Identification and Validation of Plasma Metabolomic Signatures in Precancerous Gastric Lesions That Progress to Cancer

JAMA Netw Open. 2021 Jun 1;4(6):e2114186. doi: 10.1001/jamanetworkopen.2021.14186.

Abstract

Importance: Metabolic deregulation plays an important role in gastric cancer (GC) development. To date, no studies have comprehensively explored the metabolomic profiles along the cascade of gastric lesions toward GC.

Objective: To draw a metabolic landscape and define metabolomic signatures associated with the progression of gastric lesions and risk of early GC.

Design, setting, and participants: A 2-stage, population-based cohort study was initiated in 2017 in Linqu County, Shandong Province, China, a high-risk area for GC. Prospective follow-up was conducted during the validation stage (June 20, 2017, to May 27, 2020). A total of 400 individuals were included based on the National Upper Gastrointestinal Cancer Early Detection Program in China. The discovery stage involved 200 individuals with different gastric lesions or GC (high-grade intraepithelial neoplasia or invasive GC). The validation stage prospectively enrolled 152 individuals with gastric lesions who were followed up for 118 to 1063 days and 48 individuals with GC.

Exposures: Metabolomic profiles and metabolite signatures were examined based on untargeted plasma metabolomics assay.

Main outcomes and measures: The risk of GC overall and early GC (high-grade intraepithelial neoplasia), and progression of gastric lesions.

Results: Of the 400 participants, 124 of 200 (62.0%) in the discovery set were men; mean (SD) age was 56.8 (7.5) years. In the validation set, 136 of 200 (68.0%) were men; mean (SD) age was 57.5 (8.1) years. Distinct metabolomic profiles were noted for gastric lesions and GC. Six metabolites, including α-linolenic acid, linoleic acid, palmitic acid, arachidonic acid, sn-1 lysophosphatidylcholine (LysoPC)(18:3), and sn-2 LysoPC(20:3) were significantly inversely associated with risk of GC overall and early GC (high-grade intraepithelial neoplasia). Among these metabolites, the first 3 were significantly inversely associated with gastric lesion progression, especially for the progression of intestinal metaplasia (α-linolenic acid: OR, 0.42; 95% CI, 0.18-0.98; linoleic acid: OR, 0.43; 95% CI, 0.19-1.00; and palmitic acid: OR, 0.32; 95% CI, 0.13-0.78). Compared with models including only age, sex, Helicobacter pylori infection, and gastric histopathologic findings, integrating these metabolites significantly improved the performance for predicting the progression of gastric lesions (area under the curve [AUC], 0.86; 95% CI, 0.70-1.00 vs AUC, 0.69; 95% CI, 0.50-0.88; P = .02) and risk of early GC (AUC, 0.83; 95% CI, 0.58-1.00 vs AUC, 0.61; 95% CI, 0.31-0.91; P = .03).

Conclusions and relevance: This study defined metabolite signatures that might serve as meaningful biomarkers for assessing high-risk populations and early diagnosis of GC, possibly advancing targeted GC prevention and control.

Publication types

  • Research Support, Non-U.S. Gov't

MeSH terms

  • Aged
  • China
  • Cohort Studies
  • Female
  • Helicobacter Infections / diagnosis
  • Helicobacter Infections / genetics
  • Helicobacter Infections / metabolism
  • Helicobacter pylori / drug effects
  • Helicobacter pylori / pathogenicity
  • Humans
  • Male
  • Metabolomics / methods*
  • Metabolomics / statistics & numerical data
  • Middle Aged
  • Precancerous Conditions / diagnosis*
  • Precancerous Conditions / genetics
  • Precancerous Conditions / metabolism
  • Prospective Studies
  • Stomach Neoplasms / genetics
  • Stomach Neoplasms / metabolism*