Overcoming EGFR(T790M) and EGFR(C797S) resistance with mutant-selective allosteric inhibitors (original) (raw)

Structure-activity exploration of a small-molecule allosteric inhibitor of T790M/L858R double mutant EGFR

Journal of Enzyme Inhibition and Medicinal Chemistry, 2022

EGFR is a protein kinase whose aberrant activity is frequently involved in the development of non-small lung cancer (NSCLC) drug resistant forms. The allosteric inhibition of this enzyme is currently one among the most attractive approaches to design and develop anticancer drugs. In a previous study, we reported the identification of a hit compound acting as type III allosteric inhibitor of the L858R/T790M double mutant EGFR. Herein, we report the design, synthesis and in vitro testing of a series of analogues of the previously identified hit with the aim of exploring the structure-activity relationships (SAR) around this scaffold. The performed analyses allowed us to identify two compounds 15 and 18 showing improved inhibition of double mutant EGFR with respect to the original hit, as well as interesting antiproliferative activity against H1975 NSCLC cancer cells expressing double mutant EGFR. The newly discovered compounds represent promising starting points for further hit-to-lead optimisation.

Faculty of 1000 evaluation for Novel mutant-selective EGFR kinase inhibitors against EGFR T790M

F1000 - Post-publication peer review of the biomedical literature, 2010

The clinical efficacy of epidermal growth factor receptor (EGFR) kinase inhibitors in EGFR mutant non-small cell lung cancer (NSCLC) is limited by the development of drug resistance mutations, including the gatekeeper T790M mutation 1-3. Strategies aimed at targeting EGFR T790M with irreversible inhibitors have had limited success and are associated with toxicity due to concurrent inhibition of wild type EGFR4 , 5. All current EGFR inhibitors possess a structurally related quinazoline based core scaffold and were identified as ATP-competitive inhibitors of wild type EGFR. Here we identify a covalent pyrimidine EGFR inhibitor by screening an irreversible kinase inhibitor library specifically against EGFR T790M. These agents are 30-100 fold more potent against EGFR T790M, and up to 100 fold less potent against wild type EGFR, than quinazoline based EGFR inhibitors in vitro and are effective in murine models of lung cancer driven by EGFR T790M. Cocrystallization studies reveal a structural basis for the increased potency and mutant selectivity of these agents. These mutant selective irreversible EGFR kinase inhibitors may be clinically more

Structural signature of the G719S-T790M double mutation in the EGFR kinase domain and its response to inhibitors

Scientific Reports, 2014

Some individuals with non-small-cell lung cancer (NSCLC) benefit from therapies targeting epidermal growth factor receptor (EGFR), and the characterization of a new mechanism of resistance to the EGFR-specific antibody gefitinib will provide valuable insight into how therapeutic strategies might be designed to overcome this particular resistance mechanism. The G719S and T790M mutations and their combination were involved in causing different conformational redistribution of EGFR. In the present computational study, we analyzed the impact and structural influence of G719S/T790M double mutation (DM) in EGFR with ligand (gefitinib) through molecular dynamic simulation (50 ns) and docking analysis. We observed the escalation in distance between the functional loop and activation loop with respect to T790M mutation compared to the G719S mutation. Furthermore, we confirmed that the G719S mutation causes the ligand to move closer to the hinge region, whereas T790M makes the ligand escape from the binding pocket. Obtained results provide with an explanation for the resistance induced by T790M and a vital clue for the design of drugs to combat gefitinib resistance.

Abstract 2585: Discovery of a potent covalent mutant-selective EGFR inhibitor - the journey from high throughput screening to EGF816

Cancer Research, 2015

Epidermal growth factor receptor (EGFR) is a validated therapeutic target for lung cancer. First and second generation EGFR inhibitors (e.g., gefitinib, erlotinib and afatinib) have revolutionized treatment paradigms of non-small cell lung cancer (NSCLC) patients with oncogenic EGFR mutations. The use of EGFR tyrosine kinase inhibitors (TKI) provides superior efficacy compared to chemotherapy in patients with EGFR L858R or exon 19 deletion tumors. However, resistance inevitably develops after 8-12 months of treatment; most commonly via a secondary T790M point mutation at the gatekeeper residue of EGFR. Furthermore, responses are hindered due to treatment intolerance in the form of rash and diarrhea that are mediated by simultaneous inhibition of wild-type (WT) EGFR at doses required for mutant EGFR suppression. To overcome these limitations, we initiated a project to identify mutant-selective EGFR inhibitors that potently inhibit both activating and T790M resistance EGFR mutations w...

Development of triple mutant T790M/C797S allosteric EGFR inhibitors: a computational approach

Journal of Biomolecular Structure and Dynamics, 2020

The mutations concerned with non-small cell lung cancer involving epidermal growth factor receptor of tyrosine kinase family have primarily targeted. EGFR inhibitors binding allosterically to C797S mutant EGFR enzyme have been developed. Here, database building, library screening performing R-group enumeration and scaffold hopping technique for increasing the EGFR binding affinity of compounds have been carried out. Virtual screening was performed subjecting to HTVS, SP and XP docking protocol along with its relative binding free energy calculations. Molecular docking studies provided the information about binding pockets and interactions of molecules on mutant (PDB: 5D41) as well as wild type (PDB: 4I23) EGFR enzyme. This was supported with ADMET and molecular simulation studies. On the basis of glide score and protein-ligand interactions, highest scoring molecule was selected for molecular dynamic simulation providing a complete insight into the conformational stability. The virtually screened molecules can act as potential EGFR inhibitors in the management of drug resistance.

Discovery of Selective and Noncovalent Diaminopyrimidine-Based Inhibitors of Epidermal Growth Factor Receptor Containing the T790M Resistance Mutation

Journal of Medicinal Chemistry, 2014

Activating mutations within the epidermal growth factor receptor (EGFR) kinase domain, commonly L858R or deletions within exon 19, increase EGFR-driven cell proliferation and survival and are correlated with impressive responses to the EGFR inhibitors erlotinib and gefitinib in nonsmall cell lung cancer patients. Approximately 60% of acquired resistance to these agents is driven by a single secondary mutation within the EGFR kinase domain, specifically substitution of the gatekeeper residue threonine-790 with methionine (T790M). Due to dose-limiting toxicities associated with inhibition of wild-type EGFR (wtEGFR), we sought inhibitors of T790M-containing EGFR mutants with selectivity over wtEGFR. We describe the evolution of HTS hits derived from Jak2/Tyk2 inhibitors into selective EGFR inhibitors. X-ray crystal structures revealed two distinct binding modes and enabled the design of a selective series of novel diaminopyrimidine-based inhibitors with good potency against T790M-containing mutants of EGFR, high selectivity over wtEGFR, broad kinase selectivity, and desirable physicochemical properties.

Single and dual targeting of mutant EGFR with an allosteric inhibitor

Cancer Discovery

Allosteric kinase inhibitors offer a potentially complementary therapeutic strategy to ATP-competitive kinase inhibitors due to their distinct sites of target binding. In this study, we identify and study a mutant-selective EGFR allosteric inhibitor, JBJ-04-125-02, which as a single agent can inhibit cell proliferation and EGFR L858R/T790M/C797S signaling in vitro and in vivo. However, increased EGFR dimer formation limits treatment effi cacy and leads to drug resistance. Remarkably, osimertinib, an ATP-competitive covalent EGFR inhibitor, uniquely and signifi cantly enhances the binding of JBJ-04-125-02 for mutant EGFR. The combination of osimertinib and JBJ-04-125-02 results in an increase in apoptosis, a more effective inhibition of cellular growth, and an increased effi cacy in vitro and in vivo compared with either single agent alone. Collectively, our fi ndings suggest that the combination of a covalent mutant-selective ATP-competitive inhibitor and an allosteric EGFR inhibitor may be an effective therapeutic approach for patients with EGFR-mutant lung cancer. SIGNIFICANCE: The clinical effi cacy of EGFR tyrosine kinase inhibitors (TKI) in EGFR-mutant lung cancer is limited by acquired drug resistance, thus highlighting the need for alternative strategies to inhibit EGFR. Here, we identify a mutant EGFR allosteric inhibitor that is effective as a single agent and in combination with the EGFR TKI osimertinib.

Molecular Design of a “Two-in-One” Orthosteric-Allosteric Chimeric Mutant Selective EGFR Inhibitor

Inhibitors developed to target the epidermal growth factor receptor (EGFR) are an effective therapy for patients with non-small cell lung cancer harbouring drug-sensitive activating mutations in the EGFR kinase domain. Drug resistance due to treatment-acquired mutations within the receptor itself has motivated development of successive generations of inhibitors that bind in the ATP-site, and third-generation agent osimertinib is now a first-line treatment for this disease. More recently, allosteric inhibitors have been developed to overcome the C797S mutation that confers resistance to osimertinib. In this study, we present the rational structure-guided design and synthesis of a mutant-selective EGFR inhibitor that spans the ATPand allosteric sites. The lead compound consists of a pyridinyl imidazole scaffold that binds irreversibly in the orthosteric site fused with a benzylisoindolinedione occupying the allosteric site. The compound potently inhibits enzymatic activity in L858R/T7...

Recent progress on third generation covalent EGFR inhibitors

First generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (gefitinib and erlotinib) demonstrate excellent clinical efficacy for NSCLC patients carrying EGFR oncogenic mutations (L858R, del exon 19 deletions between amino acids 746 and 750). Invariable, drug resistance occurs with around 60% of it driven by the EGFR-T790M gatekeeper mutation. To counter the T790M-dependent resistance, third generation covalent EGFR inhibitors have been developed with high potency toward T790M containing mutants and selectivity over WT EGFR. This review provides an overview of the third generation drugs currently in clinical trials and also encompasses novel methodologies developed to discover third generation covalent EGFR drugs. Lung cancer is the leading cause of cancer-related death worldwide. 1 A subset of these patients harbor oncogenic mutations in the epidermal growth factor receptor (EGFR) which constitute 10–30% of the non-small cell lung cancer (NSCLC) population resulting in an estimated 100–200 thousand newly diagnosed cases per year globally. The two frequent and mutually-exclusive primary mutations are EGFR L858R and EGFR Del (exon 19 deletions between amino acids 746 and 750) which account for approximately 85% of all mutant EGFR NSCLC cases. 2 Patients with these somatic mutants can be treated with first generation EGFR kinase inhibitor drugs (gefitinib 3 and erlotinib 4) (Fig. 1) which have excellent response rates and disease control for 11–14 months. Invariably, responsive patients develop resistance to these therapies with approximately 60% driven by a single second-site EGFR kinase domain mutation (gatekeeper residue, T790M) which restores constitutive EGFR dependent signaling. 5,6 The mechanism of T790M-derived resistance remains controversial but is believed to be from three contributions—(i) increased ATP binding affinity for T790M mutants, (ii) steric clash between Met790 gatekeeper side chain and the aniline moiety of first generation EGFR TKIs that binds in the kinase back-pocket, and (iii) altered catalytic domain conformational dynamics. 5,7 Taken together, there is an unmet medical need with an expectation that blocking signaling from the drug resistant form of EGFR is clinically effective. Covalent inhibitors can have advantages over reversible compounds because they achieve complete and sustained target engagement in the presence of high intracellular concentrations of the competitive ligand (ATP) in the cells which requires the physical turnover of the targeted protein to restore inhibited sig-naling pathways. 8–12 Indeed, second generation, covalent inhibi-tors of EGFR (dacomitinib, 13 afatinib 14) based on earlier studies of canertinib (CI-1033) 15 (Fig. 1) demonstrate increased cellular potency against EGFR oncogenic variants (e.g., EGFR-L858R/ T790M). These compounds utilize an aniline moiety to bind in the back pocket, and the aniline moiety clashes with Met790 side chain thus making their EGFR-T790M activity less potent than their WT activity. A liability of the second generation of EGFR drugs is that they potently inhibit wild-type EGFR and cause epithelium-based toxicities such as rash and diarrhea which limit their clinical dose. A third generation of EGFR TKI drugs is emerging with high potency against T790M-containing mutants and selectivity over WT EGFR. A wealth of preclinical studies has been reported and clinical findings are becoming available. The current review is designed to highlight the novel methodologies developed to enable discovery of the third generation EGFR drugs and provide an overview of those currently in clinical trials. Inhibitor bound cocrystal structures: The development of third generation drugs has been facilitated by advances in EGFR structural insight. The first published crystal structure of EGFR T790M mutant is EGFR T790M bound with WZ4002, 16 determined at