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.

  • Article
  • Published:

Diet-driven microbial ecology underpins associations between cancer immunotherapy outcomes and the gut microbiome

Abstract

The gut microbiota shapes the response to immune checkpoint inhibitors (ICIs) in cancer, however dietary and geographic influences have not been well-studied in prospective trials. To address this, we prospectively profiled baseline gut (fecal) microbiota signatures and dietary patterns of 103 trial patients from Australia and the Netherlands treated with neoadjuvant ICIs for high risk resectable metastatic melanoma and performed an integrated analysis with data from 115 patients with melanoma treated with ICIs in the United States. We observed geographically distinct microbial signatures of response and immune-related adverse events (irAEs). Overall, response rates were higher in Ruminococcaceae-dominated microbiomes than in Bacteroidaceae-dominated microbiomes. Poor response was associated with lower fiber and omega 3 fatty acid consumption and elevated levels of C-reactive protein in the peripheral circulation at baseline. Together, these data provide insight into the relevance of native gut microbiota signatures, dietary intake and systemic inflammation in shaping the response to and toxicity from ICIs, prompting the need for further studies in this area.

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: Associations between gut microbiota and treatment response and irAEs in Australian patients with melanoma.
Fig. 2: Microbial functions and dietary nutrient intake that promote gut integrity are associated with protection from irAEs and lack of response.
Fig. 3: Geographical differences in microbial communities in cohorts from Australia and the Netherlands.
Fig. 4: Ruminococcaceae-associated microbial signatures support the response to checkpoint inhibitor immunotherapy.

Similar content being viewed by others

Data availability

Sequencing data are available from the European Nucleotide Archive under accession number PRJEB54666. Supporting de-identified metadata have been provided. Publicly available datasets were attained from The NCBI Sequence Read Archive (SRA) under accession number SRP116709 and the European Nucleotide Archive under accession numbers PRJEB22894 and PRJNA770295. Further details, data and code are available upon request from the authors.

Code availability

No unique software or computational code was created for this study. Implementation of established tools and pipelines are described in the methods.

References

  1. Seidel, J. A., Otsuka, A. & Kabashima, K. Anti-PD-1 and anti-CTLA-4 therapies in cancer: mechanisms of action, efficacy, and limitations. Front Oncol. 8, 86 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Larkin, J. et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 381, 1535–1546 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Larkin, J. et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N. Engl. J. Med. 373, 23–34 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Wolchok, J. D. et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 377, 1345–1356 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Long, G. V. et al. Standard-dose pembrolizumab in combination with reduced-dose ipilimumab for patients with advanced melanoma (KEYNOTE-029): an open-label, phase 1b trial. Lancet Oncol. 18, 1202–1210 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Long, G. V. et al. Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: a multicentre randomised phase 2 study. Lancet Oncol. 19, 672–681 (2018).

    Article  CAS  PubMed  Google Scholar 

  7. Rozeman, E. A. et al. Identification of the optimal combination dosing schedule of neoadjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma (OpACIN-neo): a multicentre, phase 2, randomised, controlled trial. Lancet Oncol. 20, 948–960 (2019).

    Article  CAS  PubMed  Google Scholar 

  8. Menzies, A. M. et al. Pathological response and survival with neoadjuvant therapy in melanoma: a pooled analysis from the International Neoadjuvant Melanoma Consortium (INMC). Nat. Med. 27, 301–309 (2021).

    Article  CAS  PubMed  Google Scholar 

  9. Rozeman, E. A. et al. Survival and biomarker analyses from the OpACIN-neo and OpACIN neoadjuvant immunotherapy trials in stage III melanoma. Nat. Med. 27, 256–263 (2021).

    Article  CAS  PubMed  Google Scholar 

  10. Luke, J. J., Flaherty, K. T., Ribas, A. & Long, G. V. Targeted agents and immunotherapies: optimizing outcomes in melanoma. Nat. Rev. Clin. Oncol. 14, 463–482 (2017).

    Article  CAS  PubMed  Google Scholar 

  11. Grainger, J., Daw, R. & Wemyss, K. Systemic instruction of cell-mediated immunity by the intestinal microbiome. F1000Res 7, (2018).

  12. Harkiolaki, M. et al. T cell-mediated autoimmune disease due to low-affinity crossreactivity to common microbial peptides. Immunity 30, 348–357 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Horai, R. et al. Microbiota-dependent activation of an autoreactive t cell receptor provokes autoimmunity in an immunologically privileged site.Immunity 43, 343–353 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Parada Venegas, D. et al. Short chain fatty acids (scfas)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 10, 277 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Steed, A. L. et al. The microbial metabolite desaminotyrosine protects from influenza through type I interferon. Science 357, 498–502 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Bachem, A. et al. Microbiota-derived short-chain fatty acids promote the memory potential of antigen-activated CD8(+) T cells. Immunity 51(2), 285–297.e5, https://doi.org/10.1016/j.immuni.2019.06.002 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Marchesi, J. R. et al. The gut microbiota and host health: a new clinical frontier. Gut 65, 330–339 (2016).

    Article  PubMed  Google Scholar 

  18. 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 

  19. 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 

  20. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Chaput, N. et al. Baseline gut microbiota predicts clinical response and colitis in metastatic melanoma patients treated with ipilimumab. Ann. Oncol. 28, 1368–1379 (2017).

    Article  CAS  PubMed  Google Scholar 

  23. Coutzac, C. et al. Systemic short chain fatty acids limit antitumor effect of CTLA-4 blockade in hosts with cancer. Nat. Commun. 11, 2168 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Smith, M. et al. Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat. Med. 28, 713–723 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Andrews, M. C. et al. Gut microbiota signatures are associated with toxicity to combined CTLA-4 and PD-1 blockade. Nat. Med. 27, 1432–1441 (2021).

    Article  CAS  PubMed  Google Scholar 

  26. McCulloch, J. A. et al. Intestinal microbiota signatures of clinical response and immune-related adverse events in melanoma patients treated with anti-PD-1. Nat. Med. 28, 545–556, https://doi.org/10.1038/s41591-022-01698-2 (2022).

    Article  CAS  PubMed  Google Scholar 

  27. Lee, K. A. et al. Cross-cohort gut microbiome associations with immune checkpoint inhibitor response in advanced melanoma. Nat. Med. 28, 535–544, https://doi.org/10.1038/s41591-022-01695-5 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Gharaibeh, R. Z. & Jobin, C. Microbiota and cancer immunotherapy: in search of microbial signals. Gut 68, 385–388, https://doi.org/10.1136/gutjnl-2018-317220 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Tetzlaff, M. T. et al. Pathological assessment of resection specimens after neoadjuvant therapy for metastatic melanoma. Ann. Oncol. 29, 1861–1868 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Amaria, R. N. et al. Neoadjuvant systemic therapy in melanoma: recommendations of the International Neoadjuvant Melanoma Consortium. Lancet Oncol. 20, e378–e389 (2019).

    Article  PubMed  Google Scholar 

  31. Frankel, A. E. et al. Metagenomic shotgun sequencing and unbiased metabolomic profiling identify specific human gut microbiota and metabolites associated with immune checkpoint therapy efficacy in melanoma patients. Neoplasia 19, 848–855 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Peters, B. A. et al. Relating the gut metagenome and metatranscriptome to immunotherapy responses in melanoma patients. Genome Med 11, 61 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Yu, L. C. Microbiota dysbiosis and barrier dysfunction in inflammatory bowel disease and colorectal cancers: exploring a common ground hypothesis. J. Biomed. Sci. 25, 79 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Rajca, S. et al. Alterations in the intestinal microbiome (dysbiosis) as a predictor of relapse after infliximab withdrawal in Crohn’s disease. Inflamm. Bowel Dis. 20, 978–986 (2014).

    PubMed  Google Scholar 

  35. Bang, C. & Schmitz, R. A. Archaea associated with human surfaces: not to be underestimated. FEMS Microbiol. Rev. 39, 631–648 (2015).

    Article  CAS  PubMed  Google Scholar 

  36. Smith, N. W., Shorten, P. R., Altermann, E. H., Roy, N. C. & McNabb, W. C. Hydrogen cross-feeders of the human gastrointestinal tract. Gut Microbes 10, 270–288 (2019).

    Article  CAS  PubMed  Google Scholar 

  37. Belkaid, Y. & Hand, T. W. Role of the microbiota in immunity and inflammation. Cell 157, 121–141 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Llewellyn, S. R. et al. Interactions between diet and the intestinal microbiota alter intestinal permeability and colitis severity in mice. Gastroenterology 154, e1032 (2018).

    Article  Google Scholar 

  39. Nestel, P. et al. Indications for omega-3 long chain polyunsaturated fatty acid in the prevention and treatment of cardiovascular disease. Heart Lung Circ. 24, 769–779 (2015).

    Article  PubMed  Google Scholar 

  40. Macia, L. et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat. Commun. 6, 6734 (2015).

    Article  CAS  PubMed  Google Scholar 

  41. Chiba, M., Nakane, K. & Komatsu, M. Westernized diet is the most ubiquitous environmental factor in inflammatory bowel disease. Perm. J. 23, 18–107 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Watson, H. et al. A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiota. Gut 67, 1974–1983 (2018).

    Article  CAS  PubMed  Google Scholar 

  43. Lam, Y. Y. et al. Effects of dietary fat profile on gut permeability and microbiota and their relationships with metabolic changes in mice. Obes. (Silver Spring) 23, 1429–1439 (2015).

    Article  CAS  Google Scholar 

  44. Mager, L. F. et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science 369, 1481–1489 (2020).

    Article  CAS  PubMed  Google Scholar 

  45. Dubin, K. et al. Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat. Commun. 7, 10391 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Chen, J., Zhao, K.N. & Vitetta, L. Effects of intestinal microbial(-)elaborated butyrate on oncogenic signaling pathways. Nutrients 11(5), 1026 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  47. He, Y. et al. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat. Med. 24, 1532–1535 (2018).

    Article  CAS  PubMed  Google Scholar 

  48. Arumugam, M. et al. Enterotypes of the human gut microbiome. Nature 473, 174–180 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Holmes, I., Harris, K. & Quince, C. Dirichlet multinomial mixtures: generative models for microbial metagenomics. PLoS One 7, e30126 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wu, G. D. et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 334, 105–108 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Costea, P. I. et al. Enterotypes in the landscape of gut microbial community composition. Nat. Microbiol 3, 8–16 (2018).

    Article  CAS  PubMed  Google Scholar 

  52. Singh, R. K. et al. Influence of diet on the gut microbiome and implications for human health. J. Transl. Med 15, 73 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Chung, D. & Keles, S. Sparse partial least squares classification for high dimensional data. Stat Appl Genet Mol Biol 9(1), (2010).

  54. Spencer, C. N. et al. Dietary fiber and probiotics influence the gut microbiome and melanoma immunotherapy response. Science 374, 1632–1640 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Kovatcheva-Datchary, P. et al. Dietary fiber-induced improvement in glucose metabolism is associated with increased abundance of prevotella. Cell Metab. 22, 971–982 (2015).

    Article  CAS  PubMed  Google Scholar 

  56. Zeevi, D. et al. Personalized nutrition by prediction of glycemic responses. Cell 163, 1079–1094 (2015).

    Article  CAS  PubMed  Google Scholar 

  57. Zmora, N. et al. Personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features. Cell 174, 1388–1405 e1321 (2018).

    Article  CAS  PubMed  Google Scholar 

  58. Lam, K. C. et al. Microbiota triggers STING-type I IFN-dependent monocyte reprogramming of the tumor microenvironment. Cell https://doi.org/10.1016/j.cell.2021.09.019 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Baruch, E. N. et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 371(6529), 602–609, https://doi.org/10.1126/science.abb5920 (2020).

    Article  CAS  PubMed  Google Scholar 

  60. Davar, D. et al. Fecal microbiota transplant overcomes resistance to anti-PD-1 therapy in melanoma patients. Science 371, 595–602 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. 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 

  62. McMurdie, J. & Holmes, S. Phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One 8, e61217 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  64. Sidhu, P. et al. Radiological manifestations of immune-related adverse effects observed in patients with melanoma undergoing immunotherapy. J. Med Imaging Radiat. Oncol. 61, 759–766 (2017).

    Article  PubMed  Google Scholar 

  65. Franzosa, E. A. et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat. Methods 15, 962–968 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Suzek, B. E. et al. UniRef clusters: a comprehensive and scalable alternative for improving sequence similarity searches. Bioinformatics 31, 926–932 (2015).

    Article  CAS  PubMed  Google Scholar 

  67. Caspi, R. et al. The MetaCyc database of metabolic pathways and enzymes. Nucleic Acids Res. 46, D633–D639 (2018).

    Article  CAS  PubMed  Google Scholar 

  68. Gloor, G. B., Macklaim, J. M., Pawlowsky-Glahn, V. & Egozcue, J. J. Microbiome Datasets Are Compositional: And This Is Not Optional. Front Microbiol 8, 2224 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Rohart, F., Gautier, B., Singh, A. & Le Cao, K. A. mixOmics: An R package for ‘omics feature selection and multiple data integration. PLoS Comput. Biol. 13, e1005752 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  70. Zoll, J. et al. Fecal microbiota transplantation from high caloric-fed donors alters glucose metabolism in recipient mice, independently of adiposity or exercise status. Am. J. Physiol. Endocrinol. Metab. 319, E203–E216 (2020).

    Article  CAS  PubMed  Google Scholar 

  71. Siebelink, E., Geelen, A. & de Vries, J. H. Self-reported energy intake by FFQ compared with actual energy intake to maintain body weight in 516 adults. Br. J. Nutr. 106, 274–281 (2011).

    Article  CAS  PubMed  Google Scholar 

  72. Metsalu, T. & Vilo, J. ClustVis: a web tool for visualizing clustering of multivariate data using Principal Component Analysis and heatmap. Nucleic Acids Res. 43, W566–W570 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Shanahan, E. R. et al. Influence of cigarette smoking on the human duodenal mucosa-associated microbiota. Microbiome 6, 150 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Ramirez-Farias, C. et al. Effect of inulin on the human gut microbiota: stimulation of Bifidobacterium adolescentis and Faecalibacterium prausnitzii. Br. J. Nutr. 101, 541–550 (2009).

    Article  CAS  PubMed  Google Scholar 

  75. Mackie, R. I. et al. Ecology of uncultivated Oscillospira species in the rumen of cattle, sheep, and reindeer as assessed by microscopy and molecular approaches. Appl. Environ. Microbiol. 69, 6808–6815 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Yanagita, K. et al. Flow cytometric sorting, phylogenetic analysis and in situ detection of Oscillospira guillermondii, a large, morphologically conspicuous but uncultured ruminal bacterium. Int J. Syst. Evol. Microbiol 53, 1609–1614 (2003).

    Article  CAS  PubMed  Google Scholar 

  77. Hook, S. E., Northwood, K. S., Wright, A. D. & McBride, B. W. Long-term monensin supplementation does not significantly affect the quantity or diversity of methanogens in the rumen of the lactating dairy cow. Appl. Environ. Microbiol. 75, 374–380 (2009).

    Article  CAS  PubMed  Google Scholar 

  78. Ohnishi, A. et al. Development of a 16S rRNA gene primer and PCR-restriction fragment length polymorphism method for rapid detection of members of the genus Megasphaera and species-level identification. Appl. Environ. Microbiol. 77, 5533–5535 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Layton, A. et al. Development of Bacteroides 16S rRNA gene TaqMan-based real-time PCR assays for estimation of total, human, and bovine fecal pollution in water. Appl. Environ. Microbiol. 72, 4214–4224 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Schneeberger, M. et al. Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice. Sci. Rep. 5, 16643 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. & Geirnaert, A. et al. Interindividual differences in response to treatment with butyrate-producing Butyricicoccus pullicaecorum 25-3 T studied in an in vitro gut model. FEMS Microbiol Ecol 91, (2015).

  82. Hermann-Bank, M. L., Skovgaard, K., Stockmarr, A., Larsen, N. & Molbak, L. The Gut Microbiotassay: a high-throughput qPCR approach combinable with next generation sequencing to study gut microbial diversity. BMC Genomics 14, 788 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank G. Giles of the Cancer Epidemiology Division, Cancer Council Victoria, for permission to use the Cancer Council Victoria Dietary Questionnaire for Epidemiological Studies (DQES v3.2), Melbourne, Australia, 1996. We acknowledge the technical assistance provided by the Sydney Informatics Hub, a core research facility of the University of Sydney. G.V.L and R.A.S are supported by NHMRC Program Grant R.A.S is supported by an NHMRC Practitioner Fellowship and G.V.L by an NHMRC Investigator Grant. G.V.L is also supported by the University of Sydney Medical Foundation. Support from the Cameron Family and Ainsworth Foundation, as well as from colleagues at Melanoma Institute Australia, Royal Prince Alfred Hospital and NSW Health Pathology is gratefully acknowledged. E.R.S. acknowledges financial support from the The William Arthur Martin à Beckett Cancer Research Trust (University of Sydney Fellowship). Funding support was provided by a Tour de Cure Australia project grant (RSP-00054-19/20) (R.A.S, M.B., E.R.S).

Author information

Authors and Affiliations

Authors

Contributions

G.V.L., R.A.S, R.C.S., E.R.S. and M.B. conceived the study. R.C.S., E.R.S., M.B., M.R., A.S.A., J.T., I.L.M.R. and J.V. conducted the experiments. R.C.S., E.R.S., M.B. and M.R. analyzed the data. I.P.S., R.R., C.A., A.M.M., R.P.S., M.G., K.R.S., A.J.S., R.V., A.J.L., A.V.D., A.K.M., M.C., A.B., N.J.A., L.M., and A.J.H. contributed to patient recruitment, biospecimen processing, data analysis and/or critical review of data. J.A.W., C.U.B., R.A.S. and G.V.L. contributed resources to the study. R.C.S., E.R.S. and M.B. wrote the original manuscript draft. All authors wrote, reviewed and edited the manuscript. G.V.L., R.A.S. and J.S.W. supervised the study. G.V.L., R.A.S., M.B. and E.R.S. obtained funding for the study.

Corresponding author

Correspondence to Georgina V. Long.

Ethics declarations

Competing interests

A.M.M has received fees for advisory board membership from BMS, MSD, Novartis, Roche, Pierre Fabre and Qbiotics. R.P.M.S. has received honoraria for advisory board participation from MSD, Novartis and Qbiotics, and speaking honoraria from BMS and Novartis. A.J.S has received fees for professional services from Eli Lily Australia. G.V.L. is a consultant advisor for Agenus, Amgen, Array Biopharma, Boehringer Ingelheim International GmbH, Bristol-Myers Squibb, Evaxion Biotec, Hexal AG (Sandoz Company), Highlight Therapeutics S.L., Innovent Biologics USA, Merck Sharpe and Dohme, Novartis, OncoSec, PHMR Limited, Pierre Fabre, Provectus, Qbiotics and Regeneron. R.A.S has fees for professional services from Roche, Evaxion, Provectus Biopharmaceuticals Australia, Qbiotics, Novartis, Merck Sharp and Dohme, NeraCare, AMGEN, Bristol-Myers Squibb, Myriad Genetics and GlaxoSmithKline. All compensation was provided for work completed outside of the current work. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Medicine thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editors: Javier Carmona and Joao Monteiro, in collaboration with the Nature Medicine team.

Additional information

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

Extended data

Extended Data Fig. 1

Cohort and project schematic outlining clinical trial timeline and analyses conducted at baseline.

Extended Data Fig. 2

(a) Rarefaction analysis shows sequencing depth was sufficient to comprehensively catalogue the unique microbial strains present. (b) Composition of each Australian patient sample classified at the ‘family’ taxonomic level (n = 71). (c-e) Inverse Simpson’s index of alpha diversity for individual patients grouped by (c) maximum irAE grade experienced by each patient split by severe irAEs, (d) response/low irAE (R/G0-G2) (pink) and non-response/severe irAE (NR) (NR/G3- G5)) (orange) or (e) maximum irAE grade experienced by each patient split by severe gastrointestinal irAEs and non-gastrointestinal irAEs (NR with severe irAE ‘Adverse’ indicated in orange). (AUS, n = 7) (f & g) Tumour mutational burden (TMB) and tumour IFN-gamma signature split by response. Subset of patients Rozeman etal (2021)17 (n = 25). (h) Inverse Simpson’s index of alpha diversity for individual patients with high TMB grouped by response and non-response. (i-j) Absolute bacterial/archaeal faecal loads assessed using qPCR, with patients grouped by irAEs and ‘Adverse’ outcome groups (n = 71). (k) Correlation of diversity with 16 S rRNA gene number/mg faeces for each patient (n = 71). Each symbol represents an individual patient, bars indicate the median. Mann-Whitney U rank sum test (c-j). For linear regressions, p value was calculated on Spearman’s rank correlation (k). All statistical tests are two-sided where appropriate.

Extended Data Fig. 3

(a-f) Linear discriminant analysis (LDA) scores for differentially abundant taxa in (a) response, (c) irAE or (e) ‘Benefical/benign’ and ‘Adverse’ outcome groupings, as determined by LEfSe analysis (Australian cohort n = 71). LDA score indicates the confidence of the association, p < 0.05 for the Kruskal-Wallis H statistic, LDA score >3. (b, d & f) Dot plots show the relative abundance of sequence reads corresponding to each taxa for individual patients, where each dot represents a patient, colours correspond to the legend as indicated. (g-h) Quantitative PCR using taxa specific primers was used to determine bacterial copy number per mg faecal matter grouped according to response, irAEs and ‘Adverse’ outcome groups (n = 71). (g) Taxa associated with response or mild irAE based off LefSe analysis (h) taxa associated with non-response or severe irAE. Each symbol represents an individual patient, bars indicate the median. Mann-Whitney U rank sum test (g-h). All statistical tests are two-sided where appropriate.

Extended Data Fig. 4

(a-c) Archaeal ASV in pre-treatment faecal samples were classified using 16 S rRNA gene sequencing. Relative abundance of Archaeal ASVs (Methanobacteriaceae) from 16 S rRNA amplicon sequencing was compared by response, maximum irAE grade and ‘Beneficial/benign’ vs ‘Adverse’ outcomes (n = 71). (d) Quantitative PCR on faecal DNA using methanogen specific primers, grouped by response (left) or maximum irAE grade (right) (‘Adverse’ patients are indicated in orange). Each symbol represents an individual patient, bars indicate the median. Mann-Whitney U rank sum test (a-d). All statistical tests are two-sided where appropriate.

Extended Data Fig. 5

(a-e) Consumption of key dietary nutrients were estimated from food intake surveys. Patients were categorised as low or high according to the Australian dietary recommendations and grouped according to response. (f-i) Estimated total (f) protein (g/day), (g) fat (g/day), (h) carbohydrates (g/day) and (i) fibre (g/day) consumption from dietary surveys of food intake grouped by response (responder = R (yellow), non-responder = NR (green)) (AUS, n = 63). Each symbol represents an individual patient, bars indicate the median. Mann-Whitney U rank test (f-i). All statistical tests are two-sided where appropriate.

Extended Data Fig. 6

(a) MetaCyc pathways were predicted in the metagenomes of faecal samples of a subset of 38 Australian patients. Linear discriminant analysis (LDA) scores for differentially abundant pathway in the ‘Beneficial/benign’ (all R or NR irAE < G3) verses ‘Adverse’ (NR, irAE≥3) outcome group was determined by LEfSe analysis. LDA (log10) score on the left-hand side of the panel indicates the confidence of the association. The heat map indicates relative abundance (%) of each outcome-associated pathway in individual patients. Clinical groupings are indicated by coloured bars at the top. (b) Dot plots show the relative abundance of metabolic pathways identified by LefSe analysis as indicative of ‘Beneficial/benign’ or ‘Adverse’ outcome groups, where each dot represents a patient (subset of Australian cohort n = 38).

Extended Data Fig. 7

(a) Correlation of the relative abundance of outcome-associated butyrate pathways (metagenomic data) versus faecal butyrate concentrations, as assessed by NMR. For linear regressions, p values were calculated on Spearman’s rank correlation. (b-c) Serum butyrate levels (uM) detecting using NMR grouped according to (b) Beneficial/benign’ or ‘Adverse’ outcome or (c) response groupings (subset AUS, n = 38). Mann-Whitney U rank test. (d-g) Linear discriminant analysis (LDA) scores for differentially abundant taxa in (d) response and (f) irAE groupings (Dutch cohort, n = 32), as determined by LEfSe analysis. LDA score indicates the confidence of the association, p < 0.05 for the Kruskal-Wallis H statistic, LDA score >3. (e & g) Dot plots show the relative abundance of sequence reads corresponding to each taxa for individual patients, where each dot represents a patient. All statistical tests are two-sided where appropriate.

Extended Data Fig. 8

(a-b) Assessing clustering of samples (a) by country or (b) by DMM community type (combined AUS & NL, n = 103), by contrasting distances between samples within the same groups and between groups; distribution of distances shown. Significance assessed through the Kolmogorov-Smirnov statistic (D = effect magnitude value); p-values were corrected for multiple comparisons using the Bonferroni method. (c) Average relative abundance of Bacteroidaceae and Ruminococcaceae per community type. (d) Inverse Simpson’s index of alpha diversity for individual patients grouped by DMM community type (combined AUS & NL, n = 103). Each symbol represents an individual patient, bars indicate the median. (e-l) Relative abundance of genus level taxa by DMM community type (combined AUS & NL, n = 103). Each symbol represents an individual patient, bars indicate the median. Kruskal-Wallis with post hoc Dunn test. FDR adjusted p-values presented (d-l). All statistical tests are two-sided where appropriate.

Extended Data Fig. 9

(a) Correlation between Bacteroidaceae with Diversity in the Australian cohort (n = 71). (b-c) Correlation between (b) Bacteroidaceae or (c) Ruminococcaceae with Diversity in the Dutch cohort (n = 32). Each symbol represents an individual patient. For linear regressions, p value was calculated on Spearman’s rank correlation. (d-e) Estimated fibre consumption (g/day) from food intake surveys for (d) Australian (n = 63) and (e) Dutch patients (n = 32), grouped by DMM community type. Each symbol represents an individual patient, bars indicate the median. Mann-Whitney U rank sum test. (f-g) Baseline microbial signatures that distinguish patient outcomes (‘classes’) were determined based on microbial variables through sparse partial least squares discriminant analysis (sPLS-DA). Models were constructed from 16 S rRNA gene profiles to discriminate (f) responders (R) and non-responders (NR) to immunotherapy and (g) absent/mild (G0-G2) and severe (G3-G5) irAE development. Models were developed for the combined cohort (n = 103) and per community type: community type 1 (n = 37); community type 2 (n = 36) and community type 3 (n = 30). Classes are not equally populous; thus, plots depict the classification accuracy per class, with the percentage correctly assigned in black and the mean across classes. Colours indicate p-values derived under permutation testing. All statistical tests are two-sided where appropriate.

Extended Data Fig. 10

(a-d) Estimated fibre (g/day) from food intake surveys and Inverse Simpson diversity grouped by dominant taxa grouping (NL cohort (n = 32) & US neo-adjuvant cohort (Spencer et al n = 31). (e) Relative abundance of Faecalibacterium prausnitzii (top) or Ruminococcaceae (bottom), for patients by response (R vs NR) with in dominant taxa groups (Ba & Ru) (presented by country and combined n = 218). Symbols indicate mean, bars indicate standard error (f) Relative abundance of Ruminococcaceae, for individual patients within dominant taxa groups (Ba & Ru) by response (combined AUS/NL/US, n = 218). (g-h) Relative abundance of Ruminococcaceae (left) or Faecalibacterium prausnitzii (right) for patients by (g) maximum irAE grade (G0-G2 vs G3-G5) or (h) ‘Beneficial/benign’ (all R or NR irAE < G3) and ‘Adverse’ (NR, irAE≥ G3) outcomes (AUS/N, n = 103). (i) Inverse Simpson’s index of alpha diversity for individual patients grouped by ‘Beneficial/benign’ verses ‘Adverse’ outcome groups (n = 103, AUS circle, NL square). Each symbol represents an individual patient, bars indicate the median. Mann-Whitney U rank sum test (a-i). All statistical tests are two-sided where appropriate.

Supplementary information

Supplementary Information

Supplementary InformationSupplementary Tables 1-12. - Supplementary Table 1: Baseline patient characteristics.- Supplementary Table 2: Clinical outcomes by treatment arm combined Australia and Netherland.- Supplementary Table 3: Clinical outcomes and safety by country (Australia & Netherlands).- Supplementary Table 4- Univariate analysis for associations with pathological response.- Supplementary Table 5- Univariate analysis for associations with country.- Supplementary Table 6- Genus CLR abundance comparison of key taxa across studies.- Supplementary Table 7- Model components community type 1 response.- Supplementary Table 8- Model components community type 1 irAE.- Supplementary Table 9- Model components community type 2 response.- Supplementary Table 10- Model components community type 2irAE.- Supplementary Table 11- Model components community type 3 response.- Supplementary Table 12- Model components community type 3 irAE

Reporting Summary

Supplementary Table 1

Supplementary Table 13- 16 S sequencing metadata

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Simpson, R.C., Shanahan, E.R., Batten, M. et al. Diet-driven microbial ecology underpins associations between cancer immunotherapy outcomes and the gut microbiome. Nat Med 28, 2344–2352 (2022). https://doi.org/10.1038/s41591-022-01965-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-022-01965-2

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing