Your browser doesn't support javascript.
loading
SETD2 Loss and ATR Inhibition Synergize to Promote cGAS Signaling and Immunotherapy Response in Renal Cell Carcinoma.
Liu, Xian-De; Zhang, Yan-Ting; McGrail, Daniel J; Zhang, Xuesong; Lam, Truong; Hoang, Anh; Hasanov, Elshad; Manyam, Ganiraju; Peterson, Christine B; Zhu, Haifeng; Kumar, Shwetha V; Akbani, Rehan; Pilie, Patrick G; Tannir, Nizar M; Peng, Guang; Jonasch, Eric.
Afiliação
  • Liu XD; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Zhang YT; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • McGrail DJ; Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic, Cleveland, Ohio.
  • Zhang X; Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
  • Lam T; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Hoang A; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Hasanov E; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Manyam G; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Peterson CB; Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Zhu H; Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Kumar SV; Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Akbani R; Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Pilie PG; Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Tannir NM; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Peng G; Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Jonasch E; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
Clin Cancer Res ; 29(19): 4002-4015, 2023 10 02.
Article em En | MEDLINE | ID: mdl-37527013
ABSTRACT

PURPOSE:

Immune checkpoint blockade (ICB) demonstrates durable clinical benefits in a minority of patients with renal cell carcinoma (RCC). We aimed to identify the molecular features that determine the response and develop approaches to enhance it. EXPERIMENTAL

DESIGN:

We investigated the effects of SET domain-containing protein 2 (SETD2) loss on the DNA damage response pathway, the cytosolic DNA-sensing pathway, the tumor immune microenvironment, and the response to ataxia telangiectasia and rad3-related (ATR) and checkpoint inhibition in RCC.

RESULTS:

ATR inhibition activated the cyclic GMP-AMP synthase (cGAS)-interferon regulatory factor 3 (IRF3)-dependent cytosolic DNA-sensing pathway, resulting in the concurrent expression of inflammatory cytokines and immune checkpoints. Among the common RCC genotypes, SETD2 loss is associated with preferential ATR activation and sensitizes cells to ATR inhibition. SETD2 knockdown promoted the cytosolic DNA-sensing pathway in response to ATR inhibition. Treatment with the ATR inhibitor VE822 concurrently upregulated immune cell infiltration and immune checkpoint expression in Setd2 knockdown Renca tumors, providing a rationale for ATR inhibition plus ICB combination therapy. Setd2-deficient Renca tumors demonstrated greater vulnerability to ICB monotherapy or combination therapy with VE822 than Setd2-proficient tumors. Moreover, SETD2 mutations were associated with a higher response rate and prolonged overall survival in patients with ICB-treated RCC but not in patients with non-ICB-treated RCC.

CONCLUSIONS:

SETD2 loss and ATR inhibition synergize to promote cGAS signaling and enhance immune cell infiltration, providing a mechanistic rationale for the combination of ATR and checkpoint inhibition in patients with RCC with SETD2 mutations.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Carcinoma de Células Renais / Neoplasias Renais Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Clin Cancer Res Assunto da revista: NEOPLASIAS Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Carcinoma de Células Renais / Neoplasias Renais Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Clin Cancer Res Assunto da revista: NEOPLASIAS Ano de publicação: 2023 Tipo de documento: Article