Your browser doesn't support javascript.
loading
Trapping all ERBB ligands decreases pancreatic lesions in a murine model of pancreatic ductal adenocarcinoma.
Hedegger, Kathrin; Blutke, Andreas; Hommel, Theresa; Auer, Kerstin E; Nataraj, Nishanth B; Lindzen, Moshit; Yarden, Yosef; Dahlhoff, Maik.
Affiliation
  • Hedegger K; Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU München, Germany.
  • Blutke A; Institute of Veterinary Pathology, Center for Clinical Veterinary Medicine, LMU München, Germany.
  • Hommel T; Institute of in vivo and in vitro Models, University of Veterinary Medicine, Vienna, Austria.
  • Auer KE; Institute of in vivo and in vitro Models, University of Veterinary Medicine, Vienna, Austria.
  • Nataraj NB; Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
  • Lindzen M; Bugworks Research Inc, CCAMP, Bengaluru, India.
  • Yarden Y; Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
  • Dahlhoff M; Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
Mol Oncol ; 17(11): 2415-2431, 2023 Nov.
Article in En | MEDLINE | ID: mdl-37341059
ABSTRACT
Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest of cancers. Attempts to develop targeted therapies still need to be established. Some oncogenic mechanisms in PDAC carcinogenesis harness the EGFR/ERBB receptor family. To explore the effects on pancreatic lesions, we attempted simultaneous blockade of all ERBB ligands in a PDAC mouse model. To this end, we engineered a molecular decoy, TRAP-FC , comprising the ligand-binding domains of both EGFR and ERBB4 and able to trap all ERBB ligands. Next, we generated a transgenic mouse model (CBATRAP/0 ) expressing TRAP-FC ubiquitously under the control of the chicken-beta-actin promoter and crossed these mice with KRASG12D/+ mice (Kras) to generate Trap/Kras mice. The resulting mice displayed decreased emergence of spontaneous pancreatic lesion areas and exhibited reduced RAS activity and decreased activities of ERBBs, with the exception of ERBB4, which showed increased activity. To identify the involved receptor(s), we employed CRISPR/Cas9 DNA editing to singly delete each ERBB receptor in the human pancreatic carcinoma cell line Panc-1. Ablation of each ERBB family member, especially the loss of EGFR or ERBB2/HER2, altered signaling downstream of the other three ERBB receptors and decreased cell proliferation, migration, and tumor growth. We conclude that simultaneously blocking the entire ERBB receptor family is therapeutically more effective than individually inhibiting only one receptor or ligand in terms of reducing pancreatic tumor burden. In summary, trapping all ERBB ligands can reduce pancreatic lesion area and RAS activity in a murine model of pancreatic adenocarcinoma; hence, it might represent a promising approach to treat PDAC in patients.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Pancreatic Neoplasms / Adenocarcinoma / Carcinoma, Pancreatic Ductal Limits: Animals / Humans Language: En Journal: Mol Oncol Journal subject: BIOLOGIA MOLECULAR / NEOPLASIAS Year: 2023 Document type: Article Affiliation country: Germany

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Pancreatic Neoplasms / Adenocarcinoma / Carcinoma, Pancreatic Ductal Limits: Animals / Humans Language: En Journal: Mol Oncol Journal subject: BIOLOGIA MOLECULAR / NEOPLASIAS Year: 2023 Document type: Article Affiliation country: Germany
...