Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

Bifidobacterium bifidum strains synergize with immune checkpoint inhibitors to reduce tumour burden in mice

Abstract

The gut microbiome can influence the development of tumours and the efficacy of cancer therapeutics1,2,3,4,5; however, the multi-omics characteristics of antitumour bacterial strains have not been fully elucidated. In this study, we integrated metagenomics, genomics and transcriptomics of bacteria, and analyses of mouse intestinal transcriptome and serum metabolome data to reveal an additional mechanism by which bacteria determine the efficacy of cancer therapeutics. In gut microbiome analyses of 96 samples from patients with non-small-cell lung cancer, Bifidobacterium bifidum was abundant in patients responsive to therapy. However, when we treated syngeneic mouse tumours with commercial strains of B. bifidum to establish relevance for potential therapeutic uses, only specific B. bifidum strains reduced tumour burden synergistically with PD-1 blockade or oxaliplatin treatment by eliciting an antitumour host immune response. In mice, these strains induced tuning of the immunological background by potentiating the production of interferon-γ, probably through the enhanced biosynthesis of immune-stimulating molecules and metabolites.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Marker gene sequence (16S rRNA) profiling of the gut microbiome in patients with NSCLC.
Fig. 2: Synergistic antitumour effect of B. bifidum strains in combination with oxaliplatin or anti-PD-1 in syngeneic mouse tumour models.
Fig. 3: Transcriptomics and metabolomics analysis of syngeneic tumour models treated with B. bifidum strains.
Fig. 4: Comparative genomics and transcriptomics analyses of B. bifidum strains.

Similar content being viewed by others

Data availability

16s rRNA and WGS data from human stool samples, the mouse intestinal RNA sequencing data, the bacterial WGS data and the bacterial RNA sequencing data have been deposited in the European Nucleotide Archive (accession number PRJEB26531). Source data are provided with this paper.

References

  1. Viaud, S. et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 342, 971–976 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Daillère, R. et al. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity 45, 931–943 (2016).

    Article  PubMed  CAS  Google Scholar 

  3. Iida, N. et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 342, 967–970 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sivan, A. et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 350, 1084–1089 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Gopalakrishnan, V. et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 359, 97–103 (2018).

    Article  CAS  PubMed  Google Scholar 

  6. Thaiss, C. A., Zmora, N., Levy, M. & Elinav, E. The microbiome and innate immunity. Nature 535, 65–74 (2016).

    Article  CAS  PubMed  Google Scholar 

  7. Honda, K. & Littman, D. R. The microbiota in adaptive immune homeostasis and disease. Nature 535, 75–84 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Rosshart, S. P. et al. Wild mouse gut microbiota promotes host fitness and improves disease resistance. Cell 171, 1015–1028 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Vétizou, M. et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 350, 1079–1084 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Routy, B. et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359, 91–97 (2018).

    Article  CAS  PubMed  Google Scholar 

  11. Matson, V. et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 359, 104–108 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Eisenhauer, E. A. et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Guerrero-Ros, I. et al. The negative effect of lipid challenge on autophagy inhibits T cell responses. Autophagy 16, 223–238 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Gao, J. et al. Loss of IFN-γ pathway genes in tumor cells as a mechanism of resistance to anti-CTLA-4 therapy. Cell 167, 397–404.e9 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Patel, S. J. et al. Identification of essential genes for cancer immunotherapy. Nature 548, 537–542 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Medzhitov, R. Toll-like receptors and innate immunity. Nat. Rev. Immunol. 1, 135–145 (2001).

    Article  CAS  PubMed  Google Scholar 

  17. Kohwi, Y., Imai, K., Tamura, Z. & Hashimoto, Y. Antitumor effect of Bifidobacterium infantis in mice. GANN 69, 613–618 (1978).

    CAS  PubMed  Google Scholar 

  18. Rafter, J. Probiotics and colon cancer. Best. Pract. Res. Clin. Gastroenterol. 17, 849–859 (2003).

    Article  PubMed  Google Scholar 

  19. Zhang, L. S. & Davies, S. S. Microbial metabolism of dietary components to bioactive metabolites: opportunities for new therapeutic interventions. Genome Med. 8, 46 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Desbonnet, L., Garrett, L., Clarke, G., Bienenstock, J. & Dinan, T. G. The probiotic Bifidobacteria infantis: an assessment of potential antidepressant properties in the rat. J. Psychiatr. Res. 43, 164–174 (2008).

    Article  PubMed  Google Scholar 

  21. Xiao, J. et al. Effects of milk products fermented by Bifidobacterium longum on blood lipids in rats and healthy adult male volunteers. J. Dairy Sci. 86, 2452–2461 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. An, H. M. et al. Antiobesity and lipid-lowering effects of Bifidobacterium spp. in high fat diet-induced obese rats. Lipids Health Dis. 10, 116 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Wang, K. et al. Bifidobacterium bifidum TMC3115 can characteristically influence glucose and lipid profile and intestinal microbiota in the middle-aged and elderly. Probiotics Antimicrob. Proteins 11, 1182–1194 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Howie, D., Ten Bokum, A., Necula, A. S., Cobbold, S. P. & Waldmann, H. The role of lipid metabolism in T lymphocyte differentiation and survival. Front. Immunol. 8, 1949 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Long, J. et al. Lipid metabolism and carcinogenesis, cancer development. Am. J. Cancer Res. 8, 778–791 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Cruceriu, D., Baldasici, O., Balacescu. & Berindan-Neagoe, I. The dual role of tumor necrosis factor-alpha (TNF-α) in breast cancer: molecular insights and therapeutic approaches. Cell. Oncol. 43, 1–18 (2020).

    Article  CAS  Google Scholar 

  27. Chen, X., Bäumel, M., Männel, D. N., Howard, O. Z. & Oppenheim, J. J. Interaction of TNF with TNF receptor type 2 promotes expansion and function of mouse CD4+ CD25+ T regulatory cells. J. Immunol. 179, 154–161 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Schioppa, T. et al. B regulatory cells and the tumor-promoting actions of TNF-α during squamous carcinogenesis. Proc. Natl Acad. Sci. USA 108, 10662–10667 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zhao, X. et al. TNF signaling drives myeloid-derived suppressor cell accumulation. J. Clin. Invest. 122, 4094–4104 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zheng, L. et al. Induction of apoptosis in mature T cells by tumour necrosis factor. Nature 377, 348–351 (1995).

    Article  CAS  PubMed  Google Scholar 

  31. Bertrand, F. et al. Blocking tumor necrosis factor α enhances CD8 T-cell-dependent immunity in experimental melanoma. Cancer Res. 75, 2619–2628 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Klindworth, A. et al. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res. 41, e1 (2013).

    Article  CAS  PubMed  Google Scholar 

  33. Davis, M. P., van Dongen, S., Abreu-Goodger, C., Bartonicek, N. & Enright, A. J. Kraken: a set of tools for quality control and analysis of high-throughput sequence data. Methods 63, 41–49 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet.journal 17, 10–12 (2011).

    Article  Google Scholar 

  35. Bolyen, E. et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 37, 852–857 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Callahan, B. J. et al. DADA2: high-resolution sample inference from Illumina amplicon data. Nat. Methods 13, 581–583 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. DeSantis, T. Z. et al. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl. Environ. Microbiol. 72, 5069–5072 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Segata, N. et al. Metagenomic biomarker discovery and explanation. Genome Biol. 12, R60 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Hill, D. A. et al. Metagenomic analyses reveal antibiotic-induced temporal and spatial changes in intestinal microbiota with associated alterations in immune cell homeostasis. Mucosal Immunol. 3, 148–158 (2010).

    Article  CAS  PubMed  Google Scholar 

  40. Segata, N. et al. Metagenomic microbial community profiling using unique clade-specific marker genes. Nat. Methods 9, 811–814 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Abubucker, S. et al. Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLoS Comput. Biol. 8, e1002358 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Parks, D. H., Tyson, G. W., Hugenholtz, P. & Beiko, R. G. STAMP: statistical analysis of taxonomic and functional profiles. Bioinformatics 30, 3123–3124 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Ferrer-Font, L. et al. High-dimensional analysis of intestinal immune cells during helminth infection. eLife 9, e51678 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  45. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Bindea, G. et al. ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks. Bioinformatics 25, 1091–1093 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Bang, D. Y., Byeon, S. K. & Moon, M. H. Rapid and simple extraction of lipids from blood plasma and urine for liquid chromatography–tandem mass spectrometry. J. Chromatogr. A 1331, 19–26 (2014).

    Article  CAS  PubMed  Google Scholar 

  49. Lim, S., Byeon, S. K., Lee, J. Y. & Moon, M. H. Computational approach to structural identification of phospholipids using raw mass spectra from nanoflow liquid chromatography–electrospray ionization–tandem mass spectrometry. J. Mass Spectrom. 47, 1004–1014 (2012).

    Article  CAS  PubMed  Google Scholar 

  50. Horai, H. et al. MassBank: a public repository for sharing mass spectral data for life sciences. J. Mass Spectrom. 45, 703–714 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. Ivanova, P. T., Milne, S. B., Myers, D. S. & Brown, H. A. Lipidomics: a mass spectrometry based systems level analysis of cellular lipids. Curr. Opin. Chem. Biol. 13, 526–531 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Bankevich, A. et al. SPAdes: a new genome assembly algorithm and its applications to single-cell sequencing. J. Comput. Biol. 19, 455–477 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gurevich, A., Saveliev, V., Vyahhi, N. & Tesler, G. QUAST: quality assessment tool for genome assemblies. Bioinformatics 29, 1072–1075 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Seemann, T. Prokka: rapid prokaryotic genome annotation. Bioinformatics 30, 2068–2069 (2014).

    Article  CAS  PubMed  Google Scholar 

  55. Page, A. J. et al. Roary: rapid large-scale prokaryote pan genome analysis. Bioinformatics 31, 3691–3693 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Pritchard, L., Glover, R. H., Humphris, S., Elphinstone, J. G. & Toth, I. K. Genomics and taxonomy in diagnostics for food security: soft-rotting enterobacterial plant pathogens. Anal. Methods 8, 12–24 (2016).

    Article  Google Scholar 

  57. Bardou, P., Mariette, J., Escudié, F., Djemiel, C. & Klopp, C. jvenn: an interactive Venn diagram viewer. BMC Bioinformatics 15, 293 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  58. Förstner, K. U., Vogel, J. & Sharma, C. M. READemption—a tool for the computational analysis of deep-sequencing-based transcriptome data. Bioinformatics 30, 3421–3423 (2014).

    Article  PubMed  CAS  Google Scholar 

  59. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This research was supported by the Bio & Medical Technology Development Program of the National Research Foundation (NRF), funded by the Ministry of Science & ICT (NRF-2017M3A9F3046536 to H.P.); a GIST Research Institute (GRI) grant, funded by the GIST in 2020; Genome and Company (grant number GNC GR 17-01 to H.P.). This work was also supported by grants from National Cancer Centre, Korea (NCC-1911267 to H.P); the Post-Genome Technology Development Program (10067758, Business model development driven by clinico-genomic database for precision immuno-oncology to S.-H.L.) funded by the Ministry of Trade, Industry and Energy (MOTIE, Korea). S.-Y.C. is supported by the Bio & Medical Technology Development Program of the NRF, funded by the Ministry of Science & ICT (NRF-2018M3A9F3056902). This research was also supported by a grant from the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (grant number HI17C1076 to K.W.Y). This work was supported by the NRF grant funded by the Korea government (MSIT) (number 2017R1C1B2011196 to K.W.Y). M.H.M. is supported by grant NRF-2018R1A2A1A05019794 from the NRF of Korea. B.-N.J. is supported by the Basic Science Research Program through the NRF of Korea, funded by the Ministry of Education, Science and Technology (MEST) (2018R1C1B6005768). The International Research & Development Program of the NRF of Korea, funded by the Ministry of Science, ICT & Future Planning (grant no. 2015K1A4A3047851) also funded C.L. The Ewha Womans University Professorship is supported in part by the Ewha Womans University research grant of 2017-2019. This study is also supported in part by operational funds from The First Affiliated Hospital of Xi’an Jiaotong University.

Author information

Authors and Affiliations

Authors

Contributions

S.-H.L. and H.S.K. collected and analysed the clinical data. S.-H.L., S.-Y.C., Y.Y., C.P. and K.W.Y. wrote the manuscript. J.Y.K., S.W., J.-S.P., G.M.W., C.L. and H.P. revised the manuscript. S.-H.L., S.-Y.C., C.P. and G.K. analysed the 16S rRNA sequences and transcriptomics data. J.-J.J., B.-N.J., H.-S.Y. and Sarang Kim performed the cytometry analysis. Y.Y., J.S., S.L., Y.Y.K., Sujeong Kim, Yunjae Kim and S.G.K. performed the syngeneic mouse model experiments. Seonggon Kim. and J.-S.P. performed the orthotopic mouse model experiments. C.A., E.J.L., Yeongmin Kim and H.K. cultured the bacteria. H.-S.Y. and Sarang Kim performed the in vitro T-cell assays. M.J., H.C. and M.H.N. analysed metabolomics data. G.B.L. and M.H.M. analysed the lipidomics data. K.W.Y. and H.P. designed and supervised all experiments and analyses.

Corresponding authors

Correspondence to Kyoung Wan Yoon or Hansoo Park.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Microbiology thanks the anonymous reviewers for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Comparison of gut microbiome composition between non–small cell lung cancer (NSCLC) patients and healthy controls.

a, Scheme for human, syngeneic tumour model, and bacterial study. b, c Box-and-whisker plot illustrating alpha diversity (b) and principal coordinate analysis plot illustrating beta diversity (c) of gut microbiomes in NSCLC patients (red, n = 96) and healthy controls (blue, n = 139). p value of alpha diversity calculated using Kruskal-Wallis test. Box represents first, third quartiles, and median, and whiskers are range up to 1.5×Interquartile Ranges (IQR). d, A plot of linear discriminant analysis (LDA) scores from the linear discriminant analysis effect size (LEfSe) method illustrating the differential abundance of the indicated taxa in the gut microbiomes of NSCLC patients (red) and healthy controls (blue) (LDA score > 4). e, The abundance of B. bifidum in NSCLC patients except epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI)-treated patients was determined by qPCR (n = 80). Data show means ± SEM. p value calculated using two-sided unpaired Mann-Whitney U test, p = 0.0128. f, Effect of B. bifidum and Leuconostoc strains on in vitro stimulation of CD8+ T cells by monocytes. Secreted IFN-γ was measured by ELISA in co-cultures of human autologous T cells and monocytes treated with B. bifidum and Leuconostoc strains (Supplementary Table 5). 3 technical replicates. Data show means ± SEM. p values calculated using one-way ANOVA with Tukey for multiple comparison. B. bif_M31 versus L. mes_G16, p = 0.0040; B. bif_M31 versus L. mes_K20, p = 0.0245; B. bif_M31 versus L. gar_K21, p = 0.0301. For all graphs, *p < 0.05, and **p < 0.01.

Source data

Extended Data Fig. 2 Synergistic anti-tumor effect of B. bifidum strains in combination with oxaliplatin or anti-PD-1 in syngeneic mouse tumor models.

a, Abundance of B. bifidum in the mouse intestine after administration. Mice were administered a onetime dose of 1 × 109 CFU of B. bif_K57 and stool samples were collected at 4, 8, 12, and 24 hr after administration. B. bifidum abundance was determined using a qPCR assay with B. bifidum specific primers and normalized to 16S rRNA. n = 5 independent biological replicates. Data show means ± SEM. b, Flow cytometry analyses were performed on mouse spleen and tumor tissue. Upper: Heatmap represents the p values of differences in immune cell profile between oxaliplatin treatment and combined treatment with oxaliplatin and B. bifidum strains. p values calculated using two-sided unpaired t test. The exact p values are provided in Supplementary Table 14. Lower: Ratio of CD8+ T/Treg and effector CD8+ T/Treg in spleen and tumor. Ctrl, n = 9; Oxp or Oxp + B. bif_K57, n = 7; Oxp + B. bif_B06, n = 8 mice per group. Data show means ± SEM. p values calculated using one-way ANOVA with Tukey for multiple comparison. Oxp versus Oxp + B. bif_K57, p = 0.0002 (Ratio of CD8+ T/Treg and effector CD8+ T/Treg, tumor); Oxp versus Oxp + B. bif_B06, p = 0.0124 (Ratio of effector CD8+ T/Treg, tumor); Oxp versus Oxp + B. bif_K57, p = 0.0011 (Ratio of CD8+ T/Treg, spleen); Oxp versus Oxp + B. bif_K57, p = 0.0039 (Ratio of effector CD8+ T/Treg, spleen). c, Cytokine expression profiles in tumours from mice treated with oxaliplatin or both oxaliplatin and B. bifidum were measured by qPCR. Ctrl or Oxp groups, n = 6 independent biological replicates/group; oxaliplatin + B. bif_K57 or B. bif_ B06 groups, n = 8 independent biological replicates/group. Data show means ± SEM. p values calculated using one-way ANOVA with Tukey for multiple comparison within oxaliplatin treated groups. IFN-γ of Oxp versus Oxp + B. bif_K57, p = 0.0478; IL-2 of Oxp versus oxp + B. bif_K57, p < 0.0001; IL-2 of Oxp versus Oxp + B. bif_B06, p = 0.0006; TNF-α of Oxp versus Oxp + B. bif_K57, p = 0.0004; TNF-α of Oxp versus Oxp + B. bif_B06, p = 0.0040; IL-10 of Oxp versus Oxp + B. bif_K57, p < 0.0001; IL-10 of Oxp versus Oxp + B. bif_B06, p = 0.0063. d, Representative tumor growth curves after administration of B. bif_M31, with or without anti-PD-1 treatment. IgG, n = 8; anti-PD-1, n = 5; B. bif_M31, n = 10; anti-PD-1 + B. bif_M31, n = 9 mice per group. Data show means ± SEM. p value calculated using two-way ANOVA with Tukey for multiple comparison. anti-PD-1 versus anti-PD-1 + B. bif_M31, p < 0.0001. e, Representative tumor growth curves after administration of B. bif_C01, with or without anti-PD-1 treatment. IgG, n = 6; anti-PD-1 or B. bif_C01 or and anti-PD-1 + B. bif_C01, n = 7 mice per group. Data show means ± SEM. p value calculated using two-way ANOVA with Tukey for multiple comparison. f, Flow cytometry analysis of CD4+ T, NK and Treg cells in lamina propria of the intestine. n = 7 mice per group. Data show means ± SEM. p values calculated using one-way ANOVA with Tukey for multiple comparison. CD4+ T cells of anti-PD-1 versus anti-PD-1 + B. bif_K57, p = 0.0015; NK cells of anti-PD-1 versus anti-PD-1 + B. bif_K57, p = 0.0037. For all graphs, *p < 0.05, **p < 0.01, ***p < 0.001, and ns = not significant.

Source data

Extended Data Fig. 3 Transcriptomic and Lipidomic analysis of syngeneic tumor model treated with B. bifidum strains.

a, Network representation of enriched Gene Ontology (GO) biological pathways among differentially up-regulated genes in mice treated with anti-PD-1 + B. bif_K57 mice, determined using ClueGO. b, Principal component analysis of syngeneic mouse serum lipid profiles, based on lipids differential abundance among mice treated with anti-PD-1, anti-PD-1 + B. bif_B06, and anti-PD-1 + B. bif_K57 (>1.5-fold & p < 0.01). c, Heatmap of lipids showing significant differences (>1.5-fold & p < 0.01) in serum of syngeneic mice, as revealed by the lipidomic analysis. PE, phosphatidylethanolamine; PI, phosphoinositol; PC, phosphatidylcholine; LPG, lysophosphatidylglycerol; DG, diacylglycerol; TG, triglycerol.

Source data

Extended Data Fig. 4 Synergistic anti-tumor effect of B. bifidum strains in combination with anti-PD-1 in various syngeneic mouse tumor models.

a, Effect of B. bifidum strains on in vitro stimulation of CD8 T+ cells by monocytes. Secreted IFN-γ was measured by ELISA in co-cultures of human autologous T cells and monocytes treated with B. bif_K57 or B. bif_B06. 2 technical replicates. Data show means ± SEM. b, Upper: Experimental timeline: Animals were treated with the antibiotic cocktail (ABX) for 14 days. B. bif_K57 was then orally administered for 14 days before inoculation with MC38 colon cancer cells, followed by administration of anti-PD-1 via intraperitoneal injection twice a week for 21 days. Lower: MC38 tumor growth curves are shown for animals subjected to the indicated experimental treatments. IgG after PBS or anti-PD-1 after PBS, n = 5; anti-PD-1 + B. bif_K57 after PBS, n = 6; anti-PD-1 after ABX, n = 5; anti-PD-1 + B. bif_K57 after ABX, n = 7 mice per group. Data show means ± SEM. p values calculated using two-way ANOVA with Tukey for multiple comparison. anti-PD-1 after PBS versus anti-PD-1 + B. bif_K57 after PBS, p = 0.0053; anti-PD-1 after PBS versus anti-PD-1 + B. bif_K57 after ABX, p = 0.0004. c, Effect of ABX treatment on the abundance of gut microbiome. Stool 16S rRNA genes were analyzed by qPCR. n = 5 mice per group. Data show means ± SEM. p value calculated using two-sided unpaired t test, p < 0.0001. d, Upper: Experimental timeline: 14 days after initial oral administration of B. bif_K57, mice were inoculated with LLC1. anti-PD-1 was administered via intraperitoneal injection eight times, twice a week for 28 days. Lower: LLC1 tumor growth curves were generated for animals subjected to the indicated experimental treatments. n = 6 mice per group. Data show means ± SEM. p values calculated using two-way ANOVA with Tukey for multiple comparison. IgG versus anti-PD-1 + B. bif_K57, p < 0.0001. e, Macroscopic findings of tumor-inoculated lungs at day 23. The implanted tumors are indicated by yellow arrows. Scale bar, 50 mm. f, Upper: Experimental timeline: 14 days after initial oral administration of B. bif_K57, mice were inoculated with 4T1 breast cancer cells. anti-PD-1 was administered via intraperitoneal injection six times, twice a week for 21 days. Lower: 4T1 tumor growth curves were generated for animals subjected to the indicated experimental treatments. n = 10 mice per group. Data show means ± SEM. IgG versus anti-PD-1 + B. bif_K57, p < 0.0001. For all graphs, **p < 0.01, ***p < 0.001, and ns = not significant.

Source data

Extended Data Fig. 5 The abundance of peptidoglycan in B. bifidum strains.

The abundance of peptidoglycan of B. bif_K57 and B. bif_B06 was estimated using ELISA. Data show means ± SEM. 3 biological replicates. p value calculated using two-sided unpaired t test, p < 0.0006. For all graphs, ***p < 0.001.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–5.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–14.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data and metabolomics source data.

Source Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Lipidomics source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lee, SH., Cho, SY., Yoon, Y. et al. Bifidobacterium bifidum strains synergize with immune checkpoint inhibitors to reduce tumour burden in mice. Nat Microbiol 6, 277–288 (2021). https://doi.org/10.1038/s41564-020-00831-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-020-00831-6

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer