Oops, you're using an old version of your browser so some of the features on this page may not be displaying properly.

MINIMAL Requirements: Google Chrome 24+Mozilla Firefox 20+Internet Explorer 11Opera 15–18Apple Safari 7SeaMonkey 2.15-2.23

Cocktail & Poster Display session

86P - Regulation of cancer progression through the gut microbiome and immuno-nutrition

Date

04 Oct 2023

Session

Cocktail & Poster Display session

Presenters

Anikka Swaby

Citation

Annals of Oncology (2023) 8 (suppl_1_S5): 1-55. 10.1016/esmoop/esmoop101646

Authors

A. Swaby1, L. Desharnais2, M.W. Yu3, S. Doré2, V. Breton4, L. Wilson5, M. Sorin2, A. Arabzadeh4, B. Fiset4, L.A. Walsh2, D.F. Quail3

Author affiliations

  • 1 Medicine, Goodman Cancer Institute, McGill University, H3A 1X1 - Montreal/CA
  • 2 Human Genetics, Goodman Cancer Institute, McGill University, H3A 1X1 - Montreal/CA
  • 3 Physiology, Goodman Cancer Institute, McGill University, H3A 1X1 - Montreal/CA
  • 4 Goodman Cancer Institute, McGill University, H3A 1X1 - Montreal/CA
  • 5 Medicine, Goodman Cancer Institute, McGill University, H3A 1A3 - Montreal/CA

Resources

This content is available to ESMO members and event participants.

Abstract 86P

Background

Obesity rivals smoking as a leading modifiable risk factor for cancer mortality, accounting for up to 20% of adult cancer-related deaths. Diet is a crucial factor in obesity development and significantly influences cancer growth. Paradoxically, high BMI has been linked to improved immune checkpoint inhibitor (ICI) efficacy in various cancers, challenging the notion that obesity is universally detrimental in cancer contexts. To address this paradox, we devised a panel of 12 diets that mimic human dietary patterns in mouse models, observing vastly different rates of cancer growth and ICI response. Surprisingly, not all obesity-inducing diets were beneficial for ICI, prompting an investigation into the cause of these disparities. The microbiome's pivotal role in regulating cancer and therapy through its profound influence on the immune system is well-supported, with gut dysbiosis and antibiotic use increasing cancer risk and blunting ICI response in patients and fecal microbial transplant from ICI-responsive patients shown to enhance ICI therapy. We propose that the interplay between diet and systemic inflammatory responses to gut microbiota contributes to the variations in ICI response across obesity-inducing diet models.

Methods

To track the gut microbial composition changes in mice during the development of our 12-diet model, we collected stool samples and performed 16s rRNA sequencing. The analyses focused on four obesity-inducing diets: American, High Fat, Ketogenic and Mediterranean.

Results

Analysis unveiled differences in gut bacterial composition at the phylum level across all four obesity inducing diets. However, family-level changes were associated with ICI response in a diet-specific manner. To delve deeper into these findings, we conducted metagenomic sequencing, revealing specific species associated with ICI response in preliminary analyses.

Conclusions

These findings suggest that diet-induced gut microbial modifications, in the context of obesity, may influence ICI efficacy, and offers promising avenues for the development of new therapeutic approaches that utilize the microbiome to enhance cancer treatment outcomes.

Editorial acknowledgement

Clinical trial identification

Legal entity responsible for the study

The authors.

Funding

Canadian Cancer Society.

Disclosure

All authors have declared no conflicts of interest.

This site uses cookies. Some of these cookies are essential, while others help us improve your experience by providing insights into how the site is being used.

For more detailed information on the cookies we use, please check our Privacy Policy.

Customise settings
  • Necessary cookies enable core functionality. The website cannot function properly without these cookies, and you can only disable them by changing your browser preferences.