Fibroblast growth factor (FGF) signaling plays critical roles in key biological processes ranging from embryogenesis to wound healing and has strong links to several hallmarks of cancer. Genetic alterations in FGF receptor (FGFR) family members are associated with increased tumor growth, metastasis, angiogenesis, and decreased survival. JNJ-42756493, erdafitinib, is an orally active small molecule with potent tyrosine kinase inhibitory activity against all four FGFR family members and selectivity versus other highly related kinases. JNJ-42756493 shows rapid uptake into the lysosomal compartment of cells in culture, which is associated with prolonged inhibition of FGFR signaling, possibly due to sustained release of the inhibitor. In xenografts from human tumor cell lines or patient-derived tumor tissue with activating FGFR alterations, JNJ-42756493 administration results in potent and dose-dependent antitumor activity accompanied by pharmacodynamic modulation of phospho-FGFR and phospho-ERK in tumors. The results of the current study provide a strong rationale for the clinical investigation of JNJ-42756493 in patients with tumors harboring FGFR pathway alterations.
Fibroblast growth factors (FGFs) and their receptors (FGFR1 through 4) regulate a variety of key cellular processes, including proliferation, migration, survival, and differentiationa. Aberrant activation of FGF/FGFR is strongly implicated in oncogenic signalling in many tumor types. This has stimulated the development of a number of FGFR inhibitors, with diverse kinase inhibition and pharmacological profiles that are currently being evaluated in clinical studies. We conducted a fragment screening campaign and this resulted in identification of a 6-aminoquinoxalinyl fragment with a binding affinity in the micromolar range. Structure-guided medicinal chemistry led to the identification of a novel quinoxaline-based chemical series with nanomolar affinity for FGFR1, 2, 3, and 4, activity in cells, and selectivity with respect to VEGFR-2. Further optimisation resulted in the generation of JNJ-42756493, a compound with favourable drug-like properties that demonstrated strong anti-tumoral activity in a FGFR2-dependent SNU-16 human gastric carcinoma xenograft model. This report represents the first disclosure of the structure-activity relationships as well as the chemical synthesis pathway of the JNJ-42756493 series and illustrates how a fragment-based drug discovery approach has been efficiently used to discover FGFR1-4 inhibitors with nanomolar affinity. aTurner, N. and Grose, R. Nat. Rev. Cancer, 2010, 10, 116-129. Citation Format: Patrick R. Angibaud, Laurence Mevellec, Gordon Saxty, Christophe Adelinet, Rhalid Akkari, Valerio Berdini, Pascal Bonnet, Marine Bourgeois, Xavier Bourdrez, Anne Cleasby, Helene Colombel, Imre Csoka, Werner Embrechts, Eddy Freyne, Ronaldus Gilissen, Eleonora Jovcheva, Peter King, Jean Lacrampe, Delphine Lardeau, Yannick Ligny, Steve Mcclue, Lieven Meerpoel, David R. Newell, Martin Page, Alexandra Papanikos, Elisabeth Pasquier, Isabelle Pilatte, Virginie Poncelet, Olivier Querolle, David C. Rees, Sharna Rich, Bruno Roux, Elodie Sement, Yvan Simonnet, Matthew Squires, Virginie Tronel, Tinne Verhulst, Jorge Vialard, Marc Willems, Steven J. Woodhead, Berthold Wroblowski, Christopher W. Murray, Timothy Perera. Discovery of JNJ-42756493, a potent fibroblast growth factor receptor (FGFR) inhibitor using a fragment based approach. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4748. doi:10.1158/1538-7445.AM2014-4748
It is becoming increasingly clear that many solid tumors are not a homogeneous mass, but instead consist of highly heterogeneous microsystems comprised of multiple tumor subpopulations together with stromal cells. Such heterogeneity is often attributed to genetic and epigenetic alterations that promote functional specialization of different tumor subpopulations, thereby supporting the growth and differentiation of the primary tumor and subsequent invasion and metastases. This intratumoral complexity serves as a base for a range of intrinsic and acquired resistance mechanisms that are responsible for the failure of targeted therapies, especially in highly heterogeneous tumors such as lung cancer. JNJ-42756493 is a nanomolar inhibitor of fibroblast growth factor receptors (FGFRs) 1, 2, 3 and 4, that is currently being evaluated in Phase I clinical trials. As part of our ongoing preclinical program to elucidate potential mechanisms of resistance to JNJ-42756493, we have developed and characterized a range of cell-based models, representing different solid tumor histologies which show resistance to JNJ-42756493 and other FGFR inhibitors. Here, we describe the outgrowth of pre-existing subpopulations of cells that were intrinsically resistant to FGFR inhibition, from different FGFR1-amplified lung cancer subtypes: NCI-H1581, large cell (LCLC), and DMS114, small cell lung carcinoma (SCLC) cells. Following expansion of these resistant clones, a range of comparative ‘omics’ approaches (microarray analysis, RNA sequencing, shRNA knockdown screening, exome sequencing, and phospho RTK array analysis) were employed to explore the molecular mechanisms of resistance. Comprehensive profiling of these intrinsically resistant subpopulations has revealed that the LCLC and SCLC models have divergent mechanisms of resistance. Met pathway activation was found in LCLC model whilst IGF1R was found to be activated in the SCLC model resistant to FGFR inhibition. This is a novel observation of IGF1R pathway activation in FGFR1 amplified small cell lung cancer models that are resistant to FGFR inhibition. Blocking Met or IGF1R pathways with targeted therapies was observed to prevent the emergence of intrinsic resistance observed in these FGFR driven tumors. Follow-up experiments were performed in vitro and in vivo to confirm these intrinsic mechanisms of resistance and these data help build a rationale for combinations of targeted therapies in the clinic. Finally, our data indicates that pre-existing heterogeneity is likely to be a key driver for the appearance of resistance to targeted therapy in independent lung cancer models. Citation Format: Eleonora Jovcheva, Souichi Ogata, Kelly Van De Ven, Caroline Paulussen, Inez Van de Weyer, Hans De Wolf, Hugo Ceulemans, Steve McClue, Jorge Vialard, Timothy Perera. Identification of alternative mechanisms of resistance to FGFR inhibitor treatment in FGFR1-amplified large cell compared to FGFR1-amplified small cell lung cancer models. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr LB-329. doi:10.1158/1538-7445.AM2014-LB-329
<p>JNJ-42756493 anti-proliferative activity against cancer cells lines from multiple origins.. Detailed data supporting Figure 2.</p>
<p>A) Structure of JNJ-42541707, a structurally related compound to JNJ-42756493. B) 72h growth inhibition (IC50) of JNJ-42541707 against 236 cancer cell lines from multiple origins color coded based on FGFR1,2,4 mRNA overexpression and FGFR WT.</p>
scite is a Brooklyn-based organization that helps researchers better discover and understand research articles through Smart Citations–citations that display the context of the citation and describe whether the article provides supporting or contrasting evidence. scite is used by students and researchers from around the world and is funded in part by the National Science Foundation and the National Institute on Drug Abuse of the National Institutes of Health.
customersupport@researchsolutions.com
10624 S. Eastern Ave., Ste. A-614
Henderson, NV 89052, USA
This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.
Copyright © 2024 scite LLC. All rights reserved.
Made with 💙 for researchers
Part of the Research Solutions Family.