<<

Published OnlineFirst February 4, 2019; DOI: 10.1158/0008-5472.CAN-18-1281

Cancer Review Research

Novel Third-Generation EGFR Inhibitors and Strategies to Overcome Therapeutic Resistance in Lung Ayesha Murtuza, Ajaz Bulbul, John Paul Shen, Parissa Keshavarzian, Brian D. Woodward, Fernando J. Lopez-Diaz, Scott M. Lippman, and Hatim Husain

Abstract

EGFR-activating mutations are observed in approximately (PF-0647775), and AC0010. However, therapeutic resistance 15% to 20% of patients with non–small cell lung cancer. after the administration of third-generation inhibitors is com- Tyrosine kinase inhibitors have provided an illustrative exam- plex and not fully understood, with significant intertumoral ple of the successes in targeting oncogene addiction in cancer and intratumoral heterogeneity. Repeat tissue and plasma and the role of tumor-specific adaptations conferring thera- analyses on therapy have revealed insights into multiple peutic resistance. The compound is a third- mechanisms of resistance, including novel second site generation tyrosine kinase inhibitor, which was granted full EGFR mutations, activated bypass pathways such as MET FDA approval in March 2017 based on targeting EGFR T790M amplification, HER2 amplification, RAS mutations, BRAF resistance. The compound has received additional FDA mutations, PIK3CA mutations, and novel fusion events. approval as first-line therapy with improvement in progres- Strategies to understand and predict patterns of mutagenesis sion-free survival by suppressing the activating mutation are still in their infancy; however, technologies to under- and preventing the rise of the dominant resistance clone. stand synthetically lethal dependencies and track cancer Drug development has been breathtaking in this space with evolution through therapy are being explored. The expan- other third-generation compounds at various stages of devel- sion of combinatorial therapies is a direction forward opment: (CO-1686), (HM61713), targeting minimal residual disease and bypass pathways nazartinib (EGF816), naquotinib (ASP8273), mavelertinib early based on projected resistance.

Introduction Historical Perspective EGFR-mutant lung cancer has been a model of drug develop- Initially approved as second line therapy in advanced non– ment for oncogene addiction in cancer. The receptor belongs to small cell lung cancer (NSCLC) after , first- the ERBB family of receptors that bind ligands including EGF, generation reversible anti-EGFR tyrosine kinase inhibitors (TKI) TGFa, , , , heparin-binding EGF, and gefitinib entered clinical guidelines based on a body and . Upon ligand binding to EGFR, homodimerization of clinical trials that demonstrated efficacy in some patients after or heterodimerization with other ERBB family members triggers initial lines of chemotherapy (2–6). In 2004, three groups of receptor phosphorylation and activation of downstream signaling researchers identified a subset of patients with NSCLC with through RAS-RAF-MEK-ERK and RAS-PI3K-PTEN-AKT-mTOR activating mutations in the EGFR tyrosine kinase domain (2, 5, activation, leading to proliferation, differentiation, and migration 6). The most common EGFR mutations are exon 19 deletions of cancer cells (Supplementary Fig. S1; ref. 1). Herein, we describe (54%), comprising of five amino acids from codons 746-750, and some of the important lessons learned from an over a decade of substitution of leucine with arginine at codon 858 (L858R, 41%; experience with EGFR inhibitors in lung cancer, drug resistance, ref. 7). Other less common aberrations include G719X (4%), and some of the projections forward with new strategies in first- L861X (1%), S768I (1%) mutations, and exon 20 insertions line clinical management. (2%–9%; ref. 8). Patients with activating EGFR mutations (except exon 20 insertions) have been exquisitely sensitive to first- generation reversible EGFR TKIs, and a true paradigm shift emerged when these compounds showed improved efficacy to fi University of California San Diego, Moores Cancer Center, La Jolla, California. chemotherapy as initial therapy. For the rst time, a solid tumor was treated with oral anticancer therapy in the first-line and Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). improved PFS from approximately 5 months with chemotherapy to 11 months (3–4). The lung cancer clinical community was A. Murtuza and A. Bulbul contributed equally to this article. intellectually inspired, and the adoption of tolerable, precision Corresponding Author: Hatim Husain, University of California San Diego, oriented, oral anticancer therapy emerged to replace standard Moores Cancer Center, 3855 Health Sciences Dr #3011, La Jolla, CA 92093. intravenous chemotherapy for a subset of patients. Phone: 858-534-1590; Fax: 858-822-6186; E-mail: [email protected] Despite revolutionary advancements, patients who were sen- doi: 10.1158/0008-5472.CAN-18-1281 sitive to EGFR TKIs eventually progressed after approximately 9 2019 American Association for Cancer Research. to 11 months. More than 60% of patients develop resistance to

www.aacrjournals.org 689

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst February 4, 2019; DOI: 10.1158/0008-5472.CAN-18-1281

Murtuza et al.

first- or second-generation inhibitors with an acquired second site of development and include nazartinib (EGFR 816), rociletinib mutation in the ATP binding pocket of EGFR exon 20 leading to a (CO-1686), olmutinib (HM61713), naquotinib (ASP8273), threonine-to-methionine substitution at the amino acid position mavelertinib (PF-06747775), and AC0010 (Supplementary 790 (T790M; ref. 7). This mutation prevents drug binding by Fig. S2; Supplementary Table S1). Following over a decade of increasing the affinity of the ATP binding site for ATP. Second- active drug development, osimertinib is the only third-generation generation, irreversible TKIs including and dacomitinb inhibitor that is currently approved by the FDA, with data that were developed with intent to overcome these resistant tumor demonstrate significant improvement in clinical endpoints, clones (9, 10). However, despite encouraging in vitro data, clinical including PFS, tolerability with limited wild-type inhibition, and trials with these compounds did not improve overall survival in response with central (CNS) disease (11, 12). T790M mutant patients likely due to the inability to escalate Other third-generation inhibitors as of yet have not demonstrated dosing because of potent effects on wild-type EGFR at the ther- superiority to osimertinib despite antitumor efficacy, and at this apeutic window (9). Third-generation inhibitors with pyrimidine time it remains unclear where they will be clinically positioned. core structures were created to target the T790M clone with maintained activity against the original exon19del and L858R mutations. Binding at the C797 locus and having selectivity Osimertinib against the T790M mutation, third-generation inhibitors have Osimertinib, a pyrimidine-based compound that binds irre- had responses for patients with T790M resistance after first- versibly to the cysteine-797 residue at the ATP binding site of the generation inhibitors (Fig. 1A–E). First-line data have demon- receptor, preclinically displayed significant tumor strated superior clinical outcomes compared with first-generation cytoreduction in xenograft and transgenic mice with activity inhibitors with trends toward overall survival improvement (11). against activating mutations exon19del and L858R with and without detectable T790M mutations and less activity for the wild-type EGFR target (13). Clinically, the phase II AURA2 trial Third-Generation TKIs was restricted to EGFR T790M-positive NSCLC patients with The race to discover a next-generation EGFR TKI effective disease progression after initial first line therapy and showed a against resistant tumors has been both illustrative and ground 71% (140/199) objective response with six patients (3%) having breaking. Osimertinib initially gained full FDA approval for complete response and 134 (67%) with partial response (14). Full patients with metastatic EGFR T790M mutant NSCLC after pro- FDA approval was given after the phase III AURA3 study dem- gressing on first- or second-generation EGFR TKIs and has sub- onstrated an overall response rate (ORR) of 71% compared with sequently gained approval in the first-line treatment of EGFR- 31% with platinum- therapy (OR 5.39; P < 0.001) and mutant lung cancer (11, 12). Other third-generation inhibitors a median PFS of 101 months versus 44 months (HR 030; P < designed to target the T790M clone have been at various stages 0001; ref. 12). The most common all grade drug-related adverse

© 2018 American Association for Cancer Research

Figure 1. In silico modeling of EGFR C797S interaction with osimertinib in the complex of EGFR derived from its crystal structure (PDB4ZAU) using Chimera software. A, Space filling hydrophobicity surface model of EGFR with osimertinib. B and C, Osimertinib-EGFR interaction with C797wt (B) or C797S-mutant (C) residues pan-view with relevant residues to TKI resistance G719, T790, and L858 indicated on the model. D and E, Osimertinib covalent binding to the SH-group on C797 showing both the covalent binding (red) and the separation distance to C797 wild type. E, In silico modeling of the most stable rotamer substitution by the less nucleophilic serine side chain at position 797, which cannot undergo covalent bond formation. A higher distance between the C797S and osimertinib closest carbon atom is indicated in yellow.

690 Cancer Res; 79(4) February 15, 2019 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst February 4, 2019; DOI: 10.1158/0008-5472.CAN-18-1281

EGFR Third-Generation TKI Therapy

events included diarrhea (43%) and rash (40%), however impres- Combinational strategies with osimertinib sively less than 1% were grade three or higher. Interstitial lung Studies evaluating the safety and efficacy of additional targeted disease (ILD) was observed in eight patients and, although rare therapies added to osimertinib are underway and include com- with a mechanism that has not yet been described, did have at binations with agents that target pathways including MET, MAPK, least one fatal event (12, 14). In a larger study of 1,217 patients BCL-2, and JAK activation among others. Based on kinome (ASTRIS, NCT02474355), the investigator-assessed response rate screens and given that upregulation of other oncogenic pathways was 64% at 4 months and safety parameters reflected the original is a common resistance mechanism to tyrosine kinase inhibition, study (15). Pooled analysis has demonstrated a median survival multiple clinical studies are evaluating targets in combination of 26.8 months with second-line osimertinib (16). The success of with osimertinib (20, 21). The TATTON phase Ib study the compound in the second line prompted evaluation in the first- (NCT02143466) involves combinations of osimertinib at line therapy for EGFR-mutant disease (11). increasing doses in combination with either (AZD6094, MET inhibitor), (MEK inhibitor), or First-line data (anti-PDL1 antibody; ref. 20). The evaluation of osimertinib in the first-line space hinged on Early work with a gefitinib-sensitive lung cancer cell line reveal- the hypothesis that suppressing the T790M clone early in treat- ed focal amplification of the MET proto-oncogene in cells that ment course may improve clinical outcomes. The phase III developed resistance to gefitinib. Inhibition of MET signaling FLAURA trial (NCT02296125) directly compared osimertinib in these cells restored their sensitivity to gefitinib and MET ampli- (80 mg daily) with first-generation therapy (either gefitinib 250 fication was detected in 4 of 18 (22%) lung cancer specimens mg or erlotinib 150 mg daily) in a total of 556 patients with that had developed resistance to gefitinib or erlotinib. It was NSCLC with newly diagnosed EGFR exon 19 deletions or L858R found that MET amplification causes gefitinib resistance by driv- mutations. Osimertinib had a superior PFS relative to the erlo- ing ERBB3 (HER3)–dependent activation of PI3K (21). In clinical tinib or gefitinib arm with median progression-free survival of samples obtained after treatment with the third generation inhib- 18.9 months compared with 10.2 months for standard therapy itor osimertinib, plasma circulating tumor DNA analyses have [HR 0.46; 95% confidence interval (CI), 0.37–0.57; P < 0.0001; identified MET copy number gain after first line osimertinib in ref. 11]. The median duration of response was 17.2 months in approximately 19% of patients on the FLAURA trial and 15% after patients treated with osimertinib compared with 8.5 months in second-line osimertinib (22, 23). Updated analysis of TATTON the first-generation TKI cohort. The incidence of grade three or phase Ib study combining osimertinib and the MET TKI savoli- higher was lower with osimertinib (32%) than stan- tinib showed an ORR of (i) 33% in prior third-generation TKI dard treatment (41%; ref. 11). Although OS data have not yet exposed and (ii) 61% in patients without T790M mutation and matured, the compound has been adopted into first-line with no prior third-generation EGFR-TKI exposed cohorts (24). Second FDA approval. site mutations in the MET oncogene (D1228V) have been seen at resistance on the osimertinib and savolitinib trial demonstrating Role in brain metastasis the dependency on the pathway (25). Grade 3 adverse events Brain recurrence for patients on therapy is an unmet need with were experienced by 50% of patients, with the most common limited therapeutic options and contributed by challenging phar- all-cause events being vomiting (8%), AST/ALT increase (6%), macokinetics, drug efflux transporters, and molecular weight (17). and rash (6%). Savolitinib was discontinued for 30% of patients In preclinical models, osimertinib demonstrated the ability to because of adverse events (24). cross the blood–brain barrier (BBB) with a higher ratio of Osimertinib is also being combined with inhibitors in the unbound tissue concentration to unbound plasma concentration MAPK pathway. Resistance to the third generation EGFR inhibitor and higher substrate activity against permeability glycoprotein tool compound WZ4002 has been seen through amplification of (Pgp) and breast cancer resistance protein (BCRP) efflux trans- ERK through activation of the MAPK1 pathway or downregula- porters as compared with gefitinib (17, 18). Measurement of AUC tion of negative regulators of ERK signaling (26). The MEK tissue to plasma ratios showed osimertinib more highly distrib- inhibitor CI-1040 downregulates ERK1/2 phosphorylation and, uted in the brain (1.7–2.0) than gefitinib (0.21), rociletinib when combined with WZ4002, PC9 WZ4002-resistant cells were (<0.08), and afatinib (<0.36). Data from the BLOOM study responsive to the combination in vitro (27). In post-progression (NCT02228369) suggested encouraging activity with osimertinib patient samples, resistance involving the RAS–MAPK pathway has (160 mg daily) with a median treatment duration of 6 months been seen in patients treated with osimertinib, with acquired and manageable in patients with leptomeningeal disease; KRAS mutations and V600E BRAF mutations observed after 10 patients (43%) have had intracranial radiologic response and acquired resistance (28). Furthermore, preclinical work demon- 13 patients (56%) with stable disease (SD; ref. 18). Among strated that the concomitant inhibition of EGFR and ERK1/2 by patients in the AURA3 randomized phase III study, the median the MEK inhibitor prevents ERK1/2 reactivation, CNS PFS was significantly longer with osimertinib than with enhances anti-EGFR–induced , and inhibits the emer- chemotherapy (11.7 months vs. 5.6 months; HR 0.32; P ¼ gence of resistance in EGFR-sensitive models known to acquire 0.004). CNS ORR was 70% (21/30) with osimertinib compared resistance via both T790M-dependent and T790M-independent with 31% ORR with chemotherapy pointing to the intracranial mechanisms. Resistance to the combination with trametinib may efficacy of the compound (12). In the first-line FLAURA study, emerge due to AKT/mTOR reactivation (29). In another study events of CNS progression were observed in 12 of 61 patients combining osimertinib with selumetinib in ex vivo primary cell (20%) in the osimertinib group and 26 of 67 (39%) in the cultures obtained from osimertinib plus selumetinib-resistant standard EGFR TKI group (11). CNS response rates in measurable tumors, the Hedgehog pathway was activated and a smoothened brain metastases were 91% with osimertinib and 68% with first- (SMO) inhibitor plus osimertinib and selumetinib inhibited generation EGFR TKIs (19). proliferation and migration of resistant cells (30). These collective

www.aacrjournals.org Cancer Res; 79(4) February 15, 2019 691

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst February 4, 2019; DOI: 10.1158/0008-5472.CAN-18-1281

Murtuza et al.

data form the basis of the study of osimertinib and selumetinib onstrated significant tumor regression using in vitro models (35). (NCT02143466) with some responses seen at interim data anal- Ongoing clinical trials have combined EGF816 100 mg daily with ysis in the phase I study. The most commonly reported grade 400 mg twice daily in patients with advanced NSCLC >3 adverse events were diarrhea (21%), dermatitis acneiform with EGFR L858R, ex19del, or T790M in several lines of therapy (15%), and hypokalemia (10%; ref. 20). including T790M negative or treatment naive patients Osimertinib in combination with navitoclax is an ongoing (NCT02335944). The addition of either capmatinib or phase Ib trial in patients with disease progression on an anti- after progression on gefitinib has been encouraging (ORR 47% EGFR TKI (NCT02520778). Inhibition of Bcl-2, a prosurvival and 45%, respectively, in MET amplified patients; refs. 39, 40). protein, prompts cells to undergo apoptosis by upregulating and dephosphorylating BIM, a proapoptotic protein from Bcl-2 family Rociletinib (CO-1686) components. ABT263 (navitoclax) is a dual inhibitor of BCL-xL Rociletinib is an irreversible inhibitor of mutant EGFR includ- and Bcl-2, which induces BIM by decreasing the capacity of BCL-xL ing exon 19 deletion, L858R, and T790M binding at the C797 to neutralize BIM and facilitates Bax and Bak to initiate caspases, locus. Enrollment of trials with rociletinib ceased in May 2016 leading to cell death. In a drug screen with EGFR T790M patient after updated results in November 2015 showed a less than derived cell lines, navitoclax in combination with the tool com- expected ORR of 33.9% and PFS of 5.7 months for patients with pound WZ4002 induced significantly more apoptosis compared confirmed T790M-positive NSCLC (41). The FDA formed an with the EGFR inhibitor WZ4002 alone (31). independent expert panel and voted 12–1 against approval of Downstream targeting of the mTOR pathway is being studied in rociletinib based on existing data. When retrospec- a phase II trial of sapanisertinib (INK128), a potent mTORC1/2 tively analyzing the profound difference in ORR between April inhibitor, in combination with osimertinib for patients with and November 2015, over half of the patients had unconfirmed advanced EGFR-mutated NSCLC (NCT02503722). Preclinically, partial responses (41). Post-analysis revealed 22 of 69 patients inhibition of TORC1 and TORC2 decreased cell growth, metab- (32%, 15 patients with T790Mþ disease and 7 patients with olism, and angiogenesis. mTOR kinase inhibition via INK128, T790M disease) had brain metastases that developed before which involves ATP-dependent phosphorylation of proteins S6 discontinuing rociletinib likely due to the poor BBB penetration and 4EBP1 downstream of TORC1, leads to selective inhibition of of the compound. In patients who progressed after rociletinib and AKT phosphorylation (32). Novel mTOR mutations have been were subsequently treated with osimertinib, there was favorable seen on EGFR-resistant patient samples, and in mutant cells response with osimertinib in five of nine patients (two PR and continuation of EGFR and mTOR inhibition fully abrogated three SD). Three patients who developed brain metastases on EGFR downstream signaling (33). Additionally, osimertinib rociletinib had demonstrated improved CNS disease responses on has been combined with JAK 1 inhibitors interrupting signaling osimertinib (42). The experience with rociIetinib has demonstrat- of the JAK/STAT pathway in an ongoing second-line study ed that each EGFR inhibitor may have different off-target effects in T790M mutant patients (INCB39110, NCT02917993 and (i.e., IGFR activation leading to hyperglycemia attributed to a AZD4205, NCT03450330). metabolite of rocelitinib and occurring in 22% of patients), A phase I dose-escalation study of osimertinib combined with differential selectivity against the target, and different effects in , a humanized EGFR , is CNS metastases affecting overall response rate. underway for patients who progressed on previous first-line TKI therapy (NCT02789345). Recent work demonstrates that ampli- Olmutinib (HM61713) fication of EGFR wild-type alleles can confer resistance to EGFR Olmutinib (HM61713) began clinical evaluation in May 2015 TKIs (34). By binding to extracellular domain of the EGFR for T790M-positive mutant NSCLC and was approved for treat- receptor and competing with ligand binding, downsteam signal- ment of in South Korea. In phase II ELUXA trials, 76 patients ing leads to impaired tumor cell growth. included in the phase II expansion after first- or second-generation TKIs received 800 mg/day with median treatment duration of 7.0 Nazartinib (EGF816) months. Of the 71 evaluable patients, 54% demonstrated an EGF816 is an irreversible, mutant-selective EGFR inhibitor that objective response, 90% achieved disease control, mean duration targets L858R, exon 19 deletion, T790M mutations, and is also of response was 8.3 months, and median PFS of 6.9 months (95% being explored for efficacy against exon 20 insertion mutations, CI, 5.5–8.3 months; ref. 43). The most common adverse effects which are intrinsically resistant to first- and second-generation included diarrhea (59%), rash (41%), pruritus (42%), and inhibitors (35). A phase I study of EGF816 showed an ORR of (39%). Further ELUXA trials have been stopped due to incidence 44% and DCR of 91% with a median duration of response of of skin toxicity including toxic epidermolysis necrosis (n ¼ 2, one 9.2 months and grade 3 or 4 adverse events including diarrhea fatal) and Stevens-Johnson syndrome (n ¼ 1; ref. 43). The side- (6%), rash (14%), and anemia (6%; ref. 36). In 14 patients who effect profile of this compound is unfavorable relative to osimer- received EGF816 and then osimertinib, the ORR to osimertinib tinib, which has affected its use although the compound had been was 14% (one CR, one PR, eight SD, four PD) with a median approved in South Korea. continuation of osimertinib therapy for 9 months (37). Documented mechanisms of acquired resistance to EGF816 Naquotinib (ASP8273) include EGFR C797S mutations, deletion in mTOR, BRAF fusions, Not all third-generation compounds have shown superiority to c-MET amplification, and concurrent TP53 and RB1 truncating first- and second-generation inhibitors in the first-line. ASP8273 is mutations associated with small cell transformation (38). It is a small irreversible TKI that binds at C797 and targets the unclear if the prevalence of resistance alterations with this com- common EGFR mutations including T790M while sparing pound differs significantly to osimertinib. Dual inhibition of wild-type EGFR. First-line ASP8273 resulted in an ORR of EGFR and cMET with EGF816 and capmatinib (INC280) dem- 52%, disease control rate (DCR) of 94% and PFS of 11.3

692 Cancer Res; 79(4) February 15, 2019 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst February 4, 2019; DOI: 10.1158/0008-5472.CAN-18-1281

EGFR Third-Generation TKI Therapy

months (44). Among patients treated as second-line therapy, the inhibits activation by displacing the C-helix. With the addition confirmed ORR was 30% and median PFS for T790M-positive of , dimerization is inhibited creating uniform vulner- patients treated was 6.0 months (95% CI, 5.3–9.8 months; ability within the kinase. EGFR del 19 or L858R/T790M/C797S ref. 45). Grade 3 adverse events included diarrhea (4%), hypo- cells are resistant to gefitinib, afatinib, WZ4002, rocelitinib, and natremia (17%), and a grade 2 peripheral neuropathy (n ¼ 1) osimertinib; however, triple mutant tumor cells were partially was noted (45). The decision to discontinue development in sensitive to cetuximab (50). These data provide preclinical ratio- the SOLAR first-line study was based on the recommendation nale for the combination of a next-generation inhibitor in com- of the Independent Data Monitoring Committee comparing bination with an EGFR monoclonal antibody for patients with lower PFS and ORR to the comparators, erlotinib or gefitinib. NSCLC with the triple mutation profile. The development of Differential selectivity with ASP8273 against the original EGFR inhibitors that do not bind C797 will be important for patients activating mutations compared with T790M may have contrib- who develop this resistance. One such compound AC0010 is a uted to the efficacy results compared with first- or second- structurally distinct pyrrolopyrimidine-based irreversible inhibi- generation TKIs in the first-line. tor, and in a xenograft model oral administration of AC0010 at a daily dose of 500 mg/kg resulted in remission of tumors with Mavelertinib (PF-0647775) EGFR-active and T790M mutations for over 143 days with no PF-0647775 is a third-generation EGFR TKI currently being weight loss (51). Objective responses have been observed in studied in patients with mutated EGFR ex19del or L858R with or patients with NSCLC with EGFR T790M mutation on this com- without T790M mutation and has completed dose escalation pound, although it has of yet to be tested after osimertinib for from 25 to 600 mg. RECIST responses observed include 11 PRs C797S resistance. (42.3%, five T790Mþ plasma), six stable disease (23.1%, four Additional EGFR second site mutations have been detected on T790Mþ plasma), two PD (7.7%, one T790Mþ plasma), and six therapy. EGFR-mutant cells in vitro exposed to increasing quan- indeterminate (23.1%, three T790Mþ plasma) after unconfirmed tities of WZ4002 acquired resistance through several additional investigator review. Grade 3 adverse events were seen primarily at second-site EGFR mutations including EGFR L718Q, L844V, greater than 150 mg dose and included diarrhea (12.4%) and skin L798I, L692V, E709K. The full impact and biologic consequences toxicities (30.8%; ref. 46). Further analyses will evaluate the of the second site mutations is not clear at this time. EGFR L718Q efficacy and tolerability of the compound in combination with and L844V have been seen after pyrimidine based inhibitors, , an inhibitor of the cyclin-dependent kinases CDK4 however have occurred more frequently after rociletinib and and CDK6 based on preclinical work demonstrating synergy WZ4002 than osimertinib (52). In the clinic, resistance due to (NCT02349633). the L718Q was noted after a patient with an activating EGFR L858R mutation was treated with gefitinib and subsequently with Mechanisms of TKI Resistance with Second osimertinib for T790M progression (53). Additional case reports describe other second site mutations such as G724S, G796D, and Site Mutations in EGFR L792H on osimertinib therapy. The second-generation inhibitor Given that third-generation EGFR inhibitors irreversibly bind afatinib has been responsive to the acquired G724S muta- at the C797 location, mutations at this locus within the EGFR tion (54). E709K and L682V, both activating missense substitu- gene confer resistance and estimated to be present in 7% to 20% tions, were seen after exposure to rociletinib. EGFR L798I is a of cases (Fig. 1A–E; refs. 22, 23). In patients who progress novel mutation observed in cis conformation with T790M that on osimertinib circulating tumor DNA analyses have shown affects the covalent bond formation of rociletinib at C797 (55). that T790M may be retained with C797S acquisition (47). When Although sensitivity of EGFR TKIs to some uncommon mutations C797S and T790M are in a cis conformation on the same allele are known (L861Q, G719X, S768I), further work will be needed to (82%), EGFR TKI single agents or combinations are unable to determine the sensitivity of different anti-EGFR TKIs across a suppress activity. When the mutations are in the trans confor- diversity of second-site EGFR TKI mutations and the impact of mation on different alleles, patients have seen responses to the sequence of therapy on outcome (56). Genome editing using combination of first- and third-generation EGFR TKIs (48, 49). CRISPR-Cas9 technologies has been used to create preclinical C797S and T790M mutations were sensitive in preclinical mod- models for drug testing and will improve our ability to investigate els to a combination of gefitinib with WZ4002, a tool com- effective therapy combinations (57). pound developed as a third-generation TKI (49). There have been case reports of clinical efficacy with third- and first-gener- Mechanisms of Resistance in Alternate ation inhibitors with clinical trials ongoing (NCT03122717). Furthermore, as third-generation compounds have moved to the Bypass Pathways first line, clinical trials revisiting reversible first-generation inhi- A recent neoadjuvant study of erlotinib for locally advanced bitors to combat C797 resistance after third-generation inhibi- resectable EGFR-mutant patients has demonstrated that only a tors are being explored. few patients (3/25 pts) had a major pathologic response (<10 Additional novel drug combinations are being explored to viable tumor cells) after first-generation inhibitors (58). This overcome EGFR C797S resistance. EA1045 is an allosteric EGFR highlights that resistance may occur early with mechanisms of inhibitor that in combination with cetuximab prevents effective resistance that are diverse and independent of the EGFR pathway. EGFR dimerization. This compound has been shown to be Continued cell growth and proliferation despite the reduction effective in mouse models of NSCLC with EGFR (L858R/ and disappearance of T790M mutant clones may signify reliance T790M/C797S) mutation by displacing the regulatory C-helix on a different mechanism of resistance or bypass pathways apart inactive conformation of the kinase (50). The allosteric pocket is from EGFR (Fig. 2; refs. 28, 55). Escape mechanisms following accessible in two subunits of the dimer, and the compound therapy with third-generation EGFR TKIs are heterogeneous and

www.aacrjournals.org Cancer Res; 79(4) February 15, 2019 693

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst February 4, 2019; DOI: 10.1158/0008-5472.CAN-18-1281

Murtuza et al.

Clonal Evolution of EGFR Mutant Lung Cancer on Therapy

Chemotherapy Progression

Osimertinib T790M+/– 2nd site mutation T790M+/– 2nd site mutation (i.e C797S) (i.e C797S, G724S)

Erlotinib eted therapy Gefitinib Tar g Afatinib T790M+ against bypass Pathway

Chemotherapy

Osimertinib Progression Progression T790M– Alternate bypass pathways

T790M–

EGFR activating mutation clone ∗MET amplification, HGF expression, HER2 amplification, Alternate bypass mechanism∗ PIK3CA mutations, PTEN loss, KRAS mutations, NRAS mutations, BRAF mutation, MAPK1/AKT3 overexpression, EGFR T790M mutant clone FGF20FGFR1 loop mutation, IGFR1 activation, RB1/p53 loss, EGFR additional second site mutation (ie C797S) small cell histologic switch, fusion events

© 2018 American Association for Cancer Research

Figure 2. Clonal heterogeneity through EGFR-. Visual representation of minimal residual disease and malignant cell progression after anti-EGFR TKIs demonstrating genomic patterns of selection observed in clinical samples through different lines of therapy.

may include EGFR amplification, HER2 amplification, MET a monoclonal anti-IGF1/2 antibody sensitized WZ4002 resis- amplification, (HGF) expression, tant, PC-9/GR/WR cells to WZ4002 in in vitro and xenograft PIK3CA mutations (E545K and E542K), PTEN loss, KRAS muta- mice (63). FGR receptor (FGFR1) amplification has been seen tions, NRAS mutations, BRAF mutation, MAPK1/AKT3 overex- as an acquired resistance mechanism for first-generation EGFR pression, FGF2-FGFR1 loop mutations, IGF1R activation, fusion TKIs, and a patient who had disease progression on osimertinib events, and RB1/p53 loss associated with histologic switch to had FGFR amplification at re-biopsy with disappearance of the small cell cancer (Fig. 2; Supplementary Fig. S1; refs. 26, 28, 59– EGFR T790M clone (59). Several novel fusions have been 63). Amplification with copy number gain across other ERBB identified following first-, second-, and third-generation com- family receptors have been appreciated in samples post osimerti- pounds including RET fusions, FGFR fusions, and BRAF fusions nib compared with first- and second-generation inhibitors (22, with clinical responses seen in inhibitors that target the fusion 60). Additionally, shorter progression-free survival was identified event (28, 64). The role of polymorphisms influencing resis- in patients with coexisting oncogene drivers including cell-cycle tance profiles or third-generation drug sensitivities have yet to genes (60). Small cell transformation has been associated with remain to be explored. both Rb and p53 inactivation, and mutation in apolipoprotein B The transcriptional state change known as epithelial-to- mRNA editing enzyme, catalytic polypeptide-like (APOBEC) is mesenchymal transition (EMT) is another important resistance frequently seen (61). Patients with shorter time on therapy before mechanism to targeted therapies including EGFR inhibi- progression likely develop resistance that is independent of the tors (65). The precise mechanisms underlying EMT are still EGFR pathway (28). being investigated, but suppression of BIM and upregulation of Upregulated MET has been well characterized, and upregulated AXL, SRC, TGFb, and IFG1R have all been associated with EMT HER2, IGFR, and FGFR have been observed. In preclinical models, andresistancetotargetedtherapy(65–68). Of note small HER2 overexpression or knockout confers resistance or sensitivity molecule inhibitors of the kinase AXL have been shown to respectively in EGFR-mutant PC9, HCC827, and H3255 cells (62). re-sensitize resistant mesenchymal cells to EFGR inhibitors, Furthermore, resistance profiling against WZ4002 has demon- suggesting AXL as a potential therapeutic target in resistant strated activated -like (IGF1R) NSCLC (67). A clinical trial of DS-1205c, an orally adminis- signaling due to loss of expression of EGFBP3 (27). In preclin- tered AXL inhibitor, is currently in dose escalation in combi- ical studies, IGF1R downregulation via an IGF1R inhibitor and nation with osimertinib (NCT03255083). BGB324 is an AXL

694 Cancer Res; 79(4) February 15, 2019 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst February 4, 2019; DOI: 10.1158/0008-5472.CAN-18-1281

EGFR Third-Generation TKI Therapy

inhibitor being explored with first-generation EGFR TKIs Immune combinations with antiangiogenic strategies have (NCT02424617). been undertaken and further data is awaited to determine if In addition to the careful molecular characterization of resis- there is true synergy with this approach. Overall, antiangiogenic tant tumors, high-throughput screening with CRISPR-Cas9 tech- therapies in combination with EGFR-TKIs have shown encour- nology is a means to discover additional potential resistance aging synergy but limited overall survival benefit. Preclinical mechanisms (69). The nuclease Cas9 can be directed with great work with VEGF inhibitors including vandetinib or erlotinib/ specificity using a short RNA barcode allowing for the rapid were more efficacious than gefitinib or erlotinib knockout of any gene. These barcodes can be read using next- alone (76). The addition of bevacizumab to erlotinib resulted generation sequencing, and CRISPR knockout screens can be in an improved PFS compared with erlotinib alone as initial highly multiplexed allowing for genome-wide analyses (70). treatment (16 months vs. 10 months; HR 0.41; ref. 77). However, Synthetic lethal approaches have revealed mechanisms that can the phase II study ACCRU (NCT01532089) did not report drive both oncogene and non-oncogene EGFR-resistant NSCLC. an improvement in PFS and OS, and a phase 1/2 study Non-oncogene–dependent mechanisms include a host of factors (NCT02803203) is currently assessing osimertinib and beva- including NF-kB, STAT3, Wnt, src family kinases, and chromatin cizumab as an initial treatment for patients with EGFR-mutant remodeling proteins (71). Genome-wide knockout screens in lung . First-line osimertinib and chemotherapy combi- sensitive and resistant NSCLC cell lines can be conducted with nations have been brought forward with the goal of eradicating various EGFR inhibitors to identify de novo and acquired resistance minimal residual disease based on encouraging OS data with mechanisms that generalize and that are specific to a particular first-generation inhibitors (NEJ009). compound.

Unanswered questions Immune Combination Therapies The pattern of drug development in EGFR-mutant lung cancer Anti-PD-1:PDL1 therapies appear to be of limited benefitin has been a model for tumors driven by dominant oncogenic driver patients with EGFR-mutant NSCLC, and the role of checkpoint genes, changing multiple paradigms in clinical care. The FDA inhibitors in EGFR-mutant disease is poorly understood (72). approval of osimertinib and impressive median overall survival Larger studies with anti-PD1:PDL1 antibodies have not shown marks an important advance with simultaneous targeting of high response rates or improved overall survival in the subset of the EGFR activating mutation and the dominant resistance clone. EGFR-mutant patients. In a meta-analysis in NSCLC, immune Clinical trials are exploring targeted therapies in early stage checkpoint inhibitors did improve OS over docetaxel in the EGFR neoadjuvant and adjuvant settings (NCT 03433469 and NCT wild-type subgroup (n ¼ 1362; HR ¼ 0.66; P < 0.0001) but not in 02511106). Advances in genome sequencing technologies pro- the EGFR-mutant subgroup (n ¼ 186; HR 1.05; P ¼ 0.03; ref. 72). vide an opportunity for the serial measurement of circulating Preclinical data have shown that EGFR activation can upregulate tumor DNA to allow for detailed characterization of EGFR inhib- PD-L1 expression (73). However, even in high expressing PDL1 itor resistance through time and on therapy. The optimal order to EGFR-mutant patients, checkpoint inhibitors have had limited sequence EGFR inhibitors and whether combinations of EGFR clinical benefit and have been associated with toxicity with inhibitors or combinations with other targeted therapies can pneumonitis and transaminitis (NCT02879994). A trial of osi- prevent the development of resistance clones is an active area of mertinib combined with durvalumab (NCT02143466) has been investigation. Effective and tolerable combinations that target halted due to an increase incidence of ILD (26% in pretreated EGFR independent bypass pathways have been informed by patients and 64% in treatment na€ve patients; ref. 20). preclinical data from in vitro and patient derived xenograft exper- Hypotheses exploring the lack of response allude to lower imentation and data on resistance patterns collected from clinical tumor mutation burden (TMB) and a non-PDL1–dependent trials. Well-defined pre-clinical, pharmacokinetic, and pharma- immunosuppressive tumor microenvironment. A recent pub- codynamic testing has been employed with novel drug combina- lication has demonstrated that EGFR-mutated tumors generally tions to identify dose scheduling that allows for tolerable side have a low TMB, and higher TMB predicts worse response to effect profiles of combination therapy. anti-EGFR inhibitors (74). Mutations in p53 have been more Additional third-generation TKIs have been evaluated with common in EGFR-mutant patients with high TMB, and EGFR- mixed success, and current strategies are evaluating the first-line mutant tumors have higher TMB at the time of progression on utility of these compounds, sequencing strategies, drug combina- EGFR-TKI compared with pretreatment. Increased TMB may be tions to target cancer dependencies, and effects on brain metas- due to the emergence of subclonal mutations, and these muta- tases. Accounting for the complexity of resistance pathways, the tions may increase overall TMB but may not confer effective ability to characterize resistance clones through plasma-based immunogenicity (74). genomic strategies may assist in tracking cancer evolution through Using flow cytometry and/or quantitative PCR in cell lines, therapy. Novel technologies such as CRISPR-Cas9 gene editing median CD73 expression was increased 10-fold in EGFR- and integration of RNA sequencing analyses to identify bypass mutant NSCLC. Expression of the immunosuppressive mole- pathway activation will be important to better characterize the cule CD73 and reduced expression of the IFNg mRNA signature diversity of acquired resistance. Further understanding the biol- has been hypothesized to contribute to an immune suppressive ogy of the disease, the evolution and progression of clones on environmentinthissubsetofNSCLC(75).Atrialiscurrently therapy, the pharmacology of each inhibitor, polymorphisms, underway evaluating an anti-CD73 molecule (oleclumab, which affect selection and third-generation TKI drug sensitivity, MEDI9447) in combination with osimertinib and the adeno- and the resistance profiles seen with each compound will inform sine receptor antagonist (AZD4635) in EGFR-mutant NSCLC further clinical trials and may lead to more durable responses to (NCT03381274). targeted therapy.

www.aacrjournals.org Cancer Res; 79(4) February 15, 2019 695

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst February 4, 2019; DOI: 10.1158/0008-5472.CAN-18-1281

Murtuza et al.

Search strategy and selection criteria in Tetraphase Pharmaceuticles and Illumina. H. Husain reports receiving other We identified references for this Review through search of commercial research support from Pfizer, AstraZeneca, and Mirati; has received PubMed (MEDLINE, Web of Science, Cochrane Library, etc.) speakers bureau honoraria from AstraZeneca, Merck, and Bristol Myers Squibb; and is a consultant/advisory board member of Astrazeneca, from 2001 to 2018, using the search terms NSCLC, EGFR, TKI, Boehringer Ingelheim, Foundation Medicine, and Abbvie. No potential osimertinib, resistance, heterogeneity in various combina- conflicts of interest were disclosed by the other authors. tions. We only reviewed papers and abstracts in English and fi primarily focused on recent clinical trials. The nal reference Acknowledgments list was based on originality and relevance to this Review's scope. The authors would like to thanks Soham More and Seti Akhavan for their contributions to this work. Disclosure of Potential Conflicts of Interest J. Paul Shen reports receiving other commercial research support from Received April 27, 2018; revised September 16, 2018; accepted December 17, Idyea Biosciences and has ownership interest (including stock, patents, etc.) 2018; published first February 4, 2019.

References 1. Kan Z, Jaiswal BS, Stinson J, Janakiraman V, Bhatt D, Stern HM, et al. cell lung cancer (AURA2): a multicentre, open-label, single-arm, phase 2 Diverse somatic mutation patterns and pathway alterations in human study. Lancet Oncol 2016;17:1643–52. cancers. Nature 2010;466:869. 15. De Marinis F, Cho BC, Kim D-W, Kim S-W, Hochmair MJ, Metro G, et al. 2. Pao W, Miller V, Zakowski M, Doherty J, Politi K, Sarkaria I, et al. EGF ASTRIS: A real world treatment study of osimertinib in patients (pts) with receptor gene mutations are common in lung cancers from "never smokers" EGFR T790M positive non-small cell lung cancer (NSCLC). J Clin Oncol and are associated with sensitivity of tumors to gefitinib and erlotinib. 2017;35:9036. doi: 10.1200/JCO.2017.35.15_suppl.9036. Proc Nat Acad Sci USA 2004;101:13306–11. 16. Ahn M-J, Tsai C-M, Shepherd FA, Bazhenova L, Sequist LV, Hida T, et al. 3. Mok TS, Wu Y-L, Thongprasert S, Yang C-H, Chu D-T, Saijo N, et al. Osimertinib in patients with T790M mutation-positive, advanced non– Gefitinib or carboplatin–paclitaxel in pulmonary adenocarcinoma. N Engl small cell lung cancer: long-term follow-up from a pooled analysis of 2 J Med 2009;361:947–57. phase 2 studies. Cancer 2018. doi: 10.1002/cncr.31891. 4. Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, et al. 17. Ballard P, Yates JWT, Yang Z, Kim D-W, Yang JC-H, Cantarini M, et al. Erlotinib versus standard chemotherapy as first-line treatment for Euro- Preclinical comparison of osimertinib with other EGFR-TKIs in EGFR- pean patients with advanced EGFR mutation-positive non-small-cell lung mutant NSCLC brain metastases models, and early evidence of clinical cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. brain metastases activity. Clin Cancer Res 2016;22:5130–40. Lancet Oncol 2012;13:239–46. 18.YangJC-H,ChoBC,KimD-W,KimS-W,LeeJ-S,SuW-C,etal. 5. Lynch TJ, Bell DW, Sordella R, Gurubhagavatula S, Okimoto RA, Brannigan Osimertinib for patients (pts) with leptomeningeal metastases (LM) BW, et al. Activating mutations in the receptor from EGFR-mutant non-small cell lung cancer (NSCLC): Updated underlying responsiveness of non–small-cell lung cancer to gefitinib. results from the BLOOM study. J Clin Oncol 2017;35:2020. doi: N Engl J Med 2004;350:2129–39. 10.1200/JCO.2017.35.15_suppl.2020. 6. Paez JG, J€anne PA, Lee JC, Tracy S, Greulich H, Gabriel S, et al. EGFR 19. Reungwetwattana T, Nakagawa K, Cho BC, Cobo M, Cho EK, Bertolini A, mutations in lung cancer: correlation with clinical response to gefitinib et al. CNS response to osimertinib versus standard epidermal growth factor therapy. Science 2004;304:1497. inhibitors in patients with untreated EGFR-mutat- 7. Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, ed advanced non–small-cell lung cancer. J Clin Oncol 2018. doi: 10.1200/ Fidias P, et al. Genotypic and histological evolution of lung cancers JCO.2018.78.3118. acquiring resistance to EGFR Inhibitors. Sci Transl Med 2011;3: 20. Oxnard GR, Ramalingam SS, Ahn M-J, Kim S-W, Yu HA, Saka H, et al. 75ra26–75ra26. Preliminary results of TATTON, a multi-arm phase Ib trial of AZD9291 8. Shi Y, Au JS-K, Thongprasert S, Srinivasan S, Tsai C-M, Khoa MT, et al. A combined with MEDI4736, AZD6094 or selumetinib in EGFR-mutant prospective, molecular epidemiology study of EGFR mutations in Asian lung cancer. J Clin Oncol 2015;33:2509. doi: 10.1200/jco.2015.33. patients with advanced non–small-cell lung cancer of adenocarcinoma 15_suppl.2509. histology (PIONEER). J Thoracic Oncol 2014;9:154–62. 21. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Park JO, et al. 9. Miller VA, Hirsh V, Cadranel J, Chen Y-M, Park K, Kim S-W, et al. Afatinib MET amplification leads to gefitinib resistance in lung cancer by activating versus placebo for patients with advanced, metastatic non-small-cell lung ERBB3 signaling. Science 2007;316:1039–43. cancer after failure of erlotinib, gefitinib, or both, and one or two lines of 22. Ramalingam SS, Cheng Y, Zhou C, Ohe Y, Imamura F, Cho BC, et al. chemotherapy (LUX-Lung 1): a phase 2b/3 randomised trial. Lancet Oncol Mechanisms of acquired resistance to first-line osimertinib: preliminary 2012;13:528–38. data from the phase III FLAURA study. Ann Oncol 2018;29: 10. Reckamp KL, Giaccone G, Camidge DR, Gadgeel SM, Khuri FR, Engelman mdy424.063. JA, et al. A phase 2 trial of (PF-00299804), an oral, irreversible 23. Papadimitrakopoulou VA, Wu YL, Han JY, Ahn MJ, Ramalingam SS, John T, pan-HER (human epidermal growth factor receptor) inhibitor, in patients et al. Analysis of resistance mechanisms to osimertinib in patients with with advanced non-small cell lung cancer after failure of prior chemother- EGFR T790M advanced NSCLC from the AURA3 study. Ann Oncol 2018; apy and erlotinib. Cancer 2014;120:1145–54. 29:mdy424.064. 11. Soria J-C, Ohe Y, Vansteenkiste J, Reungwetwattana T, Chewaskulyong B, 24. Ahn M, Han J, Sequist L, Cho BC, Lee JS, Kim S, et al. TATTON Ph Ib Lee KH, et al. Osimertinib in Untreated EGFR-mutated advanced non– expansion cohort: osimertinib plus savolitinib for Pts with EGFR-mutant small-cell lung cancer. N Engl J Med 2017;378:113–25. MET-amplified NSCLC after progression on prior EGFR-TKI. J Thoracic 12. Mok TS, Wu Y-L, Ahn M-J, Garassino MC, Kim HR, Ramalingam SS, et al. Oncol 2017;12:S1768. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung can- 25. Bahcall M, Sim T, Paweletz CP, Patel JD, Alden RS, Kuang Y, et al. Acquired cer. N Engl J Med 2016;376:629–40. MET D1228V mutation and resistance to MET inhibition in lung cancer. 13. Cross DAE, Ashton SE, Ghiorghiu S, Eberlein C, Nebhan CA, Spitzler PJ, Cancer Discov 2016;6:1334–41. et al. AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated 26. Ercan D, Xu C, Yanagita M, Monast CS, Pratilas CA, Montero J, et al. resistance to EGFR inhibitors in lung cancer. Cancer Discov 2014;4: Reactivation of ERK Signaling causes resistance to EGFR kinase inhibitors. 1046–61. Cancer Discov 2012;2:934–47. 14. Goss G, Tsai C-M, Shepherd FA, Bazhenova L, Lee JS, Chang G-C, et al. 27. Cortot AB, Repellin CE, Shimamura T, Capelletti M, Zejnullahu K, Ercan D, Osimertinib for pretreated EGFR Thr790Met-positive advanced non-small- et al. Resistance to irreversible EGF receptor tyrosine kinase inhibitors

696 Cancer Res; 79(4) February 15, 2019 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst February 4, 2019; DOI: 10.1158/0008-5472.CAN-18-1281

EGFR Third-Generation TKI Therapy

through a multistep mechanism involving the IGF1R pathway. Cancer Res patients with advanced EGFRm (Del 19 or L858RT790M) NSCLC. 2013;73:834–43. J Thoracic Oncol 2017;12:S1185. 28. Oxnard GR, Hu Y, Mileham KF, Husain H, Costa DB, Tracy P, et al. 47. Thress KS, Paweletz CP, Felip E, Cho BC, Stetson D, Dougherty B, et al. Assessment of resistance mechanisms and clinical implications in patients Acquired EGFR C797S mediates resistance to AZD9291 in advanced with EGFR T790M-positive lung cancer and acquired resistance to osimer- non-small cell lung cancer harboring EGFR T790M. Nat Med 2015;21: tinib. JAMA Oncol 2018;4:1527–34. 560–2. 29. Tricker EM, Xu C, Uddin S, Capelletti M, Ercan D, Ogino A, et al. Combined 48. Piotrowska Z, Nagy R, Fairclough S, Lanman R, Marcoux N, Gettinger S, et al. EGFR/MEK inhibition prevents the emergence of resistance in EGFR Characterizing the genomic landscape of EGFR C797S in lung cancer using mutant lung cancer. Cancer Discov 2015;5:960–71. ctDNA next-generation sequencing. J Thoracic Oncol 2017;12:S1767. 30. Della Corte CM, Ciaramella V, Cardone C, La Monica S, Alfieri R, Petronini 49. Niederst MJ, Hu H, Mulvey HE, Lockerman EL, Garcia AR, Piotrowska Z, PG, et al. Antitumor efficacy of dual blockade of EGFR signaling by et al. The allelic context of the C797S mutation acquired upon treatment osimertinib in combination with selumetinib or cetuximab in activated with third generation EGFR inhibitors impacts sensitivity to subsequent EGFR Human NCLC Tumor Models. J Thorac Oncol 2018;13:810–20. treatment strategies. Clin Cancer Res 2015;21:3924–33. 31. Hata AN, Niederst MJ, Archibald HL, Gomez-Caraballo M, Siddiqui FM, 50. Jia Y, Yun C-H, Park E, Ercan D, Manuia M, Juarez J, et al. Overcoming EGFR Mulvey HE, et al. Tumor cells can follow distinct evolutionary paths to T790M and C797S resistance with mutant-selective allosteric inhibitors. become resistant to epidermal growth factor receptor inhibition. Nat Med Nature 2016;534:129–32. 2016;22:262–9. 51. Ma Y, Zheng X, Zhao H, Fang W, Zhang Y, Ge J, et al. First-in-human phase I 32. Janes MR, Vu C, Mallya S, Shieh M, Limon JJ, Li L-S, et al. Efficacy of the study of AC0010, a mutant-selective EGFR inhibitor in non–small cell lung investigational mTOR kinase inhibitor MLN0128/INK128 in models of cancer: safety, efficacy, and potential mechanism of resistance. J Thoracic B-cell acute lymphoblastic . Leukemia 2013;27:586–94. Oncol 2018;13:968–77. 33. Yu H, Suzawa K, Jordan EJ, Zehir A, Ni A, Kim HR, et al. Concurrent 52. Ercan D, Choi HG, Yun C-H, Capelletti M, Xie T, Eck MJ, et al. EGFR alterations in EGFR-mutant lung cancers associated with resistance to EGFR mutations and resistance to Irreversible pyrimidine based EGFR inhibitors. kinase inhibitors and characterization of MTOR as a mediator of resistance. Clin Cancer Res 2015;21:3913–23. Clin Cancer Res 2018;24:3108–18. 53. Bersanelli M, Minari R, Bordi P, Gnetti L, Bozzetti C, Squadrilli A, et al. 34. Nukaga S, Yasuda H, Tsuchihara K, Hamamoto J, Masuzawa K, Kawada I, L718Q mutation as new mechanism of acquired resistance to AZD9291 in et al. Amplification of EGFR wild-type alleles in non–small cell lung EGFR-mutated NSCLC. J Thoracic Oncol 2016;11:e121–e3. cancer cells confers acquired resistance to mutation-selective EGFR 54. Fassunke J, Muller€ F, Keul M, Michels S, Dammert MA, Schmitt A, et al. tyrosine kinase inhibitors. Cancer Res 2017;77:2078–89. Overcoming EGFRG724S-mediated osimertinib resistance through 35. Jia Y, Juarez J, Li J, Manuia M, Niederst MJ, Tompkins C, et al. EGF816 exerts unique binding characteristics of second-generation EGFR inhibitors. anticancer effects in non–small cell lung cancer by irreversibly and selec- Nat Commun 2018;9:4655. doi: 10.1038/s41467-018-07078-0. tively targeting primary and acquired activating mutations in the EGF 55. Chabon JJ, Simmons AD, Lovejoy AF, Esfahani MS, Newman AM, receptor. Cancer Res 2016;76:1591–602. Haringsma HJ, et al. Circulating tumour DNA profiling reveals heteroge- 36. Tan DS-W, Yang JC-H, Leighl NB, Riely GJ, Sequist LV, Felip E, et al. neity of EGFR inhibitor resistance mechanisms in lung cancer patients. Updated results of a phase 1 study of EGF816, a third-generation, Nat Commun 2016;7:11815. doi: 10.1038/ncomms11815. mutant-selective EGFR tyrosine kinase inhibitor (TKI), in advanced 56.YangJC,SequistLV,GeaterSL,TsaiCM,MokTS,SchulerM,etal. non-small cell lung cancer (NSCLC) harboring T790M. J Clin Oncol Clinical activity of afatinib in patients with advanced non-small-cell 2016;34:9044. doi: 10.1200/JCO.2016.34.15_suppl.9044. lung cancer harbouring uncommon EGFR mutations: a combined post- 37. Arbour KC, Sequist LV, Piotrowska Z, Kris MG, Paik PK, Ni A, et al. hoc analysis of LUX-Lung 2, LUX-Lung 3, and LUX-Lung 6. Lancet Oncol Response to osimertinib following treatment with EGF816 in patients 2015;16:830–8. with T790M EGFR mutant NSLCLC. J Clin Oncol 2017;35:20673. 57. Mali P, Esvelt KM, Church GM. Cas9 as a versatile tool for engineering 38. Tan DS-W, Kim D-W, Leighl NB, Riely GJ, Yang JC-H, Wolf J, et al. Genomic biology. Nat Methods 2013;10:957–63. profiling of resistant tumor samples following progression on EGF816, a 58. Zhong WZ, Wu YL, Chen KN, Chen C, Gu CD, Wang Q, et al. Erlotinib third generation, mutant-selective EGFR tyrosine kinase inhibitor (TKI), in versus plus as neo-adjuvant treatment for stage IIIA- advanced non-small cell lung cancer (NSCLC). J Clin Oncol 2017;35:11506. N2 EGFR-mutation non-small cell lung cancer (EMERGING): a rando- 39. Wu Y-L, Zhang L, Kim D-W, Liu X, Lee DH, Yang JC-H, et al. Phase Ib/II mised study. Ann Oncol 2018;29:mdy424.058-mdy424.058. study of capmatinib (INC280) plus gefitinib after failure of epidermal 59. Terai H, Soejima K, Yasuda H, Nakayama S, Hamamoto J, Arai D, et al. growth factor receptor (EGFR) inhibitor therapy in patients with EGFR- Activation of the FGF2-FGFR1 autocrine pathway: a novel mechanism mutated, MET factor–dysregulated non–small-cell lung cancer. J Clin of acquired resistance to gefitinib in NSCLC. Mol Cancer Res 2013;11: Oncol 2018;36:3101–9. 759–67. 40. Cheng Y, Zhou J, Lu S, Zhang Y, Zhao J, Pan H, et al. Phase II study of 60. Blakely CM, Watkins TBK, Wu W, Gini B, Chabon JJ, McCoach CE, et al. tepotinib and gefitinib (TEPþGEF) in MET-positive (METþ)/epidermal Evolution and clinical impact of co-occurring genetic alterations in growth factor receptor (EGFR)-mutant (MT) non-small cell lung cancer advanced-stage EGFR-mutant lung cancers. Nat Genet 2017;49:1693–704. (NSCLC). Ann Oncol 2018;29:mdy292. 61. Lee J-K, Lee J, Kim S, Kim S, Youk J, Park S, et al. Clonal history and genetic 41. Sequist LV, Soria J-C, Camidge DR. Update to Rociletinib Data with the predictors of transformation into small-cell carcinomas from lung adeno- RECIST confirmed response rate. N Engl J Med 2016;374:2296–7. carcinomas. J Clin Oncol 2017;35:3065–74. 42. Sequist LV, Piotrowska Z, Niederst MJ, et al. Osimertinib responses after 62. Takezawa K, Pirazzoli V, Arcila ME, Nebhan CA, Song X, de Stanchina E, disease progression in patients who had been receiving rociletinib. et al. HER2 amplification: a potential mechanism of acquired resistance to JAMA Oncol 2016;2:541–3. EGFR inhibition in EGFR mutant lung cancers that lack the second-site 43. Park K, Lee J-S, Lee KH, Kim J-H, Cho BC, Min YJ, et al. BI 1482694 EGFR T790M mutation. Cancer Discov 2012;2:922–33. (HM61713), an EGFR mutant-specific inhibitor, in T790Mþ NSCLC: 63. Park JH, Choi YJ, Kim SY, Lee J-E, Sung KJ, Park S, et al. Activation of the efficacy and safety at the RP2D. J Clin Oncol 2016;34:9055. IGF1R pathway potentially mediates acquired resistance to mutant-selec- 44. Sugawara S, Azuma K, Nishio M, Hayashi H, Kiura K, Satouchi M, et al. tive 3rd-generation EGF receptor tyrosine kinase inhibitors in advanced Tolerability and antitumor activity of ASP8273 in TKI-naive Japanese non-small cell lung cancer. Oncotarget 2016;7:22005–15. subjects with EGFR mutation–positive non-small cell lung cancer. J Clin 64. PiotrowskaZ,IsozakiH,LennerzJK,GainorJF,LennesIT,ZhuVW,etal. Oncol 2017;35:9037. Landscape of acquired resistance to osimertinib in EGFR-mutant 45. Yu HA, Spira A, Horn L, Weiss J, West H, Giaccone G, et al. A phase I, dose NSCLC and clinical validation of combined EGFR and RET inhibition escalation study of oral ASP8273 in patients with non–small cell lung with osimertinib and BLU-667 for acquired RET fusion. Cancer Discov cancers with epidermal growth factor receptor mutations. Clin Cancer Res 2018;8:1529–39. 2017;23:7467–73. 65. Byers LA, Diao L, Wang J, Saintigny P, Girard L, Peyton M, et al. An 46. Husain H, Martins R, Goldberg S, Senico P, Ma W, Masters J, et al. P3.02b- epithelial-mesenchymal transition (EMT) gene signature predicts resis- 001 phase 1 dose escalation of PF-06747775 (EGFR-T790M inhibitor) in tance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target

www.aacrjournals.org Cancer Res; 79(4) February 15, 2019 697

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst February 4, 2019; DOI: 10.1158/0008-5472.CAN-18-1281

Murtuza et al.

for overcoming EGFR inhibitor resistance. Clin Cancer Res 2013;19: 72. Lee CK, Man J, Lord S, Links M, Gebski V, Mok T, et al. Checkpoint 279–90. inhibitors in metastatic EGFR-mutated non–small cell lung cancer—a 66. Song K-A, Niederst MJ, Lochmann TL, Hata AN, Kitai H, Ham J, et al. meta-analysis. J Thoracic Oncol 2017;12:403–7. Epithelial-to-mesenchymal transition antagonizes response to targeted 73. Akbay EA, Koyama S, Carretero J, Altabef A, Tchaicha JH, Christensen CL, therapies in lung cancer by suppressing BIM. Clin Cancer Res 2018;24: et al. Activation of the PD-1 pathway contributes to immune escape in 197–208. EGFR-driven lung tumors. Cancer Discov 2013;3:1355–63. 67. Zhang Z, Lee JC, Lin L, Olivas V, Au V, LaFramboise T, et al. Activation of the 74. Offin M, Rizvi H, Tenet M, Ni A, Sanchez-Vega F, Li BT, et al. Tumor mutation AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. burden and efficacy of EGFR-tyrosine kinase inhibitors in patients with Nat Genet 2012;44:852–60. EGFR-mutant lung cancers. Clin Cancer Res 2019;25:1063–69. 68. Izumchenko E, Chang X, Michailidi C, Kagohara L, Ravi R, Paz K, et al. The 75. Streicher K, Higgs BW, Wu S, Coffman K, Damera G, Durham N, et al. TGFb-miR200-Mig6 pathway orchestrates the EMT-associated kinase Increased CD73 and reduced IFNG signature expression in relation to switch that induces resistance to EGFR inhibitors. Cancer Res 2014;74: response rates to anti-PD-1(L1) therapies in EGFR-mutant NSCLC. J Clin 3995–4005. Oncol 2017;35:11505. doi: 10.1200/JCO.2017.35.15_suppl.11505. 69. Hart T, Chandrashekhar M, Aregger M, Steinhart Z, Brown Kevin R, 76. Naumov GN, Nilsson MB, Cascone T, Briggs A, Straume O, Akslen LA, MacLeod G, et al. High-Resolution CRISPR screens reveal fitness et al. Combined vascular endothelial growth factor receptor and epi- genes and genotype-specific cancer liabilities. Cell 2015;163: dermal growth factor receptor (EGFR) blockade inhibits tumor growth 1515–26. in xenograft models of EGFR Inhibitor Resistance. Clin Cancer Res 70. Shalem O, Sanjana NE, Hartenian E, Shi X, Scott DA, Mikkelson T, et al. 2009;15:3484–94. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science 77. Seto T, Kato T, Nishio M, Goto K, Atagi S, Hosomi Y, et al. Erlotinib alone or 2014;343:84–7. with bevacizumab as first-line therapy in patients with advanced non- 71. Seguin L, Kato S, Franovic A, Camargo MF, Lesperance J, Elliott KC, et al. A squamous non-small-cell lung cancer harbouring EGFR mutations b3 -KRAS-RalB complex drives tumor stemness and resistance to (JO25567): an open-label, randomised, multicentre, phase 2 study. EGFR inhibition. Nat Cell Biol 2014;16:457–68. Lancet Oncol 2014;15:1236–44.

698 Cancer Res; 79(4) February 15, 2019 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2019 American Association for Cancer Research. Published OnlineFirst February 4, 2019; DOI: 10.1158/0008-5472.CAN-18-1281

Novel Third-Generation EGFR Tyrosine Kinase Inhibitors and Strategies to Overcome Therapeutic Resistance in Lung Cancer

Ayesha Murtuza, Ajaz Bulbul, John Paul Shen, et al.

Cancer Res 2019;79:689-698. Published OnlineFirst February 4, 2019.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-18-1281

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2019/06/19/0008-5472.CAN-18-1281.DC1

Cited articles This article cites 74 articles, 27 of which you can access for free at: http://cancerres.aacrjournals.org/content/79/4/689.full#ref-list-1

Citing articles This article has been cited by 7 HighWire-hosted articles. Access the articles at: http://cancerres.aacrjournals.org/content/79/4/689.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/79/4/689. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2019 American Association for Cancer Research.