Several landmark preclinical studies have shown an association between the gut microbiota and the effectiveness of immunotherapy for cancer. These studies have sparked clinical trials aimed at modulating the gut microbiota in order to improve clinical response rates to immunotherapy. Despite this, the mechanisms through which the gut microbiota influences the effectiveness of immunotherapy are still incompletely characterized. Preclinical and preliminary clinical findings from numerous types of gut microbiota modulation studies, including fecal transplantation, probiotics, consortia, and diet, demonstrate that favorable microbiota modulation is associated with increased intratumoral infiltration of CD8+ effector T cells. This CD8+ T-cell infiltration is often associated with enhanced intratumoral activity of T-helper type 1 cells and dendritic cells and a lower density of immunosuppressive cells. Herein, we discuss how gut microbiota may affect the activity of immune cells by at least three interlacing mechanisms: activation of pattern recognition receptors, molecular mimicry, and impact of metabolites. We also discuss the therapeutic potential and limitations of the different gut microbiota modulation techniques and their putative mechanisms of immune activation.

A series of landmark studies have been published demonstrating associations between the composition of the gut microbiota and clinical response to cancer immunotherapy (1–3). In addition, the use of antibiotics by patients with cancer prior to starting immune checkpoint blockade (ICB) immunotherapy has been associated with adverse prognosis (3, 4), and strategies to manipulate gut microbiota to improve immunotherapy responses are currently being tested in clinical trials. Although the notion that gut microbiota can influence immunity is well established (5), the specific mechanisms through which the microbiota affects immunity and responses to cancer immunotherapy are still being elucidated. Here, we review the current evidence on how gut microbiota affects antitumoral immune activity. We also discuss the impact of gut microbiota modulation strategies on immunity and immunotherapy response with a summary of ongoing efforts.

The antitumor immune response is highly complex and involves multiple key players (6). Antigen-presenting cells can infiltrate tumors and present antigens to lymphocytes in the tumor microenvironment (7, 8) or in tumor-draining lymph nodes. Antigen recognition leads to activation of CD4+ T-helper type 1 (Th1) cells and CD8+ T cells and also counters activation of CD4+ T regulatory cells (Treg) and myeloid-derived suppressor cells (MDSC). Evidence that gut microbiota can affect the differentiation and activation of each of these cell types comes from studies in germ-free (GF) mice and mice treated with antibiotics, as discussed below. Some of the mechanisms through which gut microbiota can induce immune modulation include activation of pattern recognition receptors (PRR), antigen-specific activation, and metabolite-derived modulation (Fig. 1).

Figure 1.

Mechanisms of immune modulation by the gut microbiota. The gut microbiota may affect immune cells playing key roles in antitumoral immunity. The microbiota may affect maturation processes, either in the bone marrow [for dendritic cells (DC)] or in the thymus (for T cells). Microbiota may also affect the activity of mature immune cells within the tumor microenvironment or in the surrounding lymph nodes. The microbiota-derived effect is mediated by three potential pathways: activation of pathogen recognition receptors such as TLRs and NODs; molecular mimicry initiating immune responses against cancer antigens that are similar to bacterial ones; and metabolic modulation using vitamins, short-chain fatty acids, bile salt, or amino acids that promote favorable intracellular processes among host cells. LN, lymph node; NOD, nucleotide-binding oligomerization domain-containing proteins; TCR, T-cell receptor; TLR, Toll-like receptor.

Figure 1.

Mechanisms of immune modulation by the gut microbiota. The gut microbiota may affect immune cells playing key roles in antitumoral immunity. The microbiota may affect maturation processes, either in the bone marrow [for dendritic cells (DC)] or in the thymus (for T cells). Microbiota may also affect the activity of mature immune cells within the tumor microenvironment or in the surrounding lymph nodes. The microbiota-derived effect is mediated by three potential pathways: activation of pathogen recognition receptors such as TLRs and NODs; molecular mimicry initiating immune responses against cancer antigens that are similar to bacterial ones; and metabolic modulation using vitamins, short-chain fatty acids, bile salt, or amino acids that promote favorable intracellular processes among host cells. LN, lymph node; NOD, nucleotide-binding oligomerization domain-containing proteins; TCR, T-cell receptor; TLR, Toll-like receptor.

Close modal

Activation of PRRs

GF mice and mice treated with broad-spectrum antibiotics have a reduced frequency of hematopoietic stem and progenitor cells in the bone marrow (9). Interestingly, immune progenitor activity can be restored in GF mice by administration of lipopolysaccharide (LPS), sterile-filtered and boiled serum, or autoclaved cecal content from specific pathogen–free mice (10). The bacterial particles activate PRRs such as MyD88-dependent Toll-like receptors (TLR) and nucleotide-binding oligomerization domain-containing proteins (NOD; ref. 11). These PRRs are expressed by mesenchymal stromal cells in the bone marrow, and their activity is STAT1 dependent. Upon PRR activation, the stromal cells secrete growth factors that support hematopoietic stem and progenitor cell proliferation and thus enhance hematopoiesis.

GF mice have also reduced CD8+ T-cell thymic maturation, and administration of bacterial peptidoglycans, which are sensed by thymocyte NOD receptors, promotes CD8+ T-cell maturation in these animals (12). Similar findings were reported in a study in which female mice were treated with antibiotics during the final stages of pregnancy and throughout the lactation period. Infant mice from these litters were highly susceptible to viral infections, and immune-profiling studies demonstrated that CD8+ T cells were unable to sustain IFNγ production upon activation due to impaired downstream signaling of T-cell receptors (TCR). Oral administration of LPS to the infant mice was associated with increased signaling of TLR4–MyD88 in CD8+ T cells, which restored their ability to produce IFNγ compared with control mice and prolonged survival in the context of an induced systemic viral infection (13).

The gut microbiota is composed not only of bacteria, but also of viruses, protozoa, and fungi that may positively or negatively affect immunity and antitumor immune responses. These are less well studied in the context of shaping antitumor immunity, as many of the early studies published on this subject utilized 16S rRNA gene sequencing, which captures only bacterial signatures, rather than metagenomic sequencing, which captures additional signature from viruses, fungi, and protists. Nonetheless, some evidence exists regarding the impact of nonbacterial microbiota on antitumor immunity, with intratumoral fungi promoting pancreatic cancer progression (14). This relationship is further highlighted by a study using mice deficient in caspase recruitment domain family member 9 (CARD9), which is an adaptor protein in macrophages that plays a key role in antifungal immune activity. CARD9−/− mice were found to have abnormally high relative abundance of fungi in their gut microbiota compared with wild-type mice, particularly Candida tropicalis. In addition, the CARD9−/− mice had a higher colon tumor burden (15), and the tumors were characterized by upregulation of IL6, IL10, and TGFß. Fecal microbiota transplant (FMT) from CARD9−/− mice into GF mice caused a similar pattern of tumorigenesis with a significant expansion in the number of MDSCs in the colon.

Molecular mimicry

In addition to activating PRRs, gut microbiota may induce antigen-specific immune responses. The mechanism for this antigen-specific activation is a cross-reaction between a bacterial antigen and a human protein containing similar antigenic sequences. This phenomenon is known as molecular mimicry. In a murine model of type 1 diabetes mellitus, CD8+ T cells harboring TCRs specific for islet-specific glucose-6-phosphatase catalytic subunit–related protein (IGRP) can be activated by peptides from oral and gut bacteria that have proteins that share strong homology with IGRP (16). Certain Bacteroides species contain peptides that mimic myosin heavy chain 6 protein, which is expressed by cardiac myocytes. Bacteroides thetaiotaomicron–activated Th1 and Th17 cells have been reported to have a key role in lethal inflammatory cardiomyopathy in mice, and this was abrogated in GF mice and could be mitigated by antibiotic therapy (17).

Antigenic mimicry may also indirectly regulate maturation of immune cells, as plasmacytoid dendritic cells (DC) activated by the polysaccharide A component of Bacteroides fragilis were shown to migrate from the mouse colon to the thymus and promote maturation of PLZF+ lymphocytes (18). Similarly, immune responses associated with antigenic mimicry have recently been demonstrated in preclinical models of cancer. The tail length tape measure protein (TMP) of a bacteriophage found in Enterococcus hirae contains MHC class I binding epitopes, and mice bearing the E. hirae bacteriophage developed anti-TMP T-cell clones that could cross-react against melanoma cells (19). In preclinical models, administration of TMP-containing enterococci enhanced antitumoral T-cell activity and overall effectiveness of anti–PD-1 ICB. T-cell clones targeting the Bifidobacterium breve antigen SVYRYYGL also have been shown to cross-react against melanoma cells (20).

Preliminary evidence for microbiota-derived molecular mimicry has also been reported in humans. The presence of TMP-containing enterococci phage in patient stool was associated with improved clinical response to anti–PD-1 therapy in patients with kidney and lung cancer (19). In addition, studies in pancreatic cancer comparing long-term with short-term survivors demonstrated that in long-term survivors, the tumor microenvironment is characterized by a higher density of neoantigens that are similar to microbially derived epitopes and an enhanced CD8+ T-cell infiltration (21, 22). Importantly, FMT using fecal samples from long-term survivors of pancreatic cancer into GF mice conferred enhanced antitumor immunity and tumor control compared with FMT using fecal samples from short-term survivors and FMT from healthy controls (22).

Immunomodulation via microbially derived metabolites

One of the most provocative mechanisms by which gut microbiota may mediate immunomodulation is via production of metabolites. GF mice have underdeveloped lymphoid organs throughout their body, even in remote, nongastrointestinal sites of the body (23). This is at least partially related to the influence of microbes on DCs, as investigators have demonstrated that intravenous administration of microbiota-activated CD103+ DCs into GF mice restores the cellularity and structure of the peripheral lymph nodes via a vitamin A–dependent process (24).

The gut microbiota can generate metabolites from naturally occurring compounds in the gut. For example, a riboflavin (vitamin B2) derivate called 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil can rapidly cross the gut mucosa and enter the bloodstream, through which it can travel to the thymus, where it induces the expansion of mucosal-associated invariant T cells, which are lymphocytes that are absent in GF mice (25). Bile acids are also potential sources of microbially derived immunomodulators because gram-positive bacteria in the gut metabolize primary bile acids into secondary ones with different properties. Reabsorption of primary bile acids in the gut increases expression of CXCL16 in liver endothelial cells, which attracts CXCR6+ natural killer T (NKT) cells. Liver CXCR6+ NKT accumulation can inhibit tumor growth. Such accumulation was demonstrated to be bile acid–dependent in mice, as antibiotic depletion of gut gram-positive bacteria increased the number of liver CXCR6+ NKT cells, whereas feeding with secondary bile acids decreased this cell population (26).

Certain foods have been studied for their potential role in improving immunity. Plants like parsley and berries contain flavonoids, which can be degraded by the gut microbiota to desaminotyrosine (DAT). DAT has been demonstrated to protect mice from influenza virus infection by enhancing type I IFN signaling (27). Dietary fiber is one of the most studied immunomodulatory foods. Dietary fibers cannot be directly digested by humans for use as an energy source, rather they are fermented by certain gut bacteria into short-chain fatty acids (SCFA), which can then be used by as an energy source. Higher levels of fecal SCFA have been associated with improved clinical response to anti–PD-1 ICB in patients with advanced cancer (28). A high fiber diet and increased SCFA production may also be associated with increased CD103+ DC production in the bone marrow, increased phagocytic capacity of DCs in the lungs and gut, and dampened DC-mediated Th2 responses (29, 30). Bachem and colleagues reported that antigen-activated CD8+ T cells that were transferred into GF mice failed to transition into long-lived memory cells (31). The SCFA butyrate enhanced the memory potential of activated CD8+ T cells by promoting oxidative phosphorylation and mitochondrial function in the T cells. This finding may have relevance in cancer immunotherapy, as mitochondrial metabolism and oxidative phosphorylation proteomic pathways are enriched in tumor samples of patients with metastatic melanoma who respond to anti–PD-1 ICB and adoptive cell therapies (32), although this needs to be further evaluated.

Another example of the tangentiality between microbiota functional activity, diet, and the tumor microenvironment is the essential amino acid tryptophan. Tryptophan can be metabolized by the gut microbiota to serotonin, kynurenine, or indoles, which are aryl hydrocarbon receptor (AhR) agonists (33). Indoles can cross the gut–blood barrier, and then, after traveling through the bloodstream, cross the blood–brain barrier to activate astrocytes and induce immunosuppression in the brain. In a murine experimental autoimmune encephalomyelitis model, central nervous system inflammation was decreased when mice were treated with a tryptophan-rich diet but increased when the mice were treated with antibiotics (34). This may be clinically relevant as patients with multiple sclerosis have low circulating levels of tryptophan-derived metabolites (34). Kynurenine, which is also an AhR agonist, can suppress NKT cell and DC activity, arresting T-cell proliferation and inducing T-cell apoptosis (35). It is a potent immunosuppressor, and tryptophan catabolism to kynurenine by indoleamine 2,3-dioxygenase (IDO)-1 is one of the reasons that IDO-1 is known as a key immune checkpoint in the tumor microenvironment and is a therapeutic target in ongoing cancer clinical trials (36).

Together, these data serve as an example of the delicate equilibrium between the gut microbiota and host immune activity. However, in cancer immunity, systemic effects on the immune system must be taken in the context of those effects on the tumor. A microbiota-derived metabolite, gallic acid (a type of phenolic acid), has been demonstrated to modify the tumor-suppressor activity of p53 within tumor cells and promote tumor growth (37). Hence, modulation of the microbiota may represent a tractable strategy to improve antitumor immunity and responses to immunotherapy by acting in multiple ways, on both immune and tumor cells.

Based on data from preclinical studies and human cohorts suggesting that gut microbiota affects responses to cancer therapy, numerous trials are now underway to test different approaches to modulating gut microbiota. Each of these approaches differs substantially, and the mechanisms through which they may affect antitumor immune responses must be carefully considered, but much is still unknown.

Fecal microbial transplantation

FMT involves the transfer of fecal material from an identified donor to a recipient. Samples are prepared by sieve or filtration with reconstitution in a liquid form so it can be either infused via endoscopy or packaged and stored in stool capsules for oral ingestion (38). Due to these minimal preparation steps, most of the donor fecal content, including bacteria, viruses, fungi, microbial particles, and metabolites, are preserved and can be transferred to the recipient. FMT has been used for decades as a therapy for recurrent or refractory Clostridioides difficile colitis. However, FMT is just beginning to be studied in the context of cancer. Several preclinical studies have shown that FMT using feces from patients with cancer who have responded to immunotherapy enhances the effectiveness of both anti–PD-1 and anti–CTLA-4 therapies in GF mice and mice treated with antibiotics, compared with FMT using nonresponder feces (1–3, 39). These FMT-enhanced antitumor responses were associated with, and probably mediated by, an intratumoral immune microenvironment in which there is increased infiltration of CXCR3+CD4+ (Th1-related) T cells and IFNγ-producing cytotoxic CD8+ T cells and decreased infiltration of RORγ+ Th17 cells and CD11b+CD11c+ suppressive myeloid cells.

One of the most intriguing characteristics of FMT is its potential dual utility in patients with cancer; although it can boost antitumor immune responses, it can also suppress immunotherapy-related colitis (40). A reduction in CD8+ T-cell density and an increase of FoxP3+ Treg within the colonic mucosa have been seen from pre- to posttreatment colonic biopsies after patients with cancer were given healthy-donor FMT. This report only had data from a few patients, and therefore, no conclusions could be made regarding putative microbial taxa that may have contributed to therapeutic response. However, the therapeutic benefit may have been in part related to the transfer of a more diverse gut microbiome with functional redundancy. Consistent with this, other investigators have noted that low microbial diversity within the gut microbiome is associated with an increased rate of severe immune-related adverse events (irAE) and lack of response to treatment with combination anti–CTLA-4 and anti–PD-1 ICB (41).

To date, several clinical trials are assessing whether adding FMT to immunotherapy can improve outcomes for patients with various types of cancer (NCT03772899, NCT03341143, and NCT04521075). Preliminary results from one trial combining FMT and anti–PD-1 reinduction in patients with refractory metastatic melanoma demonstrated clinical responses that were associated with increased intratumoral infiltration of CD8+ T cells (42). The participants also did not develop irAEs, even though they had when previously treated with immunotherapy.

Probiotics and live biotherapeutics targeting single gut microbes

In addition to FMT, the use of probiotics and live biotherapeutics targeting specific gut microbes has also been studied in the context of treatment with ICB. Increased abundance of Akkermansia muciniphila in the gut microbiome of patients with non–small cell lung cancer and kidney cancer was shown to be associated with enhanced response to treatment with ICB. Transfer of this microbe into the gut microbiome in preclinical models of cancer increased production of IL12 by DCs, enhanced recruitment of CCR9+CXCR3+CD4+ T cells in epithelial tumors and lymph nodes, and improved efficacy of PD-1 ICB in treated mice (3). Other gut microbes have also been studied. Bifidobacterium has been found to be enriched in the gut microbiota of responding patients with metastatic melanoma being treated with anti–PD-1 ICB. Treatment with Bifidobacterium in preclinical models of cancer was associated with a higher density of MHC class IIhi DCs in melanoma tumors, increased tumor-specific CD8+ T cells in the tumor microenvironment, and enhanced responses to treatment with anti–PD-1 ICB (43). Such approaches have also been studied in the context of anti–CTLA-4 ICB in preclinical models of sarcomas and colon carcinoma. In this study, treatment with enteric B. fragilis was associated with improved responses to anti–CTLA-4, maturation and production of IL12 by DCs, and increased T-cell memory responses (39). Several human clinical trials are underway incorporating the administration of these types of approaches in patients with cancer who will subsequently be treated with anti–PD-1 ICB (NCT03595683 and NCT03637803). Data are currently not available on the safety and efficacy of such approaches, or on their ability to stimulate robust antitumor immunity. Nonetheless, this remains a potentially viable strategy and offers less complexity and perhaps an enhanced safety profile over treatment with FMT, as pathogens are easily transmittable via FMT (42).

Gut microbial consortia

A lab-produced consortium of microbes might, in theory, incorporate a probiotic-like high safety profile and an FMT-like improved functionally, as these small microbial communities can work together. Major efforts are underway to develop consortia of microbes that can be administered via the gastrointestinal tract to enhance the function of the gut microbiota and the immunotherapy response, with several trials underway or in development (NCT03817125 and NCT04208958). These consortia have been informed by human cohort studies and preclinical models (44, 45) and contain either consortia derived from donors with what are considered to be favorable gut microbiota signatures (NCT03817125) or consortia engineered based on results from studies involving transplantation of human fecal matter into mice (45). Notably, the consortia derived from FMT studies in preclinical models demonstrated that transfer of a consortia consisting of 11 bacterial strains into the gut of tumor-bearing mice was associated with a high density of IFNγ-producing CD8+ T cells, and enhanced efficacy of anti–PD-1 and anti–CTLA-4 ICB in mouse models of colon cancer and melanoma (45). Additional efforts are underway to reconstruct gut microbial consortia via culturomics and other approaches (46). However, potential complexities may exist with scalability and consistency of manufacturing of such products, as well as with regulatory aspects.

Diet and prebiotics

When contemplating strategies to modulate gut microbiota, it is important to consider other factors that affect gut microbiota, including diet and prebiotics (47). Oral intake of inosine, together with IL12 secretion by DCs, has been shown to enhance the effectiveness of anti–CTLA-4 ICB against different tumors implanted in mice by promoting antitumoral T-cell activity (48). Prebiotics are dietary compounds, such as fibers and inulin, that may support certain gut microbiota populations or modify their functionality. Administration of prebiotics has been studied across many disease types. Recent studies in preclinical cancer models suggest that administration of inulin is associated with enhanced antitumoral immune responses in melanoma (49). In these studies, mice who received treatment with prebiotics demonstrated higher intratumoral infiltration of effector IFNγ-producing CD4+ and CD8+ T cells, plasmacytoid DCs, and conventional CD8α+ DCs. Tumor-resident DCs isolated from mice treated with prebiotics expressed higher levels of MHC class I and MHC class II. Importantly, all of the effects of prebiotics on antitumor immunity and tumor growth were dependent on the gut microbiota, as GF mice failed to demonstrate these changes (49). A few clinical trials assessing the potential effect of dietary modifications and prebiotics in patients with metastatic cancer undergoing immunotherapy are currently underway (NCT04552418 and NCT04316520).

There is increasing interest in targeting gut microbiota to enhance immunity and immunotherapy response; however, optimal strategies to do so are incompletely understood. Critical insights are being gained into the mechanisms through which gut microbiota modulates immunity and will help refine strategies to target gut microbiota in cancer and other states of health and disease.

J.A. Wargo reports personal fees from Bristol-Myers Squibb, Ella Therapeutics, Peer View, Novartis, AstraZeneca, Merck, GlaxoSmithKline, and Physician Education Resource outside the submitted work and is a co-inventor on patents submitted by The University of Texas MD Anderson Cancer Center to the US Patent and Trademark Office on modulating the microbiome to enhance responses to immune checkpoint blockade (patent number PCT/US1/53717) and on another patent Targeting B Cells To Enhance Response To Immune Checkpoint Blockade UTSC.P1412US.P1 - MDA19-023. No disclosures were reported by the other authors.

J.A. Wargo is supported by the Melanoma Research Alliance (4022024), a Stand Up To Cancer Innovative Research Grant, Grant Number SU2C-AACR-IRG19-17; NIH (grant number R01 CA219896-01A1); MD Anderson Cancer Center's Melanoma Moon Shots Program; and MD Anderson's Program for Innovative Microbiome and Translational Research (PRIME-TR). Stand Up To Cancer is a division of the Entertainment Industry Foundation. The indicated Stand Up To Cancer grant is administered by the American Association for Cancer Research, the scientific partner of SU2C.

1.
Gopalakrishnan
V
,
Spencer
CN
,
Nezi
L
,
Reuben
A
,
Andrews
MC
,
Karpinets
TV
, et al
Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients
.
Science
2018
;
359
:
97
103
.
2.
Matson
V
,
Fessler
J
,
Bao
R
,
Chongsuwat
T
,
Zha
Y
,
Alegre
ML
, et al
The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients
.
Science
2018
;
359
:
104
8
.
3.
Routy
B
,
Le Chatelier
E
,
Derosa
L
,
Duong
CPM
,
Alou
MT
,
Daillere
R
, et al
Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors
.
Science
2018
;
359
:
91
7
.
4.
Hakozaki
T
,
Richard
C
,
Elkrief
A
,
Hosomi
Y
,
Benlaïfaoui
M
,
Mimpen
I
, et al
The gut microbiome associates with immune checkpoint inhibition outcomes in patients with advanced non–small cell lung cancer
.
Cancer Immunol Res
2020
;
8
:
1243
50
.
5.
Belkaid
Y
,
Hand
TW
. 
Role of the microbiota in immunity and inflammation
.
Cell
2014
;
157
:
121
41
.
6.
Gajewski
TF
,
Corrales
L
,
Williams
J
,
Horton
B
,
Sivan
A
,
Spranger
S
. 
Cancer immunotherapy targets based on understanding the t cell-inflamed versus non-t cell-inflamed tumor microenvironment
.
Adv Exp Med Biol
2017
;
1036
:
19
31
.
7.
Helmink
BA
,
Reddy
SM
,
Gao
J
,
Zhang
S
,
Basar
R
,
Thakur
R
, et al
B cells and tertiary lymphoid structures promote immunotherapy response
.
Nature
2020
;
577
:
549
55
.
8.
Petitprez
F
,
de Reyniès
A
,
Keung
EZ
,
Chen
TW
,
Sun
CM
,
Calderaro
J
, et al
B cells are associated with survival and immunotherapy response in sarcoma
.
Nature
2020
;
577
:
556
60
.
9.
Josefsdottir
KS
,
Baldridge
MT
,
Kadmon
CS
,
King
KY
. 
Antibiotics impair murine hematopoiesis by depleting the intestinal microbiota
.
Blood
2017
;
129
:
729
39
.
10.
Gorjifard
S
,
Goldszmid
RS
. 
Microbiota-myeloid cell crosstalk beyond the gut
.
J Leukoc Biol
2016
;
100
:
865
79
.
11.
Yan
H
,
Baldridge
MT
,
King
KY
. 
Hematopoiesis and the bacterial microbiome
.
Blood
2018
;
132
:
559
64
.
12.
Martinic
MM
,
Caminschi
I
,
O'Keeffe
M
,
Thinnes
TC
,
Grumont
R
,
Gerondakis
S
, et al
The bacterial peptidoglycan-sensing molecules NOD1 and NOD2 promote CD8(+) thymocyte selection
.
J Immunol
2017
;
198
:
2649
60
.
13.
Gonzalez-Perez
G
,
Lamousé-Smith
ES
. 
Gastrointestinal microbiome dysbiosis in infant mice alters peripheral CD8(+) T cell receptor signaling
.
Front Immunol
2017
;
8
:
265
.
14.
Aykut
B
,
Pushalkar
S
,
Chen
R
,
Li
Q
,
Abengozar
R
,
Kim
JI
, et al
The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL
.
Nature
2019
;
574
:
264
7
.
15.
Wang
T
,
Fan
C
,
Yao
A
,
Xu
X
,
Zheng
G
,
You
Y
, et al
The adaptor protein CARD9 protects against colon cancer by restricting mycobiota-mediated expansion of myeloid-derived suppressor cells
.
Immunity
2018
;
49
:
504
14
.
16.
Tai
N
,
Peng
J
,
Liu
F
,
Gulden
E
,
Hu
Y
,
Zhang
X
, et al
Microbial antigen mimics activate diabetogenic CD8 T cells in NOD mice
.
J Exp Med
2016
;
213
:
2129
46
.
17.
Gil-Cruz
C
,
Perez-Shibayama
C
,
De Martin
A
,
Ronchi
F
,
van der Borght
K
,
Niederer
R
, et al
Microbiota-derived peptide mimics drive lethal inflammatory cardiomyopathy
.
Science
2019
;
366
:
881
6
.
18.
Ennamorati
M
,
Vasudevan
C
,
Clerkin
K
,
Halvorsen
S
,
Verma
S
,
Ibrahim
S
, et al
Intestinal microbes influence development of thymic lymphocytes in early life
.
Proc Natl Acad Sci
2020
;
117
:
2570
8
.
19.
Fluckiger
A
,
Daillère
R
,
Sassi
M
,
Sixt
BS
,
Liu
P
,
Loos
F
, et al
Cross-reactivity between tumor MHC class I–restricted antigens and an enterococcal bacteriophage
.
Science
2020
;
369
:
936
42
.
20.
Bessell
CA
,
Isser
A
,
Havel
JJ
,
Lee
S
,
Bell
DR
,
Hickey
JW
, et al
Commensal bacteria stimulate antitumor responses via T cell cross-reactivity
.
JCI Insight
2020
;
5
:
e135597
.
21.
Balachandran
VP
,
Łuksza
M
,
Zhao
JN
,
Makarov
V
,
Moral
JA
,
Remark
R
, et al
Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer
.
Nature
2017
;
551
:
512
6
.
22.
Riquelme
E
,
Zhang
Y
,
Zhang
L
,
Montiel
M
,
Zoltan
M
,
Dong
W
, et al
Tumor microbiome diversity and composition influence pancreatic cancer outcomes
.
Cell
2019
;
178
:
795
806
.e12.
23.
Macpherson
AJ
,
Harris
NL
. 
Interactions between commensal intestinal bacteria and the immune system
.
Nat Rev Immunol
2004
;
4
:
478
85
.
24.
Zhang
Z
,
Li
J
,
Zheng
W
,
Zhao
G
,
Zhang
H
,
Wang
X
, et al
Peripheral lymphoid volume expansion and maintenance are controlled by gut microbiota via RALDH+dendritic cells
.
Immunity
2016
;
44
:
330
42
.
25.
Legoux
F
,
Bellet
D
,
Daviaud
C
,
El Morr
Y
,
Darbois
A
,
Niort
K
, et al
Microbial metabolites control the thymic development of mucosal-associated invariant T cells
.
Science
2019
;
366
:
494
9
.
26.
Ma
C
,
Han
M
,
Heinrich
B
,
Fu
Q
,
Zhang
Q
,
Sandhu
M
, et al
Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells
.
Science
2018
;
360
:
eaan5931
.
27.
Steed
AL
,
Christophi
GP
,
Kaiko
GE
,
Sun
L
,
Goodwin
VM
,
Jain
U
, et al
The microbial metabolite desaminotyrosine protects from influenza through type I interferon
.
Science
2017
;
357
:
498
502
.
28.
Nomura
M
,
Nagatomo
R
,
Inoue
K
,
Doi
K
,
Shimizu
J
,
Baba
K
, et al
Association of SCFA in gut microbiome and clinical response in solid cancer patients treated with andi-PD-1 antibody
.
Ann Oncol
2019
;
30 Suppl 5
:
v509
.
29.
Trompette
A
,
Gollwitzer
ES
,
Yadava
K
,
Sichelstiel
AK
,
Sprenger
N
,
Ngom-Bru
C
, et al
Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis
.
Nat Med
2014
;
20
:
159
66
.
30.
Tan
J
,
McKenzie
C
,
Vuillermin
PJ
,
Goverse
G
,
Vinuesa
CG
,
Mebius
RE
, et al
Dietary fiber and bacterial SCFA enhance oral tolerance and protect against food allergy through diverse cellular pathways
.
Cell Rep
2016
;
15
:
2809
24
.
31.
Bachem
A
,
Makhlouf
C
,
Binger
KJ
,
de Souza
DP
,
Tull
D
,
Hochheiser
K
, et al
Microbiota-derived short-chain fatty acids promote the memory potential of antigen-activated CD8(+) T cells
.
Immunity
2019
;
51
:
285
97
.
32.
Harel
M
,
Ortenberg
R
,
Varanasi
SK
,
Mangalhara
KC
,
Mardamshina
M
,
Markovits
E
, et al
Proteomics of melanoma response to immunotherapy reveals mitochondrial dependence
.
Cell
2019
;
179
:
236
50
.
33.
Agus
A
,
Planchais
J
,
Sokol
H
. 
Gut microbiota regulation of tryptophan metabolism in health and disease
.
Cell Host Microbe
2018
;
23
:
716
24
.
34.
Rothhammer
V
,
Mascanfroni
ID
,
Bunse
L
,
Takenaka
MC
,
Kenison
JE
,
Mayo
L
, et al
Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor
.
Nat Med
2016
;
22
:
586
97
.
35.
Cervenka
I
,
Agudelo
LZ
,
Ruas
JL
. 
Kynurenines: Tryptophan's metabolites in exercise, inflammation, and mental health
.
Science
2017
;
357
:
eaaf9794
.
36.
Le Naour
J
,
Galluzzi
L
,
Zitvogel
L
,
Kroemer
G
,
Vacchelli
E
. 
Trial watch: IDO inhibitors in cancer therapy
.
Oncoimmunology
2020
;
9
:
1777625
.
37.
Kadosh
E
,
Snir-Alkalay
I
,
Venkatachalam
A
,
May
S
,
Lasry
A
,
Elyada
E
, et al
The gut microbiome switches mutant p53 from tumour-suppressive to oncogenic
.
Nature
2020
;
586
:
133
8
.
38.
Smits
LP
,
Bouter
KE
,
de Vos
WM
,
Borody
TJ
,
Nieuwdorp
M
. 
Therapeutic potential of fecal microbiota transplantation
.
Gastroenterology
2013
;
145
:
946
53
.
39.
Vetizou
M
,
Pitt
JM
,
Daillere
R
,
Lepage
P
,
Waldschmitt
N
,
Flament
C
, et al
Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota
.
Science
2015
;
350
:
1079
84
.
40.
Wang
Y
,
Wiesnoski
DH
,
Helmink
BA
,
Gopalakrishnan
V
,
Choi
K
,
DuPont
HL
, et al
Fecal microbiota transplantation for refractory immune checkpoint inhibitor-associated colitis
.
Nat Med
2018
;
24
:
1804
8
.
41.
Batten
M
,
Shanahan
ER
,
Silva
IP
,
Adhikari
C
,
Conway
J
,
Tasker
A
, et al
Abstract 2822: Low intestinal microbial diversity is associated with severe immune-related adverse events and lack of response to neoadjuvant combination antiPD1, anti-CTLA4 immunotherapy
.
Cancer Res
2019
;
79
:
2822
.
42.
Baruch
EN
,
Youngster
I
,
Ben-Betzalel
G
,
Ortenberg
R
,
Lahat
A
,
Katz
L
, et al
Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients
.
Science
2021
;
371
:
602
9
.
43.
Sivan
A
,
Corrales
L
,
Hubert
N
,
Williams
JB
,
Aquino-Michaels
K
,
Earley
ZM
, et al
Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy
.
Science
2015
;
350
:
1084
9
.
44.
Gopalakrishnan
V
,
Helmink
BA
,
Spencer
CN
,
Reuben
A
,
Wargo
JA
. 
The influence of the gut microbiome on cancer, immunity, and cancer immunotherapy
.
Cancer Cell
2018
;
33
:
570
80
.
45.
Tanoue
T
,
Morita
S
,
Plichta
DR
,
Skelly
AN
,
Suda
W
,
Sugiura
Y
, et al
A defined commensal consortium elicits CD8 T cells and anti-cancer immunity
.
Nature
2019
;
565
:
600
5
.
46.
Fischbach
MA
. 
Microbiome: focus on causation and mechanism
.
Cell
2018
;
174
:
785
90
.
47.
McQuade
JL
,
Daniel
CR
,
Helmink
BA
,
Wargo
JA
. 
Modulating the microbiome to improve therapeutic response in cancer
.
Lancet Oncol
2019
;
20
:
e77
91
.
48.
Mager
LF
,
Burkhard
R
,
Pett
N
,
Cooke
NCA
,
Brown
K
,
Ramay
H
, et al
Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy
.
Science
2020
;
369
:
1481
9
.
49.
Li
Y
,
Elmen
L
,
Segota
I
,
Xian
Y
,
Tinoco
R
,
Feng
Y
, et al
Prebiotic-induced anti-tumor immunity attenuates tumor growth
.
Cell Rep
2020
;
30
:
1753
66
.