RT Journal Article SR Electronic T1 FGFR2 fusion protein-driven mouse models of intrahepatic cholangiocarcinoma unveil a necessary role for Erk signaling JF bioRxiv FD Cold Spring Harbor Laboratory SP 2020.05.20.106104 DO 10.1101/2020.05.20.106104 A1 Giulia Cristinziano A1 Manuela Porru A1 Dante Lamberti A1 Simonetta Buglioni A1 Francesca Rollo A1 Carla Azzurra Amoreo A1 Isabella Manni A1 Diana Giannarelli A1 Cristina Cristofoletti A1 Giandomenico Russo A1 Mitesh J. Borad A1 Gian Luca Grazi A1 Maria Grazia Diodoro A1 Silvia Giordano A1 Mattia Forcato A1 Sergio Anastasi A1 Carlo Leonetti A1 Oreste Segatto YR 2020 UL http://biorxiv.org/content/early/2020/05/22/2020.05.20.106104.abstract AB Background and aims About 15% of intrahepatic cholangiocarcinoma (iCCA) express fibroblast growth factor receptor 2 (FGFR2) fusion proteins (FFs), most often in concert with mutationally inactivated TP53, CDKN2A or BAP1. FFs span residues 1-768 of FGFR2 fused to sequences encoded by any of a long list (>60) of partner genes, a configuration sufficient to ignite oncogenic FF activation. In line, FGFR-specific tyrosine kinase inhibitors (F-TKI) were shown to provide clinical benefit in FF+ iCCA, although responses were partial and/or limited by resistance mechanisms, including the FF V565F gatekeeper mutation. Herein we present an FF-driven murine iCCA model and exploit its potential for pre-clinical studies on FF therapeutic targeting.Methods Four iCCA FFs carrying different fusion sequences were expressed in Tp53-/- mouse liver organoids. Tumorigenic properties of genetically modified liver organoids were assessed by intrahepatic/subcutaneous transplantation in immuno-deficient mice. Cellular models derived from neoplastic lesions were exploited for pre-clinical studies.Results Tumors diagnosed as CCA were obtained upon transplantation of FF-expressing liver organoids. The penetrance of this tumorigenic phenotype was influenced by FF identity. Tumor organoids and 2D cell lines derived from CCA lesions were addicted to FF signaling via Ras-Erk, regardless of FF identity or presence of V565F mutation. Double blockade of FF-Ras-Erk pathway by concomitant pharmacological inhibition of FFs and Mek1/2 provided greater therapeutic efficacy than single agent F-TKI in vitro and in vivo.Conclusions FF-driven iCCA pathogenesis was successfully modelled in murine Tp53-/- background. This model revealed biological heterogeneity among structurally different FFs. Double blockade of FF-Erk signaling deserves consideration for improving precision-based approaches against human FF+ iCCA.Competing Interest StatementM.J.B. disclosures: ADC Therapeutics:Consulting to self; Exelixis Pharmaceuticals: Consulting to self; Inspyr Therapeutics: Consulting to self; G1 Therapeutics: Consulting to self; Immunovative Therapies: Consulting to self; OncBioMune Pharmaceuticals: Consulting to self; Western Oncolytics: Consulting to self; Lynx Group: Consulting to self; Genentech: Consulting to self; Merck: Consulting to self; Huya: Consulting to self; Astra Zeneca: Travel Support to self. All other Authors declare that they have no competing interest.ANOVAanalysis of variance;Bap1BRCA1-Associated-Protein 1;Cdkn2acyclin-dependent kinase inhibitor 2A;Cftrcystic fibrosis transmembrane conductance regulator;Ck19cytokeratin 19;Cyp3Acytochrome P450, family 3, subfamily A;EGFEpidermal growth factor;EGFREpidermal growth factor receptor;EpCAMepithelial cell adhesion molecule;Erkextracellular signal–regulated kinase;FGFR2fibroblast growth factor receptor 2;FRS2fibroblast growth factor receptor substrate 2;GRB2growth factor receptor-bound 2;GSEAgene set enrichment analysis;GSVAgene set variation analysis;H&Ehematoxylin and eosin;HepPar1hepatocyte Paraffin 1;Hnf4α-7hepatocyte nuclear factor 4 alpha;Hprthypoxanthine-guanine phosphoribosyl transferase;LGR5leucine-rich repeat-containing G-protein coupled receptor 5;NTRKneurotrophic Tyrosine Kinase;ParpPoly (ADP-ribose) polymerase;RECISTresponse evaluation criteria in solid tumors;SHP2Src homology phosphatase 2;Ttrtransthyretin.