[with Marc's comment] Fecal microbiota transplantation improves anti-PD-1 inhibitor efficacy in unresectable or metastatic solid cancers refractory to anti-PD-1 inhibitor

15 views
Skip to first unread message

rael-science

unread,
Jul 26, 2024, 8:13:40 AMJul 26
to rael science

The Raelian Movement
for those who are not afraid of the future : http://www.rael.org

Get Rael-Science on Facebook: http://www.facebook.com/raelscience

Get Rael-Science on Twitter: https://twitter.com/rael_science





Comment by Marc Rivard, M.D. and raelian bishop: Immunotherapy is a very promising approach for cancers. However, some people respond less well to it. This small study demonstrates the potential role of the intestinal microbiota. Indeed, by performing a fecal transplant from healthy patients to cancer patients, the latter respond better to immunotherapy. In certain immunotherapy protocols, if a person has taken antibiotics before the sessions, a fecal transplant is often carried out. Antibiotics save lives, but you have to be certain that there is a real medical indication to take them. Always ask your doctor if it is really necessary to take antibiotics. They are often prescribed too quickly.



Fecal microbiota transplantation improves anti-PD-1 inhibitor efficacy in unresectable or metastatic solid cancers refractory to anti-PD-1 inhibitor

Published:July 25, 2024DOI:https://doi.org/10.1016/j.chom.2024.06.010

Highlights

  • FMT with anti-PD-1 shows benefits in advanced solid cancers resistant to anti-PD-1
  • FMT induced partial response to anti-PD-1 in R7 recipients with enhanced immunity
  • Lactobacillus salivarius and Bacteroides plebeius may inhibit T cell activity
  • Prevotella merdae Immunoactis boosts T cell activity and reduces tumor growth

Summary

The gut microbiome significantly influences immune responses and the efficacy of immune checkpoint inhibitors. We conducted a clinical trial (NCT04264975) combining an anti-programmed death-1 (PD-1) inhibitor with fecal microbiota transplantation (FMT) from anti-PD-1 responder in 13 patients with anti-PD-1-refractory advanced solid cancers. FMT induced sustained microbiota changes and clinical benefits in 6 of 13 patients, with 1 partial response and 5 stable diseases, achieving an objective response rate of 7.7% and a disease control rate of 46.2%. The clinical response correlates with increased cytotoxic T cells and immune cytokines in blood and tumors. We isolated Prevotella merdae Immunoactis from a responder to FMT, which stimulates T cell activity and suppresses tumor growth in mice by enhancing cytotoxic T cell infiltration. Additionally, we found Lactobacillus salivarius and Bacteroides plebeius may inhibit anti-tumor immunity. Our findings suggest that FMT with beneficial microbiota can overcome resistance to anti-PD-1 inhibitors in advanced solid cancers, especially gastrointestinal cancers.

Graphical abstract

Keywords

Introduction

Immune checkpoint inhibitors (ICIs) targeting cytotoxic T lymphocyte antigen (CTLA-4) or programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) pathways have transformed cancer treatment across a wide range of cancer types. However, ICIs have not universally demonstrated effective outcomes across all cancer types, and their efficacy in those where they have shown success, such as gastroesophageal, liver, bladder, and head and neck cancers, remains modest.  Most patients initially responsive to ICIs ultimately face disease progression due to secondary resistance.  Although recent research has progressed in elucidating predictive biomarkers, understanding resistance mechanisms in immuno-oncology, and improving treatment outcomes by combining ICIs with other therapies, overcoming primary and secondary resistance remains significantly challenging. , ,
The gut microbiome has emerged as a key player in shaping immune responses and affecting the efficacy of ICIs. Studies have shown that the presence of specific commensals in patient stool samples correlates with clinical response to ICIs. , ,  Preclinical experiments have demonstrated that a specific gut microbiome or fecal microbiota transplantation (FMT) using fecal material from patients responding to ICIs can induce tumor regression, augment T cell responses, and improve the anti-tumor efficacy of ICIs. , ,  Recent pilot clinical trials have shown that the combination of anti-PD-1 inhibitors and FMT could potentially overcome resistance to anti-PD-1 therapy by altering the gut microbiome and reprogramming the tumor microenvironment in a subset of patients with anti-PD-1-refractory melanoma. ,  In this study, we aimed to evaluate the potential of FMT in overcoming resistance to anti-PD-(L)1 inhibitors in patients with advanced solid cancers refractory to these inhibitors (NCT04264975) and identify specific commensal bacteria that play a causal role in the efficacy of FMT and ICI treatments.

Results

Study design and participant characteristics

This FMT trial was a prospective, single-arm, and single-center study of the combination of FMT and an anti-PD-(L)1 inhibitor (Figure 1A). The study included two distinct participant categories: donors and recipients (Figures 1A and S1). Donors were characterized by a durable complete response (CR) or partial response (PR) according to Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 for at least 6 months following anti-PD-(L)1 monotherapy for unresectable or metastatic solid tumors. Recipients, on the other hand, were individuals with confirmed disease progression during anti-PD-(L)1-based therapy, monotherapy, or combination therapy for unresectable or metastatic solid tumors. To eliminate the native gut microbiota, recipients were given oral antibiotics of amoxicillin clavulanate for a duration of 5 days before FMT (only prior to the first FMT). FMT procedures were conducted via colonoscopy, followed by the continuation or reintroduction of an anti-PD-(L)1 inhibitor (administered at the standard dose and schedule) until either unacceptable toxicity or disease progression occurred. Subsequent FMT sessions from the same or different donors were permitted, at the investigator's discretion, with a time interval of 2–4 weeks prior to the initial response assessment or in cases where clinical response was not evident. Sequential samples of blood, stool, and tumor biopsies were collected both before and after FMT. Response assessment was carried out every 6–8 weeks using computed tomography (CT) scans, following RECIST v1.1.
Figure thumbnail gr1
Figure 1Clinical responses to FMT combined with nivolumab in patients with advanced solid cancer refractory to nivolumab
Between January 2019 and August 2020, we recruited 13 FMT recipients with metastatic gastric cancer (GC) (n = 4), esophageal squamous cell carcinoma (ESCC) (n = 5), and hepatocellular carcinoma (HCC) (n = 4) (Tables 1 and S1). The median age was 60 years (range, 38–76), and all had undergone heavy prior treatment (median of 10 systemic treatments, range, 4–34). All patients had confirmed disease progression on nivolumab monotherapy, with six (46.2%) displaying primary resistance and seven (53.8%) displaying secondary resistance. Enrollment occurred immediately after confirming disease progression, and FMT was administered while continuing nivolumab. All recipients’ tumors were microsatellite stable (MSS), with seven (53.8%) having PD-L1-positive (combined positive score [CPS] ≥ 1) and four (30.8%) having PD-L1 CPS ≥ 5. We recruited six FMT donors: four with HCC and one each with GC and ESCC. These donors achieved and maintained a CR (n = 4) or PR (n = 2) for at least 1 year with nivolumab or pembrolizumab, except one with an ongoing response of 7.9 months (Table 2). All donors’ tumors were MSS and PD-L1-negative, except for one donor with low PD-L1 expression (CPS of 7). The median tumor mutation burden (TMB) in the donors was 10.9 mutations per megabase (range, 9.4–37.5).
Table 1Profiles of recipients in FMT study (n = 13)

Age Sex ECOG performance status Cancer type Disease status Treatment line of ICI administered before FMT No. of cycles of ICI before FMT Type of ICI before and during the FMT trial Best response to ICI before FMT PD-L1 status TMB (mutations/Mb) Microsatellite status
Recipient #1 76 male 1 ESCC metastatic 4 5 nivolumab PD TPS 0% CPS 1 12.5 MSS
Recipient #2 50 female 1 ESCC metastatic 3 4 nivolumab PD TPS 0% CPS 0 15.6 MSS
Recipient #3 67 male 1 HCC metastatic 3 7 nivolumab PD TPS 0% CPS 0 20.3 MSS
Recipient #5 55 female 1 ESCC metastatic 4 13 nivolumab PR TPS 0% CPS 0 12.5 MSS
Recipient #6 60 male 1 HCC metastatic 2 12 nivolumab SD TPS 0% CPS 0 12.5 MSS
Recipient #7 47 male 1 HCC metastatic 3 5 nivolumab PD TPS 1% CPS 3 12.5 MSS
Recipient #8 64 male 0 HCC metastatic 3 24 nivolumab PR TPS 0% CPS 0 15.6 MSS
Recipient #9 67 male 1 ESCC metastatic 4 5 nivolumab PD TPS 1% CPS 1 12.5 MSS
Recipient #10 46 male 1 GC metastatic 5 21 nivolumab PR TPS 30% CPS 60 17.2 MSS
Recipient #11 42 female 1 GC metastatic 3 10 nivolumab SD TPS 0% CPS 0 10.9 MSS
Recipient #13 38 male 1 GC metastatic 3 5 nivolumab PD TPS 30% CPS 30 15.6 MSS
Recipient #14 70 male 1 ESCC metastatic 3 21 nivolumab PR TPS 15% CPS 15 28.1 MSS
Recipient #15 64 male 1 GC metastatic 4 34 nivolumab PR TPS 0% CPS 5 20.3 MSS
ECOG, Eastern Cooperative Oncology Group; ICI, immune checkpoint inhibitor; FMT, fecal microbiota transplantation; PD-L1, programmed death-ligand 1; TMB, tumor mutation burden; Mb, megabase; ESCC, esophageal squamous cell carcinoma; PD, progressive disease; TPS, tumor proportion score; CPS, combined positive score; MSS, microsatellite stable; HCC, hepatocellular carcinoma; PR, partial response; SD, stable disease; GC, gastric adenocarcinoma.
Table 2Profiles of donors in FMT study (n = 6)

Age Sex Cancer type Anti-PD-1 Response to anti-PD-1 DoR (months) Genetic alteration TMB (/Mb) Microsatellite status PD-L1 status EBV status
Donor #1 78 male HCC pembrolizumab CR 58.7+ ARID1A S634 mutation, NFE2L2 G81C mutation, MCL1 amplification, MDM4 amplification, AKT3 amplification 9.4 MSS TPS 0% CPS 0 N/A
Donor #2 66 male GC nivolumab CR 72.7+ ERBB2 amplification, TP53 Q192 mutation 37.5 MSS TPS 7% CPS 7 negative
Donor #3 63 male HCC nivolumab PR 7.9 no oncogenic mutation 15.6 MSS TPS 0% CPS 0 N/A
Donor #4 78 male ESCC nivolumab PR 15.4 FBXW7 X327_splice alteration, TP53 P223Afs3 mutation, MYC amplification, CDKN2A deletion, CDKN2B deletion 10.9 MSS TPS 0% CPS 0 N/A
Donor #5 62 male HCC nivolumab CR 31.5+ N/A (QC fail due to low tumor cellularity) N/A MSS TPS 0% CPS 0 N/A
Donor #6 69 male HCC nivolumab CR 25.7+ TP53 P278R mutation 10.9 MSS TPS 0% CPS 0 N/A
PD-1, programmed death-1; DoR, duration of response; TMB, tumor mutation burden; Mb, megabase; PD-L1, programmed death-ligand 1; EBV, Epstein-Barr virus; HCC, hepatocellular carcinoma; CR, complete response; MSS, microsatellite stable; TPS, tumor proportion score; CPS, combined positive score; N/A, not available; GC, gastric cancer; PR, partial response; ESCC, esophageal squamous cell carcinoma; QC, quality control.

Administration of study treatment and treatment-related AEs

All patients were treated with a combination of nivolumab with FMT, which involved continuing nivolumab treatment after failure to prior nivolumab therapy. A median cycle of nivolumab after FMT was five cycles (range, 1–27). Of the 13 recipients, 7 received subsequent FMTs from the same (n = 4) and/or different donors (n = 4) after the first FMT. Among them, one recipient first underwent subsequent FMTs from the same donor and then from a different donor.
Regarding safety profiles, treatment-related adverse events (AEs) were minimal (Table S2). Out of the thirteen patients, seven patients (53.8%) experienced at least one treatment-related AE, all of which were either grade 1 or 2, except for one patient who experienced grade 3 immune-related gastritis. The most common treatment-related AEs were skin pruritus (n = 5, 38.5%), followed by skin rash (n = 2, 15.4%), and hypothyroidism (n = 2, 15.4%). Endocrine AEs were managed with hormone replacement therapy, and immune-related gastritis (CTCAE; Common Terminology Criteria for Adverse Events, grade 3) was managed with systemic corticosteroids.

Efficiency of FMT and clinical response after FMT

To evaluate the efficiency of FMT engraftment, we performed 16S rRNA sequencing analysis on stool samples obtained from both recipients and donors. Analysis using Bray-Curtis distance (BCD) revealed that the microbial compositions of most recipients underwent significant changes from their baseline composition after FMT (Figures 1B and S2). To quantify these changes, we calculated the engraftment distance (ED) based on the BCDs between baseline of recipient and donor samples (Figure S3). A decrease in the ED from baseline indicates a change in microbial composition. Since all recipients in our FMT trial exhibited reduced ED, this demonstrates that our FMT trial induced alterations in the microbial compositions of all recipients (Figure 1C). Remarkably, the microbial compositions of recipient#7 (R7) closely resembled those of the respective donor as early as 1 day after both the first and second FMT procedures, and this similarity persisted for 282 days following the second FMT procedure (Figure 1B).
Among the thirteen recipients, one achieved a PR, and five exhibited stable disease (SD) following FMT in conjunction with continued nivolumab treatment. These outcomes, representing an objective response rate of 7.7% (1/13) and a disease control rate of 46.2% (6/13), underscore the combined therapeutic impact of FMT and ongoing immunotherapy (Figure 1D). Notably, R7, who was initially metastatic HCC with primary resistance to nivolumab, achieved a PR following FMT with continued immunotherapy and exhibited a significant reduction in tumor size (Figures 1D and 1E). Our analysis demonstrates that FMT with immunotherapy can effectively modify the gut microbiome of recipients, leading to significant clinical efficacy, particularly exemplified by the remarkable case of R7.

Immune changes in systemic and tumor microenvironments and clinical outcomes following FMT

We focused on R7 and observed longitudinal changes in several factors. Initially, R7 showed progressive disease (PD) with a 22.4% increase in target lesions 6 weeks post the first FMT from donor #1 (D1) (Figures 2A and 2B). After the second FMT from donor #5 (D5), tumor progression slowed, with only a 13% increase in target lesions compared to the first PD assessment. 8 weeks after the second FMT, there was a significant 30.5% reduction in tumor size (Figures 2A and 2B, top). Eventually, R7 achieved a PR with a maximum tumor size reduction of 47.7% from baseline before first FMT (Figures 2A and 2B). Tumor markers, including serum α-fetoprotein (AFP) and protein induced by vitamin K absence or antagonist-II (PIVKA-II), also significantly decreased 8 weeks post-second FMT (Figure 2B, bottom).
Figure thumbnail gr2
Figure 2Analysis of longitudinal tumor reduction and immune-related changes in recipient #7
Using CyTOF (Cytometry by time of flight), we evaluated the systemic immune changes with the response to FMT combined with nivolumab. R7 showed gradual increase in CD8+ T cells and CD8+CCR7CD27 terminal effector T (Tte) cells and a decrease in CD4+CD25+CD127CCR4+ regulatory T (Treg) cells from baseline (Figures 2C and 2D). At 23 weeks post-second FMT, Tte cells increased by 255% and Treg cells decreased by 81% (Figure 2C). These immune changes correlated with R7’s clinical response and were more pronounced after the second FMT (Figure 2D). This immune activation was absent or modest in other patients (Figure S4). Using the Simoa HD-1 Analyzer and Simoa SP-X, we found elevated levels of interferon (IFN)-γ, tumor necrosis factor alpha (TNF-α), interleukin (IL)-2, IL-7, IL-15, and IL-6 in R7 specifically after the second FMT but not the first FMT, which correlated with the clinical response (Figure 2E). On the other hand, R6, R9, and R14, who achieved a SD, showed an upward trend in levels of some of these cytokines, including IL-6, following FMT (Figure S5). This suggests that although FMT did not lead to a significant reduction in tumor size for these patients, it still resulted in elevated level of these cytokines to some extent. In our assessment of tumor-infiltrating immune cells, which play a critical role in the response to anti-PD-1 therapy, using multiplex immunohistochemistry (IHC), we observed notable changes in R7. 4 weeks after the second FMT, R7 showed a significant increase in tumor-infiltrating cytotoxic T cells (from 14 to 361 cells/mm2) and major histocompatibility complex (MHC)-II+ M1 macrophages (from 25 to 174 cells/mm2), while the number of CD4+FOXP3+CD8CD20 Treg cells remained extremely low (from 5 to 4 cells/mm2). In contrast, no comparable immune activation was noted subsequent to the first FMT (Figures 2F and 2G). This highlights a noteworthy immune activation specifically following the second FMT. R7’s enhanced immune response was not mirrored in all patients. In R8 (best response, PD) and R14 (best response, SD), although increases in cytotoxic T cells were also observed, M1 macrophage increases were absent or minimal (Figure S6). Overall, R7 showed notable tumor reduction and enhanced immune response following the second FMT, with significant changes in systemic and tumor-infiltrating immune cells.
Following the second FMT and continued immunotherapy, R7 displayed significant tumor reduction and an enhanced immune response. The CT scan showed diffuse gastric wall thickening, while esophagogastroduodenoscopy (EGD) revealed multiple gastric ulcers, confirmed as acute lymphocytic gastritis leading to a PR at 8th week post-treatment. However, 11 weeks after the second FMT, the patient reported progressive epigastric pain and nausea, diagnosed as immune-related gastritis via EGD and abdominal CT scan (Figures S7A and S7B). The CT scan showed diffuse gastric wall thickening, while EGD revealed multiple gastric ulcers, confirmed as acute lymphocytic gastritis (Figure S7B). This suggested an overactive immune response in the intestine. In order to manage the patient’s symptoms, systemic corticosteroid treatment was initiated approximately 12 weeks after the second FMT and continued long term due to fluctuating symptoms. Despite the potential negative impact of prolonged steroid use, R7 experienced disease progression with a PFS (Progression-Free Survival) of 8.7 months post-second FMT. A subsequent FMT was planned using the same D5 fecal material. Unfortunately, the patient sustained a femoral fracture requiring surgery. After recovery, the third FMT resulted in SD, but the fourth FMT failed to halt tumor growth (Figures S7A and S7C). Multiplex IHC analysis of tumor biopsies before and after the third FMT showed a mild increase in cytotoxic T cells, MHC-II+ M1 macrophages, and helper T cells, though not as pronounced as after the second FMT (Figure S7D). Therefore, in our subsequent analysis aimed at identifying potential causative bacterial strains, we intended to focus on examining the changes that occurred during the second FMT while excluding the third and fourth FMTs.

Uncovering bacterial strains associated with clinical outcomes after FMT

To identify the specific microbiota responsible for the clinical benefits of FMT combined with an anti-PD-1 inhibitor, we analyzed microbial changes in R7 using 16s rRNA sequencing. Our analysis showed that following the first FMT from D1, Bacteroides levels increased from 2.87% to 6.4%, while Prevotella levels decreased from 19.75% to 0% (Figure 3A). Conversely, after the second FMT from D5, Bacteroides decreased from 6.4% to 0.92%, while Prevotella increased from 0% to 38.48% (Figure 3A). This shift highlighted a reciprocal trend between Prevotella and Bacteroides (Figure 3B). To pinpoint the bacteria contributing to the favorable efficacy of FMT with anti-PD-1 therapy, we identified bacteria significantly more prevalent in D5 than in D1 and those that significantly emerged in R7 post-second FMT compared to post-first FMT. This led to the identification of 47 common bacteria (Figure 3C). Excluding bacteria present in R7's baseline stool sample, we narrowed the list to 34 unique bacteria. Prevotella merdae (type strain; sp. Marseille-P4119)  and Prevotella stercorea ranked highest based on their LDA (Linear Discriminant Analysis) scores (Figure 3C; Table S3). These species were significantly more abundant in D5 and R7 post-second FMT compared to D1 and R7 post-first FMT, respectively (Figures 3D and S8A). Given the prominence of P. merdae in the stool samples after the second FMT and its high LDA score, we prioritized further investigations on this species, identifying it as P. merdae sp. Marseille-P4119 by using clade-specific marker gene analysis through shotgun metagenomic sequencing.
Figure thumbnail gr3
Figure 3Discovery of key bacteria influencing clinical outcomes post-FMT in recipient #7
To explore the possibility of discovering a new strain of P. merdae, we isolated P. merdae from R7's stool sample and performed whole genome sequencing. Comparative genomic analysis revealed that the isolated P. merdae belongs to P. merdae (Table S4), but the average nucleotide identity with P. merdae sp. Marseille-P4119 was 97.41%, suggesting a potential distinction. Phylogenetic analysis identified over 1,980 single-nucleotide polymorphisms (SNPs) in conserved markers, indicating significant dissimilarity from the reference genome of P. merdae sp. Marseille-P4119 (Figure 3E). These genomic differences in marker genes suggest the discovery of a new strain, which we designated as P. merdae Immunoactis. We hypothesize that this strain contributed to the favorable clinical and immune response in R7 after the second FMT.
Following a similar logical framework, we identified bacteria that might negatively impact clinical responses. We focused on bacteria more prevalent in D1 than in D5, and in R7’s stool after the first FMT compared to the second. This analysis identified 16 common bacteria (Figure 3F; Table S5). Among these bacteria, Bacteroides plebeius (B. plebeius) exhibited the highest LDA score and was more abundant in D1 and R7 post-first FMT than post-second FMT, respectively (Figures 3F and S8B). B. plebeius is suggested to potentially cause unfavorable clinical responses due to its immunosuppressive effects.
Despite undergoing the second FMT with fecal material from the same D5, R6 experienced only a brief duration of SD with a progression-free survival of 2.1 months. Further microbial comparison between R6 and R7 revealed that P. merdae, which was suspected to be an immunity-boosting strain, was well-established in both after the FMT from D5 (Figure S8C). B. plebeius, which potentially exhibits immunosuppressive properties, was not detected at all in R6 (Figure S8C). This prompted us to investigate why R6 did not experience the expected therapeutic effect despite the presence of P. merdae. We identified significant bacterial differences between R6 and R7. Comparing stools from R6's and R7’s second FMT from same D5, we noted a higher presence of Lactobacillus salivarius (L. salivarius) in R6’s stool samples compared to R7’s (Figure 3G; Table S6). Interestingly, L. salivarius was more abundant in D1 compared to D5 and in R7’s stool samples after the first FMT compared to the second FMT (Figures 3G and S8D). These findings suggest that L. salivarius may be linked to unfavorable clinical responses due to its potential immunosuppressive properties, similar to B. plebeius.
In our further investigation, we examined whether the ratio of three bacteria (P. merdae/(B. plebeius + L. salivarius)) is associated with clinical significance. Initially, we investigated whether this ratio had an influence on survival probability in an independent cohort of patients with unresectable or metastatic solid cancers who were treated with nivolumab monotherapy (Table S7; see STAR Methods). This cohort was specifically established for microbiome biomarker research and included a total of 72 patients. All the included patients had baseline fecal samples available and had not received antibiotics within the 4 weeks preceding treatment. Interestingly, our observations confirmed that a higher ratio was associated with prolonged survival (Figures S9A–S9C). Furthermore, we observed a tendency of increasing microbial ratios in R7 following the second FMT, which positively correlated with the ratio of effector CD8+ T cells and Tregs populations (Figures 3H and S9D). In fact, both these microbial and immunological trends intensified after the second FMT. These findings highlight the intricate impact of the gut microbiota on the efficacy of FMT and anti-PD-1 therapy. Further research is required to investigate the specific roles of these bacteria and their influence on clinical outcomes.

The influence of P. merdae Immunoactis on enhancing immune response and cytotoxic activity

Our comprehensive metagenomic analysis suggests that P. merdae Immunoactis may be pivotal in overcoming resistance to anti-PD-1 inhibitors, while B. plebeius and L. salivarius could have opposing effects. To explore the immune-related effects of these bacteria, we treated human T cells with their respective conditioned bacterial supernatants. When referring to “supernatant” in the following context, it denotes the conditioned bacterial supernatant. P. merdae Immunoactis’ supernatant significantly boosted the proliferation of CD4+ and CD8+ T cells (Figures 4A and 4B), whereas those from B. plebeius and L. salivarius hindered T cell proliferation (Figures S10A and S10B). Additionally, CD8+ T cells treated with P. merdae Immunoactis’ supernatants showed increased IFN-γ secretion compared to controls, while B. plebeius or L. salivarius supernatants led to decreased IFN-γ secretion (Figure 4C). To investigate whether the two non-efficacious strains hindered the effectiveness of P. merdae Immunoactis, we conducted a parallel experiment using a three-bacteria mixture. This mixture's proportions were derived from fecal samples of R6’s post-2nd FMT and from D5, where all three strains coexisted. Since, on average, they were present at about one-tenth the level of P. merdae, three-bacteria mixture was composed in a ratio of P. merdae Immunoactis:B. plebeius:L. salivarius = 10:1:1 (Figure S10C). The three-bacteria mixture significantly reduced T cell proliferation (Figure S10D), indicating that the non-efficacious strains curbed the T cell proliferation triggered by P. merdae Immunoactis.
Figure thumbnail gr4
Figure 4Administration of P. merdae Immunoactis suppresses tumor growth by enhancing immune cell activity
We observed significant differences in IFN-γ secretion among the three strains (Figure 4C), prompting us to investigate changes in the IFN-γ pathway’s expression. OT-1 cells, from C57BL/6-Tg(TcraTcrb)1100Mjb/Crl called by OT-1 mouse, were activated with Ova-peptide and treated with supernatants from each strain. It showed significant increases in key immune cytokines in the “Ova act + P. merdae Immunoactis” group. A comparison of IFN-γ pathway-related gene expression revealed obvious differences; there were increases with P. merdae treatment and decreases with B. plebeius treatment, except for Irf-1 and Stat-2 (Figure S10E). This indicates differential activation of the IFN-γ pathway by the two strains, leading to distinct immune response outcomes.
We found a significant increase in granzyme expression levels following treatment with P. merdae Immunoactis supernatant, suggesting a potential enhancement in cytotoxicity (Figure S11A). This led us to explore P. merdae Immunoactis’ impact on CD8+ T cell cytotoxic activity using an OT-1 cell cytotoxicity assay (Figure S11B). T cells activated by P. merdae Immunoactis’ supernatants induced significant apoptosis and death among pre-seeded MC38 cells (Figures 4D and S11C), indicating its potential to augment CD8+ T cell cytotoxicity against cancer. In subsequent in vivo investigation, the combination of P. merdae Immunoactis treatment and the anti-PD-1 inhibitor resulted in a more significant reduction in tumor volume compared to either treatment alone (Figure 4E). Conversely, the two non-efficacious strains significantly increased in vivo tumor growth, further supporting the specific anti-cancer efficacy of P. merdae Immunoactis (Figure S12A). We also tested whether the two non-efficacious strains could interfere with the synergistic effect induced by P. merdae Immunoactis with anti-PD-1. While P. merdae Immunoactis showed a significant synergistic effect, the three-bacteria mixture accelerated tumor growth (Figure S12B). Additionally, we observed significantly reduced tumor growth with the combination of P. merdae Immunoactis and anti-PD-1, even in the non-immunogenic 4T1 model (Figure S12C). In summary, P. merdae Immunoactis enhances responsiveness to anti-PD-1 therapy by boosting immunity, but the presence of non-efficacious strains may diminish these synergistic effects.
To evaluate the potential enhancement of immune responses by P. merdae Immunoactis administration, we analyzed the alterations in immune gene expression and immune cell populations in tumor tissues and lymph nodes. Tumors treated with both P. merdae Immunoactis and the anti-PD-1 inhibitor exhibited increased expression of Ifn-γ and Cxcl10 while showing decreased expression of Ccl22 compared to tumors treated with the anti-PD-1 inhibitor alone (Figure 4F). Treatment with the combination of P. merdae Immunoactis and the anti-PD-1 inhibitor resulted in a significant increase in tumor-infiltrating T cells within tumor tissues, while the proportion of Treg cells among the CD45+ T cells remained unchanged (Figure 4G). Notably, the combined treatment of the anti-PD-1 inhibitor and P. merdae Immunoactis significantly increased the population of CD8+ T cells, CD8+CD44+CD62L (CD8+Effector) T cells, and CD8+PD-1+ T cells within the tumor microenvironment, compared to anti-PD-1 treatment alone (Figures 4H–4J). Additionally, we observed an increased ratio of activated natural killer (NK) (NKAct) to exhausted NK (NKEx) with P. merdae Immunoactis treatment (Figure 4K). However, these cell populations remained unchanged in the lymph nodes (Figure S12D). Taken together, these findings collectively suggest the potential of P. merdae Immunoactis, particularly when combined with the anti-PD-1 inhibitor, to stimulate immune cell proliferation and enhance cytotoxic activity, thereby promoting a more effective anti-tumor immune response within the tumor microenvironment.

Discussion

Our findings demonstrate that FMT incorporating effective microbiota has the potential to overcome resistance to anti-PD-1 inhibitors in advanced solid cancers. This study particularly emphasizes the treatment of gastrointestinal cancers, including GC, ESCC, and HCC, which are known to be immunologically challenging to treat. Our study provides proof-of-concept for the effectiveness of FMT in combination with anti-PD-1 inhibitors in solid cancers beyond melanoma, expanding upon previous pilot studies that showed the efficacy of FMT combined with anti-PD-1 inhibitors in patients with melanoma refractory to anti-PD-1 treatment. ,
Among the 13 recipients, one with HCC achieved a PR and five had SD, resulting in a 7.7% response rate and 46.2% disease control rate. Concerns might arise whether this efficacy stemmed from FMT or continued ICI beyond progression, known for certain cancers like NSCLC (Non-small cell lung cancer) and melanoma.  ICIs can induce atypical tumor responses, including pseudoprogression or delayed response, contributing to the benefit of continued treatment. However, all patients in our study had documented disease progression on anti-PD-1 therapy, confirmed by consecutive CT scans at least 4 weeks apart, indicating ongoing PD. A tumor biopsy before FMT did not show massive immune infiltration typical of pseudoprogression, ruling out pseudoprogression in anti-PD-1 monotherapy. Of five patients showing SD post-FMT with nivolumab, one (R6) had HCC, while four (R2, R5, R9, and R14) had ESCC. In these cancer types, the literature is sparse on how commonly SD occurs and how long it persists with continued ICIs beyond progression. A small-sized retrospective study reported a 5.8% response rate, a 38% SD rate, and 3.7 months of progression-free survival in advanced HCC patients treated beyond progression (n = 34).  However, this study included not only anti-PD-(L)1 inhibitors but also an anti-PD-L1 plus an anti-VEGF (Vascular endothelial growth factor) inhibitor, and initial disease progression was not confirmed with subsequent CT scans as in our study. In esophageal cancer, there is no literature suggesting the benefit of continuing ICIs beyond progression. Our four heavily pre-treated ESCC patients (all 4th- or 5th-line setting) showed progression-free survival of 2.8, 2.8, 2.7, and 4.6 months, respectively. Tumor growth on nivolumab monotherapy, confirmed on consecutive CT scans before FMT, was slowed down after FMT plus nivolumab, suggesting an actual effect of the combination. One HCC patient (R6) receiving FMT plus nivolumab as 3rd-line therapy also showed 3.1 months of progression-free survival and tumor shrinkage after FMT plus nivolumab. These findings suggest that the clinical benefit observed after FMT in our study is not solely from the continuation of the anti-PD-1 inhibitor but from the beneficial effect of combining FMT with the anti-PD-1 inhibitor.
Previous studies on FMT and cancer immunotherapy have provided insights into the general impact of microbiome changes on therapeutic outcomes, with some, such as Routy et al.,  isolating individual bacterial strains that promote anti-tumor immunity in melanoma. ,  Our study expands this field to include non-melanoma cancers, such as HCC. However, in our study, a response was observed only in HCC, making it challenging to determine if this response is specific to HCC due to the small sample size, which includes three cancer types and only one HCC responder. Additionally, we involved six donors because it was unclear which donor might possess the causative microbiome for FMT efficacy before the FMT. Furthermore, we attempted to match cancer types between recipients and donors whenever possible. Consequently, patients with GC and ESCC did not receive FMT using stool samples from D5, the only donor who was an HCC patient and induced a tumor response in the FMT trial. While additional clinical studies are needed to further elucidate the efficacy of combining FMT with ICIs in these cancer types, our research nonetheless demonstrates the potential for applying FMT in non-melanoma cancers. This highlights an important step toward broader therapeutic applications and the need for further investigation to optimize treatment strategies.
Additionally, we identified specific bacterial strains like P. merdae Immunoactis, L. salivarius, and B. plebeius and examined their distinct immunological roles in the context of anti-PD-1 therapy. We discovered and isolated a strain, Prevotella merdae Immunoactis, from a patient (R7) who exhibited an excellent response to FMT, suggesting it as a potential causative bacterium for the therapeutic effects of FMT. Moreover, two non-efficacious strains, L. salivarius and B. plebeius, were identified and found to inhibit T cell activation, potentially impairing the efficacy of FMT and anti-PD-1 inhibitors. We also confirmed that the ratio of these three bacteria (P. merdae/(B. plebeius + L. salivarius)) has clinical implications, including survival likelihood. This nuanced understanding highlights the complex interplay between beneficial and detrimental bacteria within the gut microbiota in determining treatment outcomes. By isolating and characterizing these specific strains, we demonstrated the direct mechanisms through which these bacteria influence the efficacy of immunotherapies, offering potential targets for enhancing the immunogenicity of the tumor microenvironment. This approach allowed us to move beyond general associations to specific causal relationships, providing a more granular view of microbiota-mediated modulation of cancer treatment. This emphasizes the need for targeted manipulation of the gut microbiome to optimize therapeutic outcomes in cancer patients.
In the case of R7, although there was a notable reduction in tumor size and heightened immune response observed following the second FMT with anti-PD-1 therapy, the later stages presented significant challenges. About 11 weeks after the second FMT, the patient experienced progressive epigastric pain and nausea, diagnosed as immune-related gastritis, a rare but known immune-related AE (irAE) in ICI-treated patients.  The exact pathophysiology of irAEs, especially organ-specific ones, remains unclear but seems to involve more than just an overactive immune response. To manage these symptoms, long-term systemic corticosteroid treatment was initiated around the 12th week after the second FMT, introducing an unknown variable to the treatment’s long-term impacts. While planning the third FMT using fecal material from the same D5, the patient sustained a femoral fracture from a fall, necessitating surgery. Post-surgery, subsequent FMTs with the same donor's fecal material did not yield the anticipated results. The third FMT stabilized the disease, but the fourth could not halt tumor growth. The diminished efficacy of these FMTs can be attributed to prolonged steroid use. Steroids, known for their immunosuppressive properties, , ,  can significantly alter gut microbiome communities, , ,  impacting the gut-immune axis crucial for systemic immune responses. ,  Moreover, the synergistic interaction of multiple strains might influence the outcome, suggesting that other unidentified strains could have affected the effectiveness of the third and fourth FMTs.
In conclusion, our findings suggest the potential of FMT to boost responsiveness to ICIs in advanced solid tumors, extending beyond melanoma. By comparing responses among patients, we identified P. merdae Immunoactis as an effective strain, contrasting with the ineffective B. plebeius and L. salivarius. Our analysis of the IFN-γ signaling pathway elucidated the mechanisms of immune modulation by these strains. This study not only demonstrates the broad applicability of FMT in enhancing immunotherapy efficacy but also opens new paths for targeted microbial interventions in cancer treatment.

STAR★Methods

Key resources table

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
InVivoMAb rat IgG2a isotype control, anti-trinitrophenol (clone 2A3) BioXCell Cat# BE0089; RRID: AB_1107769
InVivoMAb anti-mouse PD-1 (CD279) (clone RMP1-14) BioXCell Cat# BE0146; RRID: AB_10949053
OPAL polymer HRP Ms+Rb Akoya Biosciences Cat# ARH1001EA; RRID: AB_2890927
anti-human CD3e antibody BioLegend Cat# 317325; RRID: AB_11147370
anti-mouse CD3e antibody BioLegend Cat# 100340; RRID: AB_11149115
anti-mouse CD28 antibody BioLegend Cat# 102116; RRID: AB_11147170
Human TruStain FcX (Fc Receptor Blocking) BioLegend Cat# 422302; RRID: AB_2818986
anti-mouse CD16/CD32 (Fc Receptor Blocking) BioLegend Cat# 101330; RRID: AB_2561482
APC/Cyanine7 anti-mouse CD45 Antibody BioLegend Cat# 103116; RRID: AB_312981
PerCP/Cyanine5.5 anti-mouse CD3 Antibody BioLegend Cat# 100218; RRID: AB_1595492
PE/Cyanine7 anti-mouse CD4 Antibody BioLegend Cat# 100422; RRID: AB_312707
APC anti-mouse CD62L Antibody BioLegend Cat# 104412; RRID: AB_313099
PE anti-mouse CD279 (PD-1) Antibody BioLegend Cat# 109104; RRID: AB_313421
APC/Cyanine7 anti-mouse/human CD45R/B220 BioLegend Cat# 103224; RRID: AB_313007
APC anti-mouse CD25 Antibody BioLegend Cat# 101910; RRID: AB_2280288
PE anti-mouse IFN-γ Antibody BioLegend Cat# 163503; RRID: AB_2890730
BV421 anti-mouse F4/80 Antibody BD Bioscience Cat# 565411; RRID: AB_2734779
APC anti-mouse TNF-α Antibody BioLegend Cat# 506308; RRID: AB_315429
BV510 anti-mouse CD8a Antibody BD Bioscience Cat# 563068; RRID: AB_2687548
BV421 anti-mouse CD44 Antibody BD Bioscience Cat# 563970; RRID: AB_2738517
APC anti-mouse NK-1.1 Antibody BD Bioscience Cat# 550627; RRID: AB_398463
BV421 anti-mouse CD11c Antibody BD Bioscience Cat# 562782; RRID: AB_2737789
BB700 anti-mouse CD11b Antibody BD Bioscience Cat# 566416; RRID: AB_2744272
BV421 anti-mouse Foxp3 Antibody BD Bioscience Cat# 562996; RRID: AB_2737940
FITC anti-mouse NKG2A/C/E Monoclonal Antibody eBioscience Cat# 11-5896-82; RRID: AB_465305
Bacterial and virus strains
P. merdae Immunoactis This work KCTC14922BP
Bacteroides plebeius KCTC5793 Korean Collection for Type Cultures (KCTC) KCTC5793
Lactobacillus salivarius KCTC43133 Korean Collection for Type Cultures (KCTC) KCTC43133
Biological samples
Human feces This study N/A
Human blood samples This study N/A
Human tumor tissues This study N/A
Chemicals, peptides, and recombinant proteins
Brain heart infusion (BHI) broth BD Biosciences Cat# 237500
Reinforced clostridial medium (RCM) broth BD Biosciences Cat# 218081
De Man, Rogosa and Sharpe (MRS) broth BD Biosciences Cat# 288130
Yeast extract BD Biosciences Cat# 212750
L-Cysteine hydrochloride monohydrate Merck Cat# C7880
Hemin Sigma-Aldrich Cat# H9039
Vitamin K1 Sigma-Aldrich Cat# 95271
Kanamycin Rd-tech Cat# HKA01
Defibrinated sheep blood KisanBio Cat# S1876
DMEM medium ThermoFisher Cat# 11965-118
RPMI 1640 medium Corning Cat# 10-040-CV
Fetal bovine serum (FBS) MP Biomedicals Cat# 92916754
PBS ThermoFisher Cat# 10010-049
Penicillin-streptomycin ThermoFisher Cat# 15140-122
NEAA (MEM Non-Essential Amino Acids Solution) ThermoFisher Cat# 11140-050
HEPES ThermoFisher Cat# 15630-080
Sodium pyruvate ThermoFisher Cat# 11360070
L-glutamine ThermoFisher Cat# 25030-081
2-mercaptoethanol ThermoFisher Cat# 21985-023
carboxyfluorescein diacetate succinimidyl ester (CFSE) ThermoFisher Cat# C34554
EDTA ThermoFisher Cat# AM9260G
Ficoll® Paque Plus Merck Cat# GE17-1440-02
RBC lysis buffer BioLegend Cat# 420301
Ovalbumin (323-339) Merck Cat# O1641
TRIzol reagent Invitrogen Cat#15596026
Collagenase type I ThermoFisher Cat# 17100-017
Collagenase type II ThermoFisher Cat# 17101-015
Collagenase type IV ThermoFisher Cat# 17104-019
DNase type I Merck Cat# 10104159001
hyaluronidase type IV-S Merck Cat# H3884
Fixation/permeabilization buffer set BioLegend Cat# 424401
Leica Bond Dewax solution Leica Biosystems Cat# AR9222
Bond Epitope Retrieval 1 Leica Biosystems Cat# AR9961
Bond Epitope Retrieval 2 Leica Biosystems Cat# AR9640
Antibody diluent/block Akoya Biosciences Cat# ARD1001EA
DAPI and Hoechst Nucleic Acid Stains ThermoFisher Cat# 62248
ProLong Gold antifade reagent ThermoFisher Cat# P36935
Maxpar Cell Staining Buffer Fluidigm Cat# 201068
Cell-ID Intercalator-Ir Fluidigm Cat# 201192A
Maxpar Fix and Perm Buffer Fluidigm Cat# 201067
Critical commercial assays
Simoa CorPlex Human Cytokine 10-plex Panel 1 kit Quanterix Cat# 85-0329
Maxpar Direct Immune Profiling Assay kit Fluidigm Cat# 201334
FastDNA® SPIN Kit for Soil MP Biomedicals Cat# 116560200-CF
2× KAPA HiFi HotStart ReadyMix Roche Cat# 07958927001
TruSeq Nano DNA sample Prep Kit, set A Illumina Cat# TG-202-1001
TruSeq Nano DNA sample Prep Kit, set B Illumina Cat# TG-202-1002
Human CD4+ T Cell Isolation Kit Miltenyi Biotec Cat# 130-096-533; RRID: AB_2916089
Human CD8+ T Cell Isolation Kit Miltenyi Biotec Cat# 130-096-495; RRID: AB_3073903
Mouse CD8+ T Cell Isolation Kit Miltenyi Biotec Cat# 130-104-075
Human IFN-γ ELISA kit ThermoFisher Cat# 88-7316-88
Annexin V–PI staining kit Enzo Life Science Cat# ALX-850-020-K101
TOPscript™ RT DryMIX kit Enzynomics Cat# RT200
TOPreal™ qPCR 2X PreMIX kit Enzynomics Cat# RT501M
Deposited data
Raw Sequencing files This Study ENA: PRJEB48251
Experimental models: Cell lines
MC38 (mouse colon cancer cell line) Lab stock ENH204-FP; RRID: CVCL_B288
4T1 (mouse breast cancer cell line) Lab stock CRL-2539; RRID: CVCL_0125
Experimental models: Organisms/strains
Female C57B6/N mice (Seven weeks-old) Orientbio RRID: MGI:5882838
Female Balb/c mice (Seven weeks-old) Orientbio RRID: MGI:6323059
OT-1 mice (Tg[TcraTcrb]1100Mjb/J) In-House Breeding RRID: IMSR_JAX:003831
Oligonucleotides
TCGTCGGCAGCGTCAGATGTGTATAA

GAGACAGCCTACGGGNGGCWGCAG
This study Forward primer for bacterial 16s rRNA V3-V4 regions
GTCTCGTGGGCTCGGAGATGTGTATAA

GAGACAGGACTACHVGGGTATCTAATCC
This study Reverse primer for bacterial 16s rRNA V3-V4 regions
GCTCAACCTGGGCATTGCA This study Forward primer for identifying P. merdae Immunoactis
CATGTTTTAGGGATTCGAGCG This study Reverse primer for identifying P. merdae Immunoactis
Software and algorithms
GraphPad Prism 10 GraphPad Software https://www.graphpad.com/
FlowJo v10.10.0 Treestar, Inc. https://www.flowjo.com/
FACS Diva 9 BD Biosciences https://www.bdbiosciences.com/en-us/products/software/instrument-software/bd-facsdiva-software
Phenochart v2.2.0 Akoya Biosciences https://www.akoyabio.com/support/software/
inForm Image Analysis software v3.0 Akoya Biosciences https://www.akoyabio.com/support/software/
FCS Express 7 Flow software De Novo Software https://denovosoftware.com/
phenoptrReport packages Akoya Biosciences https://akoyabio.github.io/phenoptrReports/index.html
Cutadapt version 4.1 Martinl. https://journal.embnet.org/index.php/embnetjournal/article/view/200
QIIME2 version 2022.8 Bolyen et al. https://qiime2.org/
DADA2 software package Callahan et al. https://github.com/benjjneb/dada2
RESCRIPt Robeson et al. https://github.com/bokulich-lab/RESCRIPt
R (version 4.2.1) R core team https://www.R-project.org/
ggpubr package (version 0.6.0) Kassambara https://rpkgs.datanovia.com/ggpubr/
Analysis of Composition of Microbiomes (ANCOM) Mandal et al. https://qiime2.org/
kneaddata (version 0.12.0) Biobakery group https://github.com/biobakery/kneaddata
HUMAnN 3 Beghini et al. https://github.com/biobakery/humann
StrainPhlAn 4 Truong et al. https://github.com/biobakery/biobakery/wiki/strainphlan4
Jalview Waterhouse et al. https://www.jalview.org/
FastTree (version 2.1.11) Price et al. http://www.microbesonline.org/fasttree/
FigTree (version 1.4.4.) Rambaut http://tree.bio.ed.ac.uk/software/figtree/
BBDuk Bushnell. https://github.com/BioInfoTools/BBMap
Unicycler(version 0.5.0) Wick et al. https://github.com/rrwick/Unicycler
Usearch (version 11.0.667) Edgar https://www.drive5.com/usearch/
JspeciesWS Richter et al. https://jspecies.ribohost.com/jspeciesws/#home
OrthoANI Lee et al. https://www.ezbiocloud.net/tools/orthoani
Other
Anaerobic jar Oxoid Cat# HP0011A
Anaeropack MGC Cat# A-06
GasPak 100 system BD Biosciences Cat# 260626
Cuvette Ratiolab Cat# HRA-2712120
0.2 μm syringe filte Satorius Cat# 17823-K
40 μm cell strainer Falcon Cat# 352340
70 μm cell strainer Falcon Cat# 352350
60 mm non-treated plate SPL Cat# 11060
96-well non-treated plates SPL Cat# 32096
Cell counting slide Logos biosystems Cat# L12001
Insulin syringe 0.5mL, 31G, 8mm BD Cat# 328821
DNA/RNA Shield Fecal Collection Tube Zymo Research Cat# R1101

Resource availability

Lead contact

Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Hansoo Park (hspa...@gist.ac.kr).

Materials availability

P. merdae Immunoactis, identified in this study is available from Korean Collection for Type Cultures (KCTC). Details in key resources table.

Data and code availability

Clinical metadata for matched sequencing files of this study are available for academic use under confirmation of lead contact.
Any of computational code was not manually changed. All original code is available from each development site listed in key resources table. Details of setting are described in method details.
Any additional information and re-analysis is available under confirmation of lead contact upon request.

Experimental Model and Subject Detail

Ethical approval and consent information

This study was conducted in accordance with the Declaration of Helsinki and was approved by the IRB for Asan Medical Center and GIST (2018-0608 and 20210416-HB-60-07-04, respectively). All participants provided written informed consent for the study, including blood, stool, and tumor tissue biopsy.

Clinical study design and study population

This study was conducted at Asan Medical Center and registered in clinicaltrials.gov; the registration number is NCT04264975.

Fecal microbiota transplantation study

This was a prospective, single-center study of concurrent fecal microbiota transplantation (FMT) alongside an anti-programmed death-1/programmed death-ligand 1 (PD-1/PD-L1) inhibitor. The primary objective was to investigate whether the combination of FMT derived from good responders to anti-PD-(L)1 inhibitors, together with an anti-PD-(L)1 inhibitor could overcome resistance to anti-PD-(L)1 inhibitors in patients with advanced solid cancer refractory to these inhibitors. Secondary objectives were to evaluate the safety profile and examine the systemic and intratumor immune effects of combining FMT with an anti-PD-(L)1 inhibitor, and identify the potential causative bacteria that contribute to the efficacy of FMT via analysis of the gut microbiota composition.
The key eligibility criteria for recipients were histologically confirmed solid tumors, except for hepatocellular carcinoma (HCC), for which a clinically confirmed diagnosis as per the American Association for the Study of Liver Diseases (AASLD) was allowed ; confirmed disease progression during anti-PD-(L)1 based therapy (as monotherapy or combination) for unresectable or metastatic solid tumors; age ≥19 years; Eastern Cooperative Oncology Group (ECOG) performance status 0‒2; at least one measurable lesion(s) per the Response Evaluation Criteria in Solid Tumors (RECIST) v1.1 ; no history of other active malignancy within the previous 3 years before study entry (with the exception of curatively treated non-melanoma skin cancer, superficial bladder cancer, or carcinoma in situ of prostate, cervix, breast, or stomach); no active infection; no history of active autoimmune disease requiring systemic treatment except for type 1 diabetes mellitus or autoimmune thyroid disease; no concurrent condition requiring immunosuppressants; and no contraindications to colonoscopy.
The key eligibility criteria for donors were histologically confirmed solid tumors, except for HCC, for which clinically confirmed diagnosis as per the AASLD was allowed ; ongoing durable complete or partial response per RECIST v1.1 for at least 6 months with anti-PD-(L)1 monotherapy for unresectable or metastatic solid tumors; age ≥19 years; no concurrent transmissible diseases; no history or risk behaviors for infectious disease such as Human Immunodeficiency Virus or other viral infection, recent travel within 6 months to countries with endemic diarrheal diseases or high risk of traveler’s diarrhea; no history of chronic gastrointestinal disease including inflammatory bowel disease; and no recent ingestion of allergen with known recipient allergy. Only donors who passed serological and stool screening tests were considered suitable to donate stool samples for FMT implant.

Biomarker study for validation

This study was a single-center, prospective, non-interventional study to investigate the association between the gut microbiome and the clinical benefits of anti-PD-(L)1 inhibitors in patients with unresectable or metastatic solid cancers. The key eligibility criteria were histologically confirmed solid tumors, except for HCC, for which a clinically confirmed diagnosis as per the AASLD was allowed ; unresectable or metastatic solid tumors treated with anti-PD-(L)1 monotherapy; age ≥19 years; ECOG performance status 0‒2; at least one measurable or evaluable lesion(s) per RECIST v1.1 ; no history of other active malignancy within the previous 3 years before study entry (with the exception of curatively treated non-melanoma skin cancer, superficial bladder cancer, or carcinoma in situ of prostate, cervix, breast, or stomach); no active infection; no history of active autoimmune disease requiring systemic treatment except for type 1 diabetes mellitus or autoimmune thyroid disease; and no concurrent condition requiring immunosuppressants.
Patient stool samples and peripheral blood samples were collected at baseline before treatment with anti-PD-(L)1 inhibitors and at every tumor response evaluation, which was performed using CT scans every 6–8 weeks according to RECIST v1.1.

Study treatment in the FMT trial

Preparation of FMT implant

Feces of 200-500 mg were collected from a donor immediately after defecation in a fecal container provided by the study personnel. The collected feces were transported to the laboratory as soon as possible within 4 hours after defecation. For each stool collected, screening stool tests including stool ova and parasites, stool culture for Salmonella, Shigella, Campylobacter, Yersinia, Enterohemorrhagic Escherichia coli, Clostridium difficile, and antibiotics resistant bacteria including vancomycin-resistant enterococci carbapenem-resistant Enterobacteriaceae, and extended-spectrum beta-lactamases-producing Enterobacteriaceae were also performed.
The feces from the donor used for FMT were processed as soon as possible within 6 hours of defecation. They were diluted with a sterile solution (normal saline 0.9%) and then homogenized manually using a pipette or in a blender with low speed for at least 30 seconds. The fecal suspension was filtered through a clean metal coffee strainer to remove food debris. The filtrate was then centrifuged for 15 minutes at 6000 × g and suspended with sterile solution (0.9% normal saline/glycerin in a 90/10 v/v ratio) in one third of the initial volume. The filtrate was packaged into sterile vials (50 mL in each vial) and stored at -80°C. On the day of FMT, the frozen suspensions were thawed using a 37°C warm water bath starting from about 2 hours before the FMT procedure.

Treatment with FMT plus anti-PD-(L)1 inhibitor

Recipients underwent an initial native microbiota depletion phase where they received oral antibiotics (Augmentin®: amoxicillin clavulanate; 625 mg po tid) for 5 days (D-5 to D-1) before FMT treatment (D0). The last dose of Augmentin was completed at least 12 hours (± 2 hours) prior to the administration of the FMT implant. FMT was performed via colonoscopy, and the gastrointestinal tract was prepared by bowel lavage using 2 L of polyethylene glycol electrolyte solution before the procedure according to the institute’s protocol. Thawed fecal suspensions were administered through colonoscopy, and patients were asked to retain the material as long as possible after the procedure.
After the administration of FMT, recipients received an anti-PD-1 inhibitor at the standard dose on the day after the FMT, which was continued until unacceptable toxicity or disease progression. Subsequent FMT sessions from the same or different donors were permitted, at the investigator's discretion, with a time interval of 2 to 4 weeks prior to the initial response assessment or in cases where clinical response was not evident. Response assessment was carried out using computed tomography (CT) scans conducted every 6 to 8 weeks, following RECIST v1.1.

Preparation of bacteria

The following bacterial strains were obtained from the Korean Collection for Type Cultures (KCTC): Bacteroides plebeius (KCTC 5793) and Lactobacillus salivarius (KCTC 43133). Prevotella merdae Immunoactis was isolated as described below. All bacterial species were cultured at 37°C under anaerobic conditions, which were created in an anaerobic jar (Oxoid) with an anaeropack (#A-06, MGC). B. plebeius was cultured in autoclaved reinforced clostridial medium (RCM) broth (#218081, BD Biosciences). L. salivarius was cultured in autoclaved De Man, Rogosa and Sharpe (MRS) broth (#288130, BD Biosciences). P. merdae Immunoactis was cultured in autoclaved brain heart infusion (BHI) broth (#237500, BD Biosciences) supplemented with 0.5% yeast extract (#212750, BD Bioscience), 0.05% L-Cysteine hydrochloride monohydrate (#C7880, Merck), 0.005% hemin (#H9039, Sigma-Aldrich), and 0.0001% vitamin K1 (#95271, Sigma-Aldrich). To eliminate the effects of culture media on T cells, we prepared conditioned bacterial supernatant; Overnight-cultured bacterial cultures (1 mL) were inoculated into 9 mL of Roswell Park Memorial Institute (RPMI) 1640 medium (#10-040-CV, Corning) and cultured for 24 h. These cultures were then centrifuged at 6,000 rpm for 5 min, and the supernatants were filtered using a 0.2 μm syringe filter (#17823-K, Sartorius). One milliliter of each supernatant was stored at -80 °C until use. The P. merdae Immunoactis culture was centrifuged at 6,000 rpm for 5 min, the supernatant was discarded, and the pellet was resuspended in PBS. The optical density of the suspension at 600 nm (OD600) was adjusted to OD600 = 6.

Mammalian Cell culture

MC38 (mouse colon cancer cell line) and 4T1 (mouse breast cancer cell line) were cultured in DMEM (#11965-118, Thermo Fisher Scientific) supplemented with 10% heat-inactivated FBS (#92916754, MP Biomedicals) and 1% penicillin-streptomycin (Thermo Fisher Scientific) at 37°C in a 5% CO2 atmosphere. Cells were sub-cultured when they reached 70–80% confluence.

Mice experiments

All animal experiments were conducted following the approved protocols of the Institutional Animal Care and Use Committee of GIST (GIST-2021-096 and GIST-2024-011). The study followed the approved policies for maintaining and handling the animals. For the syngeneic tumor model, female C57B6/N mice (Orientbio) seven weeks-old were subcutaneously injected with 2 × 105 MC38 mouse colon cancer cells on day 0, and female Balb/c mice (Orientbio) seven weeks-old were subcutaneously injected with 5 × 105 4T1 mouse breast cancer cells on day 0. Tumor size was measured three times every week until the endpoint, and tumor volume was calculated using the formula: length × width2 × 0.5. Each bacterial strain, three-bacteria mixture or PBS (#10010-049, Thermo Fisher Scientific) was orally administered daily, starting 14 days before tumor inoculation and continuing for three weeks after tumor inoculation. Each strain was prepared to be ingested in an amount corresponding to an optical density value of 10 (OD600 = 10) per mice. Three-bacteria mixture was composed with the ratio of P. merdae Immunoactis: B. plebeius: L. salivarius = 10 : 1 : 1 based on OD600 value. To investigate the effects of combination therapy, 2 mg/kg IgG isotype (clone 2A3, #BE0089, BioXCell) and 2 mg/kg anti-PD-1 monoclonal antibody (mAb) (clone RMP1-14, #BE0146, BioXCell) were intraperitoneally injected on days 7, 9, 11, 14, 16, and 18 after tumor inoculation. PBS and IgG isotypes were used as controls for bacterial administration and anti-PD-1 mAb injections, respectively.

Method Details

Multiplex immunofluorescence staining

Multiplex IHC staining, scanning, and analysis were performed by PrismCDX Co., Ltd. (Gyeonggi-do, Republic of Korea). For the staining, 4-μm thick sections were cut from formalin-fixed, paraffin-embedded (FFPE) blocks. Slides were heated for at least 1 hour in a dry oven at 60°C, followed by multiplex immunofluorescence staining using a Leica Bond Rx™ Automated Stainer (Leica Biosystems, Newcastle, UK). The list of antibodies and fluorophores used in the staining process is summarized in Table S8. Briefly, slides were dewaxed using Leica Bond Dewax solution (#AR9222, Leica Biosystems), followed by antigen retrieval using Bond Epitope Retrieval 2 (#AR9640, Leica Biosystems) for 30 min. The staining was proceeded in sequential rounds of blocking with antibody diluent/block (ARD1001EA, Akoya Biosciences, Marlborough, MA, USA), followed by primary antibody incubation for 30 min, and incubation with OPAL polymer HRP Ms+Rb (#ARH1001EA, Akoya Biosciences) for 10 min. The antigen was visualized using tyramide signal amplification (Akoya Biosciences) for 10 min, after which the slide was treated with Bond Epitope Retrieval 1 (#AR9961, Leica Biosystems) for 20 min to remove bound antibodies before the next step. The process from blocking to antigen retrieval was repeated for each antibody stain. Nuclei were counterstained with DAPI (#62248, Thermo Fisher Scientific, Waltham, MA, USA) after the final antigen retrieval step. The slides were overlaid with coverslips and ProLong Gold antifade reagent (#P36935, Invitrogen, Waltham, MA, USA).

Multispectral imaging and analysis

Multiplex-stained slides were scanned at 20× magnification using the Vectra Polaris Automated Quantitative Pathology Imaging System (Akoya Biosciences). Representative images for training were selected in a Phenochart (Akoya Biosciences) and an algorithm was created using the inForm Image Analysis software (Akoya Biosciences). Multispectral images were unmixed using the spectral library in inForm software, and tumor tissues were segmented based on the presence or absence of cytokeratin (CK) expression. Each cell was segmented based on DAPI staining, and phenotyping was performed based on the expression compartment and intensity of each marker. After designating the region of interest (ROI) to be analyzed on the tissue slide, the algorithm created in this way was applied in a batch-running manner. The exported data were consolidated and analyzed in the R software using the phenoptr (Akoya Biosciences) and phenoptrReport packages (Akoya Biosciences).

Detection of multiple cytokines (Simoa)

Simoa HD-1 Assay

Plasma levels of IL-2 and IL-15 were measured using the Simoa reagent kit (Quanterix, MA, USA) on a Simoa HD-1 Analyzer (Quanterix, MA, USA) at PrismCDX Co., Ltd. by an investigator who was blinded to clinical information. Plasma samples were diluted 1:4 according to Quanterix guidelines. All HD-1 consumables, including wash buffers, 96-well plates, discs, cuvettes, and sealing oil, were purchased from Quanterix. The assay was performed on the fully automated Simoa HD-1 using a 3-step protocol. In the 3-step assay, the target antibody coated with paramagnetic beads was incubated with the sample so that the target molecule in the sample was bound by the bead-coated antibody. After washing, they were mixed with a biotinylated detector antibody and incubated to bind the detector antibody to the captured target. After washing, the complex was mixed with streptavidin-β-galactosidase (SBG) to allow SBG to bind to the biotinylated detector antibody and generate an enzyme label on the captured target. After the final wash, the beads were resuspended in RGP substrate solution and transferred to a Simoa disc. Captured and labeled target antigens were measured while β-galactosidase hydrolyzed the RGP substrate. The average number of enzymes per bead (AEB) was calculated from the fraction of active wells that correlated with the analyte concentration. For assessing plasma IL-2 and IL-15, all samples were analyzed in duplicates, and the mean of the replicates for each sample was calculated.

Simoa SP-X Assay

Ten plasma markers were measured using Simoa CorPlex Human Cytokine 10-plex Panel 1 kit (Quanterix, MA, USA). The ten targets were interferon gamma (IFN-γ), IL-1β, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12p70, IL-22, and tumor necrosis factor alpha (TNFα). Each well of a 96-well microplate was pre-spotted with protein-specific antibodies to capture the target protein in the sample. After washing unbound proteins, biotinylated detector antibodies were added to bind to the secondary site of the target protein and washed to remove the excess detector antibody. Finally, streptavidin–horseradish peroxidase (SA–HRP) was added to allow HRP to react with the substrate and generate a luminescent signal detected by the SP-X Imaging System (Quanterix, MA, USA). The amount of protein present was quantified based on the signal intensity.

Mass flow cytometry (CyTOF)

Peripheral blood mononuclear cells (PBMCs) were stained using the Maxpar Direct Immune Profiling Assay kit (Fluidigm) according to the manufacturer’s instructions. PBMC aliquots were prepared, and the buffer was exchanged with Maxpar Cell Staining Buffer (Fluidigm). Human TruStain FcX (BioLegend, San Diego, CA, USA) was added to each tube to block Fc-receptor, followed by incubation for 10 minutes at 19-24°C. PBMCs were then directly transferred to a 5 mL tube containing a dry antibody pellet (Fluidigm) and incubated for 30 min at room temperature. After washing, PBMCs were fixed in 1.6% formalin for 10 min at room temperature. Finally, PBMCs were incubated with Cell-ID Intercalator-Ir (Fluidigm) in Maxpar Fix and Perm Buffer (Fluidigm) for approximately 48 h at 4°C. Prior to acquisition, PBMCs were washed twice with Maxpar Cell Staining Buffer (Fluidigm) and filtered through a 40 μm cell strainer before being acquired on a Helios mass cytometer (Fluidigm). Mass cytometry data files were analyzed using FCS Express 7 Flow software (CAT #402001, De Novo Software, Pasadena, CA, USA). The clone and mass of each antibody are described in Table S9.

Fecal DNA extraction and sequencing

All fecal samples were collected and stored in collection tubes (#R1101, Zymo Research) until DNA extraction. DNA was extracted from fecal samples using FastDNA® SPIN Kit for Soil (MP Biomedicals) according to the manufacturer’s instructions. The purity and quantity of DNA were estimated using NanoDrop One Spectrophotometer (Thermo Fisher Scientific). The bacterial 16S rRNA V3–V4 region was amplified using the Illumina’s 16S Metagenomic Sequencing Library Preparation guide (Illumina), using primers with adapter overhang sequences added.  Forward primer: 5’- TCGTCGGCAGCGTCAGATGTGTATAAGAGACAGCCTACGGGNGGCWGCAG -3’, Reverse primer: 5’- GTCTCGTGGGCTCGGAGATGTGTATAAGAGACAGGACTACHVGGGTATCTAATCC -3’. The PCR reaction mix included 2 μL of genomic DNA, 0.5 μL of each primer, 12.5 μL of 2× KAPA HiFi HotStart ReadyMix (Kapa Biosystems), and 9.5 μL of distilled water, for a total volume of 25 μL. The PCR conditions used were as follows: 95°C for 3 min for pre-denaturing DNA; 25 cycles at 95°C for 30 s for denaturing DNA; 50°C for 30 s for annealing; 72°C for 30 s for extension; and 72°C for 5 min for final extension. The PCR products were purified using AMPure XP Beads (Beckman Coulter). Dual index attachment and Illumina sequencing adapters were performed using PCR products (5 μL), Illumina Nextera XT Index Primer 1 (5 μL, N7xx), Nextera XT Index Primer 2 (5 μL, S5xx), 2× KAPA HiFi HotStart Ready Mix (25 μL), and nuclease-free water (10 μL) using the following protocol: 95°C for 3 min; 8 cycles of 95°C for 30 s, 55°C for 30 s, and 72°C for 30 s; and 72°C for 5 min. To remove DNA debris and obtain exact DNA fragments from PCR product, the PCR products were cleaned using AMPure XP beads (Beckman), and then quality control of the 16S metagenomic libraries was performed using Agilent Technologies 2100 Bioanalyzer (Agilent). Libraries were standardized and pooled for sequencing on MiSeq platform (Illumina), according to the standard Illumina sequencing protocol. The DNA from the fecal samples was extracted using TruSeq Nano DNA sample Prep Kit (Illumina, CA, USA) for shotgun metagenomic sequencing, following the manufacturer’s instructions by Theragene Bio (Gyeonggi-do, Republic of Korea). The sequencing was performed by Theragene Bio (Gyeonggi-do, Republic of Korea) on an Illumina platform (version 1.9), and the libraries were strandardized and according to the standard Illumina sequencing protocol.

Metagenomic analysis

Illumina adapter sequences of paired-end reads were removed using Cutadapt version 4.1.  After trimming, the reads were processed using QIIME2 version 2022.8. Briefly, the reads were first assigned to each sample according to a unique index, and pairs of reads from the original DNA fragments were merged using an import tool in QIIME2.  Quality control and trimming were performed to yield sequences with lengths of 230 bp and 210 bp for the forward and reverse reads, respectively. The DADA2 software package  wrapped in QIIME2 was used to remove low-quality bases from the reads. A consensus method implemented in DADA2 was used to remove chimeras from the FASTQ files. Alpha and beta diversities were analyzed using core-metrics-phylogenetic analysis in the QIIME2 diversity plugin. Alpha and beta diversities were calculated using alpha- and beta-group significance in the QIIME2 diversity plugin, respectively. Alpha diversity was calculated using observed species (observed features), and beta diversity was compared by principal coordinate analysis using Bray–Curtis distances (BCD). To assess FMT engraftment, we calculated the "Engraftment Distance" using the formula BCDtoDonor + (1 - BCDtoBase). The significance of the similarity between the groups was evaluated using permutational multivariate analysis of variance (PERMANOVA) with 999 permutations. Taxonomic annotation was performed by mapping the training reference set with the primers (forward, 5′-CCTACGGGNGGCWGCAG-3′; reverse, 5′-GACTACHVGGGTATCTAATCC-3′) and extracting the V3–V4 region using the NCBI reference database (https://www.ncbi.nlm.nih.gov/refseq/) with manually added microbiota by using RESCRIPt.  Correlation plots and calculations were generated using the ggpubr package in R (version 4.2.1)  and R Studio (2023.09.1+494). To address the microbial composition analysis between the groups R6 and R7, we employed the Analysis of Composition of Microbiomes (ANCOM).  Additionally, 16S rRNA and marker gene-based shotgun metagenomic analyses were performed. Briefly, quality checks of the sequenced reads were conducted using kneaddata (https://github.com/biobakery/kneaddata) with the hg37dec database (version 0.1.4) and --trimmomatic Trimmomatic-0.39 option.  Subsequently, microbial alignments and abundance calculations were performed using HUMAnN 3 with the default option.  SNP calculation of concatenated multi-sequence alignment was performed using StrainPhlAn 4 with default option.  Multi-sequence alignment was visualized using Jalview.  Phylogenetic distance of concatenated multi-sequence alignment was calculated by using FastTree  (version 2.1.11) with generalized time-reversible model. Tree visualization was performed using FigTree  (version 1.4.4.).

Isolation of P. merdae Immunoactis from donor stool

Fecal samples were suspended in anaerobic PBS and then seeded on brain heart infusion (#237500, BD) agar plats supplemented with 0.5 % yeast extract (#212750, BD), 0.05 % L-cysteine HCl (#C7880, Sigma-Aldrich), 0.0005 % hemin (#51280, Sigma-Aldrich), 0.0001 % vitamin K1 (#95271, Sigma-Aldrich), 0.01 % kanamycin (#HKA01, Rd-tech), and 5 % defibrinated sheep blood (#S1876, KisanBio). The plates were incubated at 37°C under anaerobic conditions using a GasPak 100 system (#260626, BD). Colonies were randomly picked from the plates and tested with PCR for P. merdae Immunoactis species-specific sequences with the primer set, 5’-GCTCAACCTGGGCATTGCA-3’ and 5’-CATGTTTTAGGGATTCGAGCG-3’′. DNA of isolated P. merdae Immunoactis was prepared using TruSeq Nano DNA sample Prep Kit (Illumina, CA, USA) for whole genome sequencing by Theragene Bio (Gyeonggi-do, Republic of Korea). Libraries were standardized and pooled for sequencing on the Illumina platform (version 1.9), according to the standardized Illumina sequencing protocol by Theragene Bio (Gyeonggi-do, Republic of Korea). Sequenced reads were quality-checked using BBDuk plug-in BBMap with--trimq=30 and --maq=30 options,  assembled using Unicycler with --min_dead_end_size 1500 --depth_filter 0.5 option35, and filtered for the PhiX genome using Usearch.  To identify the closest and exact species of isolated P. merdae immunoactis, assembled genome contigs were searched against GenomesDB of JSpeciesWS  by using tetra correlation search with default option. Calculation of the genomic similarity between P. merdae Immunoactis and P. merdae sp. Marseille-P4119  was performed using OrthoANI.

T cell proliferation assay

Human T cells were isolated from blood samples collected from healthy donors in their 20s with informed consent. This study was approved by the Institutional Review Board (20210416-HB-60-07-04) of the Gwangju Institute of Science and Technology (GIST), South Korea. PBMCs were isolated from blood samples of 40–50 mL volume using Ficoll® Paque Plus (#GE17-1440-02, Merck) through the process of density gradient centrifugation. The CD4+ and CD8+ T cells were isolated separately using CD4+ T Cell Isolation Kit (#130-096-533, Miltenyi Biotec) and CD8+ T Cell Isolation Kit (#130-096-495, Miltenyi Biotec), respectively. The isolated T cells were resuspended in RPMI 1640 medium (#10-040-CV, Corning) supplemented with 10% fetal bovine serum (FBS; #92916754, MP Biomedicals), 1% penicillin-streptomycin (#15140-122, Thermo Fisher Scientific), 1× NEAA (#11140-050, Thermo Fisher Scientific), 200 mM HEPES (#15630-080, Thermo Fisher Scientific), 2 mM sodium pyruvate, 2 mM L-glutamine (#25030-081, Thermo Fisher Scientific), and 1× 2-mercaptoethanol (#21985-023, Thermo Fisher Scientific). Additionally, the T cells were stained with carboxyfluorescein diacetate succinimidyl ester (CFSE; #C34554, Thermo Fisher Scientific) for 30 min at 37°C in a 5% CO2 atmosphere. 96-well non-treated plates (#32096, SPL) were pre-coated with 0.2 μg anti-CD3e antibody (#100340, BioLegend) in each well and incubated overnight at 4°C. CFSE-stained T cells were then added to the pre-coated plates at a concentration of 2×105 cells/well and incubated either alone or with 10% bacterial supernatant or three-bacteria mixture for 72 h at 37°C in a 5% CO2 atmosphere. Three-bacteria mixture was composed with the ratio of P. merdae Immunoactis: B. plebeius: L. salivarius = 10 : 1 : 1 based on OD600 value. After 72 h, T cells were harvested and washed with FACS buffer (PBS + 10% FBS + 2 mM EDTA), and their proliferation was analyzed using a BD FACS CANTO II (BD Bioscience). The supernatants from the T cells were also collected and stored at -80°C until use for ELISA. The IFN-γ levels in the supernatants were measured using a human IFN-γ ELISA kit following the manufacturer’s protocol (#88-7316-88, Thermo Fisher Scientific). FlowJo software (Treestar) was used to analyze the data.

OT-1 T cell Cytotoxicity Assay

For the cytotoxicity assay, ovalbumin-specific CD8+ T cells (OT-1 T cells) were used. Briefly, the spleen was isolated from OT-1 mice (#003831, C57BL/6-Tg[TcraTcrb]1100Mjb/J, The Jackson Laboratory) and ground on a 70 μm cell strainer (#352350, Falcon) with 3 mL PBS. Cells were resuspended in 10 mL of 1× RBC lysis buffer (#420301, BioLegend) and incubated for 10 min at room temperature. OT-1 T cells were isolated using a CD8+ T Cell Isolation Kit (#130-096-495, Miltenyi Biotec). Isolated OT-1 T cells were resuspended in the T cell medium. A 60 mm non-treated plate (#32096, SPL) was pre-coated with 5 mg anti-CD3ϵ antibody (#100340, BioLegend) and 0.5 mg anti-CD28 antibody (#102116, BioLegend), and incubated overnight at 4°C. Further, OT-1 T cells were distributed into pre-coated 60 mm plate at 1–2×106 cells/well with 10% bacterial supernatant and incubated for 48 h at 37°C in a 5% CO2 atmosphere. After 48 h of incubation, OT-1 T cells were harvested for RNA isolation, or co-cultured with Ova-treated MC38 cells at a 5:1 ratio in T cell media for 6 h. The cells were then harvested and washed with FACS buffer. Finally, the cells were stained using Annexin V–PI staining kit (#ALX-850-020-K101, Enzo Life Science) and analyzed using flow cytometry. MC38 cells were treated using ovalbumin (323-339) (#O1641, Merck) and incubated overnight at 37°C in a 5% CO2 atmosphere before co-culturing with OT-1 T cells.

Comparison of relative gene expression (RT-PCR)

Activated OT-1 cells were harvested, and RNA was extracted using TRIzol reagent (#15596026, Invitrogen) following the manufacturer's protocol. RNA quality control was conducted by assessing the A260/A280 and A260/A230 ratios. Subsequently, cDNA synthesis was performed using the TOPscript™ RT DryMIX kit (#RT200, Enzynomics), followed by RT-PCR using the TOPreal™ qPCR 2X PreMIX kit (#RT501M, Enzynomics) and a StepOnePlus (#4376600, Applied Biosystems). Gene expression levels were normalized to 1 in the "Ova act" group, and relative expression levels were compared across other experimental groups. Primer sequences and PCR conditions are provided in Table S10.

Immune cell profiling

The lymph nodes and tumor tissues were isolated from mice 21 days after inoculation with MC38 cells. The immune cells analyzed were T cells, NK cells, dendritic cells (DC), Tregs, and macrophages. To isolate the lymphocytes from the lymph nodes, they were ground on a 70 μm cell strainer (#352350, Falcon) with 1 mL PBS (#10010-049, Thermo Fisher Scientific), and the cells were washed with PBS and counted. For the tumor tissue, it was cut into small pieces and incubated in 5 mL dissociation media for 40 min at 37°C. The dissociation media was made using T cell media supplemented with 2.5 mg/mL collagenase type I (#17100-017, Thermo Fisher Scientific), 1.5 mg/mL collagenase type II (#17101-015, Thermo Fisher Scientific), 1 mg/mL collagenase type IV (#17104-019, Thermo Fisher Scientific), 50 μg/mL DNase type I (#10104159001, Merck), and 0.25 mg/mL hyaluronidase type IV-S (#H3884, Merck). After the dissociation process, the samples were centrifuged, and the supernatant was discarded. The cells were then resuspended in 5 mL of 1× RBC lysis buffer (#420301, BioLegend) and incubated for 10 min at room temperature. The samples were filtered through a 70 μm cell strainer and the number of cells was counted. To block the Fc-receptor, the samples were incubated with anti-mouse CD16/CD32 (BioLegend, #101330) for 10 min at room temperature. Antibody mixtures were then added to the samples for surface marker staining and incubated for 1 h at 4°C. In the case of Tregs and macrophages, the samples were fixed and permeabilized using a fixation/permeabilization buffer set (BioLegend, #424401), and an intra Ab mixture was added for intra-marker staining and incubated for 1 h at 4°C. The stained cells were then acquired using CANTO II (BD Biosciences) and BD-FACS Diva software v.8.0.2 (BD Bioscience) and analyzed using FlowJo software (v.10, TreeStar). The staining markers used for each cell type were as follows: T cells were stained with CD45 (#103116, BioLegend), CD3 (#100218, BioLegend), CD4 (#100422, BioLegend), CD8a (#563068, BD Bioscience), CD44 (#563970, BD Bioscience), CD62L (#104412, BioLegend), and PD-1 (#109104, BioLegend); NK cells were stained with CD45 (#103116, BioLegend), CD3 (#100218, BioLegend), NK1.1 (#550627, BD Bioscience), and NKG2A (#11-5896-82, eBioscience); DC were stained with CD11c (#562782, BD Bioscience), CD11b (#566416, BD Bioscience), and B220 (#103224, Biolegend); Tregs were stained with CD45 (#103116, BioLegend), CD3 (#100218, BioLegend), CD4 (#100422, BioLegend), CD8a (#563068, BD Bioscience), CD25 (#101910, BioLegend), Foxp3 (#562996, BD Bioscience), and IFN-γ (#163503, BioLegend); and macrophages were stained with CD45 (#103116, BioLegend), CD11b (#566416, BD Biosciences), F4/80 (#565411, BioLegend), and TNF-α (#506308, BioLegend).

Quantification and Statistical Analysis

Statistical calculations were performed using Prism (Version 9.2.0, GraphPad). Differences between two variables were analyzed using the two-sided unpaired t-test or Wilcoxon-Mann-Whitney test. Multiple variables were assessed by one-way or two-way ANOVA with Tukey's multiple comparison test. Correlation analysis was performed using R version 4.1.0. Statistical significance was considered if the p-value was lower than 0.05.

Acknowledgments

This study was supported by grants from the Asan Institute for Life Sciences, Asan Medical Center (2018IT0608 to S.R.P.), the National Cancer Centre, Korea (NCC-1911267 to H.P. and S.R.P.), the GIST Research Institute (GRI), funded by the GIST (in 2020 and 2023 to H.P.), and the Bio and Medical Technology Development Program (2022R1A2C2008976 and RS-2023-00228315) from the Ministry of Science and ICT, Korean Government.

Author contributions

Conceptualization, H.P. and S.R.P.; resources, S.-Y.K., E.-J.D., J.S.S., S.H.P., S.W.H., M.-N. Kim, S.-H.K., and SR.P.; investigation, G.K., Yunjae Kim, Sujeong Kim, B.C., Seungil Kim, and M.-N. Kweon; data curation, G.K., Yunjae Kim, Seungil Kim, D.-J.B., and SR.P.; visualization, Yunjae Kim, G.K., Sujeong Kim, and D.-J.B.; writing – original draft, G.K., Yunjae Kim, Sujeong Kim, and S.R.P.; writing – review & editing, Yunjae Kim, Sujeong. Kim, G.K., B.C., Yeongmin Kim, K.M., and S.R.P.; project administration, Yunjae Kim, G.K., Sujeong Kim, H.P., and S.R.P.; supervision, M.D.A., C.L., H.P., and SR.P.; funding acquisition, H.P. and SR.P.

Declaration of interests

The authors declare no competing interests.

Supplemental information

References

    • Ribas A.
    • Wolchok J.D.
    Cancer immunotherapy using checkpoint blockade.
    Science. 2018; 359: 1350-1355https://doi.org/10.1126/science.aar4060
    • Schoenfeld A.J.
    • Hellmann M.D.
    Acquired Resistance to Immune Checkpoint Inhibitors.
    Cancer Cell. 2020; 37: 443-455https://doi.org/10.1016/j.ccell.2020.03.017
    • Meric-Bernstam F.
    • Larkin J.
    • Tabernero J.
    • Bonini C.
    Enhancing anti-tumour efficacy with immunotherapy combinations.
    Lancet. 2021; 397: 1010-1022https://doi.org/10.1016/S0140-6736(20)32598-8
    • Keenan T.E.
    • Burke K.P.
    • Van Allen E.M.
    Genomic correlates of response to immune checkpoint blockade.
    Nat. Med. 2019; 25: 389-402https://doi.org/10.1038/s41591-019-0382-x
    • Routy B.
    • Le Chatelier E.
    • Derosa L.
    • Duong C.P.M.
    • Alou M.T.
    • Daillère R.
    • Fluckiger A.
    • Messaoudene M.
    • Rauber C.
    • Roberti M.P.
    • et al.
    Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors.
    Science. 2018; 359: 91-97https://doi.org/10.1126/science.aan3706
    • Gopalakrishnan V.
    • Spencer C.N.
    • Nezi L.
    • Reuben A.
    • Andrews M.C.
    • Karpinets T.V.
    • Prieto P.A.
    • Vicente D.
    • Hoffman K.
    • Wei S.C.
    • et al.
    Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients.
    Science. 2018; 359: 97-103https://doi.org/10.1126/science.aan4236
    • Matson V.
    • Fessler J.
    • Bao R.
    • Chongsuwat T.
    • Zha Y.
    • Alegre M.L.
    • Luke J.J.
    • Gajewski T.F.
    The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients.
    Science. 2018; 359: 104-108https://doi.org/10.1126/science.aao3290
    • Baruch E.N.
    • Youngster I.
    • Ben-Betzalel G.
    • Ortenberg R.
    • Lahat A.
    • Katz L.
    • Adler K.
    • Dick-Necula D.
    • Raskin S.
    • Bloch N.
    • et al.
    Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients.
    Science. 2021; 371: 602-609https://doi.org/10.1126/science.abb5920
    • Davar D.
    • Dzutsev A.K.
    • McCulloch J.A.
    • Rodrigues R.R.
    • Chauvin J.M.
    • Morrison R.M.
    • Deblasio R.N.
    • Menna C.
    • Ding Q.
    • Pagliano O.
    • et al.
    Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients.
    Science. 2021; 371: 595-602https://doi.org/10.1126/science.abf3363
    • Maaloum M.
    • Afouda P.
    • Lo C.I.
    • Dubourg G.
    • Nguyen T.T.
    • Levasseur A.
    • Saile R.
    • Raoult D.
    • Fournier P.-E.
    Prevotella merdae sp. nov., a new bacterial species isolated from human faeces.
    FEMS Microbiol. Lett. 2022; 369fnac066https://doi.org/10.1093/femsle/fnac066
    • Topp B.G.
    • Channavazzala M.
    • Mayawala K.
    • De Alwis D.P.
    • Rubin E.
    • Snyder A.
    • Wolchok J.D.
    • Ribas A.
    Tumor dynamics in patients with solid tumors treated with pembrolizumab beyond disease progression.
    Cancer Cell. 2023; 41: 1680-1688.e2https://doi.org/10.1016/j.ccell.2023.08.004
    • Lim M.
    • Muquith M.
    • Miramontes B.
    • Espinoza M.
    • Hsiehchen D.
    Treatment Beyond Progression After Anti-PD-1 Blockade in Hepatocellular Carcinoma.
    Cancer Res. Commun. 2023; 3: 1912-1916https://doi.org/10.1158/2767-9764.CRC-23-0025
    • Halsey T.M.
    • Thomas A.S.
    • Hayase T.
    • Ma W.
    • Abu-Sbeih H.
    • Sun B.
    • Parra E.R.
    • Jiang Z.-D.
    • DuPont H.L.
    • Sanchez C.
    • et al.
    Microbiome alteration via fecal microbiota transplantation is effective for refractory immune checkpoint inhibitor–induced colitis.
    Sci. Transl. Med. 2023; 15eabq4006https://doi.org/10.1126/scitranslmed.abq4006
    • Obeidat A.
    • Silangcruz K.
    • Kozai L.
    • Wien E.
    • Fujiwara Y.
    • Nishimura Y.
    Clinical characteristics and outcomes of gastritis associated with immune checkpoint inhibitors: scoping review.
    J. Immunother. 2022; 45: 363-369https://doi.org/10.1097/CJI.0000000000000435
    • Cain D.W.
    • Cidlowski J.A.
    Immune regulation by glucocorticoids.
    Nat. Rev. Immunol. 2017; 17: 233-247https://doi.org/10.1038/nri.2017.1
    • Barnes P.J.
    Corticosteroid effects on cell signalling.
    Eur. Respir. J. 2006; 27: 413-426https://doi.org/10.1183/09031936.06.00125404
    • Xu J.
    • Lian F.
    • Zhao L.
    • Zhao Y.
    • Chen X.
    • Zhang X.
    • Guo Y.
    • Zhang C.
    • Zhou Q.
    • Xue Z.
    • et al.
    Structural modulation of gut microbiota during alleviation of type 2 diabetes with a Chinese herbal formula.
    ISME J. 2015; 9: 552-562https://doi.org/10.1038/ismej.2014.177
    • Huang E.Y.
    • Inoue T.
    • Leone V.A.
    • Dalal S.
    • Touw K.
    • Wang Y.
    • Musch M.W.
    • Theriault B.
    • Higuchi K.
    • Donovan S.
    • et al.
    Using corticosteroids to reshape the gut microbiome: implications for inflammatory bowel diseases.
    Inflamm. Bowel Dis. 2015; 21: 963-972https://doi.org/10.1097/MIB.0000000000000332
    • Couch C.E.
    • Neal W.T.
    • Herron C.L.
    • Kent M.L.
    • Schreck C.B.
    • Peterson J.T.
    Gut microbiome composition associates with corticosteroid treatment, morbidity, and senescence in Chinook salmon (Oncorhynchus tshawytscha).
    Sci. Rep. 2023; 13: 2567https://doi.org/10.1038/s41598-023-29663-0
    • Wang M.
    • Zhu Z.
    • Lin X.
    • Li H.
    • Wen C.
    • Bao J.
    • He Z.
    Gut microbiota mediated the therapeutic efficacies and the side effects of prednisone in the treatment of MRL/lpr mice.
    Arthritis Res. Ther. 2021; 23: 240https://doi.org/10.1186/s13075-021-02620-w
    • Zuo T.
    • Ng S.C.
    The gut microbiota in the pathogenesis and therapeutics of inflammatory bowel disease.
    Front. Microbiol. 2018; 9: 2247https://doi.org/10.3389/fmicb.2018.02247
    • Belkaid Y.
    • Hand T.W.
    Role of the microbiota in immunity and inflammation.
    Cell. 2014; 157: 121-141https://doi.org/10.1016/j.cell.2014.03.011
    • Martin M.
    Cutadapt removes adapter sequences from high-throughput sequencing reads.
    EMBnet. j. 2011; 17: 3https://doi.org/10.14806/ej.17.1.200
    • Bolyen E.
    • Rideout J.R.
    • Dillon M.R.
    • Bokulich N.A.
    • Abnet C.
    • Al-Ghalith G.A.
    • Alexander H.
    • Alm E.J.
    • Arumugam M.
    • Asnicar F.
    QIIME 2: Reproducible, interactive, scalable, and extensible microbiome data science.
    PeerJ. 2018; (Preprint at)
    • Callahan B.J.
    • McMurdie P.J.
    • Rosen M.J.
    • Han A.W.
    • Johnson A.J.A.
    • Holmes S.P.
    DADA2: High-resolution sample inference from Illumina amplicon data.
    Nat. Methods. 2016; 13: 581-583https://doi.org/10.1038/nmeth.3869
    • Robeson M.S.
    • O’Rourke D.R.
    • Kaehler B.D.
    • Ziemski M.
    • Dillon M.R.
    • Foster J.T.
    • Bokulich N.A.
    RESCRIPt: reproducible sequence taxonomy reference database management.
    PLoS Comput. Biol. 2021; 17e1009581https://doi.org/10.1371/journal.pcbi.1009581
    • Mandal S.
    • Van Treuren W.
    • White R.A.
    • Eggesbø M.
    • Knight R.
    • Peddada S.D.
    Analysis of composition of microbiomes: a novel method for studying microbial composition.
    Microb. Ecol. Health Dis. 2015; 26: 27663https://doi.org/10.3402/mehd.v26.27663
    • Beghini F.
    • McIver L.J.
    • Blanco-Míguez A.
    • Dubois L.
    • Asnicar F.
    • Maharjan S.
    • Mailyan A.
    • Manghi P.
    • Scholz M.
    • Thomas A.M.
    • et al.
    Integrating taxonomic, functional, and strain-level profiling of diverse microbial communities with bioBakery 3.
    eLife. 2021; 10e65088https://doi.org/10.7554/eLife.65088
    • Truong D.T.
    • Tett A.
    • Pasolli E.
    • Huttenhower C.
    • Segata N.
    Microbial strain-level population structure and genetic diversity from metagenomes.
    Genome Res. 2017; 27: 626-638https://doi.org/10.1101/gr.216242.116
    • Waterhouse A.M.
    • Procter J.B.
    • Martin D.M.
    • Clamp M.
    • Barton G.J.
    Jalview Version 2—a multiple sequence alignment editor and analysis workbench.
    Bioinformatics. 2009; 25: 1189-1191
    • Price M.N.
    • Dehal P.S.
    • Arkin A.P.
    FastTree 2–approximately maximum-likelihood trees for large alignments.
    PLoS One. 2010; 5e9490https://doi.org/10.1371/journal.pone.0009490
    • Rambaut A.
    FigTree v1. 4.
    • Bushnell B.
    BBMap: a fast, accurate, splice-aware aligner.
    Lawrence Berkeley National Lab. [LBNL], 2014
    • Wick R.R.
    • Judd L.M.
    • Gorrie C.L.
    • Holt K.E.
    Unicycler: resolving bacterial genome assemblies from short and long sequencing reads.
    PLoS Comput. Biol. 2017; 13e1005595https://doi.org/10.1371/journal.pcbi.1005595
    • Edgar R.C.
    Search and clustering orders of magnitude faster than BLAST.
    Bioinformatics. 2010; 26: 2460-2461https://doi.org/10.1093/bioinformatics/btq461
    • Richter M.
    • Rosselló-Móra R.
    • Oliver Glöckner F.
    • Peplies J.
    JSpeciesWS: a web server for prokaryotic species circumscription based on pairwise genome comparison.
    Bioinformatics. 2016; 32: 929-931https://doi.org/10.1093/bioinformatics/btv681
    • Lee I.
    • Ouk Kim Y.
    • Park S.-C.
    • Chun J.
    OrthoANI: an improved algorithm and software for calculating average nucleotide identity.
    Int. J. Syst. Evol. Microbiol. 2016; 66: 1100-1103https://doi.org/10.1099/ijsem.0.000760
    • Marrero J.A.
    • Kulik L.M.
    • Sirlin C.B.
    • Zhu A.X.
    • Finn R.S.
    • Abecassis M.M.
    • Roberts L.R.
    • Heimbach J.K.
    Diagnosis, Staging, and Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases.
    Hepatology. 2018; 68: 723-750https://doi.org/10.1002/hep.29913
    • Eisenhauer E.A.
    • Therasse P.
    • Bogaerts J.
    • Schwartz L.H.
    • Sargent D.
    • Ford R.
    • Dancey J.
    • Arbuck S.
    • Gwyther S.
    • Mooney M.
    New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1).
    Eur. J. Cancer. 2009; 45: 228-247
    • Klindworth A.
    • Pruesse E.
    • Schweer T.
    • Peplies J.
    • Quast C.
    • Horn M.
    • Glöckner F.O.
    Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies.
    Nucleic Acids Res. 2013; 41: e1https://doi.org/10.1093/nar/gks808
    • Wickham H.
    ggplot2: Elegant Graphics for Data Analysis.
    Springer, 2016
    • Bolger A.M.
    • Lohse M.
    • Usadel B.
    Trimmomatic: a flexible trimmer for Illumina sequence data.
    Bioinformatics. 2014; 30: 2114-2120https://doi.org/10.1093/bioinformatics/btu170

Article info

Publication history

Published: July 25, 2024
Accepted: June 10, 2024
Received in revised form: April 30, 2024
Received: October 17, 2023

Publication stage

In press, corrected proof

Identification

DOI: https://doi.org/10.1016/j.chom.2024.06.010

Copyright

© 2024 Elsevier Inc. All rights are reserved, including those for text and data mining, AI training, and similar technologies.

ScienceDirect

Access this article on ScienceDirect

Figures

  • Figure thumbnail fx1
    Graphical Abstract
  • Figure thumbnail gr1
    Figure 1Clinical responses to FMT combined with nivolumab in patients with advanced solid cancer refractory to nivolumab
  • Figure thumbnail gr2
    Figure 2Analysis of longitudinal tumor reduction and immune-related changes in recipient #7
  • Figure thumbnail gr3
    Figure 3Discovery of key bacteria influencing clinical outcomes post-FMT in recipient #7
  • Figure thumbnail gr4
    Figure 4Administration of P. merdae Immunoactis suppresses tumor growth by enhancing immune cell activity

Tables

Related Articles

Reply all
Reply to author
Forward
0 new messages