Optimal management of EGFR-mutant non-small cell lung cancer with disease progression on first-line tyrosine kinase inhibitor therapy

Optimal management of EGFR-mutant non-small cell lung cancer with disease progression on first-line tyrosine kinase inhibitor therapy

Lung Cancer 110 (2017) 7–13 Contents lists available at ScienceDirect Lung Cancer journal homepage: www.elsevier.com/locate/lungcan Review Optimal...

403KB Sizes 6 Downloads 49 Views

Lung Cancer 110 (2017) 7–13

Contents lists available at ScienceDirect

Lung Cancer journal homepage: www.elsevier.com/locate/lungcan

Review

Optimal management of EGFR-mutant non-small cell lung cancer with disease progression on first-line tyrosine kinase inhibitor therapy Bin-Chi Liaoa,b,c, Chia-Chi Lina,d, Jih-Hsiang Leea,e, James Chih-Hsin Yanga,b,c,f,

MARK



a

Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan National Taiwan University Cancer Center, College of Medicine, National Taiwan University, Taipei, Taiwan c Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan d Department of Urology, College of Medicine, National Taiwan University, Taipei, Taiwan e Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan f Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan b

A R T I C L E I N F O

A B S T R A C T

Keywords: Acquired resistance Epidermal growth factor receptor mutation Non-small cell lung cancer Osimertinib T790M Tyrosine kinase inhibitor

The first-generation epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs), gefitinib and erlotinib, and the second-generation EGFR-TKI, afatinib, have all been approved as standard first-line treatments for advanced EGFR-mutant non-small cell lung cancer (NSCLC) based on superior progression-free survival results compared to platinum doublet chemotherapy regimens. Acquired resistance to an EGFR-TKI inevitably develops after a period of effective drug treatment. After tumor progression, many combination therapy regimens that include an EGFR-TKI, or EGFR-TKI monotherapy, have been tested in prospective trials with the aim of extending survival. Third-generation EGFR-TKIs such as osimertinib have been developed with the aim of overcoming the effects of EGFR T790M resistance mutation, which occurs in half of the patients with disease progression on EGFR-TKI therapy. Osimertinib has become the standard treatment in patients for whom tumor re-biopsy reveals an acquired EGFR T790M mutation following EGFR-TKI therapy. Other third-generation EGFRTKIs, such as olmutinib, EGF816, and ASP8273, are still in the trial phase.

1. Introduction EGFR-mutant non-small cell lung cancer (NSCLC) accounts for 15% and 50% of non-squamous NSCLC patients of Caucasians and East Asian ethnicity, respectively [1]. The current recommended standard of care for EGFR-mutant NSCLC in the advanced stage is epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) monotherapy, such as gefitinib, erlotinib, or afatinib. Landmark clinical trials have demonstrated superior progression free survival (PFS) times and quality-of-life compared to the former standard treatment of platinum-based doublet chemotherapy [2–8]. Erlotinib in combination with bevacizumab is also a first-line treatment of choice based on a phase II clinical trial results [9]. These treatments are beneficial, especially for patients whose tumors harbor activating EGFR mutations, such as exon 19 deletions and exon 21 L858R mutation [10]. However, acquired resistance inevitably develops after a period of 9–11 months of effective treatment. Platinum-based doublet chemotherapy was the standard treatment after progression on first-line EGFR-TKI therapy. EGFR-TKI treatment beyond progression and combination therapy strategies that incorporate an EGFR-TKI have been



proposed to extend survival, based on retrospective studies of “tumor growth rebound” during withdrawal of EGFR-TKI [11]. The EGFR exon 20 T790M mutation accounts for 50–60% of the mechanisms of acquired resistance based on repeat tumor biopsies during progression to detect this acquired mutation. Novel third-generation EGFR-TKIs, such as osimertinib, rociletinib, olmutinib, EGF816, and ASP8273 were designed to overcome acquired EGFR-TKI resistance due to EGFR T790M mutation [12–16]. Other strategies to overcome different mechanisms of acquired resistance are under development. Here, we focus on these recently developed treatment strategies for EGFR-mutant NSCLC with progression on first-line EGFR-TKI therapy. A literature review of clinical studies published between January 2013 and December 2016 was conducted using PubMed and MEDLINE, with the entry keywords ‘non-small cell lung cancer,’ ‘epidermal growth factor receptor mutation,’ ‘acquired resistance,’ ‘T790M,’ ‘osimertinib,’ ‘gefitinib,’ ‘erlotinib,’ and ‘afatinib.’ We also performed a manual search of the abstracts presented at major oncology meetings.

Corresponding author at: Department of Oncology, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, 100, Taiwan. E-mail addresses: [email protected] (B.-C. Liao), [email protected] (C.-C. Lin), [email protected] (J.-H. Lee), [email protected] (J.C.-H. Yang).

http://dx.doi.org/10.1016/j.lungcan.2017.05.009 Received 28 February 2017; Received in revised form 25 April 2017; Accepted 9 May 2017 0169-5002/ © 2017 Elsevier B.V. All rights reserved.

Lung Cancer 110 (2017) 7–13

B.-C. Liao et al.

2. Types of disease progression after first-line EGFR-TKI therapy

spective observational report from France also suggested that continuing EGFR-TKI therapy beyond PD showed a trend of superior OS, and those who continued EGFR-TKI therapy were less symptomatic compared to those who did not [23]. The ASPIRATION study (Asian Pacific trial of Tarceva as first-line in EGFR mutation) was a phase II single-arm study to evaluate the efficacy of erlotinib at 150 mg/day for stage IV EGFR-mutant NSCLC [24]. After PD, erlotinib therapy was continued at the patient’s and/or investigator’s discretion. Of the 207 intent-to-treat patients, the median PFS1 (primary endpoint) was 10.8 months (95% CI, 9.2–11.1). Of the 176 patients who experienced PFS1 event, 93 continued and 78 discontinued erlotinib therapy following progression. The median PFS1 and PFS2 (time from first study dose to off-erlotinib PD in the subset who continued erlotinib therapy beyond progression) of the 93 patients who had treatment beyond PD were 11.0 and 14.9 months, respectively. The authors concluded that the results were in line with prior efficacy results of first-line erlotinib therapy for stage IV EGFR-mutant NSCLC, and that treatment beyond progression is feasible in select patients. However, the characteristics of those patients who continued erlotinib therapy and those who did not were different. Significantly more patients who had recurrent disease, ECOG performance status 0 or 1 at PFS1, longer median PFS1, improved depth of response, and a longer median time from best overall response to PFS1 continued erlotinib therapy than those who did not. Patients who continued erlotinib therapy after PD had more new brain lesions at PFS1 than those who did not (4.3 vs. 1.3%). These different patient characteristics implied that patients with slow PD or isolated new brain metastasis could benefit from continuing EGFR-TKI therapy. The decision to continue EGFR-TKI therapy or not in this study was based on the investigators’ or the patients’ discretion, and the aforementioned different modes or subtypes of PD might have influenced the choice and led to bias [17,19]. In addition, repeat tumor biopsy at the time of PD was not mandatory in this study, hence the influence of acquired EGFR T790M mutation was not analyzed [24]. Other reasons to continue EGFR-TKI therapy beyond progression included asymptomatic progression, stable primary tumor, prior good radiologic response, bone-only progression, prior long disease control period, and new lesions suitable for local therapy (Fig. 1). In many

The types of progressive disease (PD) after first-line EGFR-TKI therapy are variable. A Chinese group proposed criteria for EGFR-TKI failure in NSCLC [17]. They classified three modes of progression: dramatic progression, gradual progression, and local progression. The median PFS were 9.3, 12.9, and 9.2 months (P = 0.007) in these three modes, respectively, and the median OS were 17.7, 39.4, and 23.1 months (P < 0.001), respectively. In patients with the gradual progression mode, continuing EGFR-TKI therapy was superior to switching chemotherapy in terms of OS (39.4 vs. 17.8 months; P = 0.02) [17]. The authors concluded that these modes of EGFR-TKI failure could favor strategies of subsequent treatment and predict survival. Gandara et al. proposed another classification of PD in patients with oncogene-driven NSCLC who experienced acquired resistance to a targeted therapy. The authors classified PD into 3 subtypes: systemic PD (multisite progression), oligo-PD (new sites or regrowth in a limited number of areas, maximum of 4 PD sites) [18], and central nervous system (CNS) sanctuary PD (excluding leptomeningeal carcinomatosis due to the lack of effective treatment options for long-term control). This classification provides the basis for integrating data of resistance mechanisms from repeat tumor biopsies into clinical decision-making and prospective trial designs [19]. An algorithm (Fig. 1) to determine the appropriate treatment for patients who experience PD is proposed here to accommodate emerging evidence from recent trials and publications. This algorithm is similar to the latest National Comprehensive Cancer Network® Guidelines for NSCLC and European Society for Medical Oncology Clinical Practice Guidelines [20,21]. Details of the treatment options shown in the algorithm are discussed below.

3. Continuing EGFR-TKI therapy beyond progression 3.1. EGFR-TKI therapy alone In a Japanese retrospective study, continuous use of EGFR-TKI therapy alone prolonged survival compared to cytotoxic chemotherapy in patients with activating EGFR-mutant NSCLC [22]. Another retro-

Fig. 1. Algorithm of managing patients who develop progressive disease on first-line EGFR-TKI therapy. Abbreviations: EGFR-TKI, epidermal growth factor receptor tyrosine kinase inhibitor.

8

Lung Cancer 110 (2017) 7–13

B.-C. Liao et al.

acquired EGFR T790M mutation and those who did not (32 vs. 25%; P = 0.341). Treatment-related adverse event (AE) rates were high and included rash (90%), diarrhea (71%), nail effects (57%), stomatitis (56%), and fatigue (47%). Grade 3 and 4 AEs were recorded in 44% and 2% of these patients, respectively [37]. With the development of thirdgeneration EGFR-TKIs, and given this regimen’s high rate of AEs, this therapy is no longer a preferred treatment of choice for patients whose tumors harbor an EGFR T790M mutation. A randomized phase II/III study is ongoing to compare afatinib monotherapy with afatinib plus cetuximab combination therapy as first-line therapy for patients with advanced EGFR-mutant NSCLC (ClinicalTrials.gov, NCT02438722).

retrospective studies, continuing EGFR-TKI therapy in combination with local therapy for isolated CNS disease or non-CNS tumors in select patients could be beneficial [18,25–27]. In patients with leptomeningeal carcinomatosis, continuing EGFR-TKI therapy or switching to a different first-generation EGFR-TKI might also be beneficial, despite the fact that no standard treatment exists for this complication [28]. Most physicians would agree that patients with rapid PD are not candidates for continued EGFR-TKI therapy. Afatinib treatment beyond progression was prospectively analyzed in the LUX-Lung 7 study. That study was a randomized phase IIb study to compare afatinib with gefitinib as first-line therapy for EGFR-mutant NSCLC. Afatinib therapy demonstrated a significantly longer PFS (median 11.0 vs. 10.9 months; HR 0.73; 95% CI, 0.57–0.95; P = 0.0165), time-to-treatment failure (median 13.7 vs. 11.5 months; HR 0.73; 95% CI, 0.58–0.92; P = 0.0073), and objective response rate (ORR) (70 vs. 56%; P = 0.0083) compared to gefitinib. Interestingly, 56 (35%) of 160 patients and 47 (30%) of 159 patients administered afatinib and gefitinib, respectively, continued EGFR-TKI treatment beyond investigator-assessed radiological PD. The median duration of treatment beyond progression was 2.7 months (95% CI, 1.94–4.3) for afatinib and 2.0 months (95% CI, 1.5–3.0) for gefitinib [29]. In conclusion, because of the lack of universal consensus and definitions of which patients could benefit from continuing EGFR-TKI therapy, it is difficult to conduct prospective clinical trials to determine the best treatment strategy. However, treatment beyond progression is still an option in select patients, and physicians should discuss this with individual patients.

5. Repeat tumor biopsy after PD The EGFR T790M mutation remains the most common mechanism of acquired resistance following first- or second- generation EGFR-TKI therapy (50–60%) [38,39]. Other mechanisms included MET amplification, HER2 amplification, BRAF mutation, epithelial to mesenchymal transformation, PIK3CA mutation, and small cell transformation, etc. [40–43]. The current trend of second-line treatment after progression on first-line EGFR-TKI therapy is to identify the mechanism of acquired resistance by repeating tumor biopsies. The most important part of this trend is the development of third-generation EGFR-TKI to target the EGFR T790M mutation, as mentioned below. The diagnostic yield and safety of repeat tumor biopsies have been discussed previously [44,45]. However, the concept of intra-tumoral and inter-metastatic tumoral molecular heterogeneity has emerged and changed clinicians’ view of cancer [46–49]. Investigators might question if a single tumor biopsy represents the entire tumor; with a single tumor biopsy, some patients with treatable driver mutations, such as the EGFR T790M mutation, might not get proper treatment. Liquid biopsy, which provides information regarding resistance mechanisms using a patient’s plasma sample (for example, by analyzing circulating tumor DNA), was developed concurrently with third-generation EGFR-TKIs. This approach is advantageous because it is less invasive and represents the mutational status of the entire tumor burden [50,51].

3.2. Combination of EGFR-TKI and cytotoxic chemotherapy In some retrospective studies, continuation of EGFR-TKI therapy in combination with cytotoxic chemotherapy beyond disease progression was beneficial in terms of the response rate or survival [30,31]. The phase III IMPRESS trial enrolled patients with advanced EGFR-mutant NSCLC who had achieved disease control with first-line gefitinib monotherapy and subsequently developed acquired resistance. These patients were randomized to receive cisplatin-pemetrexed chemotherapy with or without continued gefitinib therapy [32]. The PFS (primary endpoint) was 5.4 months in both arms (HR 0.86; 95% CI, 0.65–1.13; P = 0.27). The authors concluded that PFS was not prolonged by the continuation of gefitinib therapy in combination with cisplatin-pemetrexed chemotherapy beyond progression on first-line gefitinib monotherapy compared to chemotherapy alone. Furthermore, an updated survival analysis showed that the median OS was 13.4 months in the combination arm and 19.5 months in the control arm (HR 1.44; 95% CI, 1.07–1.94; P = 0.016) [33]. In a subgroup analysis, patients with negative plasma EGFR T790M mutation had a trend toward better PFS in the continued gefitinib therapy arm (6.7 vs. 5.4 months; HR 0.67; 95% CI, 0.43–1.03; P = 0.0745); however, the median OS was similar between the two arms (21.4 vs. 22.5 months; HR 1.15; 95% CI, 0.68–1.94) [34]. Based on these results, continuation of treatment (chemotherapy) with first-generation EGFR-TKIs beyond PD is no longer considered standard treatment. However, the study acknowledges that a certain subset of patients might benefit from a combination of EGFR-TKI and chemotherapy at the time of progression. A study combining afatinib plus weekly paclitaxel was clearly superior to single-agent chemotherapy in ORR and PFS in a large cohort of randomized, highly selected patients [35,36].

6. Recent developments of third-generation EGFR-TKIs The third-generation EGFR-TKIs (osimertinib, rociletinib, olmutinib, EGF816, ASP8273, etc.) were designed to overcome the effects of major acquired resistance due to EGFR T790M mutation by covalently binding to the cysteine residue in position 797 of EGFR (Cys797) and changing the TKI backbone from quinazoline to pyrimidine to avoid wild-type EGFR inhibition and maintain high inhibitory activity against the EGFR T790M and other activating mutations [12,15,16]. These drugs have less capacity to inhibit wild-type EGFR; therefore, they might induce less skin rash and diarrhea compared to first- and secondgeneration EGFR-TKIs, while maintaining activity against activating EGFR mutations. A detailed review of the development of these novel agents has been published previously [52,53]. Here, we focus on the recent clinical trial results of these novel agents. 6.1. Osimertinib Osimertinib (AZD9291) is a mono-anilino-pyrimidine compound that irreversibly targets tumors harboring activating EGFR mutations and the EGFR T790M mutation while having little effect on wild-type EGFR. This compound binds covalently with Cys797 and acts against other kinases, such as ErBB2 and ErBB4 [15,54]. In the first phase I/II clinical study of osimertinib (AURA), 80 mg/ day was chosen as the dose for subsequent studies, and the doselimiting toxicity and maximum tolerated dose was not reached [14]. Two phase II studies, the AURA study phase II extension component and the AURA2 study, provided the efficacy and safety evidence for the accelerated approval of osimertinib [55]. In the AURA study phase II

4. Combination of EGFR-TKI and anti-EGFR agents Cetuximab is a chimeric IgG1 monoclonal antibody against EGFR. In a phase Ib study, cetuximab (500 mg/m2 every 2 weeks) plus afatinib therapy (40 mg/day) was administered to patients who had EGFRmutant NSCLC and who developed acquired resistance to first-generation EGFR-TKIs. The ORR was similar between patients who had an 9

Lung Cancer 110 (2017) 7–13

B.-C. Liao et al.

6.2. Rociletinib

extension component, patients who had EGFR-mutant NSCLC and EGFR T790M mutation in the re-biopsy sample confirmed centrally were treated with osimertinib. The confirmed ORR (primary endpoint) was 62%, and the median duration of response was 15.2 months. The median PFS was 12.3 months (95% CI, 9.5–13.8) (n = 201). Treatmentrelated AEs included all grades of diarrhea (43%) and skin rash (40%), and only < 1% of the patients developed grade ≥3 diarrhea or skin rash. Eight (4%) patients developed interstitial lung disease (ILD), and QT interval corrected for heart rate (QTc) prolongation AEs were recorded in 6 (3%) patients [56]. In the AURA2 study, a patient population similar to that of the AURA study extension component was administered osimertinib therapy. The confirmed ORR (primary endpoint) was 70%, and the median PFS was 9.9 months (95% CI, 8.5–12.3) (n = 210). The most common grade 3 and 4 AEs were pulmonary embolism (3%), QTc prolongation (2%), and decreased neutrophil count (2%). One patient developed and died from ILD [57]. Based on the aforementioned two study results, osimertinib received US Food and Drug Administration (FDA) approval in November 2015 for EGFR–TKI-pretreated metastatic NSCLC harboring EGFR T790M mutation and is now approved in many other countries (Fig. 1) [56,57]. The companion diagnostic test (cobas® EGFR Mutation Test v2; Roche, Basel, Switzerland) that is used to detect tumor EGFR T790M mutations was also approved. A confirmatory phase III study (AURA 3) compared osimertinib with platinum-pemetrexed chemotherapy in patients with advanced EGFRmutant NSCLC, after progression following first-line EGFR-TKI therapy with a centrally confirmed EGFR T790M mutation in repeat tumor biopsy samples. A total of 419 patients were randomized in a 2:1 ratio to receive osimertinib monotherapy or standard platinum-pemetrexed chemotherapy (maintenance pemetrexed was allowed). The investigator-assessed PFS (primary endpoint) was significantly longer in the osimertinib arm compared to that in the chemotherapy arm (median 10.1 vs. 4.4 months; HR 0.30; 95% CI, 0.23–0.41; P < 0.001) [58]. The application of liquid biopsy to detect the EGFR T790M mutation in blood samples has provided a good alternative to tedious, invasive, and sometimes impossible tumor re-biopsy procedures [59–61]. The investigators collected plasma samples in the AURA study to obtain the cell-free plasma DNA, which was genotyped using the beads, emulsions, amplification, and magnetics (BEAMing) digital polymerase chain reaction technique (Sysmex Inostics, Inc., Mundelein, IL, USA) [61,62]. The plasma-based sensitivity for detecting the EGFR T790M mutation was 70%. The ORR and median PFS were similar in patients who were positive for the plasma EGFR T790M mutation and in those with the EGFR T790M mutation in 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 a positive plasma EGFR T790M mutation could avoid tumor re-biopsy for EGFR T790M mutation testing, whereas those with a negative plasma EGFR T790M mutation should undergo a repeat tumor biopsy [63]. The European Medicines Agency has approved the use of either tumor DNA derived from a tissue sample or circulating tumor DNA obtained from a plasma sample to determine the EGFR T790M mutation status [64]. Another treatment strategy for conferring better anti-tumor activity is combination therapy in addition to osimertinib. Osimertinib was combined with either a MET inhibitor (savolitinib), MEK inhibitor (selumetinib), or anti-PD-L1 monoclonal antibody (durvalumab) in the TATTON study (ClinicalTrials.gov, NCT02143466) [65]. A report revealed that the incidence of ILD was high in the osimertinib plus durvalumab arm, and the development of this combination therapy was discontinued [66]. Clinical studies of other combination therapies are ongoing, such as for osimertinib in combination with ramucirumab, necitumumab, bevacizumab, or navitoclax (ClinicalTrials.gov, NCT02789345, 02496663, 02803203 and 02520778).

Rociletinib (CO-1686) is a 2,4-disubstituted pyrimidine compound that irreversibly targets tumors harboring activating EGFR mutations and the EGFR T790M mutation, while having little effect on wild-type EGFR [16,67]. Orally administered rociletinib at 500 mg twice per day was determined as the recommended dose in an early-phase clinical study (TIGER-X) [13,68]. A study of 208 patients who received this dose of rociletinib demonstrated that the common AEs included hyperglycemia (57.2%), diarrhea (56.7%), nausea (43.8%), and QTc prolongation (26.4%). Grade ≥ 3 AEs of hyperglycemia and QTc prolongation developed in 28.8% and 7.7% of these patients, respectively. ILD was observed in 0.5% of the patients [69]. The TIGER-X study demonstrated an ORR of 33.9% for the 443 patients who had centrally confirmed EGFR T790M mutant NSCLC, and who were administered rociletinib at dose levels of 500–750 mg twice per day. The PFS was 5.7 months in 208 patients who were administered this drug at 500 mg twice per day [69]. In the TIGER-X study, plasma samples were analyzed by liquid biopsy, using the cobas® EGFR Mutation Test to detect the EGFR T790M mutation. The positive percent agreement between the cobas® plasma results and tumor results was 64% [70]. Another biomarker study that collected tissue, plasma (BEAMing), and urine specimens (Trovera Quantitative NGS assay, Trovagene, San Diego, CA, USA) to detect the EGFR T790M mutation showed sensitivities of 80.9% and 81.1% for the plasma and urine assays, respectively. The confirmed ORRs in patients with EGFR T790M mutant tissue, plasma, and urine were 33.9%, 32.1%, and 36.7%, respectively. Lower plasma sensitivity was observed in patients with M1a or M0 intrathoracic disease than in patients with M1b distant metastatic disease (56.8 vs. 88.4%, P < 0.001) [71]. Rociletinib did not receive accelerated approval by the US FDA. In May 2016, Clovis Oncology, Inc. terminated enrollment in all sponsored studies of rociletinib and withdrew its Marketing Authorization Application for rociletinib from European regulatory authorities [72]. 6.3. Olmutinib Olmutinib (HM61713) is another third-generation EGFR-TKI that irreversibly binds to a cysteine residue near the kinase domain. Olmutinib is active against cell lines and xenograft tumors harboring the EGFR L858R/T790M mutation and deletions in EGFR exon 19, but has little effect on cell lines with wild-type EGFR [73]. The first phase I/II study (ClinicalTrials.gov, NCT01588145) was conducted in South Korea, and orally administered olmutinib at 800 mg/day was chosen as the recommended dose. In that study, 76 patients who harbored an acquired EGFR T790M mutation were administered olmutinib therapy, and the median PFS was 6.9 months. The confirmed ORR was 54%, and activity against CNS metastases was also observed. The treatment-related AEs from that study included all grades diarrhea (59%), pruritus (42%), rash (41%), nausea (39%), and palmar-plantar erythrodysesthesia syndrome (30%). There were no cases of grade ≥3 diarrhea or nausea, and only 1% and 5% of patients had grade ≥3 pruritus and rash, respectively. One patient experienced ILD and discontinued therapy [74]. Olmutinib was granted a breakthrough therapy designation for NSCLC by the US FDA. In May 2016, it was approved in South Korea for patients with advanced NSCLC who were pretreated with EGFR-TKIs and whose tumor harbored the EGFR T790M mutation [75]. However, the global effort to develop olmutinib was hampered in September 2016 [76]. 6.4. EGF816 Nazartinib (EGF816) targets EGFR irreversibly by forming a covalent bond to Cys797 [12,77]. In an early-phase clinical study (Clinical10

Lung Cancer 110 (2017) 7–13

B.-C. Liao et al.

tion. These trials identify patients with MET amplification by repeated tumor biopsies after progression on first-line EGFR-TKI therapy. A MET inhibitor is combined with EGFR-TKI to overcome this resistance mechanism. Currently, none of the MET inhibitors mentioned above is available outside the clinical trial setting.

Trials.gov, NCT02108964), 152 patients were enrolled and administered oral EGF816 at 75–350 mg/day. The common AEs included skin rash (53.9%), diarrhea (36.8%), pruritus (34.2%), dry skin (25.0%), and stomatitis (24.3%). Grade ≥ 3 AEs included rash (16.4%), urticaria (2.6%), anemia (2.6%), fatigue (2.0%), and diarrhea (2.0%). Two patients developed a hepatitis B virus reactivation, and two patients experienced increased serum lipase. The confirmed ORR among 147 evaluable patients was 46.9%, and the estimated PFS was 9.7 months [78]. Studies of the combination therapy of EGF816 and nivolumab (an anti-PD-1 monoclonal antibody) or INC280 (a MET inhibitor) are ongoing (ClinicalTrials.gov, NCT02323126 and 02335944).

8. Future directions Prospective trials are ongoing that employ liquid biopsy to identify patients who have a positive plasma EGFR T790M mutation during PD to receive osimertinib therapy (ClinicalTrials.gov, NCT02811354). Investigators can obtain serial changes in circulating tumor DNA by using liquid biopsy tools to explore the mutation burden, disappearance, and reappearance during treatment; serial liquid biopsies could serve as a pharmacodynamic marker, a means of quantification, a predictor of response and progression, etc. [86]. Clinical trials of third-generation EGFR-TKI-based combination therapy aiming to extend the clinical benefit and overcome resistance to the third-generation EGFR-TKIs are ongoing. Among these combination therapy regimens, immune checkpoint inhibitor combined with a third-generation EGFR-TKI demonstrated a high incidence of ILD, as mentioned previously. Hence, investigators should treat these patients with caution, especially in early-phase clinical trials. Regarding immune checkpoint inhibitor therapy, many preclinical data suggested that upregulation of programmed death ligand 1 (PDL1) in EGFR-driven lung tumors contributed to immune escape [87,88]. High PD-L1 expression seemed to be associated with the presence of EGFR mutations in advanced lung adenocarcinoma [89,90]. A report from Japan demonstrated that low PD-L1 expression was observed in EGFR T790M mutant tumors, whereas tumors without EGFR T790M mutation were associated with higher PD-L1 expression [91]. However, immunotherapy (anti-PD1) alone seems less efficacious for patients with EGFR-mutant than EGFR wild-type tumors [92,93]. Therefore, it is advisable that immunotherapy should only be recommended for patients with EGFR-mutant NSCLC in clinical trials.

6.5. ASP8273 ASP8273 is another irreversible EGFR inhibitor that targets EGFR by forming a covalent bond to Cys797 [79]. In a 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. A total of 63 patients received treatment at this dose, and most of them harbored the EGFR T790M mutation. The confirmed ORR was 30%, and the median PFS was 6.0 months [80]. Common treatmentrelated AEs included diarrhea (48%), nausea (27%), hyponatremia (19%), paresthesia (14%), vomiting (13%), and dizziness (11%). Grade ≥ 3 AEs included hyponatremia (13%) and diarrhea (2%). 6.6. Summary of third-generation EGFR-TKIs Currently, osimertinib is the recommended standard of care for patients who have progressed on EGFR-TKI therapy and whose tumors harbor an acquired EGFR T790M mutation (Fig. 1). Olmutinib is available in South Korea only for the same patient population mentioned above. The other aforementioned third-generation EGFR-TKIs are not available outside a clinical trial setting. Other novel thirdgeneration EGFR-TKIs in early-phase clinical development, which are not mentioned here, still have great potential to change the treatment paradigm. 7. Targeting MET amplification

9. Conclusions

MET amplification is another mechanism of acquired resistance to EGFR-TKI therapy [81,82]. A phase II single-arm clinical trial conducted in Japan enrolled patients with advanced EGFR-mutant (exon 19 deletions and exon 21 L858R mutation) NSCLC who developed acquired resistance to gefitinib or erlotinib to receive tivantinib (ARQ 197, a MET inhibitor) and erlotinib combination therapy regardless of MET expression status. A total of 45 patients were enrolled, and the ORR was 6.7%. High MET expression (≥50%) by immunohistochemical stain was detected in 48.9% of the patients, including all 3 partial responders [83]. Another study enrolled patients with advanced NSCLC (enriched for EGFR-mutant disease) who developed acquired resistance to erlotinib to receive emibetuzumab (LY2875358, a humanized IgG4 monoclonal bivalent MET antibody) with or without erlotinib therapy. The ORR of patients whose re-biopsy samples harbored MET overexpression (≥60%) was 3.8% in the combination arm and 4.8% in the monotherapy arm [84]. Clinical trials of other MET inhibitors are ongoing; for example, patients with EGFR-mutant NSCLC were administered capmatinib (INC280) and gefitinib following progression with prior EGFR-TKI therapy. Only patients with post-progression tumor tissue positive for MET immunohistochemical stain were enrolled. The ORR for this combination therapy was 31%, and the best results were seen in patients with a MET copy number gain of at least six, with an ORR of 50% [85]. Capmatinib plus erlotinib (ClinicalTrials.gov, NCT 02468661) following progression on EGFR-TKI therapy and tepotinib plus gefitinib after failure of prior gefitinib therapy (ClinicalTrials.gov, NCT01982955) are also ongoing clinical trials targeting MET amplifica-

After PD on first-line EGFR TKI therapy, we suggest that all patients participate in prospective clinical trials if eligible (Fig. 1). Continuation of EGFR-TKI therapy is suitable for select patients. However, in patients with rapid PD or deterioration of symptoms, continuing EGFR-TKI is not recommended. Repeat tumor biopsies to detect the EGFR T790M mutation is the current standard of care, and osimertinib should be administered in patients with EGFR T790M-mutant disease. Liquid biopsy is an alternative method to detect plasma EGFR T790M mutation and to identify patients suitable for osimertinib therapy. Although liquid biopsy is an approved diagnostic tool in the EU for patients administered osimertinib therapy, this technique should be confirmed in prospective trials. Conflicts of interest James Chih-Hsin Yang is a member of the advisory committees and has received honoraria from AstraZeneca, Roche/Genentech, Boehringer Ingelheim, MSD, Merck Serono, Novartis, Pfizer, Clovis Oncology, Eli Lilly, Bayer, Celgene, Astellas, BMS, Ono Pharmaceutical, Yuhan Pharmaceutical, and Chugai Pharmaceutical. The authors have no conflicts of interest to declare. Funding/Support This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors. 11

Lung Cancer 110 (2017) 7–13

B.-C. Liao et al.

Acknowledgments The authors also thank Editage (editage.com) for English language editing. The authors attest that this study was not supported by external funding sources. This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

[24]

[25]

[26]

References [1] C.-H. Yang, C.-J. Yu, J.-Y. Shih, 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. 26 (16) (2008) 2745–2753. [2] L.V. Sequist, J.C.-H. Yang, N. Yamamoto, et al., Phase III study of afatinib or cisplatin plus pemetrexed in patients with metastatic lung adenocarcinoma with EGFR mutations, J. Clin. Oncol. 31 (27) (2013) 3327–3334. [3] R. Rosell, E. Carcereny, R. Gervais, 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. 13 (3) (2012) 239–246. [4] C. Zhou, Y.-L. Wu, G. Chen, et al., Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study, Lancet Oncol. 12 (8) (2011) 735–742. [5] T. Mitsudomi, S. Morita, Y. Yatabe, 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. 11 (2) (2010) 121–128. [6] M. Maemondo, A. Inoue, K. Kobayashi, et al., Gefitinib or chemotherapy for nonsmall-cell lung cancer with mutated EGFR, N. Engl. J. Med. 362 (25) (2010) 2380–2388. [7] J.C.-H. Yang, V. Hirsh, M. Schuler, 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. 31 (27) (2013) 3342–3350. [8] S.L. Geater, C.-R. Xu, C. Zhou, et al., Symptom and quality of life improvement in LUX-Lung 6: an open-label phase III study of afatinib versus cisplatin/gemcitabine in Asian patients with EGFR mutation-positive advanced non-small-cell lung cancer, J. Thorac. Oncol. 10 (6) (2015) 883–889. [9] T. Seto, T. Kato, M. Nishio, et al., Erlotinib alone or with bevacizumab as first-line therapy in patients with advanced non-squamous non-small-cell lung cancer harbouring EGFR mutations (JO25567): an open-label, randomised, multicentre, phase 2 study, Lancet Oncol. 15 (11) (2014) 1236–1244. [10] B.-C. Liao, C.-C. Lin, J.C.-H. Yang, First-line management of EGFR-mutated advanced lung adenocarcinoma: recent developments, Drugs 73 (4) (2013) 357–369. [11] J.E. Chaft, G.R. Oxnard, C.S. Sima, et al., Disease flare after tyrosine kinase inhibitor discontinuation in patients with EGFR-mutant lung cancer and acquired resistance to erlotinib or gefitinib: implications for clinical trial design, Clin. Cancer Res. 17 (19) (2011) 6298–6303. [12] Y. Jia, J. Juarez, J. Li, 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. 76 (6) (2016) 1591–1602. [13] L.V. Sequist, J.-C. Soria, J.W. Goldman, et al., Rociletinib in EGFR-mutated nonsmall-cell lung cancer, N. Engl. J. Med. 372 (18) (2015) 1700–1709. [14] P.A. Jänne, J.C.-H. Yang, D.-W. Kim, et al., AZD9291 in EGFR inhibitor-resistant non-small-cell lung cancer, N. Engl. J. Med. 372 (18) (2015) 1689–1699. [15] D.A.E. Cross, S.E. Ashton, S. Ghiorghiu, et al., AZD9291, an irreversible EGFR TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung cancer, Cancer Discov. 4 (9) (2014) 1046–1061. [16] A.O. Walter, R.T.T. Sjin, H.J. Haringsma, et al., Discovery of a mutant-selective covalent inhibitor of EGFR that overcomes T790M-mediated resistance in NSCLC, Cancer Discov. 3 (12) (2013) 1404–1415. [17] J.-J. Yang, H.-J. Chen, H.-H. Yan, et al., Clinical modes of EGFR tyrosine kinase inhibitor failure and subsequent management in advanced non-small cell lung cancer, Lung Cancer 79 (1) (2013) 33–39. [18] A.J. Weickhardt, B. Scheier, J.M. Burke, et al., Local ablative therapy of oligoprogressive disease prolongs disease control by tyrosine kinase inhibitors in oncogeneaddicted non-small-cell lung cancer, J. Thorac. Oncol. 7 (12) (2012) 1807–1814. [19] D.R. Gandara, T. Li, P.N. Lara, et al., Acquired resistance to targeted therapies against oncogene-driven non-small-cell lung cancer: approach to subtyping progressive disease and clinical implications, Clin. Lung Cancer 15 (1) (2014) 1–6. [20] NCCN Guidelines Version 4.2017 Non-small cell lung cancer, (2017) https://www. nccn.org/professionals/physician_gls/pdf/nscl.pdf (Accessed 2 February 2017). [21] S. Novello, F. Barlesi, R. Califano, et al., Metastatic non-small-cell lung cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up, Ann. Oncol. 27 (Suppl. 5) (2016) v1–v27. [22] K. Nishie, T. Kawaguchi, A. Tamiya, et al., Epidermal growth factor receptor tyrosine kinase inhibitors beyond progressive disease: a retrospective analysis for Japanese patients with activating EGFR mutations, J. Thorac. Oncol. 7 (11) (2012) 1722–1727. [23] J.B. Auliac, C. Fournier, C. Audigier Valette, et al., Impact of continuing first-line

[27]

[28]

[29]

[30]

[31]

[32]

[33]

[34]

[35]

[36]

[37]

[38]

[39]

[40] [41]

[42]

[43]

[44] [45]

[46]

[47]

[48] [49]

12

EGFR tyrosine kinase inhibitor therapy beyond RECIST disease progression in patients with advanced EGFR-mutated non-small-cell lung cancer (NSCLC): retrospective GFPC 04-13 study, Target Oncol. 11 (2) (2016) 167–174. K. Park, C. Yu, S. Kim, et al., First-line erlotinib therapy until and beyond response evaluation criteria in solid tumors progression in Asian patients with epidermal growth factor receptor mutation-positive non-small-cell lung cancer: the ASPIRATION study, JAMA Oncol. 2 (3) (2016) 305–312. T. Shukuya, T. Takahashi, T. Naito, et al., Continuous EGFR-TKI administration following radiotherapy for non-small cell lung cancer patients with isolated CNS failure, Lung Cancer 74 (3) (2011) 457–461. H.A. Yu, C.S. Sima, J. Huang, et al., Local therapy with continued EGFR tyrosine kinase inhibitor therapy as a treatment strategy in EGFR-mutant advanced lung cancers that have developed acquired resistance to EGFR tyrosine kinase inhibitors, J. Thorac. Oncol. 8 (3) (2013) 346–351. F. Conforti, C. Catania, F. Toffalorio, et al., EGFR tyrosine kinase inhibitors beyond focal progression obtain a prolonged disease control in patients with advanced adenocarcinoma of the lung, Lung Cancer 81 (3) (2013) 440–444. B.-C. Liao, J.-H. Lee, C.-C. Lin, et al., Epidermal growth factor receptor tyrosine kinase inhibitors for non-small-cell lung cancer patients with leptomeningeal carcinomatosis, J. Thorac. Oncol. 10 (12) (2015) 1754–1761. K. Park, E.-H. Tan, K. O'Byrne, et al., Afatinib versus gefitinib as first-line treatment of patients with EGFR mutation-positive non-small-cell lung cancer (LUX-Lung 7): a phase 2B, open-label, randomised controlled trial, Lancet Oncol. 17 (5) (2016) 577–589. M. Faehling, R. Eckert, T. Kamp, et al., EGFR-tyrosine kinase inhibitor treatment beyond progression in long-term Caucasian responders to erlotinib in advanced non-small cell lung cancer: a case-control study of overall survival, Lung Cancer 80 (3) (2013) 306–312. S.B. Goldberg, G.R. Oxnard, S. Digumarthy, et al., Chemotherapy with erlotinib or chemotherapy alone in advanced non-small cell lung cancer with acquired resistance to EGFR tyrosine kinase inhibitors, Oncologist 18 (11) (2013) 1214–1220. J.-C. Soria, Y.-L. Wu, K. Nakagawa, et al., Gefitinib plus chemotherapy versus placebo plus chemotherapy in EGFR-mutation-positive non-small-cell lung cancer after progression on first-line gefitinib (IMPRESS): a phase 3 randomised trial, Lancet Oncol. 16 (8) (2015) 990–998. J.-C. Soria, S.-W. Kim, Y.-L. Wu, et al., Gefitinib/chemotherapy vs chemotherapy in EGFR mutation-positive NSCLC after progression on 1st line gefitinib (IMPRESS study): final overall survival (OS) analysis, Ann. Oncol. 27 (Suppl 6) (2016). J.-C. Soria, S. Kim, Y. Wu, et al., Gefitinib/chemotherapy versus chemotherapy in EGFR mutation positive NSCLC resistant to first-line gefitinib: IMPRESS T790M subgroup analysis, J. Thorac. Oncol. 10 (Suppl. 2) (2015) abstr: 3207. M. Schuler, J.C.-H. Yang, K. Park, et al., Afatinib beyond progression in patients with non-small-cell lung cancer following chemotherapy, erlotinib/gefitinib and afatinib: phase III randomized LUX-Lung 5 trial, Ann. Oncol. 27 (3) (2016) 417–423. M. Schuler, J.C.-H. Yang, D. Planchard, Reply to the Letter to the Editor ‘What Is the Clinical Impact of the Lux-Lung 5 Trial?’ by Addeo, Ann. Oncol. 27 (6) (2016) 1172–1173. Y.Y. Janjigian, E.F. Smit, H.J.M. Groen, et al., Dual inhibition of EGFR with afatinib and cetuximab in kinase inhibitor-resistant EGFR-mutant lung cancer with and without T790M mutations, Cancer Discov. 4 (9) (2014) 1036–1045. S.-G. Wu, Y.-N. Liu, M.-F. Tsai, et al., The mechanism of acquired resistance to irreversible EGFR tyrosine kinase inhibitor-afatinib in lung adenocarcinoma patients, Oncotarget 7 (11) (2016) 12404–12413. H.A. Yu, M.E. Arcila, N. Rekhtman, 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. 19 (8) (2013) 2240–2247. D.R. Camidge, W. Pao, L.V. Sequist, Acquired resistance to TKIs in solid tumours: learning from lung cancer, Nat. Rev. Clin. Oncol. 11 (8) (2014) 473–481. S. Popat, A. Wotherspoon, C.M. Nutting, et al., Transformation to high grade neuroendocrine carcinoma as an acquired drug resistance mechanism in EGFRmutant lung adenocarcinoma, Lung Cancer 80 (1) (2013) 1–4. M.J. Niederst, L.V. Sequist, J.T. Poirier, et al., RB loss in resistant EGFR mutant lung adenocarcinomas that transform to small-cell lung cancer, Nat. Commun. 6 (2015) 6377. L.V. Sequist, B.A. Waltman, D. Dias-Santagata, et al., Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors, Sci. Transl. Med. 3 (75) (2011) 75ra26. K. Nosaki, M. Satouchi, T. Kurata, et al., Re-biopsy status among non-small cell lung cancer patients in Japan: a retrospective study, Lung Cancer 101 (2016) 1–8. B.-C. Liao, Y.-Y. Bai, J.-H. Lee, et al., Outcomes of research biopsies in clinical trials of EGFR mutation-positive non-small cell lung cancer patients pretreated with EGFR-tyrosine kinase inhibitors, J. Formos. Med. Assoc. (2017) (Published online 9 May 2017). E.C. de Bruin, N. McGranahan, R. Mitter, et al., Spatial and temporal diversity in genomic instability processes defines lung cancer evolution, Science 346 (6206) (2014) 251–256. J. Zhang, J. Fujimoto, J. Zhang, et al., Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing, Science 346 (6206) (2014) 256–259. C. Swanton, R. Govindan, Clinical implications of genomic discoveries in lung cancer, N. Engl. J. Med. 374 (19) (2016) 1864–1873. A. Hata, N. Katakami, H. Yoshioka, et al., Spatiotemporal T790M heterogeneity in individual patients with EGFR-mutant non-small-cell lung cancer after acquired resistance to EGFR-TKI, J. Thorac. Oncol. 10 (11) (2015) 1553–1559.

Lung Cancer 110 (2017) 7–13

B.-C. Liao et al.

(2014) LB-100. [74] K. Park, J.-S. Lee, K.H. Lee, et al., BI 1482694 (HM61713), an EGFR mutant-specific inhibitor, in T790M+ NSCLC: efficacy and safety at the RP2D, ASCO Meeting Abstracts 34(15_suppl) (2016) 9055. [75] E.S. Kim, Olmutinib: first global approval, Drugs 76 (11) (2016) 1153–1157. [76] Boehringer Ingelheim Returns Development and Commercial Rights of Olmutinib to Hanmi Pharmaceutical, (2016) https://www.boehringer-ingelheim.com/pressrelease/boehringer-ingelheim-returns-development-commercial-rights-olmutinibhanmi (Accessed 7 February 2017). [77] G. Lelais, R. Epple, T.H. Marsilje, et al., Discovery of (R,E)-N-(7-Chloro-1-(1-[4(dimethylamino)but-2-enoyl]azepan-3-yl)-1H-benzo[d]imidazol-2-yl)-2-methylisonicotinamide (EGF816), a novel, potent, and WT sparing covalent inhibitor of oncogenic (L858R, ex19del) and resistant (T790M) EGFR mutants for the treatment of EGFR mutant non-small-cell lung cancers, J. Med. Chem. 59 (14) (2016) 6671–6689. [78] D.S.-W. Tan, J.C.-H. Yang, N.B. Leighl, 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 34(15_suppl) (2016) 9044. [79] S. Konagai, H. Sakagami, H. Yamamoto, et al., Abstract 2586: ASP8273 selectively inhibits mutant EGFR signal pathway and induces tumor shrinkage in EGFR mutated tumor models, Cancer Res. 75 (15 Suppl) (2015) 2586. [80] H.A. Yu, A.I. Spira, L. Horn, 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 34(15_suppl) (2016) 9050. [81] J.A. Engelman, K. Zejnullahu, T. Mitsudomi, et al., MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling, Science 316 (5827) (2007) 1039–1043. [82] J. Bean, C. Brennan, J.-Y. Shih, et al., MET amplification occurs with or without T790M mutations in EGFR mutant lung tumors with acquired resistance to gefitinib or erlotinib, Proc. Natl. Acad. Sci. 104 (52) (2007) 20932–20937. [83] K. Azuma, T. Hirashima, N. Yamamoto, et al., Phase II study of erlotinib plus tivantinib (ARQ 197) in patients with locally advanced or metastatic EGFR mutation-positive non-small-cell lung cancer just after progression on EGFR-TKI, gefitinib or erlotinib, ESMO Open 1 (4) (2016) e000063. [84] D.R. Camidge, T. Moran, I. Demedts, et al., A randomized, open-label, phase 2 study of emibetuzumab plus erlotinib (LY+E) and emibetuzumab monotherapy (LY) in patients with acquired resistance to erlotinib and MET diagnostic positive (MET Dx +) metastatic NSCLC, ASCO Meeting Abstracts 34(15_suppl) (2016) 9070. [85] Y. Wu, D. Kim, E. Felip, et al., Phase (Ph) II safety and efficacy results of a singlearm ph ib/II study of capmatinib (INC280)+ gefitinib in patients (pts) with EGFRmutated (mut), cMET-positive (cMET+) non-small cell lung cancer (NSCLC), ASCO Meeting Abstracts 34(15_suppl) (2016) 9020. [86] R. Califano, O. Romanidou, G. Mountzios, et al., Management of NSCLC disease progression after first-line EGFR tyrosine kinase inhibitors: what are the issues and potential therapies? Drugs 76 (8) (2016) 831–840. [87] N. Chen, W. Fang, J. Zhan, et al., Upregulation of PD-L1 by EGFR activation mediates the immune escape in EGFR-driven NSCLC: implication for optional immune targeted therapy for NSCLC patients with EGFR mutation, J. Thorac. Oncol. 10 (6) (2015) 910–923. [88] E.A. Akbay, S. Koyama, J. Carretero, et al., Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors, Cancer Discov. 3 (12) (2013) 1355–1363. [89] Y. Tang, W. Fang, Y. Zhang, et al., The association between PD-L1 and EGFR status and the prognostic value of PD-L1 in advanced non-small cell lung cancer patients treated with EGFR-TKIs, Oncotarget 6 (16) (2015) 14209–14219. [90] A. D'Incecco, M. Andreozzi, V. Ludovini, et al., PD-1 and PD-L1 expression in molecularly selected non-small-cell lung cancer patients, Br. J. Cancer 112 (1) (2015) 95–102. [91] A. Hata, N. Katakami, S. Nanjo, et al., Correlation between programmed deathligand 1 (PD-L1) expression and T790M status in EGFR-mutant non-small cell lung cancer (NSCLC), Ann. Oncol. 27 (Suppl. 6) (2016) 1237P. [92] C.K. Lee, J. Man, S. Lord, et al., Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer—a meta-analysis, J. Thorac. Oncol. 12 (2) (2017) 403–407. [93] J.F. Gainor, A.T. Shaw, L.V. Sequist, et al., EGFR mutations and ALK rearrangements are associated with low response rates to PD-1 pathway blockade in nonsmall cell lung cancer (NSCLC): a retrospective analysis, Clin. Cancer Res. 22 (18) (2016) 4585–4593.

[50] E. Crowley, F. Di Nicolantonio, F. Loupakis, et al., Liquid biopsy: monitoring cancer-genetics in the blood, Nat. Rev. Clin. Oncol. 10 (8) (2013) 472–484. [51] L. Sorber, K. Zwaenepoel, V. Deschoolmeester, et al., Circulating cell-free nucleic acids and platelets as a liquid biopsy in the provision of personalized therapy for lung cancer patients, Lung Cancer 107 (2017) 100–107. [52] C.-S. Tan, B.-C. Cho, R.A. Soo, Next-generation epidermal growth factor receptor tyrosine kinase inhibitors in epidermal growth factor receptor −mutant non-small cell lung cancer, Lung Cancer 93 (2016) 59–68. [53] B.-C. Liao, C.-C. Lin, J.-H. Lee, et al., Update on recent preclinical and clinical studies of T790 M mutant-specific irreversible epidermal growth factor receptor tyrosine kinase inhibitors, J. Biomed. Sci. 23 (1) (2016) 86. [54] M.R.V. Finlay, M. Anderton, S. Ashton, 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. 57 (20) (2014) 8249–8267. [55] J.C.-H. Yang, S.S. Ramalingam, P.A. Jänne, et al., 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. 11 (4 Suppl) (2016) S152–S153. [56] J.C.-H. Yang, M.-J. Ahn, D.-W. Kim, et al., Osimertinib in pretreated T790Mpositive advanced non-small-cell lung cancer: AURA study phase II extension component, J. Clin. Oncol. 35 (12) (2017) 1288–1296. [57] G. Goss, C.-M. Tsai, F.A. Shepherd, et al., Osimertinib for pretreated EGFR Thr790Met-positive advanced non-small-cell lung cancer (AURA2): a multicentre, open-label, single-arm, phase 2 study, Lancet Oncol. 17 (12) (2016) 1643–1652. [58] T.S. Mok, Y.-L. Wu, M.-J. Ahn, et al., Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer, N. Engl. J. Med. 376 (7) (2017) 629–640. [59] A.G. Sacher, C. Paweletz, S.E. Dahlberg, Prospective validation of rapid plasma genotyping for the detection of EGFR and KRAS mutations in advanced lung cancer, JAMA Oncol. 2 (8) (2016) 1014–1022. [60] G.R. Oxnard, C.P. Paweletz, Y. Kuang, 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. 20 (6) (2014) 1698–1705. [61] K.S. Thress, R. Brant, T.H. Carr, 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 90 (3) (2015) 509–515. [62] K. Taniguchi, J. Uchida, K. Nishino, et al., Quantitative detection of EGFR mutations in circulating tumor DNA derived from lung adenocarcinomas, Clin. Cancer Res. 17 (24) (2011) 7808–7815. [63] G.R. Oxnard, K.S. Thress, R.S. Alden, et al., Association between plasma genotyping and outcomes of treatment with osimertinib (AZD9291) in advanced non-small-cell lung cancer, J. Clin. Oncol. 34 (28) (2016) 3375–3382. [64] European Medicines Agency: Tagrisso: EPAR – Product Information, (2017) http:// www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/ human/004124/WC500202022.pdf (Accessed 7 February 2017). [65] G.R. Oxnard, S.S. Ramalingam, M.-J. Ahn, 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 33(15_suppl.) (2015) 2509. [66] M.J. Ahn, J. Yang, H. Yu, 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. 11 (4) (2016) S115. [67] J. Engel, A. Richters, M. Getlik, et al., Targeting drug resistance in EGFR with covalent inhibitors: a structure-based design approach, J. Med. Chem. 58 (17) (2015) 6844–6863. [68] J.W. Goldman, H.A. Wakelee, S. Gadgeel, et al., Dose optimization of rociletinib for EGFR mutated NSCLC, J. Thorac. Oncol. 10 (9) (2015) S261–S406. [69] J.W. Goldman, J.-C. Soria, H.A. Wakelee, 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 34(15_suppl) (2016) 9045. [70] C. Karlovich, J.W. Goldman, J.-M. Sun, et al., Assessment of EGFR mutation status in matched plasma and tumor tissue of NSCLC patients from a phase I study of rociletinib (CO-1686), Clin. Cancer Res. 22 (10) (2016) 2386–2395. [71] K.L. Reckamp, V.O. Melnikova, C. Karlovich, et al., A highly sensitive and quantitative test platform for detection of NSCLC EGFR mutations in urine and plasma, J. Thorac. Oncol. 11 (10) (2016) 1690–1700. [72] Clovis Oncology Announces Q1 2016 Operating Results and Corporate Update, (2016) http://phx.corporate-ir.net/phoenix.zhtml?c=247187&p=irolnewsArticle&ID=2165717 (Accessed 8 July 2016). [73] K.-O. Lee, M.Y. Cha, M. Kim, et al., Abstract LB-100: Discovery of HM61713 as an orally available and mutant EGFR selective inhibitor, Cancer Res. 74 (19 Suppl)

13