Understanding the relationship between breast cancer, immune checkpoint inhibitors, and gut microbiota: a narrative review
Review Article

Understanding the relationship between breast cancer, immune checkpoint inhibitors, and gut microbiota: a narrative review

Gabriella Hrubesz#, Jennifer Leigh#, Terry L. Ng

Division of Medical Oncology, The Ottawa Hospital Cancer Center, Ottawa, Ontario, Canada

Contributions: (I) Conception and design: TL Ng, J Leigh; (II) Administrative support: None; (III) Provision of study material or patients: None; (IV) Collection and assembly of data: G Hrubesz, J Leigh; (V) Data analysis and interpretation: G Hrubesz, J Leigh; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

#These authors contributed equally to this work.

Correspondence to: Terry L. Ng, MD. Division of Medical Oncology, The Ottawa Hospital Cancer Centre, 501 Smyth Road, Ottawa ON K1H 8L6, Ontario, Canada. Email: teng@toh.ca.

Background and Objective: The composition of gut microbiota plays an important role in predicting and influencing outcomes of cancer treated with immunotherapy. Our objective is to summarize the role of gut microbiota and immunotherapy in breast cancer.

Methods: A systematic search from inception until July 2024 of key search terms including immunity, breast neoplasm, gastrointestinal microbiome/microbiota, fecal microbiota transplantation, pro- and prebiotics, antibiotics and immunotherapy using EMBASE, MEDLINE and CENTRAL was conducted. The results were screened by two reviewers independently and synthesized and presented descriptively.

Key Content and Findings: Thirteen studies (5 clinical, 8 pre-clinical) met the eligibility criteria and were published from 2020–2024. Clinical studies showed that the composition and diversity of gut microbiota was associated with patient response to immunotherapy. In pre-clinical studies, dysbiotic states induced by obesity, antibiotics, and diet were associated with immunosuppression and influenced response to programmed cell death-ligand 1 (PD-L1) inhibitors. Microbiota-modulating treatments such as probiotics showed the ability to enhance response to immunotherapy, indicating their potential use as adjunct therapies in breast cancer treatment.

Conclusions: The composition of gut microbiota could help predict the chance of response to immunotherapy, and modulating gut microbiota has the potential to enhance the efficacy of chemo-immunotherapy in breast cancer. However, the available data relating to breast cancer are limited. Larger prospective studies are required to further elucidate their role as a biomarker and treatment.

Keywords: Gut microbiota; dysbiosis; breast cancer; immunotherapy


Received: 20 March 2024; Accepted: 27 August 2024; Published online: 21 October 2024.

doi: 10.21037/tbcr-24-14


Introduction

Breast cancer is the most commonly occurring cancer and leading cause of cancer-related death for females worldwide (1). Although population-based mammography screening has led to earlier detection of newly diagnosed breast cancers and an increasing chance of cure (2,3), there is still a huge unmet need for novel therapeutics against breast cancer to improve outcomes for patients. One recent change in the treatment of breast cancer has been the incorporation of immunotherapy.

Immunotherapy has become a major pillar of cancer therapy and has been successfully implemented in many types of solid and liquid tumors. They can range from technologies like vaccines and oncolytic viruses, which are relevant in antigen presentation and cancer immune activation (2,3), to monoclonal and bispecific antibodies that modulate inhibitory and stimulatory immune check points (4,5), to gene or cell therapies that involve customization of chimeric antigen receptor (CAR) T cells or ex-vivo expansion of desired immune cell products (adoptive T cells) or molecules (e.g., cytokines) relevant in anticancer activity (6-8). However, the most used type of immunotherapy for solid tumor treatment are immune checkpoint inhibitors (ICIs), humanized monoclonal antibodies that target various cellular immune checkpoints and their ligands at the tumor-immune interface. Cells acquire cancerous properties through genetic and epigenetic modifications that allow them to proliferate uncontrollably and invade surrounding tissues (9,10). The term cancer immunosurveillance refers to the coordinated response of innate and adaptive immunity against cancer. Immune checkpoints are a potential rate-limiting step in enabling T cell infiltration and tumor cell kill. Under physiologic conditions, the immune system maintains an equilibrium between its ability to clear foreign pathogens and its capacity for immune regulation. For example, programmed cell death 1 (PD-1), which is expressed on the surface of many immune cells, acts as a molecular “brake” to prevent T cells from destroying healthy cells (11,12). When T cells infiltrate the tumor microenvironment, tumor cells evade “detection” by these infiltrating lymphocytes by overexpressing inhibitory checkpoint ligands like PD-L1, which when bound to its receptor PD-1, sends inhibitory signals to PD-1 expressing cells (e.g., CD8+ T cells) and prevent an anti-tumor response (9-13). Other clinically approved ICIs target cytotoxic T lymphocyte antigen 4 (CTLA-4), which is important earlier on in the adaptive immune cascade when the requisite antigen-mediated lymphocyte activation occurs prior to CD4+ helper and CD8+ T cell clonal expansion and migration (14,15). The currently approved agents aim to wake up the immune system by blocking the interaction between PD-L1, PD-1 and anti-CTLA-4 with their ligand or receptors (9,11,16).

For breast cancer, ICIs are only approved for the treatment of triple negative breast cancer (TNBC). TNBCs comprise approximately 15% of newly diagnosed breast cancers (17-21). Pembrolizumab has been approved in the neoadjuvant setting when it is given in combination with chemotherapy for ≥ cT2 and/or ≥ cN1 (stage I–III except pT1pN0) TNBC. It has also been approved for first-line treatment of metastatic TNBC in those with a PD-L1 score ≥10% (22-24).

While ICIs can be effective, response rates only range from 15% to 30% in most solid tumors (25-28). A recent meta-analysis of immunotherapy in metastatic breast cancer (mBC) found an overall response rate (ORR) of 19% to ICIs (29). Therefore, considering the costs of treatment-related toxicity and financial toxicity, an important avenue of research is the role of biomarkers, including the tumor- and host-related factors that predict the chance of patient response to ICIs. This continues to be an avenue of exploration to maximize clinical benefit for the patient population while sparing unnecessary toxicity (25,30,31).

Beyond PD-L1 expression score as a predictive biomarker of response in metastatic TNBC (32-35), other biomarkers such as tumor-infiltrating lymphocytes (TILs) and T cell receptor (TCR) diversity have also been explored as predictors of ICI efficacy in breast cancer (18,36-39). However, the accuracy and reliability of these biomarkers have been inconsistent in practice, limiting their clinical utility (24-26). Thus, more research is needed to identify practical tumor- and patient-specific biomarkers.

Patient factors including the diversity of the gut microbiome can influence responses to ICI therapy (40,41). The gut microbiota consists of the various microorganisms found within the digestive tract, including bacteria, viruses, fungi and protozoa. Technically, the gut microbiome refers to the metagenome of these organisms, but is often used interchangeably with microbiota as it is referring to the taxonomic classification of microbial species and overall composition (38). Gut microbiota have been linked directly to many humans diseases, including cancer (42-45). Importantly, it can influence cancer development and progression, and it can also influence response to cancer treatment (46-48). For example, gut microbiota can stimulate antitumor immune responses through the regulation of CD8+ T cells and tumor-associated myeloid cells (49-51). Additionally, reduction of gut microbiota diversity through the use of antibiotics has been shown to reduce response to cancer immunotherapy (52-54). Correlations between the gut microbiome and efficacy of immunotherapy have been studied in various cancer types, including lung cancer, prostate cancer and melanoma (55-57).

In breast cancer, gut microbiota have been shown to influence response to systemic therapies including chemotherapy and anti-human epidermal growth factor receptor 2 (HER2)-targeted therapies (49,58-60). For example, increased alpha diversity, which is a measure of microbiota diversity in an individual sample, has been shown to predict responsiveness to the anti-HER2 monoclonal antibody trastuzumab in HER2+ breast cancer (61,62). Additionally, patients who did not respond to trastuzumab showed lower alpha diversity and higher abundance of certain species of Bacteroides (62,63). Another study by Li et al. also showed higher alpha diversity being associated with response to neoadjuvant breast cancer chemotherapy (64). Other studies have shown that the relative abundance of specific gut microbiota taxa or consortia is also associated with chemotherapy response in breast cancer (64,65). The role of gut microbiota in modulating the efficacy of immunotherapy, specifically, ICIs, in breast cancer, however, is less well characterized.

Given the increased recognition of the role that the gut microbiome plays in ICI response, our objective is to summarize current data on the link between gut microbiota and ICI efficacy in breast cancer. Specifically, we discuss the role of gut microbiota signatures and dysbiosis in predicting ICI response and the use of novel microbiota-modulating treatments to enhance response to ICI therapy in breast cancer. We present this article in accordance with the Narrative Review reporting checklist (available at https://tbcr.amegroups.org/article/view/10.21037/tbcr-24-14/rc).


Methods

We used EMBASE, MEDLINE, and CENTRAL databases to conduct a search using the following terms: immunity (innate and adaptive), immune checkpoint inhibitors, immunotherapy, breast neoplasms, gastrointestinal microbiome/microbiota, fecal microbiota transplantation, pre- and probiotics and antibiotics. We reviewed all studies published until June 4, 2024. The search strategy is summarized in Table 1 and the full search strategy is available in Appendix 1.

Table 1

Search strategy summary

Items Specification
Date of search June 4, 2024
Databases and other sources searched EMBASE, MEDLINE, CENTRAL
Search terms used Innate immunity, adaptive immunity, monocytes, macrophages, myeloid-derived suppressor cells, T-lymphocytes, immune checkpoint inhibitors, immunotherapy, breast neoplasms, gastrointestinal microbiome/microbiota, fecal microbiota transplantation, probiotics, prebiotics, antibacterial agents, proton pump inhibitors, hydrocortisone
Timeframe Inception to June 4, 2024
Inclusion criteria Population: clinical studies with adult males or females with confirmed breast cancer of any stage, animal models of breast cancer and/or in vitro models of breast cancer treated with immunotherapy
Concept: the relationship between gut microbiota compositions and response to immunotherapy and/or overall outcomes
Context: original articles and abstracts in English language, any country of origin
Exclusion criteria Studies with <5 patients excluded
Selection process Dual reviewer selection based on title and abstract and full text screening; single reviewer full text analysis

Our initial search strategy of the relevant databases yielded 311 citations. However, after a subsequent expansion of our search strategy, an additional 1,644 publications were found. These articles were screened by two reviewers (G.H., J.L.), using title and abstract, according to the criteria defined in Table 1. Studies that did not meet inclusion criteria were excluded, including duplicate articles. This resulted in 137 articles eligible for full text screening conducted by two reviewers (G.H., J.L.). Review articles, ongoing clinical trials without published results and studies not including immunotherapy were removed. However, the reference lists of review articles were searched for potentially relevant studies. In total, there were 16 abstracts and articles that met all inclusion criteria. These were critically appraised, and data were extracted by a single reviewer (G.H.).


Results

Based on our systematic search strategy, two-stage citation screening, and after an updated manual search of the literature on July 6, 2024, a total of 16 abstracts and articles met the inclusion criteria—13 studies were available as published conference abstracts only and 3 studies were available as full-text peer-reviewed manuscripts. Some of these abstracts were presenting updated data to an ongoing study. In total, there were 13 unique clinical and pre-clinical studies that were included in our review. These studies were published between 2020–2024, and originated from research groups in the United States, United Kingdom, Italy, China and the Republic of Korea.

Of the 13 eligible studies, 5 (38%) were clinical trials (n=202), of which 4 studies reported microbiome-related outcomes as a post-hoc exploratory endpoint and one trial had pre-planned sample collection, but the analysis was also done retrospectively (Table 2). Stool samples were not routinely collected in all study participants and the breast cancer subtypes enrolled in those trials were heterogeneous. To summarize, two clinical trials (n=70) included estrogen-receptor positive (ER+) breast cancer only, one trial (n=10) included TNBC only and two trials included a mix of TNBC and ER+ breast cancer (n=88). The remaining 8 (62%) studies were conducted in the pre-clinical setting and were predominantly based on mouse models of breast cancer (Table 3). Four studies used TNBC mouse models (n=40), one study looked at ER+ breast cancer and the two remaining studies examined HER2+ breast cancer (n=15) and an ER+, PR+ breast cancer mouse model (n=39), respectively. In terms of scope, two of the studies addressed the impact of dysbiosis on immunotherapy efficacy in breast cancer mouse models, four studies assessed the use of novel microbiota agents to increase immunotherapy efficacy in vitro and in a mouse model, and two studies explored the role of diet and obesity in immunotherapy efficacy in a mouse model.

Table 2

Summary of clinical studies

Author Year Country Publication type Study design Clinical trial number BC type No of patients Interventions* Microbiome sample source and analysis Outcome measures
Barroso-Sousa et al. (66,67) 2023 & 2020 United States Abstract Interventional (post-hoc analysis) NCT03051659  ER+ 26 Group 1 (n=15): eribulin + pembrolizumab Sample: fecal Association between microbiome diversity, taxonomic composition and clinical outcomes; PR, CR; SD, PD; OS
Group 2 (n=11): eribulin Analysis: 16S RNA sequencing + metagenomic shotgun sequencing
Database: MetaPhlAn3
Teng et al. (68) 2022 United Kingdom Article Interventional (post-hoc analysis) KELLY trial NCT03222856 ER+ HER2- 44 Pembrolizumab + eribulin Sample: fecal Safety and efficacy of chemotherapy with ICI; microbiome composition; CB; SD; drug-related microbial toxicity
Analysis: 16S RNA sequencing + metagenomic shotgun sequencing
Database: LeFSe, MEGAHIT 
Wong et al. (69); Yuan et al. (70) 2021 United States Abstract Interventional (post-hoc analysis) (I) NCT02971761 (I) TNBC (I) 12 (I) Pembrolizumab + enobosarm Sample: fecal Association of microbiome with response to triple therapy; microbiome composition; PR, CR; SD, PD 
(II) NCT02778685 (II) ER+ (II) 10 (II) Palbociclib + pembrolizumab + letrozole5 Analysis: 16S RNA sequencing + metagenomic sequencing
Database: Illumin MiSeq 
Chun et al. (71) 2023 United States Abstract Interventional (pre-planned retrospective analysis) AMTEC trial NCT03801369 TNBC 10 Olaparib + durvalumab Sample: fecal Correlation of gut microbiome diversity with tumor biomarkers (PD-L1, tumor immune cell density, TILs, MutSig3 and IFN gene signatures; clinical response to therapy
Analysis: 16S RNA sequencing 
Kulkarni et al. (72) 2024 United States Article Interventional (post-hoc analysis) I-SPY2 trial NCT01042379 TNBC, ER+, HER2− 66 Pembrolizumab + paclitaxel + doxorubicin + cyclophosphamide +/− antibiotic use during treatment Residual cancer burden; pathologic complete response rates

*, eribulin = chemotherapy (microtubule dynamics inhibitor); pembrolizumab = ICI (PD-1 inhibitor); enobosarm = hormone therapy (non-steroidal selective androgen receptor modulator); palbociclib = targeted therapy (CKD4/6 inhibitor); letrozole = hormone therapy (aromatase inhibitor); olaparib = targeted therapy (PARP inhibitor); durvalumab = ICI (PD-L1 inhibitor); paclitaxel = chemotherapy (mitotic inhibitor); doxorubicin = chemotherapy (anthracycline); cyclophosphamide (alkylating agent). BC, breast cancer; ER+, estrogen-receptor-positive; PR, partial response; CR, complete response; SD, stable disease; PD, progressive disease; OS, overall survival; HER2, human epidermal growth factor receptor 2; ICI, immune checkpoint inhibitor; CB, clinical benefit; TNBC, triple-negative breast cancer; PD-L1, programmed cell death-ligand 1; TIL, tumor-infiltrating lymphocyte; IFN, interferon; PD-1, programmed cell death 1.

Table 3

Summary of pre-clinical studies

Author Year Country Publication type Study design BC model BC type Number of subjects Interventions Microbiome sample source and analysis Outcome measures
Rosean et al. (73) 2020 United States Abstract Interventional Mice HR+ Not reported Intervention: anti-PD-1 Efficacy of anti-PD-1 therapy when combined with PGE2 inhibitors in dysbiosis
Intervention: PGE2 inhibitor
Pingili et al. (74) 2021 United States Article Interventional Mice ER+, PR+ 39 Group 1 (n =12): high-fat diet + anti-PD-1 Sample: fecal Composition of the microbiome; tumor incidence; tumor progression; response to ICI 
Group 2 (n=11): high-fat diet + IgG control Analysis: 16S RNA sequencing
Group 3 (n=9): low-fat diet + anti-PD-1 Database: GreenGenes 
Group 4 (n=7): low-fat diet + IgG control
Bohm et al. (75) 2023 United States Abstract Interventional Mice TNBC Not reported Group 1: obese on high fat diet Sample: cecal contents Tumor burden; tumor progression; immunotherapy response
Group 2: formerly obese following surgical weight loss Analysis: 16S RNA sequencing
Intervention: antibiotic therapy followed by FMT
Intervention: anti-PD-1 immunotherapy or IgG control
Clear et al. (76) 2022 United States Abstract Interventional Mice TNBC EMT-6: n=5–7/group; E0771: n=8–10/group Group 1: low-fat control diet Sample: fecal Impact of diet on ICI response; tumor volume; tumor weight; ICI responsiveness
Group 2: high-fat Western diet Analysis: metagenomic sequencing
Group 3: mediterranean diet
Intervention 1: IgG or anti-PD-L1 antibodies
Intervention 2: FMT
Clear et al. (77) 2024 United States Abstract Interventional Mice TNBC Unknown Intervention 1: IgG or anti-PD-L1 or anti-PD-1 antibodies Tumor progression; tumor volume; tumor cell populations
Intervention 2: A. muciniphila or short-chain fatty acid supplementation
Ferrari et al. (78) 2021 Italy Abstract Interventional Mice, human cell lines HER2+,
IDC
15 Group 1: microbial derivatives + anti-PD-1 HLA expression; activation and cytotoxic levels of CTLs; response to ICI 
Group 2: microbial derivatives + vehicle control
Kim et al. (79) 2021 & 2022 Republic of Korea Abstract Interventional Mice TNBC Not reported Group 1: tumor control Sample: fecal Tumor volume; PD-L1 IHC score; PD-L1 gene expression; proportion of M2 macrophages and mRNA expression; response to ICI; pro- and anti-inflammatory cytokine expression
Group 2: anti-PD-1 Analysis: 16S RNA sequencing
Group 3: RAPO
Group 4: anti-PD-1 + RAPO
Li et al. (80) 2024 China Article Interventional Human cell lines; mice TNBC 40 Group 1 (n=8): control (no tumor) Sample: fecal Efficacy of anti-PD-1 therapy when combined with fucoidan
Group 2 (n=8): PBS treatment Analysis: 16S RNA sequencing
Group 3 (n=8): anti-PD-1 antibody
Group 4 (n=8): fucoidan
Group 5 (n=8): fucoidan + anti-PD-1 antibody

BC, breast cancer; HR+, hormone-receptor-positive; PD-1, programmed cell death 1; PGE2, prostaglandin E2; ER+, estrogen-receptor-positive; PR+, progesterone-receptor-positive; ICI, immune checkpoint inhibitor; TNBC, triple-negative breast cancer; FMT, fecal microbiota transplantation; PD-L1, programmed cell death-ligand 1; HER2, human epidermal growth factor receptor 2; IDC, invasive ductal carcinoma; HLA, human leukocyte antigen; CTL, cytotoxic T-lymphocyte; RAPO, Bifidobacterium longum RAPO; IHC, immunohistochemistry; PBS, phosphate-buffered saline.


Discussion

Given that the incorporation of immunotherapy into the treatment of breast cancer is relatively new, there is a limited understanding as to which biomarkers predict response outside of PD-L1. Several studies in other disease sites such as melanoma lung, liver, gastric and prostate, have identified the potential use of gut microbiome signatures to predict patient response to immunotherapy (57,81-89). We sought to better characterize the role of gut microbiome signatures for immunotherapy in breast cancer.

Gut microbiome signatures as predictors of immunotherapy response in ER+, HER2− breast cancer

Three studies addressed the relation of gut microbiome signatures and response to immunotherapy in ER+, HER2− mBC.

Barroso de Sousa et al., reported on the gut microbiome correlates of a previously completed randomized phase II trial (NCT03051659) of eribulin monotherapy (E) vs. eribulin plus PD-L1 inhibitor pembrolizumab (E+P) in pre-treated ER+ mBC (66). A subset (eribulin: n=11; eribulin plus pembrolizumab: n=12) of these patients had fecal samples collected at baseline (BL), post two cycles (C2), and at end of treatment (EOT). Among the 23 patients (23 BL, 22 C2, 5 EOT), subjects with progression-free survival (PFS) and overall survival (OS) below the median had comparable alpha-diversity at baseline to those above the median in both treatment groups. In this initial analysis, which used 16S(v3-4) RNA, the findings were limited by a small sample size and limited depth of strain evaluation. Nonetheless, they descriptively reported an increase in Faecalibacterium from baseline amongst patients who had a partial response (PR) to eribulin while levels remained unchanged in those who had stable disease (SD). The gut microbiome composition within this small subgroup with available fecal samples was possibly prognostic for a patient’s survival outcomes but did not seem predictive of response to eribulin or pembrolizumab. The authors subsequently performed metagenomic shotgun sequencing on baseline fecal samples that were collected from 26 patients prior to randomization for more in-depth microbiome coverage and strain elucidation. With this, they found that higher alpha diversity was associated with longer OS across all patients and in the E+P subgroup. This was not the case in the eribulin monotherapy arm (67). At the taxonomic level, longer OS was associated with baseline lower relative abundance of Blautia wexlerea and higher relative abundance of Odoribacter splanchnicus, a common short-chain fatty acid producing gut bacterium, even after controlling for age, ECOG status and prior lines of therapy. Overall, this suggests that the gut microbiome could be of prognostic value whereas the predictive value in relation to treatment-specific response and outcomes require further study (66,67).

The single-arm phase II KELLY trial (NCT03222856) similarly tested the safety and efficacy of eribulin plus pembrolizumab in patients with ER+, HER2− unresectable BC or mBC (68). In the subsequent CALADRIO sub study, Teng et al. analyzed both fecal and saliva samples collected from 44 of these patients at baseline, post three cycles of treatment, and at EOT. They found that treatment with eribulin plus pembrolizumab did not cause significant oral or gut microbiota changes from baseline. However, clinical benefit was associated with an abundance of gut-associated Bacteriodes fragilis and oral Streptococcus and increased gut microbiota richness at baseline (68).

Preliminary results from 16SRNA analysis of fecal samples collected from 10 ER+, HER2− mBC patients during a phase I/II clinical trial testing the safety and efficacy of the combination of letrozole, palbociclib and pembrolizumab (NCT02778685) found that patients with PR to treatment had ‘healthy’ gut signatures dominated by Bacteroides and short chain fatty acid-producing Firmicutes (69). In a subsequent analysis of samples from the same clinical trial, metagenomic sequencing was performed on fecal samples from 11 patients collected at baseline and during treatment, and found that a higher relative abundance of Gemmiger formicillis was associated with response to therapy whereas an increase in relative abundance of Bacteroides vulgatus was associated with resistance to therapy (69).

Gut microbiome signatures as predictors of ICI response in metastatic TNBC

Currently, TNBC is the only subtype of breast cancer with approved indications (neoadjuvant chemo-immunotherapy for stage II/III TNBC and first-line unresectable/metastatic TNBC) for using ICI as part of standard care treatment. There are two clinical studies that explored gut microbiome signatures and response to immunotherapy.

Chun et al., performed a correlative analysis of gut microbiome on patients with metastatic TNBC with baseline stool sample and tissue biopsy collected (n=10) prior to starting olaparib monotherapy for a 28-day cycle, followed by on-treatment stool collection and on-treatment tissue biopsy (paired pre-treatment and on-treatment; n=8) before adding in durvalumab in cycle 2 (NCT03801369) (71). Using 16S RNA analysis, they reported that the average baseline alpha diversity did not vary with treatment response. However, the average alpha diversity on-treatment after one cycle of olaparib monotherapy was higher in patients who subsequently derived clinical benefit (PR/SD) from the olaparib and durvalumab combination. This suggests that fecal microbiome alpha diversity prior to starting durvalumab, a PD-L1 inhibitor, may predict likelihood of response to the combination. On-treatment microbiome alpha diversity was inversely related with tumor PD-L1 expression (P=NS) and was positively associated with high TILs in the paired on-treatment biopsy. However, there was no association between alpha diversity and immune cell density by multiplex IHC, inflammatory signature, IFN gene signature, or MutSig3 score (71).

In an exploratory analysis, gut microbiome diversity was assessed in patients with androgen receptor positive (AR+) metastatic TNBC who were treated with pembrolizumab and a selective androgen receptor modulator, Enobosarm in a phase II clinical trial (70). They found that patients who had disease progression had dysbiotic gut microbiomes and displayed a relative abundance of Prevotella copri, an organism associated with inflammation. Patients who had PR to treatment had relatively healthy gut microbiota dominated by Bacteroides, Faecalibacterium prausnitzii and other short chain fatty acid producers (70).

Taken together, these studies show the potential of using gut microbiome signatures to predict response to ICI therapy in TNBC. Positive responses to immunotherapy appear to be associated with increased alpha-diversity of the gut microbiome prior to starting immunotherapy. Additionally, abundance of certain microbiota, such as Bacteroides, may be associated with improved survival in patients, while microbiota such as Prevotella could be associated with early disease progression and should be further explored.

Metabolic and dietary factors related to gut dysbiosis and ICI efficacy

Gut dysbiosis, characterized by a disruption to the microbiome relative to that found in a healthy individual and resulting in changes in their functional composition and metabolic activities, has been shown to influence response to immunotherapy in patients with cancer (46,90). Gut dysbiosis can be induced or influenced by extrinsic interventions like antibiotics, medications, and diet but also intrinsic, patient-specific factors like body composition and systemic metabolism (91,92). Five pre-clinical studies and one clinical study investigated the impact of gut dysbiosis on breast cancer immunotherapy.

Rosean et al. previously showed that meloxicam combined with PD-L1 blockade was effective in an endocrine-resistant ER+ breast tumor model by targeting prostaglandin E2 (PGE2) production via COX-2 inhibition (93). This mechanism is important since PGE2 is a mediator of systemic immune suppression in mBC. However, this therapeutic combination was ineffective in mice with antibiotic-induced gut dysbiosis. They found that mice with commensal dysbiosis had more recruitment of suppressive myeloid populations and irreversible T cell dysfunction (73). Importantly, breast glands from dysbiotic mice showed enhanced COX-1 activity, and since COX-1 also regulates PGE2 synthesis, this suggests a secondary mechanism for PGE2 synthesis and resistance to meloxicam therapy. Supporting this hypothesis, targeting COX-1 mediated PGE2 synthesis resulted in decreased tumor burden only in dysbiotic mice. These findings support that dysbiosis-induced COX-1 mediated tissue inflammation is sufficient to drive immune suppression and treatment resistance (73,93).

Obesity is another known factor that modifies the gut microbiome and induces gut dysbiosis (94,95). Obesity has also been associated with increased risk of early recurrence in patients with ER+ disease (96). Pingili et al. investigated the role obesity plays in ICI response in a syngeneic TNBC mouse model using E0771 cells (74). They found that high-fat diet induced obesity led to greater immunosuppression and tumor growth. However, anti-PD-1 therapy was able to induce tumor regression not only in lean mice but also in obese mice by downregulating immune suppression and upregulating antitumor immunity. In obese mice, the presence of breast tumor was associated with decreased relative abundance of Ruminococcus and an increased relative abundance of Bacteriodales whereas treatment with anti-PD1 reduced the abundance of Enterobacteriaceae and Bacteriodales while it led to an increase in Akkermansia, Bifidobacterium, Lactobacillus, Aidercreutzia, Odoribacter, Mogibacteriaceae, and Ruminococcus (74).

In addition, Bohm et al. investigated the role of bariatric surgery in modulating the gut microbiome and its impact on anti-PD-1 immunotherapy in a TNBC mouse model (75). The commensal microbiota of study mice were ablated with broad spectrum antibiotics prior to fecal microbial transplant (FMT) using cecal contents from obese mice on high fat diet or from formerly obese mice following surgical weight loss. Results showed that FMT from weight loss donor mice was associated with reduced tumor burden and improved anti-PD-1 immunotherapy response. FMT from weight loss donors resulted in elevated infiltration of CD8+ T cells into the tumor microenvironment. Additionally, microbes from mice with weight loss surgery combined with anti-PD-1 treatment decreased circulating levels of lithocholic acid, a secondary bile acid that drives immunosuppression (75). Thus, gut microbiota can improve response to anti-PD-1 therapy in breast cancer after bariatric surgery.

Diet is another factor known to be important in microbiome modulation. Clear et al. investigated the role of diet in potentiating immunotherapy response in a syngeneic mouse model using EMT-6 and EO771 (TNBC) bearing mice (76). They found that both mice fed with either a high-fat Western diet or Mediterranean diet experienced a significant reduction in tumor volume and weight when treated with PD-L1 inhibitor compared to mice fed with a low-fat control diet. Additionally, Western and Mediterranean-fed mice displayed a 25–45% increase in proportional abundance of gut Akkermensia muciniphila, which degrades mucin to produce short-chain fatty acid metabolites. Furthermore, fecal microbiota transplant enriched with Akkermensia muciniphila, when given in conjunction with PD-L1 therapy, enhanced responsiveness to PD-L1 blockade. In a later study, Clear et al. investigated if Akkermensia muciniphila or mucin short-chain fatty acid supplementation could directly enhance ICI therapy in EO771 tumor-bearing mice (77). Results showed that mice supplemented with Akkermensia muciniphila demonstrated a significant reduction in tumor volume and increased levels of immune cell populations in the tumor microenvironment. Supplementation with short-chain fatty acids and anti-PD-1 with chemotherapy also showed enhanced response to treatment. These results suggested that relative abundance of Akkermensia muciniphila could potentially predict, or even enhance ICI efficacy in breast cancer, as has been associated with anti-PD-1 response in other cancer models (88). Overall, the results of these studies indicate that dysbiosis, which can be induced through multiple factors including antibiotics, diet and metabolic processes such as obesity, can influence the efficacy of immunotherapy in breast cancer.

Antibiotics can also play a role in inducing gut dysbiosis and have been shown to impact ICI efficacy in cancer treatment (97-100). Kulkarni et al. investigated the impact of antibiotic exposure on residual cancer burden in HER-negative early-stage breast cancer patients treated with pembrolizumab as part of the I-SPY2 trial (72). Overall, the results showed that antibiotic use during immunotherapy treatment was associated with higher mean residual cancer burden and lower pathologic complete response rates. This data suggests that antibiotic treatment may negatively impact ICI treatment outcomes in early-stage breast cancer, possibly by changing gut microbiome composition (72).

Use of microbiota agents to increase ICI efficacy

Given the known interplay between the microbiome and systemic treatment response, there has been an interest in identifying microbial interventions that could enhance this treatment effect. One particular agent of interest has been the use of probiotics (101,102). Ferrari et al. tested two proprietary microbial derivatives on murine and human primary immortalized cells lines, including those from breast cancer, and found that in vitro treatment with microbial derivatives led to human leukocyte antigen (HLA) upregulation (78). Concordantly, this led to microbial derivative-dependent increase in cytotoxic T-lymphocyte (CTL) tumor cell recognition. In 4T1 (TNBC)-bearing Balb/c mice, intraperitoneally injected microbial derivatives in combination with anti-PD-1 significantly reduced tumor growth when compared to anti-PD-1 given with vehicle control. Microbial derivative-treated mice expressed higher levels of MHC class I compared to mice treated with vehicle control. This study suggests that microbial derivatives could be used in combination with ICIs to enhance anti-cancer immune responses (78).

Kim et al. investigated the effects of oral supplementation of Bifidobacterium longum RAPO on anti-PD-1 therapy in a 4T1 mouse model of TNBC (79). They found that tumor volume was significantly inhibited in the anti-PD-1 plus RAPO group when compared with the control, RAPO only, and anti-PD-1 only groups. Treatment with anti-PD-1 + RAPO was also associated with increased levels of spleen NKT cells and tumor NK cells. Treatment with anti-PD-1 + RAPO decreased the proportion of pro-tumor M2 macrophages and reduced mRNA expression of M2 markers IL10 and Arg1, while increasing anti-tumor cytokines IFNγ and TNFα compared with anti-PD-1 treatment alone. Additionally, the relative abundance of genus Bifidobacterium, Akkermansia and Lachnospiraceae was enriched in the anti-PD-1 + RAPO group compared with other groups. Additionally, Kim et al. analyzed tissue samples from RAPO and anti-PD-1-treated mice using qRT-PCR to determine if RAPO could reduce the risk of immune-related adverse events associated with anti-PD-1 treatment by altering the expression of pro- and anti-inflammatory cytokines (103). Functional analysis showed that expression of pro-inflammatory cytokines IL1β, IL6 and TNFα was significantly decreased, while the expression of anti-inflammatory cytokine IL10 was increased in lung, liver and kidney tissues in the anti-PD-1+RAPO group compared to the anti-PD-1 group. Similarly, TNFα expression was decreased in heart and colonic tissue, while IL10 was increased in the anti-PD-1+RAPO group compared to the PD-1 group. Finally, anti-PD-1+RAPO was associated with decreased levels of pro-inflammatory enzyme, myeloperoxidase, in the liver. This data suggests that combination therapy of B. longum RAPO with anti-PD-1 therapy could be used to improve anti-PD-1 immunotherapy response in TNBC (79). Additionally, this data suggests that RAPO could even reduce the risk of immune-related adverse events related to anti-PD-1 treatment by reducing pro-inflammatory cytokines and increasing anti-inflammatory cytokines in crucial organs, such as the heart, lung, kidney, liver and colon (103). Thus, specific microbiota agents, such as Bifidobacterium longum RAPO, could be used as adjuvants to enhance immunotherapy efficacy and reduce immunotherapy-associated side effects in breast cancer.

Li et al. investigated the role of dietary supplement, fucoidan, and its impact on the efficacy of anti-PD-1 therapy in a TNBC mouse model (80). Fucoidan is a polysaccharide isolated from brown algae and marine invertebrates (104). As a dietary supplement, fucoidan has been reported to exert anti-tumor and immunomodulatory effects and enhance response to chemotherapeutic agents (105,106). Additionally, fucoidan is a potential prebiotic that can increase the diversity of gut microbiota and increase short-chain fatty acid production (107-109). The study showed that fucoidan enhanced the anti-tumor effect of anti-PD-1 blockade by decreasing tumor size, weight and growth. Correlating to this observed anti-tumor effect, 16S sequencing of the gut microbiota showed that fucoidan, when combined with anti-PD-1 antibody, increased the abundance of Firmicutes, Bifidobacterium and Faecalibaculum, which are believed to be beneficial for anti-PD-1 therapy, and decreased the abundance of Bacteroidetes, which is not favorable for ICI therapy. These changes were associated with an increase in short-chain fatty acid content in the cecum. In addition, intervention with fucoidan and anti-PD-1 antibody increased abundance of Lactobacillus, which has been shown to increase efficacy of immunotherapy in pre-clinical and clinical trials (110,111). Additionally, treatment with antibiotics prior to fucoidan and anti-PD-1 antibody treatment impaired the antitumor effects of fucoidan and anti-PD-1 immunotherapy. Thus, the gut microbiota is likely necessary to support the antitumor effect of fucoidan. Thus, the oral administration of fucoidan has the potential to sensitize breast cancer to anti-PD-1 immunotherapy in the clinic (80).


Future directions

Many clinical studies have investigated the association between patient gut microbiome and response to cancer therapy. However, our understanding of the exact roles and mechanisms of the microbiome in relation to breast cancer immunotherapy remains limited. In this review, we summarized some of the key available data regarding gut microbiome as a prognostic and predictive marker of immunotherapy response in patients, and in pre-clinical models, the role of gut dysbiosis and ICI efficacy, as well as proof-of-concept that microbiome-modulating interventions have the potential to improve immunotherapy response in breast cancer.

The exploratory clinical data presented indicates a potential role for using gut microbiome signatures to predict ICI benefit in breast cancer, as summarized in Table 4. In particular, increased baseline alpha diversity seemed to correlate with improved response to PD-1/PD-L1 immune checkpoint blockade (67-71). Due to the small sample size of patients with stool correlatives available and due to variation in gut microbiome sequencing technology (16S RNA and metagenomic sequencing) and bioinformatic approaches, most of the reported findings are purely exploratory and hypothesis generating.

Table 4

Summary of gut microbiota analysis in clinical studies

Author Model BC type Interventions* Microbiome sample source Analysis Database Associated with positive response to ICI therapy Associated with poor response to ICI therapy
Barroso-Sousa et al. (66,67) Patients HR+ Group 1 (n=15): eribulin + pembrolizumab Fecal 16S RNA sequencing; metagenomic shotgun sequencing MetaPhlAn3 Faecalibacterium; Odoribacter splanchnicus; Increased alpha diversity Blautia wexlerea
Group 2 (n=11): eribulin
Teng et al. (68) Patients HR+ HER2- Pembrolizumab + eribulin Fecal + saliva 16S RNA sequencing Bacteriodes fragilis; Increased alpha diversity
Wong et al. (69); Yuan et al. (70) Patients (I) TNBC (I) Pembrolizumab + enobosarm 16S RNA sequencing; metagenomic shotgun sequencing LeFSe, MEGAHIT  Bacteroides; Firmicutes (metabolite: SCFAs); Gemmiger formicillis; Faecalibacterium prausnitzii (metabolite: SCFAs) Bacteroides vulgatus; Prevotella copri
(II) ER+ (II) Palbociclib + pembrolizumab + letrozole
Chun et al. (71) Patients TNBC Olaparib + durvalumab Fecal 16S RNA sequencing Increased alpha diversity
Pingili et al. (74) Mice TNBC Anti-PD-1 Fecal 16S RNA sequencing GreenGenes Akkermensia; Bifidobacterium; Lactobacillus; Aidercreutzia; Odoribacter; Mogibacteriaceae; Ruminococcus Enterobacteriaceae
Bacteroidales
Bohm et al. (75) Mice TNBC Bariatric surgery + anti-PD-1 Cecal contents 16S RNA sequencing Lithocholic acid
Clear et al. (76) Mice TNBC Anti-PD-L1 Fecal Metagenomic sequencing Akkermensia muciniphilia (metabolite: SCFAs)
Kim et al. (79) Mice TNBC Anti-PD-1 + RAPO Fecal 16S RNA sequencing Bifidobacterium; Akkermansia; Lachnospiraceae
Li et al. (80) Mice TNBC Fucoidan + anti-PD-1 antibody Fecal 16S RNA sequencing Firmicutes; Bifidobacterium; Faecalibaculum actobacillus; SCFAs Bacteroidetes

*, eribulin = chemotherapy (microtubule dynamics inhibitor); pembrolizumab = ICI (PD-1 inhibitor); enobosarm = hormone therapy (non-steroidal selective androgen receptor modulator); palbociclib = targeted therapy (CKD4/6 inhibitor); letrozole = hormone therapy (aromatase inhibitor); olaparib = targeted therapy (PARP inhibitor); durvalumab = ICI (PD-L1 inhibitor). BC, breast cancer; ICI, immune checkpoint inhibitor; HR+, hormone-receptor-positive; HER2, human epidermal growth factor receptor 2; TNBC, triple negative breast cancer; ER+, estrogen-receptor-positive; SCFAs, short chain fatty acids; PD-1, programmed cell death 1; PD-L1, programmed cell death-ligand 1; RAPO, Bifidobacterium longum RAPO.

Additionally, a number of pre-clinical studies identified that dysbiosis induced alterations in the immune system had context-dependent importance in regulating breast cancer growth (73-77,112). Modifiable drivers of gut dysbiosis, such as diet, weight management and antibiotic use, could be used to predict and potentially alter tumor response to immunotherapy (72-77). Prospective clinical studies are required to elucidate the practical application of dietary and weight management interventions. The use of microbial derivatives, probiotics, and fecal transplantation in pre-clinical breast cancer models (76-80,103) highlight that microbiome-modulating interventional trials in humans may be on the horizon.

The understanding of the interaction between the gut microbiome and tumor-related immunity and control in breast cancer remains in its infancy. As a result, one of the limitations of this review is the scarcity of published studies regarding this topic. Much of the data included in this review consisted of preliminary data from published abstracts only, which do not have complete information and thus present a higher risk of bias. All the studies included in this review explored the use of immunotherapy in advanced ER+ breast cancer and advanced TNBC, but not HER2+ breast cancer (22,24,113), reflecting the current landscape of immunotherapy development in breast cancer. Clinical data from human therapeutic trials were limited by small sample size and sometimes incomplete correlative sample sets, and the samples were analyzed retrospectively. The real-world applicability of gut microbiome signatures or gut microbiome directed interventions will need to be validated using randomized prospectively validated studies.

In terms of future directions, the interrelationship between gut microbiota and host immunity is evidently complex. As the technology and cost of metagenomic and metabolomic sequencing and bioinformatics continue to improve, it is hopeful that we may understand the mechanistic underpinnings and functional consequences of the gut microbiome on cancer immunity at the population and individual level. It is unknown yet whether the gut microbiome is the most direct resource to predict cancer response or if some downstream immune-metabolomic factor is more direct and important. It is also yet unknown if the correlation between gut microbiome species/signature and immunotherapy response is dependent on the cancer type and other host factors.

Future studies of the gut microbiome and cancer immunity should also address the role of age-related gut microbial and immune system changes. For example, low-grade inflammation and immunosenescence are hallmarks of aging in the elderly (114-116). As the gut microbiome has an important role in cancer initiation, progression, and metastases, and because there is a general trend towards increased gut dysbiosis in advanced stage disease compared to early-stage disease, we need to account for cancer stage when interpreting the results of gut microbiome studies (117,118). It is most likely that immunotherapy responses modulated by gut microbiota are going to be heterogeneous across cancer subtypes, and therefore promising gut microbial species or consortia with potential therapeutic or prognostic relevance need to be validated individually across different cancers.


Conclusions

The data presented in this review highlights the importance and potential of the gut microbiome in predicting and influencing the efficacy of ICI therapy in breast cancer. There is preliminary evidence to suggest that the microbiome composition can be used to predict response to immunotherapy, and that microbiome dysbiosis resulting from factors such as antibiotics, diet, obesity, and probiotics, modulates these responses. Preliminary data also support the development of microbial-based therapeutics (e.g., probiotics) as an adjunct to cancer therapy. While this is a promising field, this topic in breast cancer remains in its infancy. Larger prospective studies are required to better understand these relationships before incorporating them into clinical practice.


Acknowledgments

We would like to thank Risa Shorr and Nigele Langlois for Administrative support.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tbcr.amegroups.org/article/view/10.21037/tbcr-24-14/rc

Peer Review File: Available at https://tbcr.amegroups.org/article/view/10.21037/tbcr-24-14/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tbcr.amegroups.org/article/view/10.21037/tbcr-24-14/coif). T.L.N. serves as the editorial board member of Translational Breast Cancer Research from November 2022 to October 2024. T.L.N. is a post-conference abstract review speaker with AstraZeneca Canada and Lilly Canada and a speaker with the Canadian Breast Cancer Network. He has been a COMPASS Ad board member with Novartis Canada, an ad board member with Knight Therapeutics, and a CONNECT MBC panel member with Gilead Sciences. The other authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Siegel RL, Giaquinto AN, Jemal A. Cancer statistics, 2024. CA Cancer J Clin 2024;74:12-49. [Crossref] [PubMed]
  2. Lei J, Ploner A, Elfström KM, et al. HPV Vaccination and the Risk of Invasive Cervical Cancer. N Engl J Med 2020;383:1340-8. [Crossref] [PubMed]
  3. Chesney JA, Ribas A, Long GV, et al. Randomized, Double-Blind, Placebo-Controlled, Global Phase III Trial of Talimogene Laherparepvec Combined With Pembrolizumab for Advanced Melanoma. J Clin Oncol 2023;41:528-40. [Crossref] [PubMed]
  4. Klein C, Brinkmann U, Reichert JM, et al. The present and future of bispecific antibodies for cancer therapy. Nat Rev Drug Discov 2024;23:301-19. [Crossref] [PubMed]
  5. Hodi FS, O'Day SJ, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 2010;363:711-23. [Crossref] [PubMed]
  6. Sterner RC, Sterner RM. CAR-T cell therapy: current limitations and potential strategies. Blood Cancer J 2021;11:69. [Crossref] [PubMed]
  7. Zhao L, Cao YJ, Engineered T. Cell Therapy for Cancer in the Clinic. Front Immunol 2019;10:2250. [Crossref] [PubMed]
  8. Guedan S, Ruella M, June CH. Emerging Cellular Therapies for Cancer. Annu Rev Immunol 2019;37:145-71. [Crossref] [PubMed]
  9. Shiravand Y, Khodadadi F, Kashani SMA, et al. Immune Checkpoint Inhibitors in Cancer Therapy. Curr Oncol 2022;29:3044-60. [Crossref] [PubMed]
  10. Paijens ST, Vledder A, de Bruyn M, et al. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol 2021;18:842-59. [Crossref] [PubMed]
  11. Seidel JA, Otsuka A, Kabashima K. Anti-PD-1 and Anti-CTLA-4 Therapies in Cancer: Mechanisms of Action, Efficacy, and Limitations. Front Oncol 2018;8:86. [Crossref] [PubMed]
  12. Han Y, Liu D, Li L. PD-1/PD-L1 pathway: current researches in cancer. Am J Cancer Res 2020;10:727-42. [PubMed]
  13. Kciuk M, Yahya EB, Mohamed Ibrahim Mohamed M, et al. Recent Advances in Molecular Mechanisms of Cancer Immunotherapy. Cancers (Basel) 2023;15:2721. [Crossref] [PubMed]
  14. Van Coillie S, Wiernicki B, Xu J. Molecular and Cellular Functions of CTLA-4. Adv Exp Med Biol 2020;1248:7-32. [Crossref] [PubMed]
  15. Iranzo P, Callejo A, Assaf JD, et al. Overview of Checkpoint Inhibitors Mechanism of Action: Role of Immune-Related Adverse Events and Their Treatment on Progression of Underlying Cancer. Front Med (Lausanne) 2022;9:875974. [Crossref] [PubMed]
  16. Uchimiak K, Badowska-Kozakiewicz AM, Sobiborowicz-Sadowska A, et al. Current State of Knowledge on the Immune Checkpoint Inhibitors in Triple-Negative Breast Cancer Treatment: Approaches, Efficacy, and Challenges. Clin Med Insights Oncol 2022;16:11795549221099869. [Crossref] [PubMed]
  17. Adams S, Gray RJ, Demaria S, et al. Prognostic value of tumor-infiltrating lymphocytes in triple-negative breast cancers from two phase III randomized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. J Clin Oncol 2014;32:2959-66. [Crossref] [PubMed]
  18. Loi S, Drubay D, Adams S, et al. Tumor-Infiltrating Lymphocytes and Prognosis: A Pooled Individual Patient Analysis of Early-Stage Triple-Negative Breast Cancers. J Clin Oncol 2019;37:559-69. [Crossref] [PubMed]
  19. Denkert C, Loibl S, Noske A, et al. Tumor-associated lymphocytes as an independent predictor of response to neoadjuvant chemotherapy in breast cancer. J Clin Oncol 2010;28:105-13. [Crossref] [PubMed]
  20. Stanton SE, Adams S, Disis ML. Variation in the Incidence and Magnitude of Tumor-Infiltrating Lymphocytes in Breast Cancer Subtypes: A Systematic Review. JAMA Oncol 2016;2:1354-60. [Crossref] [PubMed]
  21. Thomas R, Al-Khadairi G, Decock J. Immune Checkpoint Inhibitors in Triple Negative Breast Cancer Treatment: Promising Future Prospects. Front Oncol 2020;10:600573. [Crossref] [PubMed]
  22. Cortes J, Rugo HS, Cescon DW, et al. Pembrolizumab plus Chemotherapy in Advanced Triple-Negative Breast Cancer. N Engl J Med 2022;387:217-26. [Crossref] [PubMed]
  23. Schmid P, Cortes J, Dent R, et al. Event-free Survival with Pembrolizumab in Early Triple-Negative Breast Cancer. N Engl J Med 2022;386:556-67. [Crossref] [PubMed]
  24. Schmid P, Cortes J, Pusztai L, et al. Pembrolizumab for Early Triple-Negative Breast Cancer. N Engl J Med 2020;382:810-21. [Crossref] [PubMed]
  25. Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer 2019;7:306. [Crossref] [PubMed]
  26. Valero C, Lee M, Hoen D, et al. Response Rates to Anti-PD-1 Immunotherapy in Microsatellite-Stable Solid Tumors With 10 or More Mutations per Megabase. JAMA Oncol 2021;7:739-43. [Crossref] [PubMed]
  27. Ma W, Xue R, Zhu Z, et al. Increasing cure rates of solid tumors by immune checkpoint inhibitors. Exp Hematol Oncol 2023;12:10. [Crossref] [PubMed]
  28. Patel A, Hande V, Mr K, et al. Effectiveness of Immune Checkpoint Inhibitors in Various Tumor Types Treated by Low, Per-Weight, and Conventional Doses at a Tertiary Care Center in Mumbai. JCO Glob Oncol 2024;10:e2300312. [Crossref] [PubMed]
  29. Zou Y, Zou X, Zheng S, et al. Efficacy and predictive factors of immune checkpoint inhibitors in metastatic breast cancer: a systematic review and meta-analysis. Ther Adv Med Oncol 2020;12:1758835920940928. [Crossref] [PubMed]
  30. Schneider BJ, Naidoo J, Santomasso BD, et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update. J Clin Oncol 2021;39:4073-126. [Crossref] [PubMed]
  31. Choi J, Lee SY. Clinical Characteristics and Treatment of Immune-Related Adverse Events of Immune Checkpoint Inhibitors. Immune Netw 2020;20:e9. [Crossref] [PubMed]
  32. Adams S, Loi S, Toppmeyer D, et al. Pembrolizumab monotherapy for previously untreated, PD-L1-positive, metastatic triple-negative breast cancer: cohort B of the phase II KEYNOTE-086 study. Ann Oncol 2019;30:405-11. [Crossref] [PubMed]
  33. Nanda R, Chow LQ, Dees EC, et al. Pembrolizumab in Patients With Advanced Triple-Negative Breast Cancer: Phase Ib KEYNOTE-012 Study. J Clin Oncol 2016;34:2460-7. [Crossref] [PubMed]
  34. Uliano J, Nicolò E, Corvaja C, et al. Combination immunotherapy strategies for triple-negative breast cancer: current progress and barriers within the pharmacological landscape. Expert Rev Clin Pharmacol 2022;15:1399-413. [Crossref] [PubMed]
  35. Majidpoor J, Mortezaee K. The efficacy of PD-1/PD-L1 blockade in cold cancers and future perspectives. Clin Immunol 2021;226:108707. [Crossref] [PubMed]
  36. Loi S, Adams S, Schmid P, et al. Relationship between tumor infiltrating lymphocyte (TIL) levels and response to pembrolizumab (pembro) in metastatic triple-negative breast cancer (mTNBC): Results from KEYNOTE-086. Ann Oncol 2017;28:v608. [Crossref]
  37. Manuel M, Tredan O, Bachelot T, et al. Lymphopenia combined with low TCR diversity (divpenia) predicts poor overall survival in metastatic breast cancer patients. Oncoimmunology 2012;1:432-40. [Crossref] [PubMed]
  38. Santa-Maria CA, Kato T, Park JH, et al. A pilot study of durvalumab and tremelimumab and immunogenomic dynamics in metastatic breast cancer. Oncotarget 2018;9:18985-96. [Crossref] [PubMed]
  39. Chen N, Higashiyama N, Hoyos V. Predictive Biomarkers of Immune Checkpoint Inhibitor Response in Breast Cancer: Looking beyond Tumoral PD-L1. Biomedicines 2021;9:1863. [Crossref] [PubMed]
  40. Li X, Zhang S, Guo G, et al. Gut microbiome in modulating immune checkpoint inhibitors. EBioMedicine 2022;82:104163. [Crossref] [PubMed]
  41. Indini A, Rijavec E, Ghidini M, et al. Impact of BMI on Survival Outcomes of Immunotherapy in Solid Tumors: A Systematic Review. Int J Mol Sci 2021;22:2628. [Crossref] [PubMed]
  42. Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J 2017;474:1823-36. [Crossref] [PubMed]
  43. Bull MJ, Plummer NT. Part 1: The Human Gut Microbiome in Health and Disease. Integr Med (Encinitas) 2014;13:17-22. [PubMed]
  44. Hou K, Wu ZX, Chen XY, et al. Microbiota in health and diseases. Signal Transduct Target Ther 2022;7:135. [Crossref] [PubMed]
  45. Algrafi AS, Jamal AA, Ismaeel DM. Microbiota as a New Target in Cancer Pathogenesis and Treatment. Cureus 2023;15:e47072. [Crossref] [PubMed]
  46. Sadrekarimi H, Gardanova ZR, Bakhshesh M, et al. Emerging role of human microbiome in cancer development and response to therapy: special focus on intestinal microflora. J Transl Med 2022;20:301. [Crossref] [PubMed]
  47. Sun J, Chen F, Wu G. Potential effects of gut microbiota on host cancers: focus on immunity, DNA damage, cellular pathways, and anticancer therapy. ISME J 2023;17:1535-51. [Crossref] [PubMed]
  48. Vivarelli S, Salemi R, Candido S, et al. Gut Microbiota and Cancer: From Pathogenesis to Therapy. Cancers (Basel) 2019;11:38. [Crossref] [PubMed]
  49. Iida N, Dzutsev A, Stewart CA, et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013;342:967-70. [Crossref] [PubMed]
  50. Paulos CM, Wrzesinski C, Kaiser A, et al. Microbial translocation augments the function of adoptively transferred self/tumor-specific CD8+ T cells via TLR4 signaling. J Clin Invest 2007;117:2197-204. [Crossref] [PubMed]
  51. Lu Y, Yuan X, Wang M, et al. Gut microbiota influence immunotherapy responses: mechanisms and therapeutic strategies. J Hematol Oncol 2022;15:47. [Crossref] [PubMed]
  52. von Itzstein MS, Gonugunta AS, Sheffield T, et al. Association between Antibiotic Exposure and Systemic Immune Parameters in Cancer Patients Receiving Checkpoint Inhibitor Therapy. Cancers (Basel) 2022;14:1327. [Crossref] [PubMed]
  53. Jiang S, Geng S, Chen Q, et al. Effects of Concomitant Antibiotics Use on Immune Checkpoint Inhibitor Efficacy in Cancer Patients. Front Oncol 2022;12:823705. [Crossref] [PubMed]
  54. Domzaridou E, Van Staa T, Renehan AG, et al. The Impact of Oral Antibiotics Prior to Cancer Diagnosis on Overall Patient Survival: Findings from an English Population-Based Cohort Study. Curr Oncol 2023;30:8434-43. [Crossref] [PubMed]
  55. Shah H, Ng TL. A narrative review from gut to lungs: non-small cell lung cancer and the gastrointestinal microbiome. Transl Lung Cancer Res 2023;12:909-26. [Crossref] [PubMed]
  56. Najmi M, Tran T, Witt RG, et al. Modulation of the Gut Microbiome to Enhance Immunotherapy Response in Metastatic Melanoma Patients: A Clinical Review. Dermatol Ther (Heidelb) 2022;12:2489-97. [Crossref] [PubMed]
  57. Kustrimovic N, Bombelli R, Baci D, et al. Microbiome and Prostate Cancer: A Novel Target for Prevention and Treatment. Int J Mol Sci 2023;24:1511. [Crossref] [PubMed]
  58. Li S, Zhu S, Yu J. The role of gut microbiota and metabolites in cancer chemotherapy. J Adv Res 2023;S2090-1232(23)00366-1.
  59. Viaud S, Saccheri F, Mignot G, et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 2013;342:971-6. [Crossref] [PubMed]
  60. Huang J, Liu W, Kang W, et al. Effects of microbiota on anticancer drugs: Current knowledge and potential applications. EBioMedicine 2022;83:104197. [Crossref] [PubMed]
  61. Walters KE, Martiny JBH. Alpha-, beta-, and gamma-diversity of bacteria varies across habitats. PLoS One 2020;15:e0233872. [Crossref] [PubMed]
  62. Di Modica M, Gargari G, Regondi V, et al. Gut Microbiota Condition the Therapeutic Efficacy of Trastuzumab in HER2-Positive Breast Cancer. Cancer Res 2021;81:2195-206. [Crossref] [PubMed]
  63. Wu AH, Vigen C, Tseng C, et al. Effect of Chemotherapy on the Gut Microbiome of Breast Cancer Patients During the First Year of Treatment. Breast Cancer (Dove Med Press) 2022;14:433-51. [Crossref] [PubMed]
  64. Li Y, Dong B, Wu W, et al. Metagenomic Analyses Reveal Distinct Gut Microbiota Signature for Predicting the Neoadjuvant Chemotherapy Responsiveness in Breast Cancer Patients. Front Oncol 2022;12:865121. [Crossref] [PubMed]
  65. Terrisse S, Derosa L, Iebba V, et al. Intestinal microbiota influences clinical outcome and side effects of early breast cancer treatment. Cell Death Differ 2021;28:2778-96. [Crossref] [PubMed]
  66. de Sousa RB, Ajami N, Keenan TE, et al. Abstract P3-09-16: Fecal microbiome and association with outcomes among patients (pts) receiving eribulin (E) +/- pembrolizumab (P) for hormone receptor positive (HR+) metastatic breast cancer (MBC). Cancer Res 2020;80:P3-09-16.
  67. Barroso-Sousa R, Li T, Damania AV, et al. Abstract PD11-05: PD11-05 Gut microbiome signatures correlate with overall survival among patients receiving eribulin with or without pembrolizumab for hormone receptor-positive metastatic breast cancer. Cancer Res 2023;83:PD11-05. [Crossref]
  68. Teng N, Dalby MJ, Kiu R, et al. 14P Gut and oral microbiota profiling in patients (pts) with hormone receptor-positive (HR+) metastatic breast cancer (MBC) receiving pembrolizumab (P) plus eribulin (E): CALADRIO. Ann Oncol 2022;33:S129-30. [Crossref]
  69. Wong CW, Yost SE, Lee JS, et al. Abstract 336: Gut microbiome predicts response to CDK4/6 inhibitor and immune check point inhibitor combination in patients with hormone receptor positive metastatic breast cancer. Cancer Res 2021;81:336. [Crossref]
  70. Yuan Y, Highlander S, Yost SE, et al. Abstract P5-06-26: Gut microbiome profiling of patients with metastatic breast cancer undergoing immune checkpoint inhibitor therapy. Cancer Res 2020;80:P5-06-26.
  71. Chun B, Goodyear S, Rice-Stitt T, et al. Abstract P1-04-02: Gut microbiome diversity correlates with tumor PD-L1 status in metastatic triple negative breast cancer (mTNBC): correlative analysis of gut microbiome and tumoral biomarkers. Cancer Res 2023;83:P1-04-02.
  72. Kulkarni AA, Jain A, Jewett PI, et al. Association of antibiotic exposure with residual cancer burden in HER2-negative early stage breast cancer. NPJ Breast Cancer 2024;10:24. [Crossref] [PubMed]
  73. Rosean CB, Bostic R, Feng TY, et al. Abstract A64: Commensal dysbiosis alters the tumor microenvironment in breast cancer and diminishes efficacy of PD-L1 blockade. Cancer Immunol Res 2020;8:A64. [Crossref]
  74. Pingili AK, Chaib M, Sipe LM, et al. Immune checkpoint blockade reprograms systemic immune landscape and tumor microenvironment in obesity-associated breast cancer. Cell Rep 2021;35:109285. [Crossref] [PubMed]
  75. Bohm MS, Sipe LM, Joseph SC, et al. 1325 Bariatric surgery-induced modulation of gut microbiota improves response to αPD-1 immunotherapy in preclinical model of breast cancer after bariatric surgery. J Immunother Cancer 2023;11:1476.
  76. Clear K, Stirling E, Wilson A, et al. 1304 Dietary patterns associated with increased abundance of Akkermansia muciniphila potentiates anti-PD-L1 immune checkpoint blockade response in triple-negative breast cancer. J Immunother Cancer 2022;10:A1352.
  77. Clear KYJ, Wilson AS, Stirling ER, et al. Abstract 1423: Harnessing the gut microbiome to modulate immune checkpoint blockade response in triple-negative breast cancer. Cancer Res 2024;84:1423. [Crossref]
  78. Ferrari V, Melacarne A, Algieri F, et al. 835 Harnessing the microbiota to increase response rates to immunotherapy. J Immunother Cancer 2021;9:A876.
  79. Kim H, Oh R, Park S, et al. Abstract 72: Bifidobacterium longum RAPO enhances efficacy of anti-PD-1 immunotherapy in a mouse model of triple-negative breast cancer. Cancer Res 2021;81:72. [Crossref]
  80. Li H, Dong T, Tao M, et al. Fucoidan enhances the anti-tumor effect of anti-PD-1 immunotherapy by regulating gut microbiota. Food Funct 2024;15:3463-78. [Crossref] [PubMed]
  81. Zhou CB, Zhou YL, Fang JY. Gut Microbiota in Cancer Immune Response and Immunotherapy. Trends Cancer 2021;7:647-60. [Crossref] [PubMed]
  82. Mao J, Wang D, Long J, et al. Gut microbiome is associated with the clinical response to anti-PD-1 based immunotherapy in hepatobiliary cancers. J Immunother Cancer 2021;9:e003334. [Crossref] [PubMed]
  83. Han Z, Cheng S, Dai D, et al. The gut microbiome affects response of treatments in HER2-negative advanced gastric cancer. Clin Transl Med 2023;13:e1312. [Crossref] [PubMed]
  84. McCulloch JA, Davar D, Rodrigues RR, et al. Intestinal microbiota signatures of clinical response and immune-related adverse events in melanoma patients treated with anti-PD-1. Nat Med 2022;28:545-56. [Crossref] [PubMed]
  85. Duttagupta S, Hakozaki T, Routy B, et al. The Gut Microbiome from a Biomarker to a Novel Therapeutic Strategy for Immunotherapy Response in Patients with Lung Cancer. Curr Oncol 2023;30:9406-27. [Crossref] [PubMed]
  86. Davar D, Dzutsev AK, McCulloch JA, et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science 2021;371:595-602. [Crossref] [PubMed]
  87. Baruch EN, Youngster I, Ben-Betzalel G, et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 2021;371:602-9. [Crossref] [PubMed]
  88. Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018;359:91-7. [Crossref] [PubMed]
  89. Gopalakrishnan V, Spencer CN, Nezi L, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018;359:97-103. [Crossref] [PubMed]
  90. Ma J, Zhu W, Liu B. Role of gut microbiome in the outcome of cancer immunotherapy. Int J Cancer 2021; Epub ahead of print. [Crossref] [PubMed]
  91. DeGruttola AK, Low D, Mizoguchi A, et al. Current Understanding of Dysbiosis in Disease in Human and Animal Models. Inflamm Bowel Dis 2016;22:1137-50. [Crossref] [PubMed]
  92. Hrncir T. Gut Microbiota Dysbiosis: Triggers, Consequences, Diagnostic and Therapeutic Options. Microorganisms 2022;10:578. [Crossref] [PubMed]
  93. Buchta Rosean C, Bostic RR, Ferey JCM, et al. Pre-existing commensal dysbiosis is a host-intrinsic regulator of tissue inflammation and tumor cell dissemination in hormone receptor-positive breast cancer. Cancer Res 2019;79:3662-75. [Crossref] [PubMed]
  94. Breton J, Galmiche M, Déchelotte P. Dysbiotic Gut Bacteria in Obesity: An Overview of the Metabolic Mechanisms and Therapeutic Perspectives of Next-Generation Probiotics. Microorganisms 2022;10:452. [Crossref] [PubMed]
  95. Cuevas-Sierra A, Ramos-Lopez O, Riezu-Boj JI, et al. Diet, Gut Microbiota, and Obesity: Links with Host Genetics and Epigenetics and Potential Applications. Adv Nutr 2019;10:S17-30. [Crossref] [PubMed]
  96. Lohmann AE, Soldera SV, Pimentel I, et al. Association of Obesity With Breast Cancer Outcome in Relation to Cancer Subtypes: A Meta-Analysis. J Natl Cancer Inst 2021;113:1465-75. [Crossref] [PubMed]
  97. Lurienne L, Cervesi J, Duhalde L, et al. NSCLC Immunotherapy Efficacy and Antibiotic Use: A Systematic Review and Meta-Analysis. J Thorac Oncol 2020;15:1147-59. [Crossref] [PubMed]
  98. Kulkarni AA, Ebadi M, Zhang S, et al. Comparative analysis of antibiotic exposure association with clinical outcomes of chemotherapy versus immunotherapy across three tumour types. ESMO Open 2020;5:e000803. [Crossref] [PubMed]
  99. Gopalakrishnan V, Sellman BR, Cohen TS, et al. Antibiotics and Immunotherapy: Too Much of Anything is Bad! Eur Urol 2020;78:544-5. [Crossref] [PubMed]
  100. Mohiuddin JJ, Chu B, Facciabene A, et al. Association of Antibiotic Exposure With Survival and Toxicity in Patients With Melanoma Receiving Immunotherapy. J Natl Cancer Inst 2021;113:162-70. [Crossref] [PubMed]
  101. Lu K, Dong S, Wu X, et al. Probiotics in Cancer. Front Oncol 2021;11:638148. [Crossref] [PubMed]
  102. Ting NL, Lau HC, Yu J. Cancer pharmacomicrobiomics: targeting microbiota to optimise cancer therapy outcomes. Gut 2022;71:1412-25. [Crossref] [PubMed]
  103. Kim H, Oh R, Heo JW, et al. Abstract LB532: Bifidobacterium longum RAPO alleviates the risk of immune-related adverse events of anti-PD-1 immunotherapy in a mouse model of triple-negative breast cancer. Cancer Res 2022;82:LB532. [Crossref]
  104. Luthuli S, Wu S, Cheng Y, et al. Therapeutic Effects of Fucoidan: A Review on Recent Studies. Mar Drugs 2019;17:487. [Crossref] [PubMed]
  105. Sun J, Sun J, Song B, et al. Fucoidan inhibits CCL22 production through NF-κB pathway in M2 macrophages: a potential therapeutic strategy for cancer. Sci Rep 2016;6:35855. [Crossref] [PubMed]
  106. Lin Y, Qi X, Liu H, et al. The anti-cancer effects of fucoidan: a review of both in vivo and in vitro investigations. Cancer Cell Int 2020;20:154. [Crossref] [PubMed]
  107. Xue M, Ji X, Liang H, et al. The effect of fucoidan on intestinal flora and intestinal barrier function in rats with breast cancer. Food Funct 2018;9:1214-23. [Crossref] [PubMed]
  108. Tang Y, Pu Q, Zhao Q, et al. Effects of Fucoidan Isolated From Laminaria japonica on Immune Response and Gut Microbiota in Cyclophosphamide-Treated Mice. Front Immunol 2022;13:916618. [Crossref] [PubMed]
  109. Liu X, Zhang Y, Li W, et al. Fucoidan Ameliorated Dextran Sulfate Sodium-Induced Ulcerative Colitis by Modulating Gut Microbiota and Bile Acid Metabolism. J Agric Food Chem 2022;70:14864-76. [Crossref] [PubMed]
  110. Matson V, Fessler J, Bao R, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018;359:104-8. [Crossref] [PubMed]
  111. Chung MW, Kim MJ, Won EJ, et al. Gut microbiome composition can predict the response to nivolumab in advanced hepatocellular carcinoma patients. World J Gastroenterol 2021;27:7340-9. [Crossref] [PubMed]
  112. Rutkowski MR, Rosean CM, Bostic R, et al. Commensal dysbiosis modulates the tumor microenvironment in breast cancer and diminishes efficacy of PD-L1 inhibition. J Immunol 2018;200:122-7. [Crossref]
  113. Loi S, Curigliano G, Salgado RF, et al. LBA20 A randomized, double-blind trial of nivolumab (NIVO) vs placebo (PBO) with neoadjuvant chemotherapy (NACT) followed by adjuvant endocrine therapy (ET) ± NIVO in patients (pts) with high-risk, ER+ HER2− primary breast cancer (BC). Ann Oncol 2023;34:S1259-60. [Crossref]
  114. Bosco N, Noti M. The aging gut microbiome and its impact on host immunity. Genes Immun 2021;22:289-303. [Crossref] [PubMed]
  115. Ghosh TS, Das M, Jeffery IB, et al. Adjusting for age improves identification of gut microbiome alterations in multiple diseases. Elife 2020;9:e50240. [Crossref] [PubMed]
  116. Yatsunenko T, Rey FE, Manary MJ, et al. Human gut microbiome viewed across age and geography. Nature 2012;486:222-7. [Crossref] [PubMed]
  117. Witt RG, Cass SH, Tran T, et al. Gut Microbiome in Patients With Early-Stage and Late-Stage Melanoma. JAMA Dermatol 2023;159:1076-84. [Crossref] [PubMed]
  118. Ivleva EA, Grivennikov SI. Microbiota-driven mechanisms at different stages of cancer development. Neoplasia 2022;32:100829. [Crossref] [PubMed]
doi: 10.21037/tbcr-24-14
Cite this article as: Hrubesz G, Leigh J, Ng TL. Understanding the relationship between breast cancer, immune checkpoint inhibitors, and gut microbiota: a narrative review. Transl Breast Cancer Res 2024;5:31.

Download Citation