Your browser doesn't support javascript.
loading
Tumor-derived IFN triggers chronic pathway agonism and sensitivity to ADAR loss.
Liu, Huayang; Golji, Javad; Brodeur, Lauren K; Chung, Franklin S; Chen, Julie T; deBeaumont, Rosalie S; Bullock, Caroline P; Jones, Michael D; Kerr, Grainne; Li, Li; Rakiec, Daniel P; Schlabach, Michael R; Sovath, Sosathya; Growney, Joseph D; Pagliarini, Raymond A; Ruddy, David A; MacIsaac, Kenzie D; Korn, Joshua M; McDonald, E Robert.
Afiliación
  • Liu H; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Golji J; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Brodeur LK; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Chung FS; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Chen JT; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • deBeaumont RS; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Bullock CP; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Jones MD; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Kerr G; Novartis Institutes for Biomedical Research, Oncology Disease Area, Basel, Switzerland.
  • Li L; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Rakiec DP; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Schlabach MR; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Sovath S; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Growney JD; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Pagliarini RA; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Ruddy DA; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • MacIsaac KD; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • Korn JM; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA.
  • McDonald ER; Novartis Institutes for Biomedical Research, Oncology Disease Area, Cambridge, MA, USA. rob.mcdonald@novartis.com.
Nat Med ; 25(1): 95-102, 2019 01.
Article en En | MEDLINE | ID: mdl-30559422
ABSTRACT
Interferons (IFNs) are cytokines that play a critical role in limiting infectious and malignant diseases 1-4 . Emerging data suggest that the strength and duration of IFN signaling can differentially impact cancer therapies, including immune checkpoint blockade 5-7 . Here, we characterize the output of IFN signaling, specifically IFN-stimulated gene (ISG) signatures, in primary tumors from The Cancer Genome Atlas. While immune infiltration correlates with the ISG signature in some primary tumors, the existence of ISG signature-positive tumors without evident infiltration of IFN-producing immune cells suggests that cancer cells per se can be a source of IFN production. Consistent with this hypothesis, analysis of patient-derived tumor xenografts propagated in immune-deficient mice shows evidence of ISG-positive tumors that correlates with expression of human type I and III IFNs derived from the cancer cells. Mechanistic studies using cell line models from the Cancer Cell Line Encyclopedia that harbor ISG signatures demonstrate that this is a by-product of a STING-dependent pathway resulting in chronic tumor-derived IFN production. This imposes a transcriptional state on the tumor, poising it to respond to the aberrant accumulation of double-stranded RNA (dsRNA) due to increased sensor levels (MDA5, RIG-I and PKR). By interrogating our functional short-hairpin RNA screen dataset across 398 cancer cell lines, we show that this ISG transcriptional state creates a novel genetic vulnerability. ISG signature-positive cancer cells are sensitive to the loss of ADAR, a dsRNA-editing enzyme that is also an ISG. A genome-wide CRISPR genetic suppressor screen reveals that the entire type I IFN pathway and the dsRNA-activated kinase, PKR, are required for the lethality induced by ADAR depletion. Therefore, tumor-derived IFN resulting in chronic signaling creates a cellular state primed to respond to dsRNA accumulation, rendering ISG-positive tumors susceptible to ADAR loss.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Adenosina Desaminasa / Interferones / Proteínas de Unión al ARN Tipo de estudio: Diagnostic_studies Límite: Animals / Humans Idioma: En Revista: Nat Med Asunto de la revista: BIOLOGIA MOLECULAR / MEDICINA Año: 2019 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Colección: 01-internacional Banco de datos: MEDLINE Asunto principal: Adenosina Desaminasa / Interferones / Proteínas de Unión al ARN Tipo de estudio: Diagnostic_studies Límite: Animals / Humans Idioma: En Revista: Nat Med Asunto de la revista: BIOLOGIA MOLECULAR / MEDICINA Año: 2019 Tipo del documento: Article País de afiliación: Estados Unidos