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.

  • Viewpoint
  • Published:

Can we harness the microbiota to enhance the efficacy of cancer immunotherapy?

Abstract

There is currently much interest in defining how the microbiota shapes immune responses in the context of cancer. Various studies in both mice and humans have associated particular commensal species with better (or worse) outcomes in different cancer types and following treatment with cancer immunotherapies. However, the mechanisms involved remain ill-defined and even controversial. In this Viewpoint, Nature Reviews Immunology has invited six eminent scientists in the field to share their thoughts on the key questions and challenges for the field.

The contributors

B. Brett Finlay is a professor in the Michael Smith Laboratories at the University of British Columbia. His research interests are focused on host–microorganism interactions, at the molecular level, and he has published more than 500 articles. He is a co-author of the books Let Them Eat Dirt and The Whole-Body Microbiome.

Romina Goldszmid is an Earl Stadtman Investigator and Head of the Inflammatory Cell Dynamics Section in the Laboratory of Integrative Cancer Immunology, National Cancer Institute, National Institutes of Health (NIH). She received her PhD degree from the University of Buenos Aires, Argentina, and completed her postdoctoral training at the National Institute of Allergy and Infectious Diseases, NIH. She has a long-standing interest in understanding myeloid cell development, differentiation and function in the context of cancer and infections. Current work in her laboratory aims to dissect the myeloid cell repertoire within tumours, determine their contribution to therapy efficacy and unravel the mechanisms by which the microbiota regulates their function.

Kenya Honda is a professor at Keio University School of Medicine and a team leader at the RIKEN Center for Integrative Medical Sciences, Japan. His laboratory has been aiming to elucidate and translate the mutualistic relationship between the gut microbiota and the host immune system using gnotobiotic animal models.

Giorgio Trinchieri is an NIH Distinguished Investigator and Chief of the Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, NIH. His research has focused for many years on the interplay between inflammation, innate resistance and adaptive immunity and on the role of pro-inflammatory cytokines and interferons in the regulation of haematopoiesis, innate resistance and immunity against infections and tumours. The present focus of his laboratory is on the role of inflammation, innate resistance, immunity and the commensal microbiota in carcinogenesis, cancer progression and prevention of or therapy for cancer.

Jennifer Wargo is a professor of surgical oncology and genomic medicine at the University of Texas MD Anderson Cancer Center. She leads the Program for Innovative Microbiome and Translational Research (PRIME-TR) and is internationally recognized for her contributions to cancer research in immunotherapy and the microbiome.

Laurence Zitvogel, full professor at Paris-Saclay University, is a research director at INSERM and Scientific Director of the immuno-oncology program at Gustave Roussy, France. She has contributed to the field of cancer immunology and immunotherapy, pioneering the concepts of immunogenic cell death and gut microbiota in cancer.

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

Similar content being viewed by others

References

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  5. Pushalkar, S. et al. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 8, 403–416 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Groza, D. et al. Bacterial ghosts as adjuvant to oxaliplatin chemotherapy in colorectal carcinomatosis. OncoImmunology 7, e1424676 (2018).

    PubMed  PubMed Central  Google Scholar 

  7. Anker, J. F. et al. Multi-faceted immunomodulatory and tissue-tropic clinical bacterial isolate potentiates prostate cancer immunotherapy. Nat. Commun. 9, 1591 (2018).

    PubMed  PubMed Central  Google Scholar 

  8. Bhatt, A. P., Redinbo, M. R. & Bultman, S. J. The role of the microbiome in cancer development and therapy. CA. Cancer J. Clin. 67, 326–344 (2017).

    PubMed  PubMed Central  Google Scholar 

  9. Tanoue, T. et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature 565, 600–605 (2019).

    CAS  PubMed  Google Scholar 

  10. Smith, P. M. et al. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 341, 569–573 (2013).

    CAS  PubMed  Google Scholar 

  11. Ma, C. et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science 360, eaan5931 (2018).

    PubMed  PubMed Central  Google Scholar 

  12. Linehan, J. L. et al. Non-classical immunity controls microbiota impact on skin immunity and tissue repair. Cell 172, 784–796 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Riquelme, E. et al. Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell 178, 795–806 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  15. Gil-Cruz, C. et al. Microbiota-derived peptide mimics drive lethal inflammatory cardiomyopathy. Science 366, 881–886 (2019).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Lee, C. et al. NOD2 supports crypt survival and epithelial regeneration after radiation-induced injury. Int. J. Mol. Sci. https://doi.org/10.3390/ijms20174297 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Litvak, Y. & Bäumler, A. J. Microbiota-nourishing immunity: a guide to understanding our microbial self. Immunity 51, 214–224 (2019).

    CAS  PubMed  Google Scholar 

  19. Koyama, M. et al. MHC class II antigen presentation by the intestinal epithelium initiates graft-versus-host disease and is influenced by the microbiota. Immunity 51, 885–898 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. McDonald, D. et al. American gut: an open platform for citizen science microbiome research. mSystems https://doi.org/10.1128/mSystems.00031-18 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Vetizou, M. & Trinchieri, G. Anti-PD1 in the wonder-gut-land. Cell Res. 28, 263–264 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  23. Helmink, B. A. et al. The microbiome, cancer, and cancer therapy. Nat. Med. 25, 377–388 (2019).

    CAS  PubMed  Google Scholar 

  24. Gharaibeh, R. Z. & Jobin, C. Microbiota and cancer immunotherapy: in search of microbial signals. Gut 68, 385–388 (2018).

    PubMed  Google Scholar 

  25. Batten, M. et al. Low intestinal microbial diversity is associated with severe immune-related adverse events and lack of response to neoadjuvant combination anti-PD1, anti-CTLA4 immunotherapy. Cancer Res. https://doi.org/10.1158/1538-7445.AM2019-2822 (2019).

    Article  Google Scholar 

  26. Peled, J. U. et al. Microbiota as predictor of mortality in allogeneic hematopoietic-cell transplantation. N. Engl. J. Med. 382, 822–834 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Routy, B. et al. The gut microbiota influences anticancer immunosurveillance and general health. Nat. Rev. Clin. Oncol. 15, 382–396 (2018).

    CAS  PubMed  Google Scholar 

  28. Stevenson, A. et al. Host-microbe interactions mediating antitumorigenic effects of MRX0518, a gut microbiota-derived bacterial strain, in breast, renal and lung carcinoma. J. Clin. Oncol. 36, e15006 (2018).

    Google Scholar 

  29. Manfredo Vieira, S. et al. Translocation of a gut pathobiont drives autoimmunity in mice and humans. Science 359, 1156–1161 (2018).

    CAS  PubMed  Google Scholar 

  30. Wang, Y. et al. Fecal microbiota transplantation for refractory immune checkpoint inhibitor-associated colitis. Nat. Med. 24, 1804–1808 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Shepherd, E. S. et al. An exclusive metabolic niche enables strain engraftment in the gut microbiota. Nature 557, 434–438 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Patnode, M. L. et al. Interspecies competition impacts targeted manipulation of human gut bacteria by fiber-derived glycans. Cell 179, 59–73 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Staley, C. et al. Durable long-term bacterial engraftment following encapsulated fecal microbiota transplantation to treat clostridium difficile infection. MBio 10, e01586–e01619 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Kaiser, J. Fecal transplants could help patients on cancer immunotherapy drugs. Science https://doi.org/10.1126/science.aax5960 (2019).

    Article  PubMed  Google Scholar 

  35. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/show/NCT03341143 (2020).

  36. McQuade, J. L. et al. Modulating the microbiome to improve therapeutic response in cancer. Lancet Oncol. 20, e77–e91 (2019).

    PubMed  Google Scholar 

  37. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03353402 (2019).

  38. Baruch, E. N. et al. Abstract CT042: fecal microbiota transplantation (FMT) and re-induction of anti-PD-1 therapy in refractory metastatic melanoma patients - preliminary results from a phase I clinical trial (NCT03353402). Cancer Res. https://doi.org/10.1158/1538-7445.AM2019-CT042 (2019).

    Article  Google Scholar 

  39. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03341143 (2020).

  40. Maleki, S. Combination of fecal microbiota transplantation from healthy donors with anti-PD1 immunotherapy in treatment-naïve advanced or metastatic melanoma patients. SITC https://sitc.sitcancer.org/2019/abstracts/titles/index.php?filter=Clinical+Trial+In+Progress (2019).

  41. Derosa, L. et al. Negative association of antibiotics on clinical activity of immune checkpoint inhibitors in patients with advanced renal cell and non-small-cell lung cancer. Ann. Oncol. 29, 1437–1444 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Chalabi, M. et al. Efficacy of chemotherapy and atezolizumab in patients with non-small-cell lung cancer receiving antibiotics and proton pump inhibitors: pooled post hoc analyses of the OAK and POPLAR trials. Ann. Oncol. https://doi.org/10.1016/j.annonc.2020.01.006 (2020).

    Article  PubMed  Google Scholar 

  43. Pflug, N. et al. Efficacy of antineoplastic treatment is associated with the use of antibiotics that modulate intestinal microbiota. Oncoimmunology 5, e1150399 (2016).

    PubMed  PubMed Central  Google Scholar 

  44. Kaderbhai, C. et al. Antibiotic use does not appear to influence response to nivolumab. Anticancer Res. 37, 3195–3200 (2017).

    CAS  PubMed  Google Scholar 

  45. Shono, Y. et al. Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice. Sci. Transl. Med. 8, 339ra371 (2016).

    Google Scholar 

  46. Taur Y., et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin. Infect. Dis. 55, 905-914.

  47. Geller, L. T. et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 357, 1156–1160 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Wilson, B. E., Routy, B., Nagrial, A. & Chin, V. T. The effect of antibiotics on clinical outcomes in immune-checkpoint blockade: a systematic review and meta-analysis of observational studies. Cancer Immunol. Immunother. 69, 343–354 (2020).

    PubMed  Google Scholar 

  49. Pinato, D. J. et al. Association of prior antibiotic treatment with survival and response to immune checkpoint inhibitor therapy in patients with cancer. JAMA Oncol. 5, 1774–1778 (2019).

    PubMed  PubMed Central  Google Scholar 

  50. Huang, X.-Z. et al. Antibiotic use and the efficacy of immune checkpoint inhibitors in cancer patients: a pooled analysis of 2740 cancer patients. Oncoimmunology 8, e1665973 (2019).

    PubMed  PubMed Central  Google Scholar 

  51. Elkrief, A. et al. The negative impact of antibiotics on outcomes in cancer patients treated with immunotherapy: a new independent prognostic factor. Ann. Oncol. 30, 1572–1579 (2019).

    CAS  PubMed  Google Scholar 

  52. Hill, C. et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 11, 506–514 (2014).

    PubMed  Google Scholar 

  53. Suez, J. et al. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell 174, 1406–1423 (2018).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  55. National Center for Complementary and Integrative Health. Statistics from the National Health Interview Survey. NIH https://www.nccih.nih.gov/health/statistics-from-the-national-health-interview-survey (2017).

  56. Spencer, C. N. et al. Abstract 2838: the gut microbiome (GM) and immunotherapy response are influenced by host lifestyle factors. Cancer Res. https://doi.org/10.1158/1538-7445.AM2019-2838 (2019).

    Article  Google Scholar 

  57. Arthur, J. C. et al. VSL#3 probiotic modifies mucosal microbial composition but does not reduce colitis-associated colorectal cancer. Sci. Rep. 3, 2868 (2013).

    PubMed  PubMed Central  Google Scholar 

  58. Suez, J. et al. The pros, cons, and many unknowns of probiotics. Nat. Med. 25, 716–729 (2019).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

K.H. was funded through AMED LEAP under grant number JP18gm0010003. R.G. is supported by the Intramural Research Program of the US National Institutes of Health, National Cancer Institute. The L.Z. laboratory is supported by RHU Torino Lumière (ANR-16-RHUS-0008), the ONCOBIOME project (European Union Horizon 2020 programme), the Seerave Foundation, the French Agence Nationale de la Recherche (Ileobiome), the French Ligue Contre le Cancer (Équipe Labelisées programme), the French Association pour la Recherche sur le Cancer, Cancéropôle Ile-de-France, the French Fondation pour la Recherche Médicale, a donation by Elior and Dassault Systems, the European Research Council, Fondation Carrefour, the French Institut National du Cancer INSERM (HTE programme), LabEx Immuno-Oncology, SIRIC Stratified Oncology Cell DNA Repair and Tumour Immune Elimination (SOCRATE) and the CARE network (directed by X. Mariette, Assistance Publique — Hôpitaux de Paris, Kremlin-Bicêtre).

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to B. Brett Finlay, Romina Goldszmid, Kenya Honda, Giorgio Trinchieri, Jennifer Wargo or Laurence Zitvogel.

Ethics declarations

Competing interests

B.B.F. is a cofounding scientific advisory board member for Vedanta Biosciences. K.H. is a scientific advisory board member of Vedanta Biosciences and 4BIO CAPITAL. J.W. reports compensation and honoraria from Imedex, Dava Oncology, Omniprex, Ilumina, Gilead, PeerView, Physician Education Resource, MedImmune and Bristol-Myers Squibb. J.W. serves as a consultant advisory board member for Roche/Genentech, Novartis, AstraZeneca, GlaxoSmithKline, Bristol-Myers Squibb, Merck, Biothera Pharmaceuticals and Microbiome DX. J.W. also receives research support from GlaxoSmithKline, Roche/Genentech, Bristol-Myers Squibb and Novartis. J.W. is an adviser and has stock options for Ella Therapeutics. L.Z. is a founder of everImmune, a biotechnology company that develops anticancer probiotics and diagnostic tools to define intestinal dysbiosis in cancer. L.Z. also has an active scientific collaboration (research contract) with Kaleido, Innovate Pharma and Bioaster, which are companies involved in the microbiome space. R.G. and G.T. declare no competing interests.

Additional information

Publisher’s note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Finlay, B.B., Goldszmid, R., Honda, K. et al. Can we harness the microbiota to enhance the efficacy of cancer immunotherapy?. Nat Rev Immunol 20, 522–528 (2020). https://doi.org/10.1038/s41577-020-0374-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41577-020-0374-6

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