Elsevier

The Lancet Oncology

Volume 20, Issue 2, February 2019, Pages e77-e91
The Lancet Oncology

Review
Modulating the microbiome to improve therapeutic response in cancer

https://doi.org/10.1016/S1470-2045(18)30952-5Get rights and content

Summary

Although novel therapies, including immunotherapy, have dramatically improved outcomes for many patients with cancer, overall outcomes are heterogeneous and existing biomarkers do not reliably predict response. To date, predictors of response to cancer therapy have largely focused on tumour-intrinsic features; however, there is growing evidence that other host factors (eg, host genomics and the microbiome) can substantially affect therapeutic response. The microbiome, which refers to microbiota within a host and their collective genomes, is becoming increasingly recognised for its influence on host immunity, as well as therapeutic responses to cancer treatment. Importantly, microbiota can be modified via several different strategies, affording new angles in cancer treatment to improve outcomes. In this Review, we examine the evidence on the role of the microbiome in cancer and therapeutic response, factors that influence and shape host microbiota, strategies to modulate the microbiome, and present key unanswered questions to be addressed in ongoing and future research.

Introduction

The development of novel therapies such as immune checkpoint inhibitors has resulted in dramatic improvements in the outcomes of many patients with cancer. However, outcomes are heterogeneous, with some patients achieving dramatic durable complete remissions, and others deriving no benefit at all. Beyond tumour-intrinsic features that might predict response and drive resistance, there is increasing evidence that host (ie, patient) factors, including the microbiota, might influence response to therapy.1, 2, 3, 4, 5, 6

The human microbiota is comprised of complex communities of trillions of microbes that live on and inside humans. These commensal microorganisms have co-evolved with humans to have several functions that benefit human health, including harvesting otherwise inaccessible nutrients from the diet, maintaining integrity of mucosal barriers, and contributing to immune system development and homoeostasis.7, 8

Our understanding of the microbiome has grown exponentially in the past decade with the development of high-throughput sequencing approaches.7, 8 The most common component of the human microbiome that is sequenced is the small 30S ribosome subunit, which is unique to prokaryotes and has regions that vary greatly between different species of bacteria (16S sequencing). This technique can be used to quantify alpha diversity (the number of distinct species present and whether distinct species are evenly represented) and beta diversity (differences in taxonomic abundance profiles between different samples), as well as differential abundance of specific bacterial taxa. By contrast, whole-genome or metagenomic sequencing involves sequencing the entire genomes of all microbes (including viruses, fungi, protozoa, archaea) in a given sample. Metagenomic sequencing has the added advantage of deeper resolution and allows for imputation of function, but at a substantially higher cost in terms of both time and money; however, as with all omics-based profiling, the cost is decreasing and resolution increasing. As such, some components of the human microbiota, such as viruses and fungi, as well as archaea, protozoa, and other microbes, have been less well studied to date than bacteria. Characterisation of these other components of the human microbiota is an area of deep investigation and it is likely that a growing role for these non-bacterial counterparts will be uncovered in the near future. Nonetheless, complexities exist with the characterisation of such components given the vast diversity of virotypes (and viral genomes) that are present in the microbiome, among other variables. Notably, the composition of these other microbial components might directly or indirectly affect the composition of the gut bacterial components; thus, as the field moves forward, these potential interactions must be taken into consideration.

Although novel sequencing techniques have added substantially to our understanding of the human microbiome, we cannot fully understand function and mechanism from computational analysis alone. There has been renewed interest in the field of culturomics—a high-throughput method of culturing microbial species that were otherwise previously deemed difficult or impossible to culture. Culturing bacteria in this way will enable us to study the bacteria themselves rather than their genomes only and thus will help elucidate certain mechanisms of the microbiome in a way that using computational methods alone will not.9, 10

As our understanding of the microbiota grows, it is becoming increasing clear that the microbiota plays a key role in human health and disease. Disruption of the gut microbiome (dysbiosis) has been implicated in a range of human diseases, including gastrointestinal, autoimmune, neurological, and metabolic diseases.8 For cancer, specific bacterial and viral infections have been implicated in carcinogenesis11, 12, 13, 14, 15, 16 and have also been associated with treatment-related toxicity to cancer therapy.3, 17, 18, 19 Importantly, microbiota (specifically within the gut) have been shown to affect immune responses, with studies reporting strong associations between gut microbiota and response to immune checkpoint blockade and other therapies in human cohorts and murine models.1, 2, 4, 5, 20, 21, 22, 23 There is also evidence from preclinical models that successful modulation of the gut microbiota can enhance therapeutic response.

Accordingly, strategies to modulate the microbiome are being used and developed for various human diseases, including cancer. Such strategies include the use of faecal microbiota transplant, which is a safe and effective approved therapy for recurrent Clostridium difficile,24 and is being used experimentally to treat inflammatory bowel disease,25 metabolic diseases,26 and even cancer (table 1). Additional strategies to manipulate the microbiome are also under investigation (including probiotic administration and dietary intervention) in multiple diseases, although vast heterogeneity in study design presents a challenge in interpreting the success of these approaches.

In this Review, we assess the evidence for the role of the microbiota in the therapeutic response of cancer, outline the determinants of the microbiota and potential strategies and considerations for microbiota modulation, as well as highlight the complexities with this approach, and a potential path forward for cancer treatment.

Section snippets

The role of microbiota in carcinogenesis

Microbiota have long been implicated in tumour development, with bacterial and viral infections affecting multiple cellular processes (such as metabolism and immune function), with the potential to contribute to carcinogenesis (figure 1). There is certainly evidence for this involvement in the case of luminal gastrointestinal system malignancies, in which bacteria have been shown to contribute to the development of gastric (Helicobacter pylori)11 and colorectal cancers (Fusobacterium nucleatum).

Microbiome as a biomarker in cancer therapy

Increasing evidence from preclinical models and human cohorts has shown that the diversity and composition of gut microbiota are associated with the therapeutic success of different forms of cancer therapy.1, 2, 3, 4, 20, 21, 22, 71, 77, 78 Along with providing evidence to support therapeutic targeting of the microbiome, these data also substantiate the potential use of gut microbiota as a biomarker of response to cancer therapy. Importantly, this evidence should be considered alongside other

Faecal microbiota transplant

Although faecal microbiota transplant is just beginning to be investigated in the context of cancer, this therapy has been extensively studied in dysbiotic gastrointestinal diseases, specifically Clostridium difficile infection24 and inflammatory bowel disease.25 Knowledge gained from faecal microbiota transplant in these diseases might inform trial design in the treatment of cancer.

With faecal microbiota transplant, an entire enteral microbial ecosystem is transplanted from the donor or

Conclusion

There is compelling evidence that the microbiota affects immunity and therapeutic response in cancer, and that manipulation of microbiota can augment response to immunotherapy in preclinical models. Experience in other diseases, including C difficile colitis, has shown that the microbiome can indeed be successfully modulated to alter pathophysiology and benefit patients, and clinical trials targeting this approach for patients with cancer are in development or underway. However, we must

Search strategy and selection criteria

References for this Review were identified through searches of PubMed using the search terms “microbiota”, “microbiome”, “cancer”, “immunotherapy”, “chemotherapy”, “radiotherapy”, “stem-cell transplant”, “fecal microbiota transplantation (FMT)”, “probiotics”, “prebiotics”, and “antibiotics”. No date limits were applied. Articles were also identified through searches of the authors’ own files. Only papers published in English were reviewed. The final reference list was generated on the basis of

References (116)

  • M Demers et al.

    A randomized double-blind controlled trial: impact of probiotics on diarrhea in patients treated with pelvic radiation

    Clin Nutr

    (2014)
  • A Wahlstrom et al.

    Intestinal crosstalk between bile acids and microbiota and its impact on host metabolism

    Cell Metab

    (2016)
  • T Rieckmann et al.

    HNSCC cell lines positive for HPV and p16 possess higher cellular radiosensitivity due to an impaired DSB repair capacity

    Radiother Oncol

    (2013)
  • BA Helmink et al.

    Immune checkpoint blockade across the cancer care continuum

    Immunity

    (2018)
  • V Gopalakrishnan et al.

    The influence of the gut microbiome on cancer, immunity, and cancer immunotherapy

    Cancer Cell

    (2018)
  • JZ Oh et al.

    TLR5-mediated sensing of gut microbiota is necessary for antibody responses to seasonal influenza vaccination

    Immunity

    (2014)
  • L Derosa 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

    (2018)
  • A Staffas et al.

    The intestinal microbiota in allogeneic hematopoietic cell transplant and graft-versus-host disease

    Blood

    (2017)
  • LE Colbert et al.

    Rectal microbiome diversity predicts disease response at completion of radiation therapy for squamous cell carcinoma of the cervix

    Int J Radiat Oncol Biol Phys

    (2017)
  • J Ou et al.

    Diet, microbiota, and microbial metabolites in colon cancer risk in rural Africans and African Americans

    Am J Clin Nutr

    (2013)
  • B Routy et al.

    Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors

    Science

    (2018)
  • V Gopalakrishnan et al.

    Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients

    Science

    (2018)
  • V Matson et al.

    The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients

    Science

    (2018)
  • Structure, function and diversity of the healthy human microbiome

    Nature

    (2012)
  • I Cho et al.

    The Human Microbiome: at the interface of health and disease

    Nature Rev Genet

    (2012)
  • J-C Lagier et al.

    Culturing the human microbiota and culturomics

    Nature Rev Microbiol

    (2018)
  • RM Peek et al.

    Helicobacter pylori and gastrointestinal tract adenocarcinomas

    Nat Rev Cancer

    (2002)
  • K Mima et al.

    Fusobacterium nucleatum and T Cells in Colorectal Carcinoma

    JAMA Oncol

    (2015)
  • S Pushalkar et al.

    The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression

    Cancer Discov

    (2018)
  • PS Moore et al.

    Why do viruses cause cancer? Highlights of the first century of human tumour virology

    Nat Rev Cancer

    (2010)
  • K Dubin et al.

    Intestinal microbiome analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis

    Nat Commun

    (2016)
  • M Vetizou et al.

    Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota

    Science

    (2015)
  • A Sivan et al.

    Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy

    Science

    (2015)
  • S Viaud et al.

    The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide

    Science

    (2013)
  • N Iida et al.

    Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment

    Science

    (2013)
  • E van Nood et al.

    Duodenal infusion of feces for recurrent Clostridium difficile

    N Engl J Med

    (2013)
  • P Garcia-Peris et al.

    Effect of a mixture of inulin and fructo-oligosaccharide on Lactobacillus and Bifidobacterium intestinal microbiota of patients receiving radiotherapy: a randomised, double-blind, placebo-controlled trial

    Nutri Hosp

    (2012)
  • L Gianotti et al.

    A randomized double-blind trial on perioperative administration of probiotics in colorectal cancer patients

    World J Gastroenterol

    (2010)
  • AA Hibberd et al.

    Intestinal microbiota is altered in patients with colon cancer and modified by probiotic intervention

    BMJ Open Gastroenterol

    (2017)
  • AM Sheflin et al.

    Dietary supplementation with rice bran or navy bean alters gut bacterial metabolism in colorectal cancer survivors

    Mol Nutr Food Res

    (2017)
  • L Liu et al.

    Diets that promote colon inflammation associate with risk of colorectal carcinomas that contain Fusobacterium nucleatum

    Clin Gastroenterol Hepatol

    (2018)
  • RS Mehta et al.

    Association of dietary patterns with risk of colorectal cancer subtypes classified by Fusobacterium nucleatum in tumor tissue

    JAMA Oncol

    (2017)
  • K Mima et al.

    Fusobacterium nucleatum in colorectal carcinoma tissue according to tumor location

    Clin Transl Gastroenterol

    (2016)
  • K Mima et al.

    Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis

    Gut

    (2016)
  • N Zmora et al.

    Personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features

    Cell

    (2018)
  • AP Bhatt et al.

    The role of the microbiome in cancer development and therapy

    CA Cancer J Clin

    (2017)
  • P Louis et al.

    The gut microbiota, bacterial metabolites and colorectal cancer

    Nat Rev Microbiol

    (2014)
  • DR Donohoe et al.

    A gnotobiotic mouse model demonstrates that dietary fiber protects against colorectal tumorigenesis in a microbiota- and butyrate-dependent manner

    Cancer Discov

    (2014)
  • SJ O'Keefe et al.

    Fat, fibre and cancer risk in African Americans and rural Africans

    Nat Commun

    (2015)
  • C Ma et al.

    Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells

    Science

    (2018)
  • Cited by (251)

    View all citing articles on Scopus
    View full text