baseline fecal samples. Transfer of stool samples from patients to GF mice in this study also successfully recapitulated the phenotype, with mice that received R stool growing tumors at a slower rate and having markedly improved efficacy to anti-PD-L1 immunotherapy compared with mice that received NR stool. These effects were mediated by increased densities of CD8
+T cells and reduced FoxP3
+CD4
+ Tregs in the tumor microenvironment (
).
Although immune checkpoint blockade agents have been a qualified success in the treatment of various malignancies resulting in sustained responses, a significant proportion of patients continue to experience treatment-limiting toxicity with anti-PD-1 (16%), anti-CTLA-4 (27%), and combination therapies (65%) (
). Approximately one-third of all patients undergoing anti-CTLA-4 therapy develop intestinal inflammation due to mucosal immune dysregulation (
,
). Efforts to characterize gut microbiota that contribute to toxicity to immune checkpoint blockade are underway. Pre-clinical models have demonstrated an improvement in toxicity scores in anti-CTLA-4-treated mice with oral gavage of
B. fragilis and
B. cepacia (
). The influence of the gut microbiota on toxicity has also been studied in human cohorts (
,
,
) (
Figure 2). Taxonomical and functional differences have been reported in anti-CTLA-4-treated melanoma patients who were colitis-free (with enrichment of Bacteroidetes and abundance of genetic pathways involved in polyamine transport and B vitamin synthesis) as opposed to those who developed colitis (
). This may be related to the known influence of these bacteria in Treg differentiation (
,
). Additional cohorts have also been studied, showing that patients with a higher abundance of
Faecalibacterium prausnitzii and other related Firmicutes and low abundance of Bacteroidetes had a higher risk of colitis on anti-CTLA-4 therapy (
,
). The group also reported that patients with colitis had increased expression of ICOS on the surface of effector CD4
+ T cells and low levels of Tregs and systemic inflammatory proteins such as IL-6, IL-8, and sCD25 in the blood at baseline, which may be related to the compositional differences in the microbiome (
).
Based on the available literature, there are clearly bacterial taxa that are associated with response and toxicity—with some overlap in the bacterial signatures across the studies (
Figure 2). Bacterial taxa within the Ruminococaceae family of the Firmicutes phylum (such as
F. prausnitzii) have been associated with both response and toxicity to immune checkpoint blockade across studies (
,
,
) (
Figure 2). Conversely, bacterial taxa within the Bacteroidales order of the Bacteroidetes phylum have been associated with a lack of response to immune checkpoint blockade, while a higher abundance of these taxa within the gut are also generally associated with a lower incidence of toxicity (
,
,
,
). However, at lower levels of taxonomy, these generalizations do not apply, as some taxa within Firmicutes have been associated with a lack of response (
Roseburia, Streptococcus) (
,
) and some taxa within Bacteroidetes have been associated with response (
Alistipes, Porphyromonas pasteri, and
C. aerofaciens) (
,
,
). Importantly, taxa outside of Firmicutes/Bacteroidetes have also been associated with response (such as
A. muciniphila, B. longum, Bifidobacterium adolescentis, and
C. aerofaciens) and non-response (such as
Actinomyces viscosus and
Garnderella vaginalis) (
,
,
,
). Overall, there is not a great deal of overlap between specific bacterial taxa associated with response across these published studies, although several taxa that are implicated with either response or toxicity are phylogenetically related (such as members of the Ruminococcaceae and Lachnospiraceae families and the Bacteroidales order) (
Figure 2). Importantly, differences may be related to several different factors—including differences in techniques used to analyze samples and reference databases used for analysis, which varied widely across the studies (
,
,
,
,
,
,
,
)—suggesting the importance of developing standardized approaches for microbiome analysis. Geographical influences also may exist, as these studies were performed in centers at different locations around the world. In line with this, dietary and lifestyle factors may also account for some of the differences observed. Nevertheless, the impact of the gut microbiota on therapeutic response is uncontested, and these data provide strong evidence that the gut microbiota can modulate anti-tumor immune responses and responses to immune checkpoint blockade.