Skip to main content

Update on recent preclinical and clinical studies of T790M mutant-specific irreversible epidermal growth factor receptor tyrosine kinase inhibitors

Abstract

The first- and second-generation epidermal growth factor receptor tyrosine kinase inhibitors (1/2G EGFR-TKIs) gefitinib, erlotinib, and afatinib have all been approved as standard first-line treatments for advanced EGFR mutation-positive non-small cell lung cancer. The third-generation (3G) EGFR-TKIs have been developed to overcome the EGFR T790M mutation, which is the most common mechanism of acquired resistance to 1/2G EGFR-TKI treatment. This resistance mutation develops in half of the patients who respond to 1/2G EGFR-TKI therapy. The structures of the novel 3G EGFR-TKIs are different from those of 1/2G EGFR-TKIs. Particularly, 3G EGFR-TKIs have lower affinity to wild-type EGFR, and are therefore associated with lower rates of skin and gastrointestinal toxicities. However, many of the adverse events (AEs) that are observed in patients receiving 3G EGFR-TKIs have not been observed in patients receiving 1/2G EGFR-TKIs. Although preclinical studies have revealed many possible mechanisms for these AEs, the causes of some AEs remain unknown. Many mechanisms of resistance to 3G EGFR-TKI therapy have also been reported. Here, we have reviewed the recent clinical and preclinical developments related to novel 3G EGFR-TKIs, including osimertinib, rociletinib, olmutinib, EGF816, and ASP8273.

Background

The first-generation reversible epidermal growth factor receptor tyrosine kinase inhibitors (1G EGFR-TKIs) gefitinib and erlotinib are both quinazoline derivatives, as is the second-generation (2G) irreversible EGFR-TKI afatinib. These drugs are effective for treating advanced EGFR mutation-positive non-small cell lung cancer (NSCLC), especially in patients who harbor EGFR exon 21 L858R mutation (EGFR L858R) or exon 19 deletions (EGFR del19). Accordingly, all of these drugs are currently standard first-line therapies for these patients [16]. However, these drugs also inhibit wild-type EGFR (EGFRwt), and diarrhea and skin acne/rash are common adverse events (AEs). After a period of 9 to 11 months of effective treatment, acquired resistance to 1G/2G EGFR-TKIs inevitably ensues. About 50–60% of the cases of acquired resistance are attributable to the EGFR T790M mutation, which is the substitution of threonine with methionine at amino acid position 790, EGFR T790M [712]. Novel third-generation (3G) EGFR-TKIs were designed to overcome this major mechanism of resistance while also having less capacity to inhibit EGFRwt, thereby minimizing the AEs that are seen in 1G/2G EGFR-TKI therapy. Here, we have reviewed the recent preclinical and clinical developments related to 3G EGFR-TKIs with a special focus on the unusual AEs that are associated with these novel drugs. We have also reviewed the mechanisms of acquired resistance to these drugs and the possible solutions by which these resistance mechanisms may be overcome.

A literature review of clinical studies published between January 2013 and June 2016 was conducted using PubMed and MEDLINE, with the entry keywords ‘non-small cell lung cancer,’ ‘epidermal growth factor receptor T790M mutation,’ ‘osimertinib,’ ‘rociletinib,’ ‘olmutinib,’ ‘EGF816,’ and ‘ASP8273.’ We also performed a manual search of the abstracts presented at major oncology meetings.

Main text of the review

Osimertinib

Osimertinib (AZD9291) is a mono-anilino-pyrimidine compound that irreversibly targets tumors harboring EGFR L858R, EGFR del19, and EGFR T790M, while having little effect on EGFRwt. This compound makes a covalent bond with cysteine residue in position 797 of EGFR (Cys797), and also has activity against other kinases that harbor cysteine residue in the analogous kinase domain, such as ErBB2, ErBB4, and BLK (BLK proto-oncogene, Src family tyrosine kinase; previous name: B lymphoid tyrosine kinase). Like EGFR T790M, insulin receptor and insulin-like growth factor 1 receptor also have methionine gatekeeper in their kinase domains. Nonetheless, osimertinib does not have significant activity against either of these receptors [13, 14].

In the first phase I/II clinical study of orally administered osimertinib (AURA), 80 mg/day was chosen as the dose for subsequent phase II or III studies, even though a true dose-limiting toxicity was not observed at this dose level [15]. In a pooled analysis of two studies (AURA phase II extension cohort and AURA 2), outcomes were examined for patients who had EGFR mutation-positive NSCLC, whose disease had progressed following previous EGFR-TKI therapy, whose tumors harbored EGFR T790M, and who had been treated with osimertinib at 80 mg/day. Among the 397 evaluable patients, the confirmed objective response rate (ORR) was 66% and the disease control rate (DCR) was 91%. The median progression-free survival (PFS) was 11.0 months (n = 411). The observed treatment-related AEs are listed in Table 1, and only < 1% of the patients developed grade ≥ 3 skin rash or diarrhea. Three percent of the patients developed interstitial lung disease (ILD) and QT interval corrected for heart rate (QTc) prolongation while hyperglycemia developed in less than 1% of the patients [16]. Some grade ≥ 3 laboratory abnormalities, such as neutropenia (3.4%), lymphopenia (3.3%), thrombocytopenia (1.2%), and hyponatremia (3.4%) were also reported [17].

Table 1 Selected clinical efficacy reports of third-generation EGFR-TKIs

In November 2015, osimertinib received US Food and Drug Administration (FDA) approval for EGFR–TKI-pretreated metastatic EGFR T790M-positive NSCLC, as did the companion diagnostic test (cobas® EGFR Mutation Test v2) that is used to detect tumor EGFR T790M. By July 2016, osimertinib had also received approvals in the European Union, Japan, South Korea, Canada, Switzerland, Israel, and Mexico. A confirmatory phase III study (AURA 3, ClinicalTrials.gov, NCT02151981) is comparing osimertinib with platinum-based chemotherapy in patients who have advanced EGFR mutation-positive NSCLC, whose disease progressed following first-line EGFR-TKI therapy, and whose tumors harbor EGFR T790M. This study has completed patient accrual and is ongoing.

Because osimertinib has activity against sensitizing EGFR mutations and is associated with reduced skin rash and diarrhea AEs, it has also been tested as a first-line treatment for metastatic EGFR mutation-positive NSCLC. Two expansion cohorts in the AURA study enrolled patients with metastatic EGFR mutation-positive NSCLC and tested the safety and efficacy of first-line osimertinib monotherapy. Osimertinib monotherapy was tested at 80 and 160 mg/day, and a total of 60 patients were enrolled (30 at each dose level). The ORR was 67% at 80 mg/day and 87% at 160 mg/day. The DCR was 93% at 80 mg/day and 100% at 160 mg/day. For 80 mg/day, the median PFS had not been reached at the time of the data cutoff for the analysis, and the 18-month progression-free survival rate was 57%. For 160 mg/day, the median PFS was 19.3 months, and the 18-month progression-free survival rate was 53%. All grades skin rash and diarrhea developed in 70 and 87% of patients receiving 80 mg/day, respectively, as well as 60 and 80% of patients receiving 160 mg/day, respectively. Three percent and 7% of patients developed grade ≥ 3 skin rash and grade ≥ 3 diarrhea at 160 mg/day, respectively. ILD and QTc prolongation developed in 10 and 0% of patients receiving 80 mg/day, as well as 7 and 10% of patients receiving 160 mg/day, respectively [18]. A phase III randomized study (FLAURA study, ClinicalTrials.gov, NCT02296125) is comparing osimertinib with gefitinib or erlotinib as first-line therapies in patients with advanced EGFR mutation-positive NSCLC. The study has completed patient accrual and is ongoing.

Osimertinib showed clinical activity for brain metastases in the AURA and AURA 2 studies [19]. Leptomeningeal metastasis is another detrimental complication of advanced EGFR mutation-positive NSCLC [20]. A phase I study (BLOOM study, ClinicalTrials.gov, NCT02228369) is ongoing to test the safety and efficacy of osimertinib monotherapy against brain and leptomeningeal metastasis. In a preliminary report, osimertinib at 160 mg/day showed promising activity against leptomeningeal metastasis [21].

Combination therapy is another treatment strategy for conferring better anti-tumor activity. In the TATTON study (ClinicalTrials.gov, NCT02143466), osimertinib was combined with either MET inhibitor (AZD6094, savolitinib), MEK inhibitor (selumetinib), or anti-PD-L1 monoclonal antibody (MEDI4736, durvalumab) [22]. However, a preliminary report showed that the incidence of ILD was high in the osimertinib plus durvalumab arm. A phase III study of osimertinib plus durvalumab versus osimertinib monotherapy (CAURAL study, ClinicalTrials.gov, NCT02454933), also showed a high incidence of ILD in the combination arm, and the development of osimertinib plus durvalumab combination therapy was therefore discontinued [23]. Investigations of other combination therapies are ongoing, such as for osimertinib in combination with necitumumab, ramucirumab, or bevacizumab (ClinicalTrials.gov, NCT02496663, 02789345, and 02803203). In addition to metastatic disease, clinical trials of osimertinib monotherapy for EGFR mutation-positive NSCLC are also ongoing in the adjuvant setting (ADAURA study, ClinicalTrials.gov, NCT02511106).

Another important issue in the development of 3G EGFR-TKIs is the application of liquid biopsies to detect EGFR T790M in blood or urine samples [2427]. Plasma samples were collected in the AURA study, and the cell-free plasma DNA was genotyped using the beads, emulsions, amplification, and magnetics (BEAMing) digital polymerase chain reaction technique (Sysmex Inostics, Inc., Mundelein, IL, USA) [25, 28]. The plasma-based sensitivity for detecting EGFR T790M was 70%. The ORR and median PFS were similar in patients with EGFR T790M-positive plasma and those with EGFR T790M-positive tissue, which was defined as the gold standard (ORR: 63 vs. 62%; median PFS: 9.7 vs. 9.7 months). The authors concluded that patients with EGFR T790M-positive plasma could avoid tumor re-biopsy for EGFR T790M testing, while those with EGFR T790M-negative plasma should undergo tumor re-biopsy [29]. Mechanisms of resistance to osimertinib therapy have also been reported. In a preclinical cell line study, acquired EGFR C797S mutation (the substitution of cysteine with serine at amino acid position 797, EGFR C797S) was identified as a mechanism of resistance to osimertinib therapy. This is understandable because osimertinib forms a key covalent bond with EGFR at the position of the noted cysteine residue. Resistant cells that contain sensitizing mutations (EGFR L858R/C797S and EGFR del19/C797S) retain sensitivity to quinazoline-based 1G/2G EGFR-TKIs, such as gefitinib and afatinib, but are resistant to these drugs in the presence of EGFR T790M (EGFR L858R/T790M/C797S and EGFR del19/T790M/C797S). However, resistant cells that harbor EGFR L858R/T790M/C797S remain partially sensitive to cetuximab because of the disruption of EGFR dimerization [30]. Another preclinical study also provided evidence that EGFR C797S is a resistance mechanism [31]. Furthermore, the authors of that study demonstrated that, if EGFR C797S occurred in trans (on a different allele) from EGFR T790M, then the resistant cells were sensitive to a combination of 1G/3G EGFR-TKIs. In contrast, if the two mutations occurred in cis (on the same allele), then the cells were resistant to the combination therapy. Other mechanisms of resistance to osimertinib therapy have also been identified in cell line studies, including NRAS E63K mutation and gains of copy number for wild-type NRAS and wild-type KRAS. Combination therapy with osimertinib and selumetinib prevented and delayed the developments of resistance [32].

In the clinical setting, EGFR C797S was first described in a patient who developed acquired resistance to osimertinib therapy. In this case, EGFR C797S was detected in a cell-free plasma DNA analysis that was performed using next-generation sequencing. A subsequent study collected plasma samples from 15 patients who received osimertinib therapy and had pre-existing plasma EGFR T790M that was detected using droplet digital polymerase chain reaction. Upon developing resistance, 6 (40%) patients had EGFR del19/T790M/C797S, 5 (33%) patients had EGFR T790M alone, and EGFR T790M was no longer detectable in 4 (27%) patients who retained prior sensitizing mutations [33]. Mechanistically, EGFR C797S parallels the acquired Bruton tyrosine kinase (BTK) C481S mutation, which is observed in patients with chronic lymphocytic leukemia who develop acquired resistance to therapy with ibrutinib, an irreversible BTK inhibitor. Provided in combination with cetuximab, a novel EGFR–resistance-mutation selective allosteric inhibitor (EAI045) has been observed to be effective in a mouse model of NSCLC harboring EGFR L858R/T790M/C797S. Cetuximab blocks the dimerization of EGFR and renders the kinase susceptible to this allosteric agent [34]. EGFR C797S was also detected in tumor re-biopsy samples from a patient who developed acquired resistance to osimertinib [35]. Several other acquired resistance mechanisms were reported in patients who experienced disease progression on osimertinib therapy: acquired EGFR L718Q, small cell transformation, MET amplification, HER2 amplification, BRAF V600E mutation, PIK3CA E545K mutation, loss of EGFR T790M plus alternative pathway activation, and EGFR ligand-dependent activation [3641]. In a case report, a patient who developed MET amplification responded to therapy with crizotinib, an ALK and MET inhibitor [36].

Rociletinib

Rociletinib (CO-1686) is a 2,4-disubstituted pyrimidine compound that irreversibly targets tumors harboring EGFR L858R, EGFR del19, and EGFR T790M, while having little effect on EGFRwt. There is a meta-acrylamide that points to Cys797 and forms the covalent bond. This compound also has activities against other kinases, such as FAK, CHK2, ErBB4, and JAK3 [42, 43]. A metabolite of rociletinib, M502, has potency against insulin receptor and insulin-like growth factor 1 receptor, which may lead to the AE of hyperglycemia [44].

An early-phase clinical study (TIGER X, ClinicalTrials.gov, NCT01526928) determined that orally administered rociletinib at 500 mg twice per day was the recommended dose for subsequent clinical study [45, 46]. An update report of the 208 patients who received rociletinib at 500 mg twice per day disclosed that any grade of hyperglycemia, diarrhea, nausea, and QTc prolongation developed in 57.2, 56.7, 43.8, and 26.4% of the patients, respectively. Of the patients, 28.8 and 7.7% developed grade ≥ 3 hyperglycemia and grade ≥ 3 QTc prolongation, respectively. ILD was observed in 0.5% of the patients at this dose level [47]. An unexpected AE of sudden-onset cataract developed in 21 of 40 patients (53%) who were treated with rociletinib in a single-hospital study, and most of these patients required surgical repair [48]. In the TIGER-X study, the overall incidence of cataract was 9.1% in patients who received rociletinib therapy at dose levels of 500–750 mg twice per day [47]. Additionally, this drug has limited activity against central nervous system metastases [49, 50]. In the first report of the TIGER-X study, the ORR of rociletinib in patients who harbored EGFR T790M was 59% [45]; however, those patients who achieved partial response (PR) as their best response did not have a subsequent confirmed PR at least 4 weeks apart, per the RECIST criteria (version 1.1) [51]. Rociletinib did not receive accelerated approval by the US FDA. An official report of the TIGER-X study has updated the confirmed ORR to 33.9% for the efficacy population of 443 patients who received rociletinib at dose levels of 500–750 mg twice per day, and who had centrally confirmed EGFR T790M-positive NSCLC. The PFS was 5.7 months in 208 patients who received rociletinib therapy at 500 mg twice per day. The safety profile of 548 patients from the study is shown in Table 1 [47]. A biomarker study that used tissue, plasma (BEAMing), and urine specimens (Trovera Quantitative NGS assay, Trovagene, San Diego, CA, USA) to detect EGFR T790M showed sensitivities of 80.9 and 81.1% based on plasma and urine, respectively. The confirmed ORRs in patients with EGFR T790M-positive tissue, plasma, and urine were 33.9, 32.1, and 36.7%, respectively. Patients with M1a/M0 intrathoracic disease had lower plasma sensitivity than did patients with M1b distant metastatic disease (56.8 vs. 88.4%, p < .001) [52].

Ongoing clinical studies include the TIGER-2 study (ClinicalTrials.gov, NCT02147990), which is a phase II study that seeks to test the safety and efficacy of rociletinib as a second-line treatment for advanced EGFR mutation-positive NSCLC, following progression on prior EGFR-TKI therapy. Additionally, the TIGER-3 study (ClinicalTrials.gov, NCT02322281) is a phase III study that seeks to compare rociletinib with single-agent cytotoxic chemotherapy as a third-line or later treatment for advanced EGFR mutation-positive NSCLC in patients for whom EGFR-TKI and platinum-doublet therapy have failed. Patients with EGFR T790M-positive and -negative disease were both eligible for the two aforementioned studies.

Rociletinib is also being tested in the first-line setting in TIGER-1 (ClinicalTrials.gov, NCT02186301), which is a randomized phase II/III study that is comparing rociletinib with erlotinib as first-line treatments for advanced EGFR mutation-positive NSCLC. Studies of combination therapies are ongoing, such as for rociletinib in combination with trametinib (a MEK inhibitor) (ClinicalTrials.gov, NCT02580708) or atezolizumab (MPDL3280A, an anti-PD-L1 monoclonal antibody) (ClinicalTrials.gov, NCT02630186). However, in May 2016, Clovis Oncology, Inc. announced that it had terminated enrollment in all ongoing sponsored studies of rociletinib and withdrawn its Marketing Authorization Application for rociletinib from European regulatory authorities [53].

A preclinical study identified epithelial-mesenchymal transition as a possible mechanisms of resistance to rociletinib therapy that can be overcome by AKT inhibitors [43]. A cell line study identified MET amplification with or without EGFR T790M as a mechanism of resistance to CNX-2006 (tool compound of rociletinib). In a cell line that had MET amplification without EGFR T790M, MET tyrosine kinase inhibitor was able to overcome the resistance by itself, suggesting an oncogenic shift from EGFR to MET. The authors described this phenomenon as “oncogene swap” [54].

In clinical studies, mechanisms of resistance to rociletinib were identified by using plasma circulating tumor DNA profiling (Cancer Personalized Profiling by Deep Sequencing, CAPP-Seq). MET copy number gain is the most frequent mechanism, and was observed in 43 (26%) patients in a recent study [55]. In this study, rociletinib-resistant xenografts also developed MET amplification that could be overcome using crizotinib therapy. EGFR C797S and novel EGFR L798I were each identified in one patient. Other mechanisms involving MET, EGFR, PIK3CA, ERRB2, KRAS, and RB1 were also described. Nineteen percent of the patients displayed resistance mechanisms that affected multiple genes, a finding that further emphasizes the importance of tumor heterogeneity [55]. In another study, tumor re-biopsy after progression on rociletinib therapy showed that loss of EGFR T790M plus EGFR amplification and small cell transformation were mechanisms of resistance. The authors emphasized the concept of tumor heterogeneity, in which EGFR T790M-positive and -negative tumor cells may coexist in a tumor before rociletinib therapy, and for which it is not sufficient to target only one mechanism of resistance [56]. A short clinical report demonstrated that, following the development of resistance to rociletinib, some patients still responded to osimertinib therapy [57].

Olmutinib

Olmutinib (BI 1482694 / HM61713) is an irreversible kinase inhibitor that binds to a cysteine residue near the kinase domain. Olmutinib shows activities against cell lines and xenograft tumors harboring EGFR L858R/T790M and EGFR del19, while having little effect on cell lines with EGFRwt [58].

In the first phase I/II study (ClinicalTrials.gov, NCT01588145) conducted in South Korea, orally administered olmutinib at 800 mg/day was identified as the recommended dose for subsequent studies. In that study, 76 EGFR T790M-positive patients received olmutinib therapy at a dose of 800 mg/day, and the median PFS was 6.9 months. The confirmed ORR was 54% among 70 evaluable patients, and activity against central nervous system metastases was also observed [59]. The treatment-related AEs from this study are listed in Table 1. One patient experienced ILD and discontinued therapy, but there was no AE of hyperglycemia [59].

In December 2015, olmutinib was granted the breakthrough therapy designation for NSCLC by the US FDA. In May 2016, it was approved in South Korea for advanced EGFR T790M-positive NSCLC patients who were pretreated with EGFR-TKIs [60]. Regarding the future clinical development of olmutinib, the phase II ELUXA 1 study (ClinicalTrials.gov, NCT02485652) is recruiting patients with advanced EGFR T790M-positive NSCLC after prior EGFR-TKI therapy. Other global clinical trial plans for olmutinib are available online [61]. Regarding first-line treatment, a phase II study of first-line olmutinib for advanced EGFR mutation-positive NSCLC was conducted in South Korea (ClinicalTrials.gov, NCT02444819).

Acquired resistance to olmutinib was reported in a patient who developed EGFR C797S after a period of effective olmutinib therapy [62].

EGF816

EGF816 irreversibly targets EGFR by forming a covalent bond to Cys797. Preclinical data show activities against cell lines and xenograft models harboring EGFR L858R/T790M, EGFR del19/T790M, EGFR del19, and EGFR L858R, while having little effect on cell lines harboring EGFRwt [63]. In an early-phase clinical study (ClinicalTrials.gov, NCT02108964), 152 patients were enrolled to receive orally administered EGF816 at 75–350 mg/day. The common AEs in this study are shown in Table 1. Grade ≥ 3 AEs included rash (16.4%), anemia (2.6%), urticaria (2.6%), diarrhea (2.0%), and fatigue (2.0%). The type and distribution of the skin rash were different from the acneiform rash that is observed in patients treated with 1G/2G EGFR-TKIs. Two and 2 patients developed hepatitis B virus reactivation and increased lipase level, respectively [64]. The confirmed ORR and DCR among 147 evaluable patients were 46.9 and 87.1%, respectively. The estimated PFS across all dose levels was 9.7 months [64]. In a preclinical study, EGF816 also showed activity against EGFR exon 20 insertion. Therefore, the early-phase study also enrolled patients with tumors harboring this genetic alteration [63]. A study of the combination therapy of EGF816 and nivolumab (an anti-PD-1 monoclonal antibody) is ongoing (ClinicalTrials.gov, NCT02323126).

A preclinical study demonstrated several mechanism of resistance to EGF816, including EGFR C797S, MET amplification, and epithelial-mesenchymal transition. Dual inhibition of EGFR and cMET with EGF816 and INC280 (a MET inhibitor) can overcome this resistance mechanism [63]. A phase Ib/II study of EGF816 and INC280 combination therapy for advanced EGFR mutation-positive NSCLC is in progress (ClinicalTrials.gov, NCT02335944).

ASP8273

ASP8273 is a mutant-selective irreversible EGFR inhibitor that targets EGFR by forming a covalent bond to Cys797. Preclinical data show activities against cell lines and xenograft models harboring EGFR L858R/T790M, EGFR del19/T790M, EGFR del19, and EGFR L858R, while having little effect on cell lines harboring EGFRwt [65, 66]. In a first-in-human phase I/II study (ClinicalTrials.gov, NCT02192697) conducted in Japan, ASP8273 has been well tolerated across multiple dose levels. Common treatment-related AEs have included diarrhea, vomiting, nausea, hyponatremia, increased serum alanine transaminase level, and thrombocytopenia. A few patients have experienced rash, QTc prolongation, and ILD-like events. Three hundred mg/day was chosen as the recommended dose for subsequent phase II studies [67]. An update report showed a preliminary ORR (including both confirmed and unconfirmed response) of 64% in 70 EGFR T790M-positive patients treated with ASP8273 at 300 mg/day [68]. This study is ongoing in Japan, South Korea, and Taiwan.

In another phase I study that was conducted in the US (ClinicalTrials.gov, NCT02113813), orally administered ASP8273 at 300 mg/day was chosen as the recommended dose for subsequent phase II studies. A total of 110 patients were enrolled and 63 patients received treatment at the dose of 300 mg/day. Ninety-two percent of these 63 patients harbored EGFR T790M. The confirmed ORR was 30% and the median PFS was 6.0 months [69]. Common treatment-related AEs are listed in Table 1. Grade ≥ 3 AEs included diarrhea (2%) and hyponatremia (13%). One subject developed grade 1 hyperglycemia, and no patient developed ILD or QTc prolongation [69].

ASP8273 was mainly developed as a first-line treatment for advanced EGFR mutation-positive NSCLC in a phase II study in Japan (ClinicalTrials.gov, NCT02500927). The global randomized phase III SOLAR study (ClinicalTrials.gov, NCT02588261) is comparing ASP8273 with gefitinib or erlotinib as first-line treatments for advanced EGFR mutation-positive NSCLC.

Others

Other third-generation EGFR-TKIs are currently under early clinical development, including PF-06747775 (Pfizer, ClinicalTrials.gov, NCT02349633), avitinib (Hangzhou ACEA Pharmaceutical Research Co., Ltd., ClinicalTrials.gov, NCT02330367 and NCT02274337), brigatinib (ARIAD, AP26113, ALK and EGFR inhibitor, ClinicalTrials.gov, NCT01449461), and TAS-121 (Taiho Pharma) [7075].

Discussion

The EGFR T790M mechanism of acquired resistance to 1G EGFR-TKIs was first reported in 2005 [9, 10]. EGFR T790M is also the major mechanism of acquired resistance to therapy using the 2G EGFR-TKI afatinib [7]. The prognostic role of acquired EGFR T790M was controversial before the era of 3G EGFR-TKIs [12, 26, 76, 77]. Indeed, there were many different theories about the development of drug-resistant cells, such as the pre-existence theory and evolution from drug-tolerant cells. The drug-tolerant state may provide an environment in which heterogeneous drug-resistance mechanisms can be developed [7881].

3G EGFR-TKIs have been developed to address the EGFR T790M resistance mechanism, and phase I dose finding studies of these 3G EGFR-TKIs have been conducted almost exclusively for patients with the EGFR T790M mutation. These “atypical” phase I studies have several characteristics in common: First, these studies (for example AURA and TIGER-X) enrolled large numbers of patients in their phase I components. Second, given the effectiveness of the 3G EGFR-TKIs, these studies expanded patient numbers at the same dose level and were extended to the phase II component within the same overall study, in order to accelerate accruals. Third, because of the relatively low rates of treatment-related toxicities that were observed, the recommended phase II/III doses are not necessary the true maximum tolerated doses. Fourth, because of the complexities of the trial designs, caution is warranted when interpreting their clinical outcomes. The application of liquid biopsies (plasma or urine) using various modalities is one of the attractive features of these studies, and the concordance rates between tissue and liquid biopsy results have been high [29, 52]. Nonetheless, the current diagnostic standard for EGFR T790M is still tumor re-biopsy for tissue sampling, and the role of liquid biopsy should be explored further in prospective trials [82, 83].

The resistance mechanisms to 3G EGFR-TKIs were discovered either via preclinical investigations, clinical liquid biopsy, or tumor re-biopsy. Some of the resistance mechanisms revealed in preclinical studies were later confirmed in clinical studies. EGFR C797S and MET amplification/copy number gain are the most important of the mechanisms of resistance. Novel treatment strategies are under development to overcome these resistance mechanisms [30, 31, 33, 3640, 5457, 62]. In light of tumor heterogeneity, EGFR T790M-positive and -negative cells may coexist in the same tumor, or may occur at different sites in the same patient. 3G EGFR-TKI-based combination therapies that seek to overcome the bypass pathways are a reasonable strategy for addressing the coexistence of EGFR T790M-positive and -negative cells [39, 56, 84].

Some distinct AEs have been observed in patients who received 3G EGFR-TKI therapy. For example, neutropenia, lymphopenia, thrombocytopenia, and QTc prolongation have been observed in patients receiving osimertinib; hyperglycemia and cataract have been observed in patients receiving rociletinib; distinct skin rash and hepatitis B virus reactivation have been observed in patients receiving EGF816; and hyponatremia and paresthesia have been observed in patients receiving ASP8273. Although some of these AEs are attributable to off-target effects, the causes of others remain unknown. These rare and unexpected AEs were prudently observed by the investigators in these early-phase studies, and information on their occurrence spread to all collaborators in a timely manner; the importance of tight global collaboration cannot be overemphasized [85]. The incidences of ILD were low in the clinical trials, although they excluded patients with a history of ILD.

Given their high inhibitory activity against sensitizing EGFR mutations, some 3G EGFR-TKIs are under development as first-line treatments that are being compared with 1G/2G EGFR-TKIs (FLAURA [osimertinib], TIGER-1 [rociletinib], ELUXA 1 [olmutinib], SOLAR [ASP8273] studies). Investigators should be more cautious of ILD and other AEs associated with these therapies. When administered as a first-line treatment, the combination therapy of osimertinib and durvalumab induced a high incidence of ILD, which serves as an important reminder that the same drugs can have unexpected consequences when provided in different clinical settings and combinations [23].

In summary, the development of 3G EGFR-TKIs is encouraging because they have often shown noteworthy effectiveness and reduced rates of classic AEs, such as diarrhea and skin rash. The application of these drugs in different settings and combinations should be explored in the future.

Conclusions

Osimertinib therapy is the current standard of care for patients with EGFR mutation-positive NSCLC who developed acquired EGFR T790M after prior EGFR-TKI therapy. Other 3G EGFR-TKIs are currently under development. Liquid biopsy is being investigated as a means of both detecting plasma/urine EGFR T790M prior to 3G EGFR-TKI therapy and identifying mechanisms of resistance to 3G EGFR-TKI therapy. Various combination therapies that incorporate a 3G EGFR-TKI aim to prolong the benefits of 3G EGFR-TKIs and/or overcome resistance mechanisms. Many clinical trials of first-line 3G EGFR-TKI therapies are in progress, and their findings may ultimately change the paradigm of standard first-line treatment for EGFR mutation-positive NSCLC.

Abbreviations

1G:

First-generation

2G:

Second-generation

3G:

Third-generation

AEs:

Adverse events

DCR:

Disease control rate

EGFR:

Epidermal growth factor receptor

EGFR C797S :

EGFR C797S mutation

EGFR del19 :

EGFR exon 19 deletions

EGFR L858R :

EGFR exon 21 L858R mutation

EGFR T790M :

EGFR T790M mutation

EGFRwt :

Wild-type EGFR

EGFR-TKIs:

EGFR tyrosine kinase inhibitors

FDA:

Food and Drug Administration

ILD:

Interstitial lung disease

NSCLC:

Non-small cell lung cancer

ORR:

Objective response rate

PFS:

Progression-free survival

PR:

Partial response

QTc:

QT interval corrected for heart rate

References

  1. Yang JC-H, Hirsh V, Schuler M, Yamamoto N, O’Byrne KJ, Mok TSK, et al. Symptom control and quality of life in LUX-Lung 3: a phase III study of afatinib or cisplatin/pemetrexed in patients with advanced lung adenocarcinoma with EGFR mutations. J Clin Oncol. 2013;31:3342–50.

    Article  CAS  PubMed  Google Scholar 

  2. Sequist LV, Yang JC-H, Yamamoto N, O’Byrne K, Hirsh V, Mok T, et al. Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations. J Clin Oncol. 2013;31:3327–34.

    Article  CAS  PubMed  Google Scholar 

  3. Rosell R, Carcereny E, Gervais R, Vergnenegre A, Massuti B, Felip E, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2012;13:239–46.

    Article  CAS  PubMed  Google Scholar 

  4. Mitsudomi T, Morita S, Yatabe Y, Negoro S, Okamoto I, Tsurutani J, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 2010;11:121–8.

    Article  CAS  PubMed  Google Scholar 

  5. Maemondo M, Inoue A, Kobayashi K, Sugawara S, Oizumi S, Isobe H, et al. Gefitinib or chemotherapy for non–small-cell lung cancer with mutated EGFR. N Engl J Med. 2010;362:2380–8.

    Article  CAS  PubMed  Google Scholar 

  6. Yang C-H, Yu C-J, Shih J-Y, Chang Y-C, Hu F-C, Tsai M-C, et al. Specific EGFR mutations predict treatment outcome of stage IIIB/IV patients with chemotherapy-naive non–small-cell lung cancer receiving first-line gefitinib monotherapy. J Clin Oncol. 2008;26:2745–53.

    Article  CAS  PubMed  Google Scholar 

  7. Wu S-G, Liu Y-N, Tsai M-F, Chang Y-L, Yu C-J, Yang P-C, et al. The mechanism of acquired resistance to irreversible EGFR tyrosine kinase inhibitor-afatinib in lung adenocarcinoma patients. Oncotarget. 2016;7:12404–13.

    PubMed  PubMed Central  Google Scholar 

  8. Yu HA, Arcila ME, Rekhtman N, Sima CS, Zakowski MF, Pao W, et al. Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers. Clin Cancer Res. 2013;19:2240–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Pao W, Miller VA, Politi KA, Riely GJ, Somwar R, Zakowski MF, et al. Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain. PLoS Med. 2005;2:e73.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Kobayashi S, Boggon TJ, Dayaram T, Jänne PA, Kocher O, Meyerson M, et al. EGFR mutation and resistance of non–small-cell lung cancer to gefitinib. N Engl J Med. 2005;352:786–92.

    Article  CAS  PubMed  Google Scholar 

  11. Sequist LV, Waltman BA, Dias-Santagata D, Digumarthy S, Turke AB, Fidias P, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med. 2011;3:75ra26.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Kuiper JL, Heideman DAM, Thunnissen E, Paul MA, van Wijk AW, Postmus PE, et al. Incidence of T790M mutation in (sequential) rebiopsies in EGFR-mutated NSCLC-patients. Lung Cancer. 2014;85:19–24.

    Article  CAS  PubMed  Google Scholar 

  13. Cross DAE, Ashton SE, Ghiorghiu S, Eberlein C, Nebhan CA, Spitzler PJ, et al. AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer. Cancer Discov. 2014;4:1046–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Finlay MRV, Anderton M, Ashton S, Ballard P, Bethel PA, Box MR, et al. Discovery of a potent and selective EGFR inhibitor (AZD9291) of both sensitizing and T790M resistance mutations that spares the wild type form of the receptor. J Med Chem. 2014;57:8249–67.

    Article  CAS  PubMed  Google Scholar 

  15. Jänne PA, Yang JC-H, Kim D-W, Planchard D, Ohe Y, Ramalingam SS, et al. AZD9291 in EGFR inhibitor–resistant non–small-cell lung cancer. N Engl J Med. 2015;372:1689–99.

    Article  PubMed  Google Scholar 

  16. Yang J, Ramalingam SS, Jänne PA, Cantarini M, Mitsudomi T. LBA2_PR: osimertinib (AZD9291) in pre-treated pts with T790M-positive advanced NSCLC: updated Phase 1 (P1) and pooled Phase 2 (P2) results. J Thorac Oncol. 2016;11 Suppl 4:152–3.

    Article  Google Scholar 

  17. Tagrisso (Osimertinib) Tablet Prescribing Information. AstraZeneca Pharmaceuticals, Wilmington, DE. Available at http://www.azpicentral.com/tagrisso/tagrisso.pdf. Accessed 8 July 2016.

  18. Ramalingam S, Yang JCH, Lee CK, Kurata T, Kim DW, John T, et al. LBA1_PR: Osimertinib as first-line treatment for EGFR mutation-positive advanced NSCLC: updated efficacy and safety results from two phase I expansion cohorts. J Thorac Oncol. 2016;11 Suppl 4:152.

    Article  Google Scholar 

  19. Ahn MJ, Tsai CM, Yang JCH, Shepherd FA, Satouchi M, Kim DW, et al. 3083 AZD9291 activity in patients with EGFR-mutant advanced non-small cell lung cancer (NSCLC) and brain metastases: data from phase II studies. Eur J Cancer. 2015;51 Suppl 3:625-S6.

    Google Scholar 

  20. Liao B-C, Lee J-H, Lin C-C, Chen Y-F, Chang C-H, Ho C-C, et al. Epidermal growth factor receptor tyrosine kinase inhibitors for non–small-cell lung cancer patients with leptomeningeal carcinomatosis. J Thorac Oncol. 2015;10:1754–61.

    Article  CAS  PubMed  Google Scholar 

  21. Yang JC-H, Kim D-W, Kim S-W, Cho BC, Lee J-S, Ye X, et al. Osimertinib activity in patients (pts) with leptomeningeal (LM) disease from non-small cell lung cancer (NSCLC): updated results from BLOOM, a phase I study. ASCO Meeting Abstracts. 2016;34:9002.

    Google Scholar 

  22. Oxnard GR, Ramalingam SS, Ahn M-J, Kim S-W, Yu HA, Saka H, et al. Preliminary results of TATTON, a multi-arm phase Ib trial of AZD9291 combined with MEDI4736, AZD6094 or selumetinib in EGFR-mutant lung cancer. ASCO Meeting Abstracts. 2015;33:2509.

    Google Scholar 

  23. Ahn MJ, Yang J, Yu H, Saka H, Ramalingam S, Goto K, et al. 136O: Osimertinib combined with durvalumab in EGFR-mutant non-small cell lung cancer: results from the TATTON phase Ib trial. J Thorac Oncol. 2016;11 Suppl 4:115.

    Article  Google Scholar 

  24. Sacher AG, Paweletz C, Dahlberg SE, et al. Prospective validation of rapid plasma genotyping for the detection of EGFR and KRAS mutations in advanced lung cancer. JAMA Oncol. 2016;2:1014–22.

    Article  PubMed  Google Scholar 

  25. Thress KS, Brant R, Carr TH, Dearden S, Jenkins S, Brown H, et al. EGFR mutation detection in ctDNA from NSCLC patient plasma: a cross-platform comparison of leading technologies to support the clinical development of AZD9291. Lung Cancer. 2015;90:509–15.

    Article  PubMed  Google Scholar 

  26. Zheng D, Ye X, Zhang MZ, Sun Y, Wang JY, Ni J, et al. Plasma EGFR T790M ctDNA status is associated with clinical outcome in advanced NSCLC patients with acquired EGFR-TKI resistance. Sci Rep. 2016;6:20913.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Oxnard GR, Paweletz CP, Kuang Y, Mach SL, O’Connell A, Messineo MM, et al. Noninvasive detection of response and resistance in EGFR-mutant lung cancer using quantitative next-generation genotyping of cell-free plasma DNA. Clin Cancer Res. 2014;20:1698–705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Taniguchi K, Uchida J, Nishino K, Kumagai T, Okuyama T, Okami J, et al. Quantitative detection of EGFR mutations in circulating tumor DNA derived from lung adenocarcinomas. Clin Cancer Res. 2011;17:7808–15.

    Article  CAS  PubMed  Google Scholar 

  29. Oxnard GR, Thress KS, Alden RS, Lawrance R, Paweletz CP, Cantarini M, et al. Association between plasma genotyping and outcomes of treatment with osimertinib (AZD9291) in advanced non–small-cell lung cancer. J Clin Oncol. 2016. doi:10.1200/JCO.2016.66.7162.

    Google Scholar 

  30. Ercan D, Choi HG, Yun C-H, Capelletti M, Xie T, Eck MJ, et al. EGFR mutations and resistance to irreversible pyrimidine-based EGFR inhibitors. Clin Cancer Res. 2015;21:3913–23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Niederst MJ, Hu H, Mulvey HE, Lockerman EL, Garcia AR, Piotrowska Z, et al. The allelic context of the C797S mutation acquired upon treatment with third-generation EGFR inhibitors impacts sensitivity to subsequent treatment strategies. Clin Cancer Res. 2015;21:3924–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Eberlein CA, Stetson D, Markovets AA, Al-Kadhimi KJ, Lai Z, Fisher PR, et al. Acquired resistance to the mutant-selective EGFR inhibitor AZD9291 is associated with increased dependence on RAS signaling in preclinical models. Cancer Res. 2015;75:2489–500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Thress KS, Paweletz CP, Felip E, Cho BC, Stetson D, Dougherty B, et al. Acquired EGFR C797S mutation mediates resistance to AZD9291 in non-small cell lung cancer harboring EGFR T790M. Nat Med. 2015;21:560–2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Jia Y, Yun C-H, Park E, Ercan D, Manuia M, Juarez J, et al. Overcoming EGFR (T790M) and EGFR (C797S) resistance with mutant-selective allosteric inhibitors. Nature. 2016;534:129–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Yu HA, Tian SK, Drilon AE, Borsu L, Riely GJ, Arcila ME, Ladanyi M, et al. Acquired resistance of EGFR-mutant lung cancer to a T790M-specific EGFR inhibitor: emergence of a third mutation (C797S) in the EGFR tyrosine kinase domain. JAMA Oncol. 2015;1:982–4.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Ou S-HI, Agarwal N, Ali SM. High MET amplification level as a resistance mechanism to osimertinib (AZD9291) in a patient that symptomatically responded to crizotinib treatment post-osimertinib progression. Lung Cancer. 2016;98:59–61.

    Article  PubMed  Google Scholar 

  37. Ham JS, Kim S, Kim HK, Byeon S, Sun J-M, Lee S-H, et al. Two cases of small cell lung cancer transformation from EGFR mutant adenocarcinoma during AZD9291 treatment. J Thorac Oncol. 2016;11:e1–4.

    Article  PubMed  Google Scholar 

  38. Bersanelli M, Minari R, Bordi P, Gnetti L, Bozzetti C, Squadrilli A, et al. L718Q mutation as new mechanism of acquired resistance to AZD9291 in EGFR-mutated non-small cell lung cancer. J Thorac Oncol. 2016. doi:10.1016/j.jtho.2016.05.019.

    PubMed  Google Scholar 

  39. Planchard D, Loriot Y, André F, Gobert A, Auger N, Lacroix L, et al. EGFR-independent mechanisms of acquired resistance to AZD9291 in EGFR T790M-positive NSCLC patients. Ann Oncol. 2015;26:2073–8.

    Article  CAS  PubMed  Google Scholar 

  40. Kim TM, Song A, Kim D-W, Kim S, Ahn Y-O, Keam B, et al. Mechanisms of acquired resistance to AZD9291: a mutation-selective, irreversible EGFR inhibitor. J Thorac Oncol. 2015;10:1736–44.

    Article  CAS  PubMed  Google Scholar 

  41. Oxnard GR, Thress KS, Paweletz CP, Stetson B, Dougherty ZL, Markovets A, et al. Mechanisms of acquired resistance to AZD9291 in EGFR T790M positive lung cancer. J Thorac Oncol. 2015;10:S173–260.

    Article  Google Scholar 

  42. Engel J, Richters A, Getlik M, Tomassi S, Keul M, Termathe M, et al. Targeting drug resistance in EGFR with covalent inhibitors: a structure-based design approach. J Med Chem. 2015;58:6844–63.

    Article  CAS  PubMed  Google Scholar 

  43. Walter AO, Sjin RTT, Haringsma HJ, Ohashi K, Sun J, Lee K, et al. Discovery of a mutant-selective covalent inhibitor of EGFR that overcomes T790M-mediated resistance in NSCLC. Cancer Discov. 2013;3:1404–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Simmons AD, Jaw-Tsai S, Haringsma HJ, Allen A, Harding TC. Abstract 793: Insulin-like growth factor 1 (IGF1R)/insulin receptor (INSR) inhibitory activity of rociletinib (CO-1686) and its metabolites in nonclinical models. Cancer Res. 2015;75(Suppl):793–5.

    Article  Google Scholar 

  45. Sequist LV, Soria J-C, Goldman JW, Wakelee HA, Gadgeel SM, Varga A, et al. Rociletinib in EGFR-mutated non–small-cell lung cancer. N Engl J Med. 2015;372:1700–9.

    Article  PubMed  Google Scholar 

  46. Goldman JW, Wakelee HA, Gadgeel S, Camidge DR, Solomon BJ, Yu H, et al. Dose optimization of rociletinib for EGFR mutated NSCLC. J Thorac Oncol. 2015;10(Suppl):261–406.

    Google Scholar 

  47. Goldman JW, Soria J-C, Wakelee HA, Camidge DR, Gadgeel SM, Yu HA, et al. Updated results from TIGER-X, a phase I/II open label study of rociletinib in patients (pts) with advanced, recurrent T790M-positive non-small cell lung cancer (NSCLC). ASCO Meeting Abstracts. 2016;34:9045.

    Google Scholar 

  48. Piotrowska Z, Liu E, Logan J, Chow J, Woreta FA, Muzikansky A, et al. Rociletinib-associated cataracts in EGFR-mutant NSCLC. ASCO Meeting Abstracts. 2016;34:9042.

    Google Scholar 

  49. Camidge R, Sequist LV, Soria J-C, Wakelee H, Ou S-H, Goldman JW, et al. Activity of rociletinib in EGFR mutant NSCLC patients with a history of CNS involvement. J Thorac Oncol. 2015;10(Suppl):261–406.

    Google Scholar 

  50. Sequist LV, Soria J-C, Camidge DR. Update to rociletinib data with the RECIST confirmed response rate. N Engl J Med. 2016;374:2296–7.

    Article  PubMed  Google Scholar 

  51. Eisenhauer EA, Therasse P, Bogaerts J, Schwartz LH, Sargent D, Ford R. New response evaluation criteria in solid tumours; revised RECIST guideline (version 1.1). Eur J Cancer. 2009;45:228–47.

    Article  CAS  PubMed  Google Scholar 

  52. Wakelee HA, Gadgeel SM, Goldman JW, Reckamp KL, Karlovich CA, Melnikova V, et al. Epidermal growth factor receptor (EGFR) genotyping of matched urine, plasma and tumor tissue from non-small cell lung cancer (NSCLC) patients (pts) treated with rociletinib. ASCO Meeting Abstracts. 2016;34:9001.

    Google Scholar 

  53. Clovis Oncology Announces Q1 2016 Operating Results and Corporate Update. Available at http://phx.corporate-ir.net/phoenix.zhtml?c=247187&p=irol-newsArticle&ID=2165717. Accessed 8 July 2016.

  54. Mizuuchi H, Suda K, Murakami I, Sakai K, Sato K, Kobayashi Y, et al. Oncogene swap as a novel mechanism of acquired resistance to epidermal growth factor receptor-tyrosine kinase inhibitor in lung cancer. Cancer Sci. 2016;107:461–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Chabon JJ, Simmons AD, Lovejoy AF, Esfahani MS, Newman AM, Haringsma HJ, et al. Circulating tumour DNA profiling reveals heterogeneity of EGFR inhibitor resistance mechanisms in lung cancer patients. Nat Commun. 2016;7:11815.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Piotrowska Z, Niederst MJ, Karlovich CA, Wakelee HA, Neal JW, Mino-Kenudson M, et al. Heterogeneity underlies the emergence of EGFR T790 wild-type clones following treatment of T790M-positive cancers with a third generation EGFR inhibitor. Cancer Discov. 2015;5:713–22.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Sequist LV, Piotrowska Z, Niederst MJ, et al. Osimertinib responses after disease progression in patients who had been receiving rociletinib. JAMA Oncol. 2016;2:541–3.

    Article  PubMed  Google Scholar 

  58. Lee K-O, Cha MY, Kim M, Song JY, Lee J-H, Kim YH, et al. Abstract LB-100: Discovery of HM61713 as an orally available and mutant EGFR selective inhibitor. Cancer Res. 2014;74:LB-100.

    Article  Google Scholar 

  59. Park K, Lee J-S, Lee KH, Kim J-H, Cho BC, Min YJ, et al. BI 1482694 (HM61713), an EGFR mutant-specific inhibitor, in T790M+ NSCLC: efficacy and safety at the RP2D. ASCO Meeting Abstracts. 2016;34:9055.

    Google Scholar 

  60. Kim ES. Olmutinib: first global approval. Drugs. 2016;76:1153–7.

    Article  CAS  PubMed  Google Scholar 

  61. Boehringer Ingelheim Launches Ambitious ELUXA Trial Programme to Broadly Investigate Promising Lung Cancer Compound Olmutinib. Available at https://www.boehringer-ingelheim.com/press-release/boehringer-ingelheim-launches-ambitious-eluxa-trial-programme. Accessed 8 July 2016.

  62. Song H-N, Jung KS, Yoo KH, Cho J, Lee JY, Lim SH, et al. Acquired C797S mutation upon treatment with a T790M-specific third-generation EGFR inhibitor (HM61713) in non–small cell lung cancer. J Thorac Oncol. 2016;11:e45–7.

    Article  PubMed  Google Scholar 

  63. Jia Y, Juarez J, Li J, Manuia M, Niederst MJ, Tompkins C, et al. EGF816 exerts anticancer effects in non–small cell lung cancer by irreversibly and selectively targeting primary and acquired activating mutations in the EGF receptor. Cancer Res. 2016;76:1591–602.

    Article  CAS  PubMed  Google Scholar 

  64. Tan DS-W, Yang JC-H, Leighl NB, Riely GJ, Sequist LV, Felip E, et al. Updated results of a phase 1 study of EGF816, a third-generation, mutant-selective EGFR tyrosine kinase inhibitor (TKI), in advanced non-small cell lung cancer (NSCLC) harboring T790M. ASCO Meeting Abstracts. 2016;34:9044.

    Google Scholar 

  65. Konagai S, Sakagami H, Yamamoto H, Tanaka H, Matsuya T, Mimasu S, et al. Abstract 2586: ASP8273 selectively inhibits mutant EGFR signal pathway and induces tumor shrinkage in EGFR mutated tumor models. Cancer Res. 2015;75:2586.

    Article  Google Scholar 

  66. Sakagami H, Konagai S, Yamamoto H, Tanaka H, Matsuya T, Mori M, et al. Abstract 1728: ASP8273, a novel mutant-selective irreversible EGFR inhibitor, inhibits growth of non-small cell lung cancer (NSCLC) cells with EGFR activating and T790M resistance mutations. Cancer Res. 2014;74:1728.

    Article  Google Scholar 

  67. Goto Y, Nokihara H, Murakami H, Shimizu T, Seto T, Krivoshik AP, et al. ASP8273, a mutant-selective irreversible EGFR inhibitor in patients (pts) with NSCLC harboring EGFR activating mutations: preliminary results of first-in-human phase I study in Japan. ASCO Meeting Abstracts. 2015;33:8014.

    Google Scholar 

  68. Azuma K. Interim ASP8273 clinical activity in Asian NSCLC patients with EGFR activating and T790M mutations. Presented at the 56th Annual Meeting of Japanese Lung Cancer Society. 2015

  69. Yu HA, Spira AI, Horn L, Weiss J, West HJ, Giaccone G, et al. Antitumor activity of ASP8273 300 mg in subjects with EGFR mutation-positive non-small cell lung cancer: interim results from an ongoing phase 1 study. ASCO Meeting Abstracts. 2016;34:9050.

    Google Scholar 

  70. Xu X. Parallel phase 1 clinical trials in the US and in China: accelerating the test of avitinib in lung cancer as a novel inhibitor selectively targeting mutated EGFR and overcoming T790M-induced resistance. Chin J Cancer. 2015;34:1–3.

    Article  Google Scholar 

  71. Tan C-S, Cho B-C, Soo RA. Next-generation epidermal growth factor receptor tyrosine kinase inhibitors in epidermal growth factor receptor-mutant non-small cell lung cancer. Lung Cancer. 2016;93:59–68.

    Article  PubMed  Google Scholar 

  72. Cheng H, Nair SK, Murray BW. Recent progress on third generation covalent EGFR inhibitors. Bioorg Med Chem Lett. 2016;26:1861–8.

    Article  CAS  PubMed  Google Scholar 

  73. Cheng H, Nair SK, Murray BW, Almaden C, Bailey S, Baxi S, et al. Discovery of 1-{(3R,4R)-3-[({5-Chloro-2-[(1-methyl-1H-pyrazol-4-yl)amino]-7H-pyrrolo[2,3-d]pyrimidin-4-yl}oxy)methyl]-4-methoxypyrrolidin-1-yl}prop-2-en-1-one (PF-06459988), a potent, WT sparing, irreversible inhibitor of T790M-containing EGFR mutants. J Med Chem. 2016;59:2005–24.

    Article  CAS  PubMed  Google Scholar 

  74. Rivera VM, Wang F, Anjum R, Zhang S, Squillace R, Keats J, et al. Abstract 1794: AP26113 is a dual ALK/EGFR inhibitor: characterization against EGFR T790M in cell and mouse models of NSCLC. Cancer Res. 2012;72:1794.

    Article  Google Scholar 

  75. Kato M, Miyadera K, Ito K, Aoyagi Y, Hashimoto A, Yonekura K, et al. 401 TAS-121, a highly potent and mutant-specific EGFR inhibitor, overcomes T790M-acquired resistance with promising antitumor activity through specific inhibition of mutant EGFR signaling. Eur J Cancer. 2014;50:128.

    Article  Google Scholar 

  76. Oxnard GR, Arcila ME, Sima CS, Riely GJ, Chmielecki J, Kris MG, et al. Acquired resistance to EGFR tyrosine kinase inhibitors in EGFR-mutant lung cancer: distinct natural history of patients with tumors harboring the T790M mutation. Clin Cancer Res. 2011;17:1616–22.

    Article  CAS  PubMed  Google Scholar 

  77. Sun J-M, Ahn M-J, Choi Y-L, Ahn JS, Park K. Clinical implications of T790M mutation in patients with acquired resistance to EGFR tyrosine kinase inhibitors. Lung Cancer. 2013;82:294–8.

    Article  PubMed  Google Scholar 

  78. Hata AN, Niederst MJ, Archibald HL, Gomez-Caraballo M, Siddiqui FM, Mulvey HE, et al. Tumor cells can follow distinct evolutionary paths to become resistant to epidermal growth factor receptor inhibition. Nat Med. 2016;22:262–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Oxnard GR. The cellular origins of drug resistance in cancer. Nat Med. 2016;22:232–4.

    Article  CAS  PubMed  Google Scholar 

  80. Ramirez M, Rajaram S, Steininger RJ, Osipchuk D, Roth MA, Morinishi LS, et al. Diverse drug-resistance mechanisms can emerge from drug-tolerant cancer persister cells. Nat Commun. 2016;7:10690.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Chen L-Y, Molina-Vila MA, Ruan S-Y, Su K-Y, Liao W-Y, Yu K-L, et al. Coexistence of EGFR T790M mutation and common activating mutations in pretreatment non-small cell lung cancer: a systematic review and meta-analysis. Lung Cancer. 2016;94:46–53.

    Article  PubMed  Google Scholar 

  82. Piotrowska Z, Drapkin B, Engelman JA, Nagy RJ, Lanman RB, Sequist LV. Plasma T790M result alters treatment options in a previously T790 wild-type EGFR-mutant lung cancer. J Thorac Oncol. 2016;11:e95–7.

    Article  PubMed  Google Scholar 

  83. Liao BC, Bai YY, Lin CC, Lin SY, Lee YF, Ho CC, et al. 151P: Outcomes of research biopsies in clinical trials of EGFR mutation-positive NSCLC patients pretreated with EGFR-TKIs. J Thorac Oncol. 2016;11:S123–4.

    Article  PubMed  Google Scholar 

  84. Scheffler M, Merkelbach-Bruse S, Bos M, Fassunke J, Gardizi M, Michels S, et al. Spatial tumor heterogeneity in lung cancer with acquired epidermal growth factor receptor-tyrosine kinase inhibitor resistance: targeting high-level MET-amplification and EGFR T790M mutation occurring at different sites in the same patient. J Thorac Oncol. 2015;10:e40–3.

    Article  PubMed  Google Scholar 

  85. Trimble EL, Abrams JS, Meyer RM, Calvo F, Cazap E, Deye J, et al. Improving cancer outcomes through international collaboration in academic cancer treatment trials. J Clin Oncol. 2009;27:5109–14.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank Editage (editage.com) for English language editing.

Funding

No sources of funding were used to assist with the preparation of this review.

Availability of data and materials

Not applicable (The present paper is a review article and it describes published data).

Authors’ contributions

BCL, JHL, CCL, and JCY conceptualized, organized, and wrote the manuscript together. All authors read and approved the final manuscript.

Competing interests

JCY is a consultant and has received honoraria from AstraZeneca, Roche/Genentech, Boehringer Ingelheim, MSD, Merck Serono, Novartis, Pfizer, Clovis Oncology, Eli Lilly, Bayer, Celgene, Astellas, Innopharmax, Ono Pharmaceutical, and Chugai Pharmaceutical. The remaining authors declare that they have no competing interests.

Consent for publication

Not applicable (The present paper does not contain data from any individual person).

Ethics approval and consent to participate

Not applicable (The present paper does not report on or involve the use of any animal or human data or tissue).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to James Chih-Hsin Yang.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liao, BC., Lin, CC., Lee, JH. et al. Update on recent preclinical and clinical studies of T790M mutant-specific irreversible epidermal growth factor receptor tyrosine kinase inhibitors. J Biomed Sci 23, 86 (2016). https://doi.org/10.1186/s12929-016-0305-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12929-016-0305-9

Keywords