Your browser doesn't support javascript.
loading
Genomic Landscape of Lynch Syndrome Colorectal Neoplasia Identifies Shared Mutated Neoantigens for Immunoprevention.
Bolivar, Ana M; Duzagac, Fahriye; Deng, Nan; Reyes-Uribe, Laura; Chang, Kyle; Wu, Wenhui; Bowen, Charles M; Taggart, Melissa W; Thirumurthi, Selvi; Lynch, Patrick M; You, Y Nancy; Rodriguez-Pascual, Jesus; Lipkin, Steven M; Kopetz, Scott; Scheet, Paul; Lizee, Gregory A; Reuben, Alexandre; Sinha, Krishna M; Vilar, Eduardo.
Afiliación
  • Bolivar AM; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Duzagac F; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Deng N; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Reyes-Uribe L; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Chang K; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Wu W; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Bowen CM; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Taggart MW; Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Thirumurthi S; Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas; Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Lynch PM; Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, Texas; Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • You YN; Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Colorectal Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Rodriguez-Pascual J; Department of Medical Oncology, Grupo Vithas, Madrid, Spain.
  • Lipkin SM; Division of Gastroenterology and Hepatology, Weill Cornell University, New York, New York.
  • Kopetz S; Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Scheet P; Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Lizee GA; Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Reuben A; Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Sinha KM; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Vilar E; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas; Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Hous
Gastroenterology ; 166(5): 787-801.e11, 2024 05.
Article en En | MEDLINE | ID: mdl-38244726
ABSTRACT
BACKGROUND &

AIMS:

Lynch syndrome (LS) carriers develop mismatch repair-deficient neoplasia with high neoantigen (neoAg) rates. No detailed information on targetable neoAgs from LS precancers exists, which is crucial for vaccine development and immune-interception strategies. We report a focused somatic mutation and frameshift-neoAg landscape of microsatellite loci from colorectal polyps without malignant potential (PWOMP), precancers, and early-stage cancers in LS carriers.

METHODS:

We generated paired whole-exome and transcriptomic sequencing data from 8 colorectal PWOMP, 41 precancers, 8 advanced precancers, and 12 early-stage cancers of 43 LS carriers. A computational pipeline was developed to predict, rank, and prioritize the top 100 detected mutated neoAgs that were validated in vitro using ELISpot and tetramer assays.

RESULTS:

Mutation calling revealed >10 mut/Mb in 83% of cancers, 63% of advanced precancers, and 20% of precancers. Cancers displayed an average of 616 MHC-I neoAgs/sample, 294 in advanced precancers, and 107 in precancers. No neoAgs were detected in PWOMP. A total of 65% of our top 100 predicted neoAgs were immunogenic in vitro, and were present in 92% of cancers, 50% of advanced precancers, and 29% of precancers. We observed increased levels of naïve CD8+ and memory CD4+ T cells in mismatch repair-deficient cancers and precancers via transcriptomics analysis.

CONCLUSIONS:

Shared frameshift-neoAgs are generated within unstable microsatellite loci at initial stages of LS carcinogenesis and can induce T-cell responses, generating opportunities for vaccine development, targeting LS precancers and early-stage cancers.
Asunto(s)
Palabras clave

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Neoplasias Colorrectales Hereditarias sin Poliposis / Mutación del Sistema de Lectura / Secuenciación del Exoma / Antígenos de Neoplasias Idioma: En Revista: Gastroenterology Año: 2024 Tipo del documento: Article

Texto completo: 1 Base de datos: MEDLINE Asunto principal: Neoplasias Colorrectales Hereditarias sin Poliposis / Mutación del Sistema de Lectura / Secuenciación del Exoma / Antígenos de Neoplasias Idioma: En Revista: Gastroenterology Año: 2024 Tipo del documento: Article