Your browser doesn't support javascript.
loading
CD73-Dependent Adenosine Signaling through Adora2b Drives Immunosuppression in Ductal Pancreatic Cancer.
Faraoni, Erika Y; Singh, Kanchan; Chandra, Vidhi; Le Roux, Olivereen; Dai, Yulin; Sahin, Ismet; O'Brien, Baylee J; Strickland, Lincoln N; Li, Le; Vucic, Emily; Warner, Amanda N; Pruski, Melissa; Clark, Trent; Van Buren, George; Thosani, Nirav C; Bynon, John S; Wray, Curtis J; Bar-Sagi, Dafna; Poulsen, Kyle L; Vornik, Lana A; Savage, Michelle I; Sei, Shizuko; Mohammed, Altaf; Zhao, Zhongming; Brown, Powel H; Mills, Tingting; Eltzschig, Holger K; McAllister, Florencia; Bailey-Lundberg, Jennifer M.
Afiliación
  • Faraoni EY; Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Singh K; Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Chandra V; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Le Roux O; The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, and The University of Texas Health Science Center at Houston, Houston, Texas.
  • Dai Y; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Sahin I; Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas.
  • O'Brien BJ; Department of Engineering, Texas Southern University, Houston, Texas.
  • Strickland LN; Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Li L; Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Vucic E; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Warner AN; Departments of Biochemistry and Molecular Pharmacology and Medicine, NYU Langone School of Medicine, New York, New York.
  • Pruski M; Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Clark T; The Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, and The University of Texas Health Science Center at Houston, Houston, Texas.
  • Van Buren G; Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Thosani NC; Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Bynon JS; Division of Surgical Oncology, Baylor College of Medicine, Houston, Texas.
  • Wray CJ; Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Bar-Sagi D; Center for Interventional Gastroenterology at UTHealth (iGUT), McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Poulsen KL; Department of Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Vornik LA; Department of Surgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Savage MI; Department of Engineering, Texas Southern University, Houston, Texas.
  • Sei S; Department of Anesthesiology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Mohammed A; Center for Perioperative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Zhao Z; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Brown PH; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
  • Mills T; Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland.
  • Eltzschig HK; Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland.
  • McAllister F; Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas.
  • Bailey-Lundberg JM; Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas.
Cancer Res ; 83(7): 1111-1127, 2023 04 04.
Article en En | MEDLINE | ID: mdl-36720042
ABSTRACT
The microenvironment that surrounds pancreatic ductal adenocarcinoma (PDAC) is profoundly desmoplastic and immunosuppressive. Understanding triggers of immunosuppression during the process of pancreatic tumorigenesis would aid in establishing targets for effective prevention and therapy. Here, we interrogated differential molecular mechanisms dependent on cell of origin and subtype that promote immunosuppression during PDAC initiation and in established tumors. Transcriptomic analysis of cell-of-origin-dependent epithelial gene signatures revealed that Nt5e/CD73, a cell-surface enzyme required for extracellular adenosine generation, is one of the top 10% of genes overexpressed in murine tumors arising from the ductal pancreatic epithelium as opposed to those rising from acinar cells. These findings were confirmed by IHC and high-performance liquid chromatography. Analysis in human PDAC subtypes indicated that high Nt5e in murine ductal PDAC models overlaps with high NT5E in human PDAC squamous and basal subtypes, considered to have the highest immunosuppression and worst prognosis. Multiplex immunofluorescent analysis showed that activated CD8+ T cells in the PDAC tumor microenvironment express high levels of CD73, indicating an opportunity for immunotherapeutic targeting. Delivery of CD73 small-molecule inhibitors through various delivery routes reduced tumor development and growth in genetically engineered and syngeneic mouse models. In addition, the adenosine receptor Adora2b was a determinant of adenosine-mediated immunosuppression in PDAC. These findings highlight a molecular trigger of the immunosuppressive PDAC microenvironment elevated in the ductal cell of origin, linking biology with subtype classification, critical components for PDAC immunoprevention and personalized approaches for immunotherapeutic intervention.

SIGNIFICANCE:

Ductal-derived pancreatic tumors have elevated epithelial and CD8+GZM+ T-cell CD73 expression that confers sensitivity to small-molecule inhibition of CD73 or Adora2b to promote CD8+ T-cell-mediated tumor regression. See related commentary by DelGiorno, p. 977.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Neoplasias Pancreáticas / Vacunas contra el Cáncer / Carcinoma Ductal Pancreático Tipo de estudio: Prognostic_studies Límite: Animals / Humans Idioma: En Revista: Cancer Res Año: 2023 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Neoplasias Pancreáticas / Vacunas contra el Cáncer / Carcinoma Ductal Pancreático Tipo de estudio: Prognostic_studies Límite: Animals / Humans Idioma: En Revista: Cancer Res Año: 2023 Tipo del documento: Article
...