Abstract
Gut microbiota influence the antitumour efficacy of immune checkpoint blockade1,2,3,4,5,6, but the mechanisms of action have not been fully elucidated. Here, we show that a new strain of the bacterial genus Hominenteromicrobium (designated YB328) isolated from the faeces of patients who responded to programmed cell deathâ1 (PD-1) blockade augmented antitumour responses in mice. YB328 activated tumour-specific CD8+ Tâcells through the stimulation of CD103+CD11bâ conventional dendritic cells (cDCs), which, following exposure in the gut, migrated to the tumour microenvironment. Mice showed improved antitumour efficacy of PD-1 blockade when treated with faecal transplants from non-responder patients supplemented with YB238. This result suggests that YB328 could function in a dominant manner. YB328-activated CD103+CD11bâ cDCs showed prolonged engagement with tumour-specific CD8+ Tâcells and promoted PD-1 expression in these cells. Moreover, YB238-augmented antitumour efficacy of PD-1 blockade treatment was observed in multiple mouse models of cancer. Patients with elevated YB328 abundance had increased infiltration of CD103+CD11bâ cDCs in tumours and had a favourable response to PD-1 blockade therapy in various cancer types. We propose that gut microbiota enhance antitumour immunity by accelerating the maturation and migration of CD103+CD11bâ cDCs to increase the number of CD8+ Tâcells that respond to diverse tumour antigens.
Similar content being viewed by others
Main
Immune checkpoint blockade (ICB) therapies, including monoclonal antibodies against PD-1 and PD-1 ligand (PD-L1), are used in many cancer types7. PD-1 blockade therapies have led to significant improvements in progression-free survival (PFS) and overall survival. However, the complete response rate remains low8,9,10,11. Therefore, predictive biomarkers for clinical efficacy and new approaches to overcome resistance mechanisms are needed. Notably, high ratios of PD-1+CD8+ Tâcells to PD-1+ regulatoryâT (Treg) cells in the tumour microenvironment (TME) correspond to the responsiveness to PD-1 and PD-L1 inhibitors12. Moreover, PD-1 expression by CD8+ Tâcells is induced by Tâcell activation signalling, especiallyNFATC1, through the stimulation of fully matured dendritic cells (DCs)13,14.
In gut microbiota, the abundance of certain bacteria (for example, Ruminococcus spp. and Prevotellaceae spp.) is associated with the clinical efficacy of ICB therapies1,2,3,4,5,6. Several mechanisms of microbiome-mediated antitumour immunity have been proposed, including stimulation of macrophages and monocytes15, antigenic mimics between microbiota and tumour antigens16,17,18 or direct activation of tumour-infiltrating CD8+ Tâcells by microbiota-derived metabolites19,20,21,22. Moreover, a consortium of bacteria isolated from healthy individuals reportedly stimulated CD103+ DCs, which led to the activation of effector CD8+ Tâcells23. However, the detailed mechanisms that underlie the connection between DC maturation by gut microbiota and the activation of tumour antigen-specific PD-1+CD8+ Tâcells in the distant TME have not been fully elucidated.
Here, we identified a new bacterial strain, designated YB328, that was significantly enriched in faeces of patients who responded to PD-1 blockade therapies. YB328 accelerated the differentiation of CD103+CD11bâ cDCs, which is an essential DC subset for antigen cross-presentation to CD8+ Tâcells24,25. YB328 mediated cDC differentiation through the phosphorylation of S6K and STAT3 and the induction of IRF8 through the stimulation of multiple Toll-like receptors (TLRs) in the gut. YB328-stimulated CD103+CD11bâ cDCs were further activated by other bacteria and migrated to tumour-draining lymph nodes (dLNs) and the TME. Once in these locations, the cDCs primed and activated CD8+ Tâcells and induced PD-1+CD8+ Tâcells specific for a broad range of tumour antigens. Overall, we describe in detail the mechanism of the activation of antigen-specific CD8+ Tâcells in the distant TME by gut microbiota.
Gut microbiota and antitumour efficacy
We prospectively collected stool samples from 50 Japanese patients with cancer (15 patients with non-small cell lung cancer (NSCLC) and 35 patients with gastric cancer (GC)) who received PD-1 blockade therapy. These patients were classified as responders or non-responders (Supplementary Tables 1 and 2). Clinicopathological features that are reportedly associated with a favourable clinical response to PD-1 blockade (listed in Supplementary Tables 3â5) were not significantly different in our cohort, except for PD-L1 expression by tumour cells in patients with GC. 16S rRNA gene amplicon sequencing of faecal samples showed that the Shannon diversity of gut microbiota was significantly higher in responders than in non-responders (Fig. 1a and Extended Data Fig. 1a,b,f). Receiver operating characteristic (ROC) analysis further showed that the Shannon diversity was a significant predictive biomarker for identifying responders (Fig. 1b and Extended Data Fig. 1c, left, g, left). Moreover, patients with high diversity of gut microbiota exhibited significantly prolonged PFS, regardless of the cancer type (Fig. 1c, Supplementary Table 6 and Extended Data Fig. 1c, right, g, right). Principal coordinate analysis (PCoA) and analysis of similarities (ANOSIM) based on unweighted unique fraction metric (UniFrac) distances revealed a significant difference in gut microbiota between responders and non-responders (Fig. 1d and Extended Data Fig. 1a,d,h). Linear discriminant analysis effect size (LEfSe) showed that members of several families, namely, Ruminococcaceae, Propionibacteriaceae, Dehalobacteriaceae and Turicibacteraceae, were enriched in responders (Fig. 1e,f and Extended Data Fig. 1e,i,j). Among them, Ruminococcaceae abundance was significantly correlated with prolonged PFS after PD-1 blockade therapy and showed the highest AUC score (Fig. 1g and Extended Data Fig. 1k). In contrast to previous reports1,2,3,4,18, the abundance of the genera Faecalibacterium, Enterococcus, Bifidobacterium and Akkermansia did not differ significantly between responders and non-responders (Extended Data Fig. 1l). Notably, members of the families Bacteroidaceae and Veillonellaceae, which were enriched in non-responders, were associated with a significantly shorter PFS (Fig. 1e,f,h and Extended Data Fig. 1e,iâk). We also confirmed that patients with high Shannon diversity values, a high abundance of Ruminococcaceae and a low abundance of Bacteroidaceae, when stratified by the median cut-off, had prolonged PFS (Extended Data Fig. 1m).
Faecal samples were collected from patients (nâ=â15 NSCLC, nâ=â35 GC) who received PD-1 blockade therapy. a, Comparison of Shannon diversity of gut microbiota of responders and non-responders. b, ROC analysis of the diversity (Shannon index) of microbiota in faeces. AUC, area under the curve. c, KaplanâMeier curves for PFS of patients with high or low Shannon diversity of gut microbiota. High and low Shannon diversity of gut microbiota in patients were stratified by ROC analysis (b). d, Differences between gut microbiota of responders and non-responders as visualized by PCoA based on unweighted UniFrac distances. The ellipses represent the 95% confidence intervals (CIs). e, Cladogram of differentially abundant taxa identified by LEfSe analysis. Symbol sizes are scaled by the relative abundance. f, Linear discriminant analysis (LDA) scores of the significantly different taxa between responders and non-responders. g,h, ROC analysis (left) and KaplanâMeier curves (right) for PFS in patients with high or low Ruminococcaceae (g) or Bacteroidaceae (h) abundance. iâo, Immune profiles of tumour-infiltrating lymphocytes (TILs) in patients with both biopsy and faecal samples available (nâ=â4 NSCLC, nâ=â22 GC). Correlations between subsets of TILs and bacterial families enriched in responders and non-responders (i). The frequency of PD-1+CD8+ Tâcells (representative staining (j,m) and summary (k,n)) in patients with high or low abundances of Ruminococcaceae (k) or Bacteroidaceae (n) stratified by ROC analysis in g and h. Cells were gated on CD8 TILs (j,m). The correlation between PD-1 expression by tumour-infiltrating CD8+ Tâcells and the abundance of Ruminococcaceae (l) or Bacteroidaceae (o). Significance was assessed using two-sided MannâWhitney U-test, with data presented as the median (a,k,n); two-sided log-rank test (c, g, right, h, right); ANOSIM with one-tailed significance computed using 999 permutations) (d); or two-sided Pearsonâs correlation (i,l,o).
Among the multiple parameters associated with antitumour immunity, the abundance of PD-1+CD8+ Tâcells in the TME, which is correlated with the response to PD-1 blockade therapy12,26,27,28, was significantly correlated with the abundance of Ruminococcaceae (Fig. 1iâl and Extended Data Fig. 1n). By contrast, the abundance of Bacteroidaceae was associated with a decrease in PD-1+CD8+ Tâcells and an increase in Treg cell infiltration in the TME (Fig. 1i,mâo). These findings indicate a clear correlation between specific gut microbiota compositions and immune responses.
Gut microbiota regulate antitumour responses
Germ-free (GF) mice colonized with faeces from responders, but not from non-responders, exhibited strong antitumour effects against MC38 tumours after anti-PD-1 monoclonal antibody (anti-PD-1) treatment (Extended Data Fig. 2a,b) and showed increased Shannon diversity in the faeces (Extended Data Fig. 2c). Moreover, Ruminococcaceae was enriched in mice who received faecal microbiota transplantation (FMT) derived from responders (R-FMT), whereas Bacteroidaceae was enriched in mice who received FMT derived from non-responders (NR-FMT) (Extended Data Fig. 2dâg). To exclude the influence of congenital immunodeficiency in GF mice, we performed the same experiments in specific-pathogen-free (SPF) mice preconditioned with antibiotics (ATB-SPF mice). Consistent with GF mice, anti-PD-1 treatment significantly inhibited the growth of MC38 and EMT6 tumours in ATB-SPF mice that received R-FMT (Extended Data Fig. 3aâc). Analyses of the TME demonstrated that activated CD8+ Tâcells (PD-1+CD8+ Tâcells and CD62LâCD44+ effector memory CD8+ Tâcells) and cytokine-producing CD8+ Tâcells (IFNγ+TNF+) were significantly higher in mice treated with R-FMT and anti-PD-1 than in mice treated with NR-FMT and anti-PD-1 or in control mice (Extended Data Fig. 3dâf). Moreover, tumour-infiltrating PD-1+CD8+ Tâcells from mice treated with R-FMT and anti-PD-1 exhibited a broader T cell receptor (TCR) Vβ repertoire diversity than mice treated with NR-FMT and anti-PD-1 (Extended Data Fig. 3gâi).
As PD-1 expression by Tâcells correlates with the strength of Tâcell activation signalling13,29, we examined the maturation of DCs in the TME, a process that has a crucial role in Tâcell activation. The expression levels of DC maturation markers (CD86, CD40 and CD80) were significantly higher in R-FMT-treated mice than in NR-FMT-treated mice (Extended Data Fig. 3jâl). Therefore, gut microbiota augment PD-1 blockade efficacy by promoting mature DC infiltration and PD-1+CD8+ Tâcell accumulation in the TME.
YB328 promotes antitumour immunity
Given the strong antitumour efficacy of R-FMT, we aimed to isolate bacterial strains from the faeces of responders. A previously undescribed strain, designated YB328, was isolated, and its characteristics were investigated (Fig. 2a, Supplementary Tables 7â9). Taxonomic assignment based on its full-length 16S rRNA gene sequence and the Greengenes database (v.13â8) indicated that strain YB328 belongs to the family Ruminococcaceae. Based on its whole-genome sequence and the Genome Taxonomy Database (GTDB; v.220), strain YB328 was assigned to the recently described species Hominenteromicrobium mulieris30 (Extended Data Fig. 4a), sharing an average 97.3% nucleotide identity to the GTDB species-representative genome. Notably, the family-level name depended on the taxonomic framework, namely, Ruminococcaceae (Greengenes), Oscillospiraceae (NCBI taxonomy) or Acutalibacteraceae (GTDB taxonomy). Moreover, YB328 was detected not only in the Japanese population but also in individuals worldwide (Extended Data Fig. 4b).
aâe, The morphological traits of YB328. a, Left, scanning electron microscopy image. Scale bar, 1âµm. Right, transmission electron microscopy image of negatively stained cells. The arrows indicate extracellular membrane vesicles secreted by YB328. Scale bar, 0.5âµm. b,d, Relative abundances of YB328 (b) and P.âvulgatus (d) in faecal samples of patients shown in Fig. 1aâh (nâ=â50). c,e, ROC curves (left) and KaplanâMeier curves (right) of the PFS of patients with high or low abundances of YB328 (c) or P.âvulgatus (e) in faeces. f,g, Tumour growth curves of SPF mice with B16F10 (f) or MC38 (g) tumours (left, summary; right, each mouse). nâ=â4â6 mice per group. Here and in other figures, control indicates mice treated with an isotype control antibody. hâj, The frequency (representative staining (left) and summary (right)) of PD-1 (h, nâ=â8), CD62L and CD44 (i, nâ=â7) and IFNγ and TNF production (j, nâ=â7) by CD8+ Tâcells in MC38 tumours. Cells were gated on CD8+ TILs (hâj). kâm, The mean fluorescent intensity (MFI) of CD86 (k, nâ=â6), CD40 (l, control or anti-PD-1, nâ=â3, P.âvulgatus or YB328, nâ=â4) and H-2Kb (a marker for MHCI) (m, nâ=â6) by CD11c+MHCII+ DCs in MC38 tumours. nâp, Tumour growth curves (n, nâ=â5â7 mice per group), TCR Vβ repertoire (o) and numbers of skewed TCR Vβ clones (>10% of each clone) (p) among PD-1+CD8+ Tâcells (nâ=â5 mice per group). For b and d, each dot indicates one patient, and the data are presented as the median. For f (left), g (left) and n, the average tumour sizes of the groups on a certain day are shown as dots and are presented as the meanâ±âs.d. For hâm and p, each dot in the summary graphs indicates one mouse. Data are presented as the meanâ±âs.d. (hâm) or median (p). Significance was assessed using two-sided MannâWhitney U-test (b,d,p), two-sided log-rank test (c, right, e, right), two-way analysis of variance (ANOVA) with the TukeyâKramer method (f,g,n) or one-way ANOVA with Bonferroni correction (hâm).
We further evaluated the abundance of YB328 in the gut microbiome through metagenome sequencing and species-level quantification using Kraken2 and Bracken against a custom version of the GTDB (v.220). This analysis showed that the abundance of YB328 was significantly higher in the faeces of responders than in non-responders (Fig. 2b). Patients with abundant YB328, defined by a ROC-based cut-off value, had a significantly longer PFS (Fig. 2c), even after excluding patients with high microsatellite instability (Extended Data Fig. 4c). By contrast, Phocaeicola vulgatus (represented by strain AE61, a newly isolated strain from this study belonging to the family Bacteriodeceae and enriched in non-responders), showed an increased abundance in non-responders. Moreover, patients with abundant P.âvulgatus had a shorter PFS (Fig. 2d,e). Similar data were obtained using the median cut-off value (Extended Data Fig. 4d). As P.âvulgatus is a well-known commensal bacterium31, we used the patient-isolated P.âvulgatus strain AE61 to represent gut microbiota of non-responders and evaluated its effects on immunotherapy responses in animal models together with YB328. Although YB328 administration alone did not inhibit tumour growth (Extended Data Fig. 4e), YB328 augmented the antitumour effect of anti-PD-1 treatment more than P.âvulgatus in ATB-SPF mice. This effect was observed in both poorly immunogenic B16F10 and immunogenic MC38 tumour models, with colonization beyond the gavage period (Fig. 2f,g and Extended Data Fig. 4f). Other bacterial strains enriched and isolated from the faeces of responders exhibited few or no antitumour effects compared with YB328 and PD-1 blockade therapy (Extended Data Fig. 4g).
Mice treated with YB328 and anti-PD-1 had higher abundances of activated CD8+ Tâcells and cytokine-producing CD8+ Tâcells in the TME than mice treated with P.âvulgatus and anti-PD-1 or control mice (Fig. 2hâj and Extended Data Fig. 5aâc). However, the frequencies of Treg cells and PD-1+ Treg cells were comparable across groups (Extended Data Fig. 5d,e). Nevertheless, DCs in the TME of YB328-colonized mice exhibited significantly increased expression of co-stimulatory molecules (Fig. 2k,l and Extended Data Fig. 5f) and recapitulated the phenotypes of DCs in mice that received R-FMT. Moreover, major histocompatibility classâI (MHCI) expression was significantly upregulated in these DCs (Fig. 2m). Taken together, these results suggest that YB328 colonization induces DC maturation with high efficiency antigen presentation to activate tumour-infiltrating CD8+ Tâcells. Furthermore, the administration of YB328, but not P.âvulgatus, favourably changed the Shannon diversity of gut microbiota (Extended Data Fig. 5g).
YB328 redirects treatment sensitivity
ATB-SPF mice treated with NR-FMT and colonized with YB328, but not P.âvulgatus, showed increased sensitivity to anti-PD-1 therapy (Fig. 2n and Extended Data Fig. 5h,i). Moreover, YB328 administration increased the diversity of the TCR Vβ repertoire of tumour-infiltrating PD-1+CD8+ Tâcells among skewed TCRs in these mice (Fig. 2o,p and Extended Data Fig. 5j). The administration of YB328, but not P.âvulgatus, favourably altered the Shannon diversity of gut microbiota (Extended Data Fig. 5k). Consistently, in our GC and NSCLC cohort of patients, increased levels of YB328 were positively correlated with the Shannon diversity of gut microbiota (Extended Data Fig. 5l).
However, the co-administration of YB328 and P.âvulgatus abolished the antitumour immunity induced by YB328 (Extended Data Fig. 5m). This effect was in contrast to YB328 single administration in mice treated with NR-FMT, which showed effective induction of antitumour immunity in combination with anti-PD-1 treatment. The co-administration of P.âvulgatus prevented the colonization of YB328 in ABT-SPF mice. However, YB328 sufficiently colonized the gut of mice treated with NR-FMT at a similar level to that observed with YB328 administration alone. This finding indicated that YB328 colonization is not significantly influenced by the microbial environment shaped by NR-FMT (Extended Data Fig. 5n). Indeed, P.âvulgatus administration led to significantly greater engraftment of P.âvulgatus than NR-FMT (Extended Data Fig. 5o). These observations underscore the impact of competitive dominance on the efficacy of microbiome-based therapies.
YB328-treated DCs activate CD8+ T cells
To understand the mechanisms of DC maturation, we analysed bone-marrow-derived DCs (BMDCs) treated with P.âvulgatus or YB328. Compared with P.âvulgatus-treated BMDCs, YB328-treated BMDCs were larger and presented multiple pseudopodia. Moreover, they had increased expression of CD86, CD80 and MHCI and increased levels of IL-12p70 and the chemokines CXCL9, CXCL10 and CCL5 (Fig. 3aâd and Extended Data Fig. 6aâc). When ovalbumin (OVA)-specific (OT-I) TCR transgenic CD8+ Tâcells were cultured with bacteria-treated DCs pulsed with OVA peptides, YB328-treated BMDCs were more strongly attracted and were in contact with CD8+ Tâcells for a longer time than P.âvulgatus-treated BMDCs (Fig. 3e,f and Supplementary Videos 1â3). No difference was observed in CCL22 production, which recruits Treg cells, among YB328-treated, P.âvulgatus-treated and unstimulated BMDCs (Extended Data Fig. 6c). This result is consistent with the comparable Treg cell infiltration levels observed in the TME across treatments.
aâd, BMDCs were stimulated with YB328 or P.âvulgatus. MFI summaries of the maturation markers CD86 (a, nâ=â6 wells per group) and H-2Kb (b, nâ=â5 wells per group), IL-12p70 production (c, nâ=â5 wells per group) and chemokine (CXCL9 and CXCL10) expression (d, nâ=â8 wells per group) in BMDCs are shown. e,f, Dynamic imaging of CD8+ Tâcells and BMDCs stimulated with YB328 or P.âvulgatus. e, Contact events were evaluated at 10-min intervals (right), and a representative trajectory of the indicated cell (left) was extracted from Supplementary Video 1. f, Images showing the interaction between DCs and CD8+ Tâcells. The elapsed time is shown in minutes and seconds. nâ=â4 cells per group. Scale bars, 10âμm. gâm, OT-I CD8+ Tâcells were cultured with bacteria-treated BMDCs pulsed with the indicated concentrations of antigen peptides. g,i, Expression levels of TCR signalling molecules by CD8+ Tâcells induced by N4 peptide (g) or Q4H7 peptide (i). NS, not significant. h, Representative confocal images of NFATC1 nuclear translocation (left) and MFI summary (right) Scale bars, 20âµm (first and third columns) and 10âµm (second and fourth columns). jâm, The frequency of expression of the effector molecules PD-1 (j,l) and CD62L and CD44 (k,m) in CD8+ Tâcells induced by treatment with N4 peptide (j,k) or Q4H7 peptide (l,m). Each dot in the summary graphs indicates a well (aâd,gâm), and the data are presented as the meanâ±âs.d. (aâf,h,jâm) or the mean (g,i). Significance was assessed using one-way ANOVA with Bonferroni correction (aâd) or two-sided unpaired t-test (eâm).
OT-I CD8+ Tâcells were cultured with DCs and then pulsed with different OVA peptides capable of delivering titrated levels of TCR signalling32. YB328-treated and P.âvulgatus-treated BMDCs pulsed with higher doses (100ânM to 1âμM) of high-affinity OVA peptide (N4) presented comparable levels of phosphorylated ZAP70. When BMDCs were pulsed with lower doses (0.01ânM to 10ânM) of N4, YB328-treated BMDCs, but not P.âvulgatus-treated BMDCs, exhibited ZAP70 phosphorylation. YB328-treated BMDCs also stimulated strong phosphorylation of co-stimulatory signals (pJNK, pERK1 and pERK2 (pERK1/2), pAKT and pS6K) in CD8+ Tâcells (Fig. 3g and Extended Data Fig. 7a). YB328-treated BMDCs pulsed with a low dose (10ânM) of the N4 peptide induced extensive NFATC1 nuclear translocation in CD8+ Tâcells, whereas P.âvulgatus-treated BMDCs induced only limited translocation (Fig. 3h). YB328-treated BMDCs pulsed with N4 peptides, regardless of the dose, were correlated with the successful activation of and increased cytokine production by CD8+ Tâcells, whereas P.âvulgatus-treated BMDCs induced PD-1 expression only when pulsed with high N4 doses (Fig. 3j,k and Extended Data Fig. 7bâd). Moreover, when supplied with low-affinity OVA peptide (Q4H7), YB328-treated BMDCs induced ZAP70 phosphorylation and co-stimulatory signalling (Fig. 3i and Extended Data Fig. 7e) at relatively high doses and promoted the nuclear translocation of NFATC1 in CD8+ Tâcells (Fig. 3h). YB328-treated BMDCs consistently induced increased PD-1 expression, which correlated with the activation of and cytokine production by CD8+ Tâcells, regardless of the dose (Fig. 3l,m and Extended Data Fig. 7fâh).
YB328 enhances the differentiation of cDCs
Analysis of the transcriptional profile revealed higher expression of Irf8 and Batf3 in YB328-treated DCs than in P.âvulgatus-treated DCs and untreated DCs (Fig. 4aâc). BATF3 maintains IRF8 autoactivation to commit to the terminal differentiation of CD103+CD11bâ cDCs, which have a crucial role in antitumour immunity by effectively activating CD8+ Tâcells33,34. Accordingly, the number of CD103+CD11bâ cDC progenies increased in response to YB328 treatment (Fig. 4d and Extended Data Fig. 8a). By contrast, IRF4, which is a key transcription factor of CD103âCD11b+ cDCs35, was highly expressed by P.âvulgatus-treated DCs (Fig. 4e). To further understand the mechanisms of CD103+CD11bâ cDC differentiation induced by YB328 treatment, common DC progenitors (CDPs) were cultured with YB328 or P.âvulgatus and with FLT3 ligand (FLT3L), a factor essential for the differentiation of CDPs into CD103+CD11bâ cDCs36,37. IRF8 expression was significantly upregulated in CDPs stimulated with YB328 at low concentrations of FLT3L, although IRF8 expression was comparable between the groups treated with high concentrations of FLT3L (Fig. 4f). This result suggests that YB328 activates an alternative signalling pathway in CDPs that compensates for FLT3L signalling. FLT3L treatment promotes both PI3KâmTOR and STAT3 signalling for the development of CD103+CD11bâ cDCs38,39, and these two pathways regulate Batf3 expression40,41. YB328 stimulation increased levels of S6K and STAT3 phosphorylation compared with P.âvulgatus stimulation (Fig. 4g,h). YB328-induced Irf8 expression was reduced by an S6K inhibitor (rapamycin) or a STAT3 inhibitor (HJC0152) and was more significantly inhibited by the administration of both inhibitors (Fig. 4i). Consequently, the number of CD103+CD11bâ cDCs induced by YB328 stimulation was decreased by rapamycin and/or HJC0152 treatment (Fig. 4j). The antitumour effects induced by combination treatment with YB328 and anti-PD-1 were completely abrogated in Batf3â/â mice, which lack a subset of CD103+CD11bâ cDCs42 (Fig. 4k and Extended Data Fig. 8bâd). By contrast, the depletion of macrophages with an anti-CSF1R monoclonal antibody did not affect the antitumour efficacy induced by YB328 and anti-PD-1 treatment (Extended Data Fig. 8e,f). Moreover, YB328-stimulated macrophages were less effective in activating CD8+ Tâcells than YB328-stimulated DCs (Extended Data Fig. 8g). These data provide further support for the importance of CD103+CD11bâ cDCs in YB328-driven antitumour activity.
aâe, DCs were treated with P.âvulgatus, YB328 or vehicle. Comprehensive gene expression was examined (a), as was the expression of Batf3 (b, nâ=â6), IRF8 (c, nâ=â5), IRF4 (e, nâ=â3) and changes in the progeny of the CD103+CD11bâ cDCs (d, nâ=â3). f, IRF8 expression by CDPs treated with bacteria and Flt3L as indicated, nâ=â3â4. gâj, BMDCs were treated with the indicated stimuli, and expression of pS6K (g), pSTAT3 (h) and IRF8 (i) and changes in the progeny of CD103+CD11bâ cDCs (j) were examined (nâ=â4â6). k, Tumour growth curves of wild-type (WT) and Batf3â/â mice (nâ=â6â9) treated as indicated. l, Gene expression signatures of BMDCs stimulated with YB328 were extracted. Red text indicates TLR proteins that were highly expressed in the YB328-specific signature. TPM, transcripts per million. m, Changes in the progeny of CD103+CD11bâ cDCs (nâ=â3â4). n, Tumour growth curves of WT and Myd88â/â mice (nâ=â5â6) treated as indicated. o, Representative confocal images of the engulfment of YB328 by BMDCs. Scale bars, 5âμm. p,q, The expression of MHCI (p; gated on CD11c+MHCII+ cells) and the changes in the progeny of CD103+CD11bâ cDCs (q) (nâ=â6). Max, maximum. r, Frequency of PD-1 expression in CD8+ Tâcells (nâ=â4â6). s, Tumour growth curves of WT and Tlr7â/âTlr9â/â mice (nâ=â5â6) treated as indicated. t, Experimental scheme (top) and tumour growth curves (bottom). The number of mice showing a complete response (CR) in nâ=â4â6 mice is shown in parentheses. u, Changes in the progeny of CD103+CD11bâ cDCs (nâ=â7â9). v, Experimental scheme (top) and tumour growth curves (bottom). nâ=â4â6 mice. Each dot in the summary graphs indicates a well (bâj,m,pâr,u). The average tumour size of the groups on a certain day is shown as a dot (k,n,s,t,v). The data are presented as the meanâ±âs.d. Significance was assessed using one-way ANOVA with Bonferroniâs correction (bâj,pâr,u), two-way ANOVA with the TukeyâKramer method (k,n,s,v) or two-sided unpaired Studentâs t-test (m).
YB328 stimulates multiple TLR pathways
We next investigated how YB328 stimulates multiple signalling pathways in DCs. The expression of TLR family members, particularly TLR5 and TLR7âTLR9, was markedly upregulated in YB328-treated BMDCs compared with P.âvulgatus-treated or untreated BMDCs (Fig. 4l). Similarly, YB328 administration increased the diversity of TLR expression by DCs in the gut-associated lymphoid tissues (GALTs) of mice treated with NR-FMT (Extended Data Fig. 9a).
The adaptor protein MYD88 mediates the downstream signalling pathways of TLR5 and TLR7âTLR9 (ref. 43). Myd88â/â BMDCs treated with YB328 did not lead to increases in the number of CD103+CD11bâ cDC progeny (Fig. 4m). Moreover, the increase in antitumour efficacy induced by YB328 and anti-PD-1 treatment was abrogated in Myd88â/â mice (Fig. 4n and Extended Data Fig. 9b). The frequency of CD103+CD11bâ cDCs was significantly lower in the lamina propria and Peyerâs patch, but not in the spleen, of Myd88â/â mice than in wild-type mice (Extended Data Fig. 9c). This result suggests that YB328-triggered CD103+CD11bâ cDC generation requires the activation of MYD88 signalling.
As YB328 is not flagellated (Fig. 2a), the ability of YB328 to stimulate endosomal TLRs (TLR7âTLR9) to promote the differentiation of CD103+CD11bâ cDCs was examined. YB328 was phagocytosed by BMDCs (Fig. 4o and Supplementary Video 4). The stimulation of THP-1-based TLR7, TLR8 and TLR9 reporter cells revealed that YB328 activated both the IRF and NF-ĸB signalling pathways (Extended Data Fig. 9d). As mouse TLR8 is nonfunctional44, the roles of TLR7 and TLR9 in YB328-mediated antitumour immunity were investigated. BMDCs derived from Tlr7â/â or Tlr9â/â mice partially reduced the upregulation of MHCI expression (Fig. 4p) and abrogated the differentiation of CD103+CD11bâ cDCs induced by YB328 (Fig. 4q). Furthermore, BMDCs derived from Tlr7â/âTlr9â/â mice did not respond to YB328. Therefore increased MHCI expression, differentiation into CD103+CD11bâ cDCs and induction of the production of PD-1-expressing CD8+ Tâcells were not observed in these cells (Fig. 4pâr). YB328 administration to Tlr7â/âTlr9â/â mice did not augment the antitumour efficacy of anti-PD-1 treatment (Fig. 4s and Extended Data Fig. 9e). These data suggest that endosomal TLR7âTLR9 are key mediators of YB328-induced antitumour immunity.
Multiple TLR ligands sensitize DCs
YB328 induced TLR5 expression in DCs. However, YB328 probably does not have genes encoding components of flagellum, based on whole-genome sequence annotation, and flagella were not detected by electron microscopy (Fig. 2a). Therefore, we examined whether TLR ligands such as flagellin derived from the gut microbiome could further augment the antitumour immunity induced by YB328-stimulated DCs. PD-1 expression by CD8+ Tâcells stimulated with YB328-treated DCs, but not those stimulated with P.âvulgatus, was significantly increased after flagellin treatment (Extended Data Fig. 9f). Although treatment with YB328 and anti-PD-1 inhibited tumour growth, combined treatment with NR-FMT, which contained a pool of TLR ligands, led to enhanced antitumour efficacy (Fig. 4t and Extended Data Fig. 9g). Therefore, YB328 sensitizes DCs to multiple TLR ligands derived from not only YB328 but also from other gut microbes to augment antitumour immunity.
TLR signalling mimics YB328 stimulation
As YB328 upregulated multiple TLRs in DCs, we explored whether the combined activation of TLR signalling recapitulated the phenotype of YB328-treated DCs. BMDCs treated with the TLR agonist R848 or ODN-1826 exhibited increased S6K and STAT3 phosphorylation, and combination treatment further increased the phosphorylation of both these proteins. The triple combination of flagellin, R848 and ODN-1826 led to the most effective induction of S6K and STAT3 phosphorylation, which in turn resulted in the highest levels of IRF8 expression and the strongest induction of CD103+CD11bâ cDCs out of all the treatment combinations (Fig. 4u and Extended Data Fig. 9h). Moreover, combination treatment with an anti-PD-1 and a TLR agonist cocktailâat doses in which each agonist alone was insufficient to completely control tumour growth (Extended Data Fig. 9i)âsignificantly inhibited tumour growth (Fig. 4v and Extended Data Fig. 9j).
YB328 shows specific antitumour features
We next examined whether the immunomodulatory functions of YB328 are specific to this strain. Five representative bacterial strains were chosen for analysis: one strain isolated from the faeces of responders (Ruminococcus torques AE30) and four strains from species phylogenetically close to YB328 (the type strains of the species Ruminococcus albus, Clostridium leptum, Acutalibacter muris and Neglectibacter timonensis) (Extended Data Fig. 4a). Among these, YB328 treatment led to the most significant induction of PD-1 expression in CD8+ Tâcells (Extended Data Fig. 10a) and specifically augmented antitumour effects of anti-PD-1 compared with the other selected bacteria (Extended Data Fig. 10b). We then compared the whole-genome sequences of YB328, the five bacterial strains that lacked antitumour efficacy and P.âvulgatus (Extended Data Fig. 10câf, Supplementary Table 10 and Supplementary Discussion). Different cellular fractions of YB328 were also prepared to determine which components of YB328 were responsible for its antitumour activity (Extended Data Fig. 10g and Supplementary Discussion).
We also analysed whether the bacteria Akkermansia muciniphila1,15 and Bifidobacterium longum3,4, which reportedly improve responses to cancer immunotherapy, possessed properties similar to those of YB328. A.âmuciniphila, but not B.âlongum, exhibited a significant antitumour effect when combined with anti-PD-1 treatment (Extended Data Fig. 10h). A.âmuciniphila-treated BMDCs promoted CD103+CD11bâ cDC differentiation, which in turn induced PD-1 expression by CD8+ Tâcells (Extended Data Fig. 10i,j). However, A.âmuciniphila induced the expression of IRF8 but not S6K or STAT3 phosphorylation in DCs (Extended Data Fig. 10kâm). Together, these results demonstrate the uniqueness of YB328.
YB328 mobilizes cDCs to tumour lesions
We next investigated the location of CD103+CD11bâ cDC differentiation and CD8+ Tâcell activation. The abundance of CD103+CD11bâ cDCs, but not CD103âCD11b+ DCs, in the lamina propria and Peyerâs patch was significantly higher after YB328 treatment than after P.âvulgatus treatment (Fig. 5a,b). The frequencies of migratory CD103+CD11bâ cDCs were also significantly increased in the dLNs (inguinal LNs) and the TME of mice treated with YB328 and anti-PD-1 (Fig. 5c,d). Moreover, the frequency of CD103+CD11bâ cDCs was comparable to that observed after P.âvulgatus treatment in tumour nondraining LNs (NdLNs: inguinal LNs on the opposite flank of the tumour) and the spleen (Extended Data Fig. 11a,b). CD103+CD11bâ cDCs in the mesenteric lymph nodes (MLNs) of mice treated with YB328 exhibited significantly increased expression of CCR7, a crucial chemokine receptor for migration45,46 (Fig. 5e). PD-1+CD8+ Tâcells co-expressing TCF-1 significantly proliferated in the tumour dLNs, but not in the MLNs, of YB328-treated mice, and this phenotype was not observed in P.âvulgatus-treated mice (Fig. 5f and Extended Data Fig. 11c).
aâf, ATB-SPF mice were subcutaneously injected with MC38 cells and treated with the indicated bacteria. The phenotype and frequency (representative staining (left) and frequency (right)) of DC populations in lamina propria (gated on CD11c+MHCII+ cells) (a), Peyerâs patch (gated on CD11c+MHCII+ cells) (b), dLNs (gated on live CD45+ cells) (c) and tumour (gated on CD11c+MHCII+ cells) (d). Expression (representative staining (left) and MFI summary (right)) of CCR7 by CD103+CD11bâ cDCs in the MLNs (e, nâ=â3). Expression (representative staining (left) and MFI summary (right)) of Ki67 by PD-1+CD8+ Tâcells in dLNs and MLNs (f, nâ=â5; gated on PD-1+CD8+CD3+ cells). g, Representative image (left) and staining (right) of KikG-expressing or KikR-expressing intestinal cells (gated on CD45+ cells). h, A representative image of the localization of migrated intestinal cells after the indicated bacterial treatment. i, The frequency of KikR+CD103+CD11bâ cDCs in CD45+ cells in dLNs and NdLNs. j, KikR+CD103+ DCs (representative staining (left) and number summary (right)) in tumours (per mg) (gated on CD45+ cells). k, The KikRâ/KikR+ ratio of PD-1+CD8+ Tâcells (representative staining (left) and ratio summary (right)) in tumours. Cells were gated on PD-1+CD8+CD3+ cells. l, The TCR Vβ repertoire in CD8+ Tâcells in the indicated organs. For iâl, nâ=â3â6 mice per group. For aâl, each dot in the summary graphs indicates one mouse. The data are presented as the meanâ±âs.d. mâq, Assays in human samples. m, Representative multiplex immunohistochemical staining of tumours from patients with GC. Scale bars, 200âμm. n,o, Correlation between the YB328 abundance in the faeces of patients and the density of PD-1+CD8+ Tâcells (n) or CLEC9A+IRF8+ cells (o). nâ=â20 patients. p,q, Correlation between PD-1+CD8+ Tâcells and CLEC9A+IRF8+ cells (p) and correlation coefficients between PD-1+CD8+ Tâcells and other immune cells (q). nâ=â24 patients. For nâp, each dot indicates one patient, and the relative correlation was assessed by two-sided Pearsonâs correlation. Significance was assessed using two-sided unpaired Studentâs t-test (aâc,e,f,jâk), one-way ANOVA with Bonferroni correction (d,i) or ANOSIM with one-tailed significance using 999 permutations (l).
We next used mice expressing photoconvertible Kikume green-red (KikGR) to trace intestinal immune cells (Fig. 5g). In contrast to those in P.âvulgatus-treated mice, intestinal immune cells (KikR+ cells) showed substantial infiltration in the tumour dLNs of YB328-treated mice (Fig. 5h). The intestinal infiltration of KikR+CD103+CD11bâ cDCs into the dLNs and MLNs was higher in YB328-treated mice than in P.âvulgatus-treated mice but was not higher in NdLNs, spleens or cervical lymph nodes (Fig. 5i and Extended Data Fig. 11d). Moreover, treatment with YB328, but not P.âvulgatus, increased the abundance of KikR+CD103+ DCs and the number of KikRâPD-1+CD8+ Tâcells in the TME (Fig. 5j,k). Although the frequency of KikR+PD-1+CD8+ Tâcells was slightly increased in the MLNs of YB328-treated mice, the TCR Vβ repertoire of tumour-infiltrating CD8+ Tâcells was significantly different from that of CD8+ Tâcells in MLNs (Fig. 5l and Extended Data Fig. 11e), which indicated that there was local activation of tumour-antigen-specific CD8+ Tâcells. By contrast, the frequencies of KikR+PD-1+CD8+ Tâcells and KikR+CD11b+ cells were comparable in the dLNs, NdLNs, spleens and cervical lymph nodes after YB328 or P.âvulgatus administration. However, KikR+FOXP3+ Tâcells were slightly increased in the dLNs of YB328-treated mice (Extended Data Fig. 11eâg). Moreover, YB328 itself was not detected in the tumour (Extended Data Fig. 11h). These data support the notion that YB328 promotes the migration of intestinal CD103+CD11bâ cDCs into tumour dLNs and the TME and primes and activates CD8+ Tâcells in situ, which augments the antitumour efficacy of PD-1 blockade treatment.
YB328 augments human antitumour effects
YB328 abundance was significantly associated with the infiltration of PD-1+CD8+ Tâcells and IRF8+CLEC9A+ DCs, which correspond to mouse CD103+CD11bâ cDCs25, in the TME of patients treated with PD-1 blockade therapies (Fig. 5mâo and Extended Data Fig. 12a,b). The abundance of PD-1+CD8+ Tâcells was significantly correlated with that of IRF8+CLEC9A+ DCs but not with that of CD206+ or CD14+ cells (Fig. 5p,q and Extended Data Fig. 12c).
We confirmed these findings in our validation cohort (GC, n = 14; NSCLC, n = 7; collected from January 2018 to September 2018; see Methods) (Supplementary Tables 1â5). In accordance with our discovery cohort (GC, n = 35; NSCLC, n = 15; collected from March 2017 to December 2017; see Methods), the presence of YB328 in gut microbiota showed a clear correlation with a favourable clinical outcome, and patients with abundant YB328 exhibited a significantly longer PFS (Extended Data Fig. 12d,e). In a prospective cohort of patients with head and neck squamous cell carcinoma (HNSCC) (Supplementary Table 11), the YB328 phylotype was more abundant in the faeces of the responder group than in the non-responder group (Extended Data Fig. 12f). We also analysed datasets from the Japanese nationwide MONSTAR project (MONSTAR cohort)47 (Supplementary Table 12). Patients with malignant melanoma (MM) in the MONSTAR cohort had a higher abundance of Akkermansia in the responder group than in the non-responder group, as previously observed1. Moreover, YB328 was more abundant in the responder group of not only patients with MM and in patients with renal cell carcinoma, both of which are known to be immunotherapy-favourable cancer types, but also in patients with GC (thereby validating our results) and in patients with oesophageal cancer (Extended Data Fig. 12g). Furthermore, in a public dataset48 from patients with MM treated with anti-PD-1, responders exhibited significantly higher YB328 colonization than non-responders 3âmonths after FMT and anti-PD-1 treatment (Extended Data Fig. 12h). Thus, a high abundance of YB328 in faeces correlates with responsiveness to anti-PD-1 treatment in multiple types of cancer. Similarly, YB328 treatment also significantly increased the antitumour efficacy of the adoptive cell transfer of tumour antigen-specific CD8+ Tâcells (Extended Data Fig. 12i). Overall, patients with a high abundance of YB328 in gut microbiota have a substantial number of CD103+CD11bâ cDCs, which enable CD8+ Tâcells to respond to diverse tumour antigens and consequently improving the clinical efficacy of immunotherapies (Extended Data Fig. 12j).
Discussion
One of the difficulties in elucidating the detailed mechanisms by which gut microbiota induce antitumour immunity stems from the complex composition of the microbiota. Restoring and cultivating anaerobic gut microbiota from faecal preservation is technically challenging. Currently, most studies use commercially available bacterial strains, which may limit the clinical relevance of these studies. Here, we successfully isolated a previously undescribed bacterial strain, YB328, from the faeces of patients who responded to PD-1 blockade therapy and revealed the mechanism by which YB328 improves responsiveness to immunotherapy. On the basis of publicly available 16S rRNA gene sequence data, YB328 was estimated to be in the gastrointestinal tract of approximately one-fourth of Japanese individuals. As YB328 abundance ranges from 0.01% to 1%, which indicates that YB328 constitutes a relatively small fraction of microbiota, intervention with YB328 provides a large window to increase its abundance and to improve antitumour immunity (Extended Data Fig. 4b).
The occupation of immunodominant antigen-specific CD8+ Tâcells in the TME rapidly exhausts CD8+ Tâcells, which hampers the antitumour efficacy of ICB therapies in patients49. YB328 optimizes the range of tumour antigens that contribute to the antitumour efficacy of PD-1 blockade therapy. YB328-stimulated cDCs provided enhanced Tâcell stimulation signalling to reduce the threshold required for CD8+ Tâcell antigen recognition. Consequently, PD-1+CD8+ Tâcell clones with diverse TCRs against both immunodominant and subdominant tumour antigens were primed and activated, which resulted in a favourable PD-1 blockade treatment outcome (Extended Data Fig. 12j and Supplementary Discussion).
Our findings, together with previous reports2,50,51,52, suggest that gut microbiota diversity can serve as a pan-ethnic predictive biomarker for response to PD-1 blockade therapy across different cancer types. One can then envision a plausible explanation for the association between high diversity of the gut microbiota and clinical responses to ICB53. That is, bacteria that are beneficial for antitumour immunity, such as YB328, first induce CD103+CD11bâ cDC differentiation and activation and further sensitize these cDCs to TLR ligands. We propose a two-step activation system that involves YB328 for CD103+CD11bâ cDC progenies in the gut to activate CD8+ Tâcells in the TME, namely, primary direct activation and secondary activation induced by other bacteria (Extended Data Fig. 12j,k).
In summary, we demonstrated that a new bacterial strain, YB328, stimulates DCs in GALTs to augment antitumour immunity in the TME. We also presented the mechanism of spatiotemporal regulation of DCs by the microbiome from GALTs to the distant TME for the priming and activation of tumour antigen-specific CD8+ Tâcells.
Methods
Patients and samples
Patients with advanced NSCLC (nâ=â22) and patients with GC (nâ=â49) who received PD-1 blockade monotherapy (nivolumab or pembrolizumab) from March 2017 to September 2018 were enrolled in this study. Patients were separated into two independent cohorts in this study: the discovery cohort, in which patients with NSCLC or GC received PD-1 blockade monotherapy from March 2017 to December 2017 (nâ=â50; NSCLC, nâ=â15; GC, nâ=â35), and the validation cohort, in which patients with NSCLC or GC received PD-1 blockade monotherapy from January 2018 to September 2018 (nâ=â21; NSCLC, nâ=â7; GC, nâ=â14). Patients with HNSCC (nâ=â16) who received PD-1 blockade monotherapy (nivolumab or pembrolizumab) from June 2022 to October 2023 were also enrolled in this study. Patients with HNSCC who tested positive for human papillomavirus were excluded. In the MONSTAR cohort47, patients who received PD-1 blockade monotherapy (nivolumab or pembrolizumab) were enrolled from July 2019 to February 2022. Cancer types with nâ>â15 patients (advanced MM (nâ=â16), renal cell carcinoma (nâ=â44), GC (nâ=â32) and oesophageal cancer (nâ=â29)) were analysed in this study. Patients who were treated with antibiotics or microbiome intervention therapy within 1âmonth (the NSCLC and GC cohort and the HNSCC cohort) or 3âmonths (MONSTAR cohort) before the initiation of treatment were excluded from this study. Patients who achieved a complete or partial response or stable disease lasting >6âmonths were classified as responders and those who progressed on therapy or had stable disease <6âmonths were classified as non-responders per the Response Evaluation Criteria in Solid Tumors (RECIST; v.1.1). Forty-seven patients (discovery cohort: NSCLC, nâ=â4 and GC, nâ=â22; validation cohort: NSCLC, nâ=â7 and GC, nâ=â14) had an available quantity of TIL isolated; these samples were subjected to multiple immunological analyses. Stool samples were collected within 1âweek before initial treatment with a DNA stabilizer (FS-0009, TechnoSuruga Laboratory) according to the manufacturerâs instructions and then stored at â80â°C. Fresh tumour samples were obtained from primary or metastatic tumours by needle or endoscopic biopsy within 2âweeks before initial administration and subjected to immunological analyses. Clinical information of patients was obtained from their medical records.
PD-L1 immunohistochemistry
Anti-PD-L1 monoclonal antibody (22C3, Dako; SP142 or SP263, Roche) was used for immunohistochemistry (IHC) with an automatic staining instrument (BenchMark ULTRA, Roche) as previously described54. PD-L1 positivity was defined as staining in 1% or more of the tumour cells.
Evaluation of mismatch repair status
Anti-mutL homologueâ1 (MLH1; ES05) monoclonal antibody, anti-mutS homologueâ2 monoclonal antibody (MSH2; FE11), anti-postmeiotic segregation increasedâ2 monoclonal antibody (PMS2; EP51) and anti-mutS homologueâ6 monoclonal antibody (MSH6; EP49) (all from Dako) were used for IHC. Tumours were considered negative for MLH1, MSH2, PMS2 or MSH6 expression if there was a complete absence of nuclear staining in tumour cells. Normal epithelial cells and lymphocytes were used as internal controls. Tumours lacking MLH1, MSH2, PMS2 or MSH6 expression were considered deficient in mismatch repair, whereas tumours that expressed all these markers were considered proficient in mismatch repair.
Evaluation of EpsteinâBarr virus status
Chromogenic in situ hybridization for RNA encoding EpsteinâBarr virus was performed with fluorescein-labelled oligonucleotide probes (INFORM EBER probe) with enzymatic digestion (ISH proteaseâ3, Roche) and an iViewBlue detection kit (Roche) with the BenchMark ULTRA staining system.
Analysis of human TILs
Tumour tissues were minced and treated within 72âh after surgery according to a TIL preparation protocol using an optimized tissue preservation reagent (Tumor & Tissue Preservation Reagent (TTPR)) and a TIL isolation reagent (Tumor & Tissue Dissociation Reagent (TTDR)), which were codeveloped by BD Biosciences55. Alternatively, tumour tissues were treated immediately with a gentleMACS Dissociator (Miltenyi Biotec) as previously described56. These samples were analysed using a BD LSRFortessa X-20 (BD Biosciences) or a FACSymphony system (BD Biosciences).
Flow cytometry analysis
Cells were washed with PBS containing 2% FCS and stained with the indicated antibodies and fixable viability dye. After staining for cell surface markers, the cells were intracellularly stained with antibodies with a FOXP3 staining buffer set (Thermo Fisher Scientific) according to the manufacturerâs instructions. After washing, cells were analysed with a BD LSRFortessa X-20 (BD Biosciences) or a FACSymphony system (BD Biosciences). The data were analysed with FlowJo software (v.10.8.1; BD Biosciences). The staining antibodies were diluted according to the manufacturerâs instructions. To analyse the similarity of the CD8+ TCR Vβ repertoires, the expression of TCR Vβ was evaluated and presented as an ordination plot by uniform manifold approximation and projection. To analyse the diversity of TLR expression on DCs, the Shannon index of TLR expression was calculated based on the expression of TLR1âTLR9.
DNA extraction from faecal samples
Total DNA extraction was conducted using a QIAamp DNA Stool Mini kit (Qiagen) according to the manufacturerâs instructions. To increase the recovery of bacterial DNA, particularly from Gram-positive bacteria, pretreatment with lytic enzymes was performed before extraction using a stool kit. In brief, 100âmg faecal sample was suspended in 10âml Tris-EDTA buffer (pHâ7.5), and then 50âμl of 100âmgâmlâ1 lysozyme typeâVI purified from chicken egg white (MP Biomedicals) and 50âμl of 1âmgâmlâ1 purified achromopeptidase (Fujifilm Wako Pure Chemical) were added. The solution was incubated at 37â°C for 1âh with mixing. Next, 0.12âg SDS (final concentration of 1%) was added, and the suspension was mixed until it became clear. Next, 100âμl of 20âmgâmlâ1 proteinaseâK (Fujifilm Wako Pure Chemical) was added, followed by incubation at 55â°C for 1âh with mixing. The cell lysate was then subjected to ethanol precipitation. The precipitate was dissolved in 1.6âml ASL buffer from the stool kit and subsequently purified using a QIAamp DNA Stool Mini kit (Qiagen). For the HNSCC cohort, DNA was extracted using an ISOSPIN Fecal DNA kit (Nippon Gene) following protocolâN as previously described57.
16S rRNA gene sequencing
Each library was prepared according to the âIllumina 16S Metagenomic Sequencing Library Preparation Guideâ with a primer set (27Fmod: 5â²-AGR GTT TGA TCM TGG CTC AG-3â² and 338R: 5â²-TGC TGC CTC CCG TAG GAG T-3â²) targeting the V1âV2 region of the 16S rRNA gene. Next, 251-bp paired-end sequencing of the amplicon was performed using MiSeq (Illumina) with a MiSeq v.2 500 cycle kit. The raw sequences were demultiplexed and quality-trimmed with a FASTX-Toolkit (http://hannonlab.cshl.edu/fastx_toolkit/index.html, v.0.0.14) and BBtrim (http://bbmap.sourceforge.net, v.33.21). A total of 20,000 reads per sample were randomly selected using seqtk (v.1.3-r106) for further analyses. The processed sequences were clustered into operational taxonomic units (OTUs) defined at a 97% similarity cut-off using UCLUST (v.1.2.22q). Representative sequences for each OTU were then classified taxonomically by using RDP Classifier (v.2.2) with the Greengenes (v.13â8) database. The bioinformatics pipeline QIIME (v.1.9.1) was used as the informatics environment for all relevant processing of raw sequencing data and the determination of relative bacterial abundances. For the HNSCC cohort, sequencing libraries of the V4 hypervariable region of the 16S rRNA gene were prepared as previously described58 and sequenced on an Illumina MiSeq instrument with V2 chemistry (2âÃâ251âbp reads).
Bacterial diversity analysis
Alpha diversity was estimated using the Shannon index, which captures both microbiota richness and evenness, after subsampling to an even depth (10,000 reads per sample). Beta diversity was assessed by unweighted UniFrac distance matrices and visualized by PCoA. The PCoA of unweighted UniFrac distances was performed with QIIME, and the results were plotted using R (v.4.02). ANOSIM was used to evaluate the significant differences between groups.
LEfSe
KruskalâWallis and pairwise Wilcoxon tests were performed, followed by LDA to assess the effect size of each differentially abundant taxon59. Bacteria with markedly increased abundance were defined as those with a log10[LDA score]â>â3.
Estimation of YB328 phylotype abundance
Phylotype analysis, as delineated on the basis of the YB328 amplicon sequence variant (ASV) using 16S rRNA gene amplicon sequencing data, was performed for the following datasets: (1) prospective cohort of patients with HNSCC (V4 region, sequencing performed in this study); (2) MONSTAR cohort (V3V4 region, sequencing data obtained with permission from the MONSTAR alliance); and (3) two publicly available sequencing datasets48,60 (both V3V4 region). For ref. 60, data captured two healthy Japanese cohorts, namely the MORINAGA cohort (nâ=â704 for study accession DRP005906) and the NIBIOHN cohort (nâ=â1,280 for study accessions DRP007219, DRP007221, DRP007218, DRP007222 and DRP007220); the data were downloaded from the DDBJ Sequence Read Archive (DRA) in fastq format. For ref. 48, sequence data were obtained from the NCBI Sequence Read Archive (SRA) under BioProject PRJNA928744. In all cases, sequencing reads were denoised to obtain ASVs using DADA2 (v.1.26.0)61. For this purpose, primers in the reads were trimmed using Cutadapt (v.4.2)62, and read pairs without identifiable primers and undetermined bases (Ns) were discarded. Reads were then further trimmed by length (truncLen option) and filtered on the basis of their expected error (maxEE option) using the filterAndTrim function of DADA2. For the publicly available sequencing data, forward and reverse reads were denoised on a per-sample basis using the dada function in DADA2 with the options errâ=âNULL and selfConsistâ=âTRUE. For the data generated in this study (that is, the prospective cohort of patients with HNSCC) and the MONSTAR cohort data, error models for the forward and reverse reads were generated using the learnErrors function in DADA2 on a per-sequencing run basis. After denoising using the dada function with the run-specific error models, ASV tables were merged using mergeSequenceTables function in DADA2 and chimeras were removed using the removeBimeraDenovo function with methodâ=ââconsensusâ. For all datasets, a single ASV that perfectly matched the 16S rRNA gene sequence of YB328 was retained and used to calculate the relative abundance of YB328 (phylotype). To verify the specificity of detection of YB328, the V4 and V3V4 ASVs of YB328, as extracted from its whole-genome sequence, were compared against the Greengenes2 database (release 2024.09; Supplementary Data 1).
Metagenomic sequencing
The shotgun sequencing library for each microbial sample was constructed using Illumina DNA Prep (Illumina). The Illumina library was converted to a library for DNBSEQ using an MGIEasy Universal Library Conversion kit (App-A, MGI). Sequencing was performed on a DNBSEQ-G400RS (MGI) in 150-bp paired-end mode. All procedures were performed according to the manufacturerâs instructions.
Whole-genome sequencing of bacteria
The genomic DNA of YB328 and P.âvulgatus AE61 was extracted using an enzymatic lysis method with Qiagen Genomic-tip 100/G columns according to the manufacturerâs protocol. The complete genome sequence of strain YB328 and a high-quality draft genome of P.âvulgatus AE61 were determined using PacBio HiFi (high-fidelity) reads (Supplementary Table 7). PacBio sequencing, including library preparation and library quality control, were performed by Bioengineering Lab. In brief, short DNA fragments were removed using a Short Read Eliminator kit (PacBio) and retained DNA was sheared using a Covaris instrument to a size of 10â20âkbp. Sequencing libraries were then prepared using a PacBio SMRTbell Prep kit 3.0 and a SMRTbell gDNA Sample Amplification kit and quantified using a QuantiFluor dsDNA system (Promega) and an Agilent HS Genomic DNA 50âkb kit (Agilent Technologies) for quantification and size distribution, respectively. The library was then treated with a Revio Polymerase kit (PacBio) and sequenced on a Revio instrument. HiFi reads (Q20) were generated using SMRT Link (v.13.0.0.207600) to remove overhang adapter sequences and to create consensus sequences based on the resultant subreads. Lima (v.2.9.0) and pbmarkdup (v.1.0.3) were used for removing ultra-low PCR adapter sequences and PCR duplex reads, respectively. The resulting reads were assembled using Flye (v.2.9.5) and specifying the parameters --genome-size 3âm --asm-coverage 40. A phylogenetic tree showing the phylogenetic placement of YB328 and related taxa was constructed based on 120 single-copy marker genes using GTDB-Tk (v.2.4.0) with GTDB release 220 reference data. Protein sequences of identified marker genes (using the identify function of GTDBTk) were aligned using the align function of GTDBTk with the option skip_trimming. Aligned protein sequences were then loaded into an ARB (v.7.0) database, representative genomes were selected and the data were exported with custom masks for uninformative sites. Approximate maximum-likelihood phylogenetic trees were inferred using FastTree (v.2.1.1053), and the trees were visualized in ARB. The average nucleotide identity (ANI) values were then calculated using fastANI63 through comparisons of YB328 and closely related species. Genome gene identification was performed with Prodigal (v.2.6.3) with the default settings64. The predicted protein sequences were further analysed by matching against the Clusters of Orthologous Groups (COGs) database (release 3.10 in the Conserved Domain Database)65 using RPS-Blast (BLAST v.2.9.0+; Eâvalue cut-off of 0.01), the Kyoto Encyclopedia of Genes and Genomes (KEGG) database (release 2019-11-18)66 and the Virulence Factor Database (VFDB; release 2022.04)67 and by protein alignment using DIAMOND (v.2.0.13)68. Coding sequences unique to strain YB328 (see Extended Data Fig. 4a for genome accessions) were identified using CompareM (v.0.1.2; https://github.com/dparks1134/CompareM) with default settings.
Quantification of species abundances based on metagenome sequencing data
Taxonomic profiling using metagenome sequencing data was performed through read-level taxonomic assignment with Kraken2 (v.2.1.3)69, followed by estimation of relative abundances using Bracken (v.2.9)70. For this purpose, paired-end reads were processed using fastp (v.0.23.2 or v.0.24.0)71 with the following settings: --trim_tail1 1 --trim_tail2 1 --cut_right --cut_right_window_size 4 --cut_right_mean_quality 15 --qualified_quality_phred 15 --unqualified_percent_limit 40 --trim_poly_x --poly_x_min_len 10 --n_base_limit 0 --length_required 75. Reads were then filtered using NCBIâs BMTagger (v.3.101) to remove reads derived from the human genomic DNA. Taxonomic assignment of the reads was performed using Kraken2 against a customized version of the GTDB (v.220), with options --confidence 0.30 --use-names --paired. Species abundances were then estimated based on the Kraken2 output using Bracken, with options -r 150 -t 10.
To construct the custom GTDB-based database, we built Kraken2 and Bracken databases using all species-representative genomes in the GTDB (v.220; nâ=â113,104 total bacterial and archaeal genomes), after replacing the corresponding genomes of the species P.âvulgatus (representative: GCF_000012825.1) and H.âmulieris (representative: GCF_020687165.1) by the two genome sequences generated in this study. Furthermore, to reduce the potential of false-positive assignments, a representative human genome sequence (GRCh38) was also included in the database. Construction of the Kraken2 and Bracken databases largely followed the scheme implemented in Struo2 (ref. 72), with a default k-mer length of 35 and specifying a read length of 150 for the bracken-build command.
To investigate the abundance of YB328 on a global scale, we analysed 1,188 human faecal metagenomic datasets from NCBIâs SRA. In this study, we focused on samples from healthy individuals; these were identified on the basis of the metadata available in GMrepo2 (ref. 73) and HumGut74 by retaining entries marked as âD006262â and âHealthyâ for GMrepo2 and HumGut, respectively. Data were retrieved in FASTQ format using the SRA Toolkitâs prefetch and fasterq-dump commands, and then uniformly processed and analysed using fastp and Kraken2 as described above for the in-house generated sequencing data.
Bacterial identification and culture
All bacterial strains were isolated from fresh human faecal samples. The collected faecal samples were transported under 100% CO2 using a deoxygenating agent (Mitsubishi Gas Chemical) to a vinyl anaerobic chamber (COY) with an atmosphere composed of N2, H2 and CO2 (8:1:1). The faecal samples were diluted with pre-reduced dilution buffer to 1.0âÃâ10â8âgâmlâ1 in a tenfold dilution series. Fifty microlitres of 1.0âÃâ10â6âgâmlâ1, 1.0âÃâ10â7âgâmlâ1 and 1.0âÃâ10â8âgâmlâ1 diluted faecal fluid was inoculated on EggerthâGagnon (EG) agar plates75 supplemented with 5% (v/v) horse blood and M98-5 agar plates76. After incubation for 48âh at 37â°C, every colony grown on the plates was picked and subcultured on new plates for purification and growth. The obtained strains were preserved in pre-reduced nutrient broth (BD Biosciences) supplemented with 10% glycerol at â80â°C, and a portion of the colony was used for bacterial taxon identification. For bacterial taxon identification, the 16S rRNA gene sequence of the strains was amplified using the universal primers 27F (5â²-AGAGTTTGATCCTGGCTCAG-3â²) and 1492R (5â²-GGTTACCTTGTTACGACTT-3â²), and almost-full-length 16S rRNA genes were sequenced with the universal primers using a previously described procedure77. The species of the strains were determined by NCBI BLAST searches if the similarity between the query and the results exceeded 99%. Bacteroides vulgatus AE61 (renamed P.âvulgatus), Sellimonas intestinalis AE3, Clostridium colicanis AE66, Eggerthella lenta AE7, Ruminococcus torques AE30 and YB328 were isolated from EG agar plates. Erysipelatoclostridium ramosum AM28 was isolated using M98-5 agar plates. Ruminococcus albus JCM 14654T was obtained from the Japan Collection of Microorganisms (JCM) (RIKEN). Acutalibacter muris DSM 26090T, Neglectibacter timonensis DSM 102082T and Clostridium leptum DSM 753T were obtained from the German Collection of Microorganisms and Cell Cultures. A.âmuciniphila BAA-835T and B.âlongum BAA-999 were obtained from the American Type Culture Collection (ATCC). The culture method for the commercial bacteria was performed according to the manufacturerâs instructions. For inoculation of these bacterial strains into mice, the cells were collected using degassed GAM broth (Nissui) in an anaerobic chamber and were then suspended at a concentration of approximately 1.0âÃâ109 cells per ml in pre-reduced PBS containing 0.05% (w/v) l-cysteine·HCl·H2O or 15% (w/v) glycerol solution for storage. Until inoculation, the suspended bacterial strains were kept at â80â°C.
Cell culture
The mouse colon cancer cell line MC38 (ENH204, Kerafast) and the mammary carcinoma cell line EMT6 (CRL-2755, ATCC) were cultured in high-glucose DMEM (Fujifilm Wako Pure Chemical) supplemented with 10% FBS (Biosera) and 2âmM l-glutamine. Mouse B16F10 (CRL-6475, ATCC) and B16F10-OVA (stable transfectant derived from B16F10 cells, which were generated in house) melanoma cell lines were cultured in RPMI-1640 (Fujifilm Wako Pure Chemical) supplemented with 10% FBS (Biosera) and 2âmM l-glutamine. All cell lines are confirmed to be free of mycoplasma contamination. For the induction of BMDCs from bone marrow (BM), 4âÃâ106 BM cells were cultured in tissue-culture-treated 6-well plates in 4âml complete medium (RPMI-1640 (Fujifilm Wako Pure Chemical) supplemented with 2âmM l-glutamine and 10% FBS) containing 20ângâmlâ1 GM-CSF (Peprotech) as previously described78. Half of the medium was removed on dayâ2, and fresh complete medium containing 40ângâmlâ1 GM-CSF was added. The culture medium was entirely discarded on dayâ3 and replaced with complete medium containing 20ângâmlâ1 GM-CSF. For the bacterial stimulation experiments, the multiplicity of infection (m.o.i.) of bacteria used to infect BMDCs was 100 or indicated in each figure legend. For the TLR stimulation experiment, flagellin (25ângâmlâ1, InvivoGen), R848 (25ângâmlâ1, Selleck Chemicals) and/or ODN-1826 (250ângâmlâ1, InvivoGen) were administered for 24âh in complete medium containing 20ângâmlâ1 GM-CSF. CDPs were isolated from BM cells by cell sorting and cultured with FLT3L (100ângâmlâ1, Peprotech) and IMDM (Merck) supplemented with 2âmM GlutaMax (Merck), 55âμM 2-mercaptoethanol (Merck), 25âmM HEPES (Merck), 1âmM sodium pyruvate (Merck) and 10% FBS. CDPs were identified as LINâCD117intCD135+CD115+CD11câMHCIIâ BM cells, which lacked the markers of other cell lineages, including CD19, B220, CD3, NK1.1 and TER-119 (refs. 79,80,81). Bone marrow-derived macrophages were induced from BM cells as previously described82 and cultured in DMEM containing 10ângâmlâ1 M-CSF (Peprotech). THP-1-derived reporter cell lines expressing human TLRs (hTLR7, hTLR8 and hTLR9) were cultured according to the manufacturerâs instructions (InvivoGen). Cells were stimulated with YB328 for 24âh, and supernatants were collected for downstream analyses. For YB328 fractionation, YB328 was cultured with GAM broth (Nissui) at 37â°C for 16âh. Using the collected cells, the cell membrane fraction and cytoplasm fraction were prepared using the bead-beating method as previously described83,84. Using the culture supernatant, the membrane vesicle fraction and supernatant fraction (without membrane vesicles) were prepared by ultracentrifugation previously as described85,86. For the stimulation of fractions of YB328, each fraction was added to the culture medium at a concentration of 10% (v/v). The MC38, EMT6 and B16F10 cell lines were pre-authenticated by ATCC using short tandem repeat sequencing. OVA expression in B16F10-OVA cells was confirmed by flow cytometry. THP1-Dual⢠hTLR7, THP1-Dual⢠hTLR8 and THP1-Dual⢠hTLR9 cells were not authenticated but were functionally validated for responsiveness to their respective TLR agonists, serving as positive controls.
Animal models
C57BL/6 mice and BALB/cJ mice (6â10âweeks old) were purchased from CLEA Japan. OT-I TCR transgenic mice87 (provided by W.âR. Heath, University of Melbourne), Myd88â/â mice88, Tlr7â/â, Tlr9â/â and Tlr7â/âTlr9â/â mice (Oriental Bio Service, provided by S.âAkira, WPI Immunology Frontier Research Center, Osaka University), Batf3â/â mice89 (The Jackson Laboratory, provided by K.âMurphy, Washington University School of Medicine) and Kikume GreenâRed (KikGR) mice90 (RIKEN BioResource Research Center, provided by M.âTomura, Kyoto University Graduate School of Medicine) were used. GF mice were bred and maintained in the gnotobiotic facilities of the Central Institute for Experimental Animals or purchased from CLEA Japa or Sankyo Labo and maintained within the gnotobiotic facilities of the National Institute of Advanced Industrial Science and Technology. Other mice were housed in cages under SPF conditions, provided with standard food, given free access to hypochlorous weak-acid water and housed with a 12â12-h lightâdark cycle with lights on at 8:00. The temperature was maintained at 22â°C (20â26â°C), and the humidity was 45% (40â60%). In tumour challenge experiments, syngeneic mice were subcutaneously inoculated with 1âÃâ106 MC38 colon cancer cells, 1âÃâ106 EMT6 mammary carcinoma cells, 5âÃâ105 B16F10 melanoma cells or 5âÃâ105 B16F10-OVA melanoma cells in a total of 100âμl of PBS. The mice were randomized into different groups, ensuring that the tumour size was similar across all groups before the initiation of treatment. Anti-PD-1 monoclonal antibody (200âμg per mouse; clone: RMP1-14; BioLegend), anti-CSF1R monoclonal antibody (100âμg per mouse; clone: AFS98; Bio X Cell) or isotype control antibody (clone RTK2758; BioLegend) was intraperitoneally administered 3âtimes at 3-day intervals. Tumour length and width were measured on the indicated days, and tumour size was calculated as Vâ=â½ (tumour lengthâÃâ tumour width2). The immunological profile of the TME was analysed on daysâ13 or 15 after tumour inoculation. For TLR agonist injection, TLR agonists (0.5âμg flagellin, 5âμg R848 and/or 5âμg per ODN-1826 per mouse) were intraperitoneally administered. Mice were monitored twice a week and euthanized when the subcutaneous tumour diameter exceeded 20âmm. In some FMT and bacterial administration experiments, mice were preconditioned with antibiotics (ampicillin 500âmgâlâ1 (Merck), neomycin 500âmgâlâ1 (Merck), metronidazole 1âgâlâ1 (Merck) and vancomycin 500âmgâlâ1 (Merck)) dissolved in sterile drinking water. Mice received bacterial or faecal transplantation by oral gavage using feeding needles. For FMT, we were able to collect faecal samples sufficient for in vivo animal models after examining gut microbiota from a total of 6 patientsâ3 responders and 3 non-respondersâand used the faecal samples from these patients for each independent experiment. Faecal samples (0.1âg) were dissolved in 10âml PBS and administered to mice by oral gavage (100âμl per mouse). For the bacterial administration experiments, we prepared the bacterial stock at 108 cells per ml and administered it to mice by oral gavage (100âμl per mouse). Intestinal-cell-labelling experiments were performed as previously described90. In brief, the small intestine was drawn from the abdominal cavity and photoconverted by exposure to violet light (435ânm, 40â50âmWâcmâ2) for 90âs by laparotomy. Nontargeted regions were protected from light using aluminium foil. For adoptive transfer experiments, splenocytes were collected from OT-I TCR transgenic mice, and CD8+ Tâcells were purified with CD8a microbeads (Miltenyi Biotec). SPF mice pretreated with antibiotics were subcutaneously injected with B16F10-OVA cells, and the sorted CD8+ Tâcells (1âÃâ106 cells per mouse) were transferred by intravenous injection. No blinding was performed. Sample sizes for animal experiments were chosen according to preliminary pilot studies12,91,92, and are in line with standards in the field.
Isolation of mouse immune cells
To isolate TILs and lymphocytes from the lymph nodes, tissue samples were collected and minced into small pieces, followed by digestion with BD Horizon Dri TTDR reagent at room temperature for 20âmin. After dissociation, the cell suspension was filtered through a 70-μm strainer and washed with PBS supplemented with 1% BSA. For intestinal tissues, small intestines (duodenum, jejunum and ilium) were excised. The intestinal contents were removed by washing with HBSS. Fat tissue was also removed. Peyerâs patches and mesenteric tissues were carefully removed and digested in BD Horizon Dri TTDR according to the manufacturerâs instructions. The intestines were further opened longitudinally and cut into 5-mm long segments. Tissues were digested using a Lamina Propria Dissociation kit (Miltenyi Biotec) according to the manufacturerâs instructions.
ELISAs
BMDCs were stimulated with the indicated bacteria for 12âh, and chemokines and cytokines were measured using ELISA kits according to the manufacturerâs instructions (CXCL9, Abcam; CXCL10, Thermo Fisher Scientific; Proteome Profiler Mouse Chemokine Array kit, R&D Systems; and IL12p70, Thermo Fisher Scientific). For profiling chemokine expression with a Proteome Profiler Mouse Chemokine Array kit, the membrane was visualized using an LAS-4000 instrument (GE Healthcare), and the raw imaging data were processed and analysed with ImageJ software (Fiji, v.2.0.0).
Features of YB328
The cell structure of YB328 was observed by scanning electron microscopy (S-4500; Hitachi) and transmission electron microscopy (H-7600; Hitachi) as previously described93,94. In brief, for scanning electron microscopy observation, YB328 cells were fixed with 2% glutaraldehyde, post-fixed with 1% osmium tetroxide and dehydrated through a graded ethanol series followed by 3-methylbutyl acetate. For transmission electron microscopy observation, the cells were fixed with 2.5% glutaraldehyde and then post-fixed with 1% osmium tetroxide. The fixed cells were suspended in 1% aqueous uranyl acetate. The biochemical properties of YB328 were evaluated using an API ZYM system (bioMérieux) according to the manufacturerâs instructions.
Confocal microscopy
CD8+ Tâcells from OT-I TCR transgenic mice were prestained with 1âmgâmlâ1 Hoechst 33342 (DÅjindo Laboratories) at 37â°C for 10âmin, washed and then cultured with the indicated bacteria-stimulated DCs pulsed with OVA peptides. After 4âh of culture, the cells were collected. Bacteria were prestained with SYTO Green Fluorescent Nucleic Acid stains (Thermo Fisher Scientific) and cultured with BMDCs prestained with CellTracker Deep Red dye (Thermo Fisher Scientific). All staining procedures were performed according to the manufacturerâs instructions. After 15âmin of culture, the cells were collected. Cells were fixed with 4% formaldehyde solution for 30âmin, permeabilized with 0.5% Triton X-100 for 10âmin and blocked with PBS supplemented with 10% BSA for 1âh. The fixed cells were stained with an anti-NFATC1 (Thermo Fisher Scientific) monoclonal antibody overnight and then with a DyLight 488 secondary antibody (Thermo Fisher Scientific). All the antibodies were diluted in 10% BSA in PBS. Coverslips were mounted with ProLong Diamond Antifade mountant (Thermo Fisher Scientific). Images were captured by fluorescence microscopy (Carl Zeiss LSM-880, Zeiss) and analysed using ZEN software (Zeiss).
Time-lapse microscopy
The DCs and Tâcells were prestained with 0.5âμM CellTracker Green CMFDA dye, 0.5âμM CellTracker Red CMTPX dye, 0.5âμM CellTracker Deep Red dye (Thermo Fisher Scientific) or 1âmgâmlâ1 Hoechst 33342 at 37â°C for 10âmin. Bacteria were prestained with SYTO Green Fluorescent Nucleic Acid stains (Thermo Fisher Scientific) according to the manufacturerâs instructions. Prestained DCs (2âÃâ105) were mixed with prestained CD8+ Tâcells from OT-I TCR transgenic mice (1âÃâ105) or bacteria (m.o.i.â=â100) and seeded into a culture dish. The videos were recorded using an A1R MP+ multiphoton microscope system (Nikon) or a fluorescence microscope (Carl Zeiss LSM-880, Zeiss). The raw imaging data were processed and analysed with ImageJ software (Fiji, v.2.0.0).
IVIS spectrum in vivo imaging
Mice were depilated by hair removal cream 1âweek before observation using an IVIS spectrum imaging system (IVIS kinetics, PerkinElmer), and the iVid-neo diet (Oriental Bio) was applied 2âweeks before observation to reduce autofluorescence from the diet. After 4âdays of photoconversion, in vivo fluorescence images were obtained (for KikG, excitation filterâ=â465ânm, emission filterâ=âGFP; for KikR, excitation filterâ=â570ânm, emission filterâ=âCy5.5). Analysis was conducted using Living Image software (v.4.7.3, PerkinElmer).
RNA sequencing
Total RNA was extracted from BMDCs using a RNeasy Micro kit (Qiagen) and further processed with a SMART-Seq v4 Ultra Low Input RNA kit (Takara Bio). cDNA library preparation was conducted with a Nextera XT DNA Library Preparation kit (Illumina). The prepared RNA sequencing libraries were subjected to next-generation sequencing with a NovaSeq 6000 (Illumina) using 150-bp read lengths in paired-end mode. The reads generated for each RNA sample were analysed and compared using the Illumina DRAGEN Bio-IT Platform. Sequencing reads were aligned and annotated to the UCSC mouse reference genome (GRCm38.p6) from Gencode. The number of transcripts per kilobase million was calculated and used for downstream analyses. Expression levels across samples were normalized by zâscore transformation.
Quantitative PCR
To determine the gene expression of BMDCs, total RNA was extracted from BMDCs using a RNeasy Micro kit (Qiagen). cDNA synthesis was performed with SuperScript IV VILO master mix (Thermo Fisher Scientific). The TaqMan gene assay probe was purchased from Thermo Fisher Scientific (assay ID: Batf3/Mm01318273_m1; 18S rRNA/Hs03003631_g1). TaqMan Fast Advanced master mix (Thermo Fisher Scientific) was used to perform PCR, and the results were examined with a QuantStudio 7 Flex Real-Time PCR system (Thermo Fisher Scientific). The PCR conditions were as follows: 50â°C for 2âmin, 95â°C for 20âs, and 40 cycles of 95â°C for 3âs and 60â°C for 30âs. Gene expression was normalized to that of the endogenous control gene 18S rRNA. Relative gene expression between groups was determined using the comparative threshold cycle method. YB328 and P.âvulgatus were quantified in faeces and tumour tissues by real-time PCR with a primer set targeting the recA gene of YB328 (forward primer: 5â²-CCTCTTGGACCTTGCCGAAA-3â²; reverse primer: 5â²-ATACGCGTGCCGTTATACGA-3â²) and the 16S rRNA gene of P.âvulgatus (5â²-forward primer: CGGGCTTAAATTGCAGATGA; reverse primer: 5â²-CATGCAGCACCTTCACAGAT-3â²) as previously described95 using a QuantStudio 5 Real-Time PCR instrument. The reactions (20âµl) contained 1âÃâPower SYBR Green PCR master mix and 500ânM each of the forward and reverse primers, and the thermal cycling conditions were as follows: 95â°C for 1âmin, 40 cycles of 95â°C for 30âs, 55â°C for 30âs, and 72â°C for 15âs. A standard curve was prepared using purified genomic DNA of YB328 and P.âvulgatus.
Multiplex IHC staining
Formalin-fixed and paraffin-embedded blocks of tumour specimens were sliced into 4-μm-thick sections on adhesion microscope slides (Matsunami). The tissue slides were deparaffinized and rehydrated for multiplex IHC staining. Antigen retrieval and subsequent staining were performed using an Opal 7-Colour IHC kit (Akoya Biosciences) according to the manufacturerâs instructions.
Statistical analysis
Statistical analyses were performed with Prism software (v.8; GraphPad), SPSS (v.21.0; IBM) and R (v.4.02; R Foundation for Statistical Computing). Patient characteristics were compared between the two groups using Pearsonâs chi-square test and Fisherâs exact test. ROC curves for continuous variables were created by plotting the true-positive rate against the false-positive rate at each threshold. The AUC shown in each plot summarizes the performance of continuous variables. The cut-off values of continuous variables were determined as the maximum sum of the sensitivity and specificity in ROC analyses. Survival curves were estimated using the KaplanâMeier method and compared using the log-rank test. No statistical methods were used to predetermine sample size. Data were analysed for a normal distribution using the Shapiro-Wilk test before comparisons. The relationships between two groups were compared using a two-sided Studentâs t-test for normally distributed data or the nonparametric MannâWhitney U-test. For multiple testing, significance was determined by one-way ANOVA followed by Bonferroniâs correction. Tumour volume curves were compared using two-way ANOVA with the TukeyâKramer method. The correlation coefficient was evaluated by Pearsonâs correlation. Univariate or multivariate analyses were performed with the Cox regression model. All graphs of animal and in vitro experiments show representative data from at least two independent experiments. Pâ<â0.05 was considered to indicate significance. NS, not significant.
Inclusion and ethics
This study was conducted in Japan with the involvement of local researchers throughout the research process, from conceptualization to data analyses and manuscript preparation. All patients provided written informed consent before sampling, according to the Declaration of Helsinki. Analyses of cohort samples were conducted in a blinded manner and approved by the National Cancer Center Ethics Committee (IRB protocol numbers: GC and NSCLC cohort: 2015-048 and 2017-007; HNSCC cohort: 2022-346; MONSTAR cohort: 2018-367). The GC and NSCLC cohort and the MONSTAR cohort are registered in the UMIN Clinical Trials Registry (https://www.umin.ac.jp/ctr/, IDs: UMIN000019129 and UMIN000036749, respectively). The study was not designed to evaluate outcomes based on participant characteristics such as sex or ethnicity. Animal care and experiments were conducted according to the guidelines of the animal committee of the National Cancer Center/Institutional Animal Experiment Committee of National Institute of Advanced Industrial Science and Technology after approval by the Ethics Review Committee for Animal Experimentation of the National Cancer Center (protocol numbers: K24-007 and K24-010) and the Institutional Animal Experiment Committee of National Institute of Advanced Industrial Science and Technology (protocol number: 2022-0413).
Reporting summary
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.
Data availability
The whole genome sequences of YB328 and P.âvulgatus AE61 have been deposited in the NCBI BioProject under accession PRJDB17635. The 16S rRNA gene sequences of the bacterial strains isolated in this study have been deposited in DDBJ, EMBL and GenBank under the accession numbers LC719261 and LC744884âLC744889, corresponding to strain YB328, P.âvulgatus strain AE61, S.âintestinalis strain AE3, C.âcolicanis strain AE66, E.âlenta strain AE7, R.âtorques strain AE30 and E.âramosum strain AM28. The metagenome and 16S rRNA gene amplicon sequencing data generated in this study are available under BioProject accession PRJDB17628. The datasets used in this study, including the HNSCC cohort and the MONSTAR cohort, are not publicly available because of their association with unpublished studies. Access to these datasets is therefore restricted in accordance with institutional policies and data protection requirements. Researchers interested in accessing these data may submit a formal request to the corresponding author H.N. The datasets for the Healthy Japanese cohort (the data have been deposited in DNA Databank of Japan (DDBJ) under accession numbers DRP005906, DRP007219, DRP007221, DRP007218, DRP007222 and DRP007220), the MM cohort (BioProject PRJNA928744)48 and the faecal metagenomics analysis of global abundance of YB328 in healthy donors (BioProjects PRJEB6456, PRJEB21528, PRJNA422434, PRJNA397112, PRJDB4176, PRJNA517801, PRJNA529400, PRJEB27005, PRJNA529124, PRJEB8094, PRJEB17896, PRJNA328899, PRJEB7369, PRJEB6092, PRJNA268964, PRJNA447983, PRJEB1220, PRJEB1690, PRJNA289586, PRJNA421881, PRJEB17784, PRJEB5224, PRJNA504891, PRJEB9576, PRJEB6070, PRJNA438847, PRJEB27928, PRJEB12357 and PRJEB1786) were downloaded through the SRA. The RNA sequences of bacteria-stimulated BMDCs are available from the Gene Expression Omnibus under accession number GSE285376. The predicted protein sequences were analysed by matching against the COGs database (release 3.10 in the Conserved Domain Database, https://www.ncbi.nlm.nih.gov/research/cog-project/)65, the KEGG database (release 2019-11-18, https://www.kegg.jp/kegg/download/)66 and the VFDB database (release 2022.04, https://www.mgc.ac.cn/VFs/download.htm)67. Source data are provided with this paper.
Code availability
No custom code was developed in this study.
References
Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359, 91â97 (2018).
Gopalakrishnan, V. et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359, 97â103 (2018).
Matson, V. et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 359, 104â108 (2018).
Sivan, A. et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 350, 1084â1089 (2015).
Vetizou, M. et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 350, 1079â1084 (2015).
Lee, K. A. et al. Cross-cohort gut microbiome associations with immune checkpoint inhibitor response in advanced melanoma. Nat. Med. 28, 535â544 (2022).
Ribas, A. & Wolchok, J. D. Cancer immunotherapy using checkpoint blockade. Science 359, 1350â1355 (2018).
Robert, C. et al. Seven-year follow-up of the phase III KEYNOTE-006 study: pembrolizumab versus ipilimumab in advanced melanoma. J. Clin. Oncol. 41, 3998â4003 (2023).
Robert, C. et al. Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. Lancet Oncol. 20, 1239â1251 (2019).
Larkin, J. et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 381, 1535â1546 (2019).
Reck, M. et al. Five-year outcomes with pembrolizumab versus chemotherapy for metastatic non-small-cell lung cancer with PD-L1 tumor proportion score â§ 50. J. Clin. Oncol. 39, 2339â2349 (2021).
Kumagai, S. et al. The PD-1 expression balance between effector and regulatory T cells predicts the clinical efficacy of PD-1 blockade therapies. Nat. Immunol. 21, 1346â1358 (2020).
Oestreich, K. J., Yoon, H., Ahmed, R. & Boss, J. M. NFATc1 regulates PD-1 expression upon T cell activation. J. Immunol. 181, 4832â4839 (2008).
Agata, Y. et al. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int. Immunol. 8, 765â772 (1996).
Lam, K. C. et al. Microbiota triggers STING-type I IFN-dependent monocyte reprogramming of the tumor microenvironment. Cell 184, 5338â5356 (2021).
Fluckiger, A. et al. Cross-reactivity between tumor MHC class I-restricted antigens and an enterococcal bacteriophage. Science 369, 936â942 (2020).
Bessell, C. A. et al. Commensal bacteria stimulate antitumor responses via T cell cross-reactivity. JCI Insight 5, e135597 (2020).
Griffin, M. E. et al. Enterococcus peptidoglycan remodeling promotes checkpoint inhibitor cancer immunotherapy. Science 373, 1040â1046 (2021).
Hezaveh, K. et al. Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity 55, 324â340 (2022).
Singh, N. et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 40, 128â139 (2014).
Kim, O. Y. et al. Bacterial outer membrane vesicles suppress tumor by interferon-γ-mediated antitumor response. Nat. Commun. 8, 626 (2017).
Mager, L. F. et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science 369, 1481â1489 (2020).
Tanoue, T. et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature 565, 600â605 (2019).
Gutierrez-Martinez, E. et al. Cross-presentation of cell-associated antigens by MHC class I in dendritic cell subsets. Front. Immunol. 6, 363 (2015).
Cabeza-Cabrerizo, M., Cardoso, A., Minutti, C. M., da Costa, M. P. & Sousa, C. R. E. Dendritic cells revisited. Annu. Rev. Immunol. 39, 131â166 (2021).
Gros, A. et al. PD-1 identifies the patient-specific CD8+ tumor-reactive repertoire infiltrating human tumors. J. Clin. Invest. 124, 2246â2259 (2014).
Kansy, B. A. et al. PD-1 status in CD8+ T cells associates with survival and anti-PD-1 therapeutic outcomes in head and neck cancer. Cancer Res. 77, 6353â6364 (2017).
Thommen, D. S. et al. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat. Med. 24, 994â1004 (2018).
Wherry, E. J. & Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 15, 486â499 (2015).
Afrizal, A. et al. Anaerobic single-cell dispensing facilitates the cultivation of human gut bacteria. Environ. Microbiol. 24, 3861â3881 (2022).
Wexler, A. G. & Goodman, A. L. An insiderâs perspective: Bacteroides as a window into the microbiome. Nat. Microbiol. 2, 17026 (2017).
Zehn, D., Lee, S. Y. & Bevan, M. J. Complete but curtailed T-cell response to very low-affinity antigen. Nature 458, 211â214 (2009).
Spranger, S., Dai, D., Horton, B. & Gajewski, T. F. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 31, 711â723 (2017).
Grajales-Reyes, G. E. et al. Batf3 maintains autoactivation of Irf8 for commitment of a CD8α+ conventional DC clonogenic progenitor. Nat. Immunol. 16, 708â717 (2015).
Suzuki, S. et al. Critical roles of interferon regulatory factor 4 in CD11bhighCD8αâ dendritic cell development. Proc. Natl Acad. Sci. USA 101, 8981â8986 (2004).
Bogunovic, M. et al. Origin of the lamina propria dendritic cell network. Immunity 31, 513â525 (2009).
Murphy, T. L. et al. Transcriptional control of dendritic cell development. Annu. Rev. Immunol. 34, 93â119 (2016).
Sathaliyawala, T. et al. Mammalian target of rapamycin controls dendritic cell development downstream of Flt3 ligand signaling. Immunity 33, 597â606 (2010).
Laouar, Y., Welte, T., Fu, X. Y. & Flavell, R. A. STAT3 is required for Flt3L-dependent dendritic cell differentiation. Immunity 19, 903â912 (2003).
Vrzalikova, K. et al. S1PR1 drives a feedforward signalling loop to regulate BATF3 and the transcriptional programme of Hodgkin lymphoma cells. Leukemia 32, 214â223 (2018).
Lollies, A. et al. An oncogenic axis of STAT-mediated BATF3 upregulation causing MYC activity in classical Hodgkin lymphoma and anaplastic large cell lymphoma. Leukemia 32, 92â101 (2018).
Edelson, B. T. et al. Peripheral CD103+ dendritic cells form a unified subset developmentally related to CD8α+ conventional dendritic cells. J. Exp. Med. 207, 823â836 (2010).
Kawai, T. & Akira, S. Signaling to NF-κB by toll-like receptors. Trends Mol. Med. 13, 460â469 (2007).
Kugelberg, E. Innate immunity: making mice more human the TLR8 way. Nat. Rev. Immunol. 14, 6 (2014).
Jang, M. H. et al. CCR7 is critically important for migration of dendritic cells in intestinal lamina propria to mesenteric lymph nodes. J. Immunol. 176, 803â810 (2006).
Liu, J., Zhang, X., Cheng, Y. & Cao, X. Dendritic cell migration in inflammation and immunity. Cell. Mol. Immunol. 18, 2461â2471 (2021).
Hashimoto, T. et al. The SCRUM-MONSTAR cancer-omics ecosystem: striving for a quantum leap in precision medicine. Cancer Discov. 14, 2243â2261 (2024).
Routy, B. et al. Fecal microbiota transplantation plus anti-PD-1 immunotherapy in advanced melanoma: a phase I trial. Nat. Med. 29, 2121â2132 (2023).
Burger, M. L. et al. Antigen dominance hierarchies shape TCF1+ progenitor CD8 T cell phenotypes in tumors. Cell 184, 4996â5014 (2021).
Zitvogel, L., Ma, Y., Raoult, D., Kroemer, G. & Gajewski, T. F. The microbiome in cancer immunotherapy: diagnostic tools and therapeutic strategies. Science 359, 1366â1370 (2018).
Peters, B. A. et al. Relating the gut metagenome and metatranscriptome to immunotherapy responses in melanoma patients. Genome Med. 11, 61 (2019).
Jin, Y. et al. The diversity of gut microbiome is associated with favorable responses to anti-programmed death 1 immunotherapy in Chinese patients with NSCLC. J. Thorac. Oncol. 14, 1378â1389 (2019).
Matson, V., Chervin, C. S. & Gajewski, T. F. Cancer and the microbiome-influence of the commensal microbiota on cancer, immune responses, and immunotherapy. Gastroenterology 160, 600â613 (2021).
Tada, Y. et al. Targeting VEGFR2 with ramucirumab strongly impacts effector/activated regulatory T cells and CD8+ T cells in the tumor microenvironment. J. Immunother. Cancer 6, 106 (2018).
Kobayashi, T. et al. Isolation of tumor-infiltrating lymphocytes from preserved human tumor tissue specimens for downstream characterization. STAR Protoc. 3, 101557 (2022).
Saito, T. et al. Two FOXP3+CD4+ T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat. Med. 22, 679â684 (2016).
Tourlousse, D. M. et al. Validation and standardization of DNA extraction and library construction methods for metagenomics-based human fecal microbiome measurements. Microbiome 9, 95 (2021).
Tourlousse, D. M. et al. Characterization and demonstration of mock communities as control reagents for accurate human microbiome community measurements. Microbiol. Spectr. 10, e0191521 (2022).
Segata, N. et al. Metagenomic biomarker discovery and explanation. Genome Biol. 12, R60 (2011).
Park, J. et al. Comprehensive analysis of gut microbiota of a healthy population and covariates affecting microbial variation in two large Japanese cohorts. BMC Microbiol. 21, 151 (2021).
Callahan, B. J. et al. DADA2: high-resolution sample inference from Illumina amplicon data. Nat. Methods 13, 581â583 (2016).
Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 10 (2011).
Jain, C., Rodriguez, R. L., Phillippy, A. M., Konstantinidis, K. T. & Aluru, S. High throughput ANI analysis of 90 K prokaryotic genomes reveals clear species boundaries. Nat. Commun. 9, 5114 (2018).
Seemann, T. Prokka: rapid prokaryotic genome annotation. Bioinformatics 30, 2068â2069 (2014).
Galperin, M. Y. et al. COG database update: focus on microbial diversity, model organisms, and widespread pathogens. Nucleic Acids Res. 49, D274âD281 (2021).
Kanehisa, M., Furumichi, M., Sato, Y., Ishiguro-Watanabe, M. & Tanabe, M. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res. 49, D545âD551 (2021).
Liu, B., Zheng, D., Zhou, S., Chen, L. & Yang, J. VFDB 2022: a general classification scheme for bacterial virulence factors. Nucleic Acids Res. 50, D912âD917 (2022).
Buchfink, B., Reuter, K. & Drost, H. G. Sensitive protein alignments at tree-of-life scale using DIAMOND. Nat. Methods 18, 366â368 (2021).
Wood, D. E., Lu, J. & Langmead, B. Improved metagenomic analysis with Kraken 2. Genome Biol. 20, 257 (2019).
Lu, J., Breitwieser, F. P., Thielen, P. & Salzberg, S. L. Bracken: estimating species abundance in metagenomics data. PeerJ Comput. Sci. 3, e104 (2017).
Chen, S., Zhou, Y., Chen, Y. & Gu, J. fastp: an ultra-fast all-in-one FASTQ preprocessor. Bioinformatics 34, i884âi890 (2018).
Youngblut, N. D. & Ley, R. E. Struo2: efficient metagenome profiling database construction for ever-expanding microbial genome datasets. PeerJ 9, e12198 (2021).
Dai, D. et al. GMrepo v2: a curated human gut microbiome database with special focus on disease markers and cross-dataset comparison. Nucleic Acids Res. 50, D777âD784 (2022).
Hiseni, P., Rudi, K., Wilson, R. C., Hegge, F. T. & Snipen, L. HumGut: a comprehensive human gut prokaryotic genomes collection filtered by metagenome data. Microbiome 9, 165 (2021).
Tanaka, Y. & Benno, Y. Application of a single-colony coculture technique to the isolation of hitherto unculturable gut bacteria. Microbiol. Immunol. 59, 63â70 (2015).
Salanitro, J. P., Fairchilds, I. G. & Zgornicki, Y. D. Isolation, culture characteristics, and identification of anaerobic bacteria from the chicken cecum. Appl. Microbiol. 27, 678â687 (1974).
Shigeno, Y., Kitahara, M., Shime, M. & Benno, Y. Phascolarctobacterium wakonense sp. nov., isolated from common marmoset (Callithrix jacchus) faeces. Int. J. Syst. Evol. Microbiol. 69, 1941â1946 (2019).
Helft, J. et al. GMâCSF mouse bone marrow cultures comprise a heterogeneous population of CD11c+MHCII+ macrophages and dendritic cells. Immunity 42, 1197â1211 (2015).
Onai, N. et al. Identification of clonogenic common Flt3+M-CSFR+ plasmacytoid and conventional dendritic cell progenitors in mouse bone marrow. Nat. Immunol. 8, 1207â1216 (2007).
Liu, K. et al. In vivo analysis of dendritic cell development and homeostasis. Science 324, 392â397 (2009).
Kim, S. et al. High amount of transcription factor IRF8 engages AP1-IRF composite elements in enhancers to direct type 1 conventional dendritic cell identity. Immunity 54, 1622 (2021).
Toda, G., Yamauchi, T., Kadowaki, T. & Ueki, K. Preparation and culture of bone marrow-derived macrophages from mice for functional analysis. STAR Protoc. 2, 100246 (2021).
Barel, M. et al. A novel receptorâligand pathway for entry of Francisella tularensis in monocyte-like THP-1 cells: interaction between surface nucleolin and bacterial elongation factor Tu. BMC Microbiol. 8, 145 (2008).
Jalal, N. & Lee, S. F. The MsrAB reducing pathway of Streptococcus gordonii is needed for oxidative stress tolerance, biofilm formation, and oral colonization in mice. PLoS ONE 15, e0229375 (2020).
Morinaga, K., Yamamoto, T., Nomura, N. & Toyofuku, M. Paracoccus denitrificans can utilize various long-chain N-acyl homoserine lactones and sequester them in membrane vesicles. Environ. Microbiol. Rep. 10, 651â654 (2018).
Yasuda, M. et al. Phage genes induce quorum sensing signal release through membrane vesicle formation. Microbes Environ. 37, ME21067 (2022).
Hogquist, K. A. et al. T cell receptor antagonist peptides induce positive selection. Cell 76, 17â27 (1994).
Adachi, O. et al. Targeted disruption of the MyD88 gene results in loss of IL-1- and IL-18-mediated function. Immunity 9, 143â150 (1998).
Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8α+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097â1100 (2008).
Tomura, M. et al. Tracking and quantification of dendritic cell migration and antigen trafficking between the skin and lymph nodes. Sci. Rep. 4, 6030 (2014).
Kumagai, S. et al. Lactic acid promotes PD-1 expression in regulatory T cells in highly glycolytic tumor microenvironments. Cancer Cell 40, 201â218 (2022).
Itahashi, K. et al. BATF epigenetically and transcriptionally controls the activation program of regulatory T cells in human tumors. Sci. Immunol. 7, eabk0957 (2022).
Tamaki, H. et al. Armatimonas rosea gen. nov., sp. nov., of a novel bacterial phylum, Armatimonadetes phyl. nov., formally called the candidate phylum OP10. Int. J. Syst. Evol. Microbiol. 61, 1442â1447 (2011).
Katayama, T. et al. Isolation of a member of the candidate phylum âAtribacteriaâ reveals a unique cell membrane structure. Nat. Commun. 11, 6381 (2020).
Tong, J., Liu, C., Summanen, P., Xu, H. & Finegold, S. M. Application of quantitative real-time PCR for rapid identification of Bacteroides fragilis group and related organisms in human wound samples. Anaerobe 17, 64â68 (2011).
Acknowledgements
We thank members of the SCRUM-Japan MONSTAR-SCREEN alliance for providing the cohort study data; T.âKobayashi, T.âOwari, H.âNishinakamura, D.âSugiyama and M.âTomura for experimental guidance and discussion; and Y.âKa, T.âOgura, R.âTakahashi, Y.âTada, T.âTakaku, M.âNakai, K.âOnagawa, M.âTakemura, M.âHoshino, C.âOzawa, A.âTakakura, K.âYoshida, Y.âOsada, M.âOzawa, Y.âOhira, S.âYoshimatsu, the animal facility team at the National Cancer Center Japan, K.âMorinaga, M.âTakashino and R.âNorimine for their technical assistance. This study was supported by the Grants-in-Aid for Scientific Research (A grant no. 22H00455 to H.N., S grant no. 25H12345 and 17H06162 to H.N., and B grant no. 24K02306 to S.âKoyama) from the Ministry of Education, Culture, Sports, Science and Technology of Japan; the Japan Agency for Medical Research and Development (AMED)-CREST grant (JP18gm1010005 to H.N., H.M. and S.N.); the Moonshot Research and Development Program (grant no. 22zf0127009h0001 to H.N., H.M. and S.âKoyama); the Next Generation Drug Discovery and Development Technology on Regulating Intestinal Microbiome (no. 21ae0121037h0001 to H.N. and S.âKoyama); the Project for Cancer Research and Therapeutic Evolution (P-CREATE, no. 16cm0106301h0002 to H.N.); the program âThe next-generation drug discovery and development technology on regulating intestinal microbiomeâ (NeDDTrim) from Japan Agency for Medical Research and Development (AMED) (under the Grant Number JP 21ae0121037h0001 to H.N. and S.âKoyama); the Development of Technology for Patient Stratification Biomarker Discovery (grant no. 19ae0101074s0401 to H.N.); the Project for Promotion of Cancer Research and Therapeutic Evolution (P-PROMOTE, no. 22ama221301h0001 to H.N. and S.âKoyama, and no. 23ama221329h0001 to S.âKoyama and K.S.); the Practical Research for Innovative Cancer Control (no. 22ck0106724h0001 to H.N. and S.âKoyama, and no. 23ck0106796h001 to S.âKoyama and K.S.) from AMED; the National Cancer Center Research and Development Fund (no. 28-A-7 and no. 31-A-7 to H.N., no. 2022-S-7 to S.âKumagai, no. 2022-A-4 to K.W. and no. 2024-A-03 to H.N.); the Canon Foundation (S.âKoyama), the Mitsubishi Foundation (S.âKoyama); the Takeda Science Foundation (S.âKumagai); the Naito Foundation (S.âKumagai); the Kobayashi Foundation for Cancer Research (H.N., S.âKoyama and S.âKumagai); the Astellas Foundation for Research on Metabolic Disorders (S.âKoyama); and the Yasuda Foundation (H.N.).
Author information
Authors and Affiliations
Contributions
N.Y.-T.L., S.F., S.âKoyama, D.M., D.M.T., Y.âShigeno, T.I., Y.âMatsumoto, H.Y., K.M., H.T., Y.âSekiguchi, E.S., S.âKumagai, K.I., T.âTanegashima, K.F., S.I., M.S., T.âTsuji, H.W., K.W., Y.âMaeda, H.M., S.N. and H.N. performed the experiments and analysed the data. T.E., M.T., R.Y., T.F., M.N., A.K., K.G., T.D. and K.S. collected the clinical specimens and performed analyses of the clinical data. Y.âShigeno and Y.B. isolated YB328 and developed the culture system. N.Y.-T.L., S.F., S.âKoyama, Y.B. and H.N. conceived the project and wrote the paper.
Corresponding authors
Ethics declarations
Competing interests
S.âKoyama receives research funding from Otsuka Pharmaceutical and Chugai Pharmaceutical outside this study and receives honoraria from MSD and Chugai Pharmaceutical. K.I. receives honoraria from MSD and Chugai Pharmaceutical. M.N. receives honoraria (lecture fees) from MSD, Ono Pharmaceutical and Bristol-Myers Squibb (BMS). A.K. receives personal fees from Daiichi-Sankyo, Lilly, Ono Pharmaceutical, Taiho Pharmaceutical, BMS, Merck Pharmaceutical, Sumitomo Dainippon Pharma and AstraZeneca outside this study. K.W. serves as a board member and a founder of ARC Therapies outside of this study. T.D. receives personal fees for advisory roles from Sumitomo Dainippon Pharma, Taiho Pharmaceutical, Takeda Pharmaceutical, Chugai Pharmaceutical, AbbVie, Bayer, Rakuten Medical, Otsuka Pharmaceutical, KAKEN Pharmaceutical, Kyowa Kirin, SHIONOGI, PRA Health Science, A2 Health Care, Noile-Immune Biotech, MSD, Daiichi-Sankyo, Amgen, Novartis, Boehringer Ingelheim, Janssen Pharmaceutical and Astellas Pharmaceutical; receives honoraria (lecture fees) from BMS, Rakuten Medical, Ono Pharmaceutical, Daiichi-Sankyo and AstraZeneca; and receives research funding from Lilly, MSD, Daiichi-Sankyo, Sumitomo Dainippon Pharma, Taiho Pharmaceutical, Novartis, Merck Pharmaceutical, Janssen Pharmaceutical, Boehringer Ingelheim, Pfizer, BMS, AbbVie, Eisai, IQVIA, Chugai Pharmaceutical and SHIONOGI outside this study. K.S. reports receiving personal fees for consulting and advisory roles from BMS, Takeda, Ono Pharmaceutical, Novartis, Daiichi Sankyo, Amgen, Boehringer Ingelheim, Merck Pharmaceutical, Astellas, Guardant Health Japan, Janssen, AstraZeneca, Zymeworks Biopharmaceuticals, ALX Oncology and Bayer; receiving honoraria from BMS, Ono Pharmaceutical, Janssen, Eli Lilly, Astellas and AstraZeneca; and receiving research funding (all to the institution) from Astellas, Ono Pharmaceutical, Daiichi Sankyo, Taiho Pharmaceutical, Chugai, Merck Pharmaceutical, Amgen, Eisai, PRA Health Sciences and Syneos Health, outside the submitted work. H.M. receives research grants from Ono Pharmaceutical, Daiichi Sankyo, PFDeNA, Konica-Minolta and Ambry Genetics and serves as a board member of CureGene outside this study. H.N. receives research funding and honoraria (lecture fees) from Ono Pharmaceutical, BMS, Chugai Pharmaceutical, BD Japan and MSD; receives honoraria (lecture fees) from Amgen; receives research funding from Taiho Pharmaceutical, Daiichi-Sankyo, Kyowa Kirin, Zenyaku Kogyo, Oncolys BioPharma, Debiopharma, Asahi-Kasei, Sysmex, Fujifilm, SRL, Astellas Pharmaceutical, Sumitomo Dainippon Pharma, ARC Therapies and RIKAKEN Holdings; and serves as a scientific advisor and a founder of ARC Therapies and a scientific advisor of LTZ Therapeutics outside this study. S.F., Y.B. and H.N. are the primary inventors on pending patents 2020-165470 belonging to RIKEN and the National Cancer Center Japan. The other authors declare no competing interests.
Peer review
Peer review information
Nature thanks Ben Boursi and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.
Additional information
Publisherâs note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data figures and tables
Extended Data Fig. 1 The microbiota significantly impacts the clinical outcome of PD-1 blockade therapy.
a-i, Stacked bar plot of the bacterial abundance aggregated at the family-level. Only families with an abundance of >0.1% are shown. (a, nâ=â50 patients). Shannon diversity of the faecal microbiota in responder (Resp) and non-responder (N-Resp) patients (b, nâ=â15 patients; f, nâ=â35 patients). High/low Shannon diversity of the gut microbiota in patients was stratified by ROC analysis (left) KaplanâMeier curves (right) for PFS of patients with high/low Shannon diversity of the gut microbiota (c, g). In NSCLC patients, the median PFS was 266 days (95% CI, 0 to 696) for those with high diversity, and 23 days (95% CI, 20 to 25) for those with low diversity. In patients with GC, the median PFS was 92 days (95% CI, 17 to 166) for those with high diversity, and 31 days (95% CI, 15 to 47) for those with low diversity (c, g). PCoA of the composition of the faecal microbiota based on unweighted Unifrac distances (d, h). Bar plots of the LDA scores of differentially abundant taxa in the Resp and N-Resp groups as determined by LEfSe analysis (e, i). j-l, Abundances of selected bacterial taxa. (j: family enriched in Resp/N-Resp represented in Fig. 1f; l: bacterial genera previously associated with response to immunotherapy). The ROC analysis (left) and KaplanâMeier curves for PFS (right) for the enriched bacterial families in Fig. 1f were examined. High/low abundances of the indicated bacterial families in patients were stratified by ROC analysis (k). nâ=â50 patients. m, KaplanâMeier curves for the PFS of patients stratified by the median cut-off for each indicated factor. nâ=â50 patients. n, The correlations between bacteria that were significantly correlated with the clinical course and PD-1 expression by CD8+ T cells are shown. nâ=â26 patients. (c, g, k, m) KaplanâMeier curves were analysed by the two-sided log-rank test. (b, f) Two-sided unpaired t test, and data are presented as the median. (j, l) Two-sided MannâWhitney U test, and data are presented as the median. (d, h) ANOSIM, with one-tailed significance computed by permutation (nâ=â999 permutations). (n) The correlation was evaluated by two-sided Pearsonâs correlation analysis.
Extended Data Fig. 2 FMT from patients influences the diversity and composition of the gut microbiota.
a-g, GF mice transplanted with faeces from Resps or N-Resps were subcutaneously injected with MC38 tumour cells. Experimental scheme (a). Tumour growth curves (b, left: summary, right: each mouse). nâ=â4â8 mice per group. c-g, The faecal samples were subjected to 16S rRNA gene sequencing to analyse the bacterial composition. The alpha diversity of the gut microbiota was scored by the Shannon index (c). Stacked bar plot of the family-level taxonomic composition of the faeces of Resp-FMT/N-Resp FMT mice. Only families with an abundance of >0.1% are shown (d). Differentially abundant taxa were analysed by LEfSe, the symbol sizes are scaled by relative abundance (e). The bar plot showed the LDA scores of differentially abundant taxa between the Resp and N-Resp groups (f). The relative abundance of Ruminococcaceae (left) and Bacteroidaceae (right)(g). nâ=â14 mice per group. (b, left), The average tumour sizes of the groups on a certain day of the experiment are shown as dots and are presented as the meanâ±âSD, two-way ANOVA with the TukeyâKramer method. Each dot indicates one mouse, and the data are presented as the median (c, g). (c, g) Two-sided unpaired t test.
Extended Data Fig. 3 Faecal transplantation from patients who responded to PD-1 blockade therapy inhibits tumour progression and augments antitumour immunity.
a-l, ATB-SPF mice were transplanted with faeces from Resp or N-Resp mice following the subcutaneous injection of tumour cells. Experimental scheme (a). Tumour growth curves of MC38 (b, left: summary, right: each mouse) and EMT6 (c, left: summary, right: each mouse) tumour models. nâ=â5â6 mice per group. d-f, The frequency [representative staining (left) and % summary (right)] of effector molecule expression [PD-1 (d, nâ=â10) and CD62L/CD44 (e, nâ=â12)] and cytokine (IFN-γ/TNFα) production (f, nâ=â12) by CD8+ T cells. g-i, TCR Vβ repertoire of PD-1+CD8+ T cells (g, h). Numbers of skewed TCR Vβ clones (percentages of clones >10%) of PD-1+CD8+ T cells (i). nâ=â12 mice per group. j-l, The expression [representative staining (left) and MFI summary (right)] of maturation markers [CD86 (j), CD40 (k) and CD80 (l)] by DCs. nâ=â6 mice per group. (b, left; c, left), The average tumour sizes of the groups on a certain day of the experiment are shown as dots and are presented as the meanâ±âSD. (d-l) Each dot indicates one mouse, and the data are presented as the mean (i) or meanâ±âSD (d-f, h, j-l). (b, c) Two-way ANOVA with the TukeyâKramer method. (d-f, j-l), One-way ANOVA with Bonferroni correction. (i) Two-sided MannâWhitney U test.
Extended Data Fig. 4 A bacterium identified from the faeces of Resps is classified as the novel strain YB328.
a, Genome-based phylogenetic tree showing the position of strain YB328 in the family âAcutalibacteriaceaeâ in the GTDB v220. Species names, strain identifiers and genome sequence accession numbers are presented in the tip labels, if applicable. Scale bar represents 10% amino-acid substitutions per site. b, Abundance of the YB328-related bacteria in: (upper panel) healthy Japanese individuals based on publicly available 16S rRNA gene sequencing data of two cohorts (MORINAGA cohort: nâ=â704 patients; NIBIOHN cohort: nâ=â1280 patients), and (lower panel) individuals from 21 countries across the world based on shotgun metagenomics data. For the upper panel, each symbol represents a different dataset. For the lower panel, abundances were calculated as the percentage of metagenomic reads assigned to the YB328 reference genome in our custom GTDB-based database. Text labels on the y-axis indicate the country of origin, and in parentheses the BioProject and the number of datasets included in the analysis. For the box plots, the thick line within each box shows the median, the boxes represent the interquartile range (IQR), and the whiskers extend to the furthest data point within 1.5 times the IQR of the box. c, Faecal samples from patients, excluding MSI-H patients (NSCLC: nâ=â15; GC: nâ=â32), were subjected to shotgun metagenomic sequencing. Analyses of the relative abundances of the YB328 using Kraken2+Bracken with a custom version of the GTDB (left panel). High/low bacterial abundance was stratified by a ROC curve analysis (middle panel). KaplanâMeier curves (right panel) of the PFS of patients with high/low faecal bacterial abundance are shown. d, High/low bacterial abundance of all GC/NSCLC patients (NSCLC: nâ=â22; GC: nâ=â49), stratified by the median cut-off value, was used to plot KaplanâMeier curves of the PFS of patients. e, Experimental scheme (left) and tumour growth curves (middle: summary, right: each mouse). Average tumour sizes of the groups on a certain day of the experiment are shown as dots and are presented as the meanâ±âSD. nâ=â4 mice per group. f, YB328 colonisation detected in faecal sample by qPCR, nâ=â3 mice per group. g, The tumour growth curves are shown. Each line represents one mouse. nâ=â5â6 mice per group. (c, left panel) Two-sided MannâWhitney U test and data are presented as the median. (c, right panel; d) Two-sided log-rank test.
Extended Data Fig. 5 YB328 administration changes the immunological landscape of the TME.
a-g, Experimental scheme (a). The frequency [representative staining (left) and % summary (right)] of effector molecule [perforin (b) and granzyme B (c)] production by CD8+ T cells. The frequency [representative staining (left) and % summary (right)] of Foxp3+ cells in CD4+ T cells (d) and PD-1 expression by Foxp3+CD4+ T cells (e). Expression [representative staining (left) and MFI summary (right)] of the maturation marker CD80 (f) by CD11c+MHCII+ DCs. nâ=â5â12 mice per group. g, Shannon diversity of microbiota after the administration of the indicated treatment. nâ=â4â6 mice per group. h-k, Experimental scheme (h). Tumour growth curves (i, nâ=â5â7 mice per group). Profile of the TCR Vβ repertoire in PD-1+CD8+ T cells (j, nâ=â5 mice per group). Changes in the Shannon index of microbiota diversity (k, nâ=â11â12 mice per group). l, The correlation between the abundance of YB328 and the Shannon index of microbiota diversity in all GC/NSCLC patients (NSCLC: nâ=â22; GC: nâ=â49) in this study. m, The experimental scheme is shown (left panel). Tumour growth curves (middle panel: summary; right panel: each mouse). nâ=â3â5 mice per group. n-o, ATB-SPF or N-Resp-FMT recipient mice received the indicated bacterial treatment. Bacterial engraftment was assessed. nâ=â3â4 mice per group. (b-g, j, k, n-o) Each dot in the summary graphs indicates one mouse in the in vivo experiment. (b-g, j, o) The data are presented as the meansâ±âSD. (m, middle) The average tumour size of the groups on a certain day is shown as a dot. (i, m left panel) Each line indicates one mouse for the in vivo experiment. (l) Each dot indicates one patient. (b-g) One-way ANOVA with Bonferroniâs correction. (k) Two-sided paired t test. (l) The correlation coefficient was evaluated by two-sided Pearsonâs correlation analysis. (m) Two-way ANOVA with the TukeyâKramer method. (o) Two-sided unpaired t test.
Extended Data Fig. 6 YB328-stimulated BMDCs differentiate into mature CD103+CD11bâ cDCs and produce CXCL10 to attract CD8+ T cells.
a, BMDCs were stimulated with P. vulgatus or YB328. The expression [representative staining (left) and MFI summary (right)] of the maturation marker CD80. nâ=â6 wells per group. b, The complexity and size of DCs (nâ=â30 in each group from Supplementary Video 1) stimulated with YB328 or P. vulgatus were evaluated. A representative correlation model for DCs is shown in the binary image. Scale bar: 10 μm. c, Chemokine production by YB328-, P. vulgatus- or vehicle-treated BMDCs was examined with a proteome profiler chemokine antibody array. Representative data (left) and quantified as fold changes (right) are shown. nâ=â2 technical replicates. (a) One-way ANOVA with Bonferroni correction. Each dot in the summary graphs indicates one well, and the data are presented as the meanâ±âSD. (b) Each dot indicates one cell. (c) Data are presented as the mean.
Extended Data Fig. 7 YB328-stimulated BMDCs deliver robust TCR signals to activate CD8+ T cells.
a-h, OT-I CD8+ T cells were cocultured with bacteria-treated BMDCs pulsed with the indicated antigen peptides (a-d, N4 peptide; e-h, Q4H7 peptide). (a, e), Representative data showing the expression of TCR signalling molecules (p-ZAP70, p-JNK, p-Erk1/2, p-Akt and p-S6K) by CD8+ T cells after stimulation with DCs pulsed with the indicated concentrations of peptides. (b, f), Representative data of PD-1 expression by CD8+ T cells after stimulation with DCs pulsed with the indicated concentrations of peptides. (c, g), Representative data of CD62L/CD44 expression by CD8+ T cells after stimulation with DCs pulsed with the indicated concentrations of peptides. (d, h), Cytokine (IFN-γ and TNFα) production [representative staining (left) and % summary (right)] by CD8+ T cells after stimulation with DCs pulsed with the indicated concentrations of peptides. nâ=â3â4 wells per group. The data were analysed by a two-sided unpaired t test and presented as the meanâ±âSD.
Extended Data Fig. 8 YB328 treatment induces CD103+CD11bâ cDC generation to augment antitumour immunity of anti-PD-1 mAb.
a, A representative gating strategy for evaluating the progeny of CD103+CD11bâ cDCs in BMDCs stimulated with YB328, P. vulgatus, or vehicle, corresponding to Fig. 4d. b-c, Representative staining (b) of the phenotype of CD103+CD11bâ cDCs (% summary: c, left panel) or CD103+ DCs (% summary: c, right panel) in the indicated organs of WT or Batf3â/â MC38 tumour-bearing mice (nâ=â3). d, Tumour growth curves (nâ=â6â9). e-f, ATB-SPF mice were subcutaneously injected with MC38 cells and administered an anti-PD-1 mAb, anti-CSF1R mAb or control Ab with/without YB328 treatment. Changes in the immunological landscape are shown (e, nâ=â3). The experimental scheme (f, upper panel) and tumour growth curves (f, lower-left: summary, lower-right: each mouse) (f, nâ=â4â7). g, OT-I CD8+ T cells were cocultured with bacteria-treated BMDCs (DC) or BMDMs (MF) pulsed with antigen peptides. The frequency of PD-1 expression [representative staining (left) and % summary (right)] by OT-I CD8+ T cells is shown (nâ=â5 biologically replicates). (d; f, lower-right) Each line indicates one mouse for the in vivo experiment. The average tumour size of the groups on a certain day is shown as a dot and are presented as the meanâ±âSD (f, lower-left). Each dot in the summary graphs indicates one mouse in the in vivo experiment, and the data are presented as the meanâ±âSD (c, e). Each dot in the summary graphs indicates a well, and the data are presented as the meanâ±âSD (g). (c, e), Two-sided unpaired t test. (f) Two-way ANOVA with the TukeyâKramer method. (g) One-way ANOVA with Bonferroniâs correction.
Extended Data Fig. 9 YB328 treatment induces CD103+CD11bâ cDC generation and sensitises DCs to multiple TLR ligands via MyD88 signalling.
a, The expression of TLRs (TLR1âTLR9) by GALT-isolated DCs was examined (upper panel, experimental scheme). The diversity of the expression of TLRs by DCs was calculated by the Shannon index (lower panel), nâ=â3â4. IEL: Intraepithelial lymphocyte. b, Tumour growth curves of each mouse in Fig. 4n are shown, nâ=â5â6. c, The frequency of CD103+CD11bâ cDCs [representative staining (left panel) and % summary (right panel)] in the indicated organs of WT or MyD88â/â mice (nâ=â3). d, Assessment of IRF signalling (upper panel) and NF-ĸB activity (lower panel) in TLR reporter THP-1 cells treated with/without YB328 (nâ=â3â4). e, Tumour growth curves of each mouse in Fig. 4s are shown. (nâ=â5â6). f, BMDCs were stimulated with YB328 or P. vulgatus at an MOI of 25 and then treated with flagellin. OT-I CD8+ T cells were cocultured with the stimulated BMDCs pulsed with 1ânM N4 peptides. PD-1 expression by CD8+ T cells [representative staining (left panel) and % summary (right panel)] is shown, nâ=â5. g. Tumour growth curves of each mouse in Fig. 4t. are shown. nâ=â4â6 mice per group. h, BMDCs were treated with the indicated TLR ligands. The levels [representative staining (left panel) and MFI summary (right panel)] of p-S6K, p-STAT3 and IRF8 are shown (nâ=â4). i, Tumour growth curves (nâ=â3â4). j, Tumour growth curves of each mouse in Fig. 4v. is shown. nâ=â4â6. (b, e, g, i-j) Each line indicates one mouse for the in vivo experiment. (a, c) Each dot in the summary graphs indicates one mouse in the in vivo experiment, and the data are presented as the meanâ±âSD. (d, f, h) Each dot in the summary graphs indicates a well, and the data are presented as the meanâ±âSD. (a, c-d), Two-sided unpaired t test. (f, h) One-way ANOVA with Bonferroniâs correction.
Extended Data Fig. 10 Specific genomic traits and phenotypic features of YB328 are involved in antitumour effects and their responsible factors.
a, BMDCs were stimulated with bacterial strains that were isolated from Resps or were genomically similar to YB328. OT-I CD8+ T cells were cocultured with bacteria-treated BMDCs pulsed with N4 peptides (10ânM). The frequency [representative staining (left) and % summary (right)] of PD-1 expression by CD8+ T cells. nâ=â3 wells per group. b, Tumour growth curves (left: summary, right: each mouse). nâ=â3â4 mice per group. c-f, Comparative genomics of YB238 and other six strains (referred to as âReference strainsâ) Venn diagram indicating the number of genes unique to YB328 (c). Bar plots of the distribution of genes annotated to different categories according to COG (d), KEGG (e), and VFDB (f). Values were calculated for all genes of YB328 (red bars), genes unique to YB328 (green bars), and the other six strains (blue bars show the meanâ±âSD). g, BMDCs were treated with each of four cell culture fractions of YB328 [G1: cytoplasm, G2: cell membrane, G3: culture supernatant (without membrane vesicle) or G4: membrane vesicle] and cocultured with OT-I CD8+ T cells stimulated with N4 peptide-pulsed DCs. The expression of the indicated proteins was measured by flow cytometry (nâ=â3) and normalized between samples. cDC1: CD103+CD11bâ cDCs. h, Tumour growth curves (upper: summary, lower: each mouse). nâ=â4â6 mice per group. i-m, BMDCs were stimulated with the indicated bacterial strains. OT-I CD8+ T cells were cocultured with bacteria-treated BMDCs pulsed with N4 peptides (10ânM). Changes in the progeny of the CD103+CD11bâ population (i). The frequency [% summary (left) and representative staining (right)] of PD-1 expression by CD8+ T cells (j). The expression of IRF8 (k), p-S6K (l) and p-STAT3 (m) in BMDCs. The MFI summary (left) and representative staining (right) are shown. nâ=â3â4 wells per group. (a, i-m) Each dot indicates one well for the in vitro experiment in the summary data. (b, left; h, upper) The average tumour sizes of the groups on the day of the experiment are shown as dots and are presented as the meanâ±âSD. (b, right; h, bottom) Each line represents one mouse. (a, i-m) Data are presented as the meanâ±âSD. (i-m) One-way ANOVA with Bonferroni correction. (h) Two-way ANOVA with the TukeyâKramer method.
Extended Data Fig. 11 YB328 treatment increases CD103+CD11bâ cDCs in tumour-related tissues to activate CD8+ T cells.
a-c, ATB-SPF mice were subcutaneously injected with MC38 cells and treated with the indicated bacteria. The phenotype and frequency [representative staining (left) and % summary (right)] of DC populations in the indicated organs were examined: NdLNs (a) and SPs (b). nâ=â4â6 mice per group. TCF-1 expression by PD-1+CD8+ T cells in the indicated organs after the indicated treatment. Representative staining (upper) and % summary (lower) (c). d-g, KikGR mice pretreated with ATB were injected subcutaneously with MC38 cells and treated with the indicated bacteria. Before bacterial treatment, KikGR mice were surgically photoconverted. Migratory intestinal cells that expressed KikR+ were profiled in the indicated organs. The frequency [representative staining (left) and % summary (right)] of KikR+CD103+CD11bâ DCs (d), KikR+PD-1+CD8+ T cells (e), KikR+Foxp3+ T cells (f) and KikR+CD11b+ cells (g) among CD45+ cells. nâ=â3â6 mice per group. h, WT GF mice were subcutaneously injected with MC38 cells and were administered an anti-PD-1 mAb with YB328 treatment. Faeces were collected after the 24âh of the first administration of YB328. Tumour were collected at days 20. The presence of YB328 was examined. n.d., not detected. nâ=â7 mice per group. Each dot in the summary graphs indicates one mouse. All the data are presented as the meanâ±âSD. (a, b, d-g) Two-sided unpaired Studentâs t test.
Extended Data Fig. 12 The abundance of YB328 is associated with the response to anti-PD-1 therapy in human cancers.
a-c, Representative images of multiplex immunohistochemical staining of tumours from GC patients (a) and the calculated density of immune cells. Correlations between the relative abundance of YB328 in the gut microbiota and the density of CD206+ cells (left) or CD14+ cells (right) (b, nâ=â20). Correlations between PD-1+CD8+ T cells and CD206+ cells (left panel) or CD14+ cells (right panel) infiltration (c, nâ=â24). The correlation coefficient was evaluated by two-sided Pearsonâs correlation. Scale bar: 200 μm. d, YB328 abundance in faeces from patients in the validation cohort (nâ=â21; NSCLC:7; GC:14). e, High/low YB328 abundance was stratified by a ROC curve analysis (left panel) KaplanâMeier curves for the PFS of patients with high/low YB328 abundance in the validation cohort (right panel). f, Relative abundance of the YB328 phylotype in the HNSCC cohort (nâ=â16). g, h, Relative abundance of the YB328 phylotype (g, h), and Akkermansia (g) in faecal samples across datasets from multiple cohorts. [g: MONSTAR cohort47 (MM nâ=â16; RCC nâ=â44; GC nâ=â32; EC nâ=â29), h: Routy et al. 48 (nâ=â20)]. A pseudocount was added for visualization purposes, and the boxplots denote the median with a interquartile range, and the whiskers extend to the furthest data point within 1.5 times the interquartile range (g). i, ATB-SPF mice were subcutaneously injected with B16F10-OVA cells, and OT-I CD8+ T cells were transferred with/without YB328 treatment. Experimental scheme (upper). Tumour growth curves (lower left: summary, meanâ±âSD; lower right: each mouse). The graph shows representative data from two independent experiments. nâ=â3â4 mice per group. j, k, Graphical summaries of the mechanism by which the gut microbiota augments the antitumour efficacy of PD-1 blockade therapy. (k) was created using BioRender (https://www.biorender.com). Data are presented as the median (d) or meanâ±âSD (f). (b-d, f-h) Each dot indicates one patient. (d, f, g) Two-sided MannâWhitney test. (e, right) Two-sided log-rank test. (h) Two-sided paired t test. (i) Two-way ANOVA with the TukeyâKramer method.
Supplementary information
Supplementary Information
Supplementary Discussion, Supplementary Tables 1â7 and 11, descriptions for Supplementary Tables 8â10 and 12 (tables supplied separately), Supplementary Data 1 and Supplementary Figs. 1 and 2.
Supplementary Table 8
Genomic proximity (% ANI) between strain YB328 and other strains of the H.âmulieris species. Closeness of strain YB328 to other strains within the H.âmulieris species.
Supplementary Table 9
Phenotypic traits and biochemical properties of YB328 and the species of YB328-related genera. Phenotypic traits and biochemical properties of YB328 compared with H.âmulieris, N.âtimonensis, A.âmuris and Scatolibacter rhodanostii.
Supplementary Table 10
List of the CDS annotations for the genome of strain YB238. Unique genes in YB238 in comparison with the genomes of Acutalibacter muris DSM 26090, N.âtimonensis DSM 102082, C.âleptum DSM 753, R.âalbus JCM 14654, R.âtorques strain AE30, and P.âvulgatus AE61.
Supplementary Table 12
Correlations of clinicopathological features with the clinical responses in MONSTAR cohort. Clinicopathological features, including age, sex, the number of previous chemotherapy treatments and the number of metastatic organ sites, are reported.
Supplementary Video 1
The interaction of Tâcells with YB328-stimulated BMDCs and P.âvulgatus-stimulated BMDCs. Dynamic imaging of CD8+ Tâcells (cyan) and BMDCs stimulated with YB328 (green) or P.âvulgatus (magenta). Elapsed time is shown as mm:ss. Scale bar, 10âμm. Lines indicate cell tracks.
Supplementary Video 2
The interaction of Tâcells with YB328-stimulated BMDCs. Dynamic imaging of CD8+ Tâcells (cyan) and BMDCs stimulated with YB328 (green). Elapsed time is shown as mm:ss. Scale bar, 10âμm.
Supplementary Video 3
The interaction of Tâcells with P.âvulgatus-stimulated BMDCs. Dynamic imaging of CD8+ Tâcells (cyan) and BMDCs stimulated with P.âvulgatus (magenta). Elapsed time is shown as mm:ss. Scale bar, 10âμm.
Supplementary Video 4
The engulfment of YB328 by BMDCs. Dynamic imaging of BMDCs (red) co-cultured with YB328 (green). Elapsed time is shown as mm:ss. Scale bar, 5âμm.
Source data
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the articleâs Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the articleâs Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Lin, N.YT., Fukuoka, S., Koyama, S. et al. Microbiota-driven antitumour immunity mediated by dendritic cell migration. Nature (2025). https://doi.org/10.1038/s41586-025-09249-8
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41586-025-09249-8