Metabolic dysfunction and early-onset colorectal cancer - how macrophages build the bridge

Cancer Med. 2020 Sep;9(18):6679-6693. doi: 10.1002/cam4.3315. Epub 2020 Jul 23.

Abstract

Background: The incidence of colorectal cancer (CRC) among patients <50 years of age has increased dramatically over the last decades. At the same time, the growing proportion of obese children and adolescents and the increasing proportion of young and obese patients with CRC suggests an association between metabolic dysfunction and carcinogenesis. Tumor-associated macrophages (TAMs) are able to orchestrate tumor promoting and suppressing mechanisms in CRC. The aim of this review was to discuss the different roles of TAMs in CRC and their phenotype-specific metabolic pathways to identify potential new targets for CRC treatment.

Methods: A literature search was performed using PubMed, Cochrane and Embase to identify studies on TAMs and their metabolism in CRC. The following search terms were used in various combinations: (obesity OR adiposity OR obese) AND (macrophage OR polarization OR macrophage metabolism) AND ((colon cancer*) OR (colon carcinoma) OR (colonic tumor*) OR (colonic neoplasm[MeSH]) OR (rectal cancer*) OR (rectal carcinoma) OR (rectal tumor*) OR (rectal neoplasm[MeSH]) OR (colorectal cancer*) OR (colorectal carcinoma) OR (colorectal tumor*) OR (colorectal neoplasm[MeSH])). Studies including data on the phenotype and metabolism of TAMs in CRC were analyzed.

Results: Evidence for the prognostic utility of macrophage markers in CRC is currently evolving, with a particular role of stage-dependent cellular metabolism profiles of TAMs. Itaconate is one of the metabolites produced by proinflammatory subtypes of TAMs and it is known to have tumor promoting effects. Metabolic pathways that are involved in macrophage activation and reprogramming play a role in a chronic inflammatory setting, consequently affecting the onset and development of CRC.

Conclusions: Tumor-promoting metabolites, such as itaconate, are directly regulating these mechanisms, thereby triggering carcinogenesis. Metabolic reprogramming in TAMs can build a bridge between metabolic dysfunction and the onset and progression of CRC through inflammatory pathways, particularly in younger patients with early-onset CRC.

Keywords: adiposity; colonic neoplasms; colorectal neoplasms; obesity; rectal neoplasms; tumor‐associated macrophages.

Publication types

  • Research Support, Non-U.S. Gov't
  • Review

MeSH terms

  • Adiposity
  • Adolescent
  • Adult
  • Cell Polarity
  • Cellular Reprogramming
  • Child
  • Colonic Neoplasms / diagnosis
  • Colonic Neoplasms / etiology*
  • Humans
  • Inflammation / immunology
  • Macrophage Activation
  • Middle Aged
  • Obesity / epidemiology
  • Pediatric Obesity / epidemiology
  • Phenotype
  • Prognosis
  • Rectal Neoplasms / diagnosis
  • Rectal Neoplasms / etiology*
  • Succinates / metabolism
  • Tumor-Associated Macrophages / immunology
  • Tumor-Associated Macrophages / metabolism*
  • Young Adult

Substances

  • Succinates
  • itaconic acid