Next Article in Journal
Prognostic Relevance of Circulating 25OHD Fractions for Early Recovery and Survival in Patients with Hip Fracture
Next Article in Special Issue
CXCR4 Based Therapeutics for Non-Small Cell Lung Cancer (NSCLC)
Previous Article in Journal
Brief Review: Ergospirometry in Patients with Obstructive Sleep Apnea Syndrome
Previous Article in Special Issue
Diagnosing Lung Cancer: The Complexities of Obtaining a Tissue Diagnosis in the Era of Minimally Invasive and Personalised Medicine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeted Molecular Treatments in Non-Small Cell Lung Cancer: A Clinical Guide for Oncologists

1
Chris O’Brien Lifehouse, 119-143 Missenden Road Camperdown, Camperdown, NSW 2050, Australia
2
Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
3
School of Medicine, Western Sydney University, Sydney, NSW 2560, Australia
4
Royal Prince Alfred Hospital, 50 Missenden Road Camperdown, Camperdown, NSW 2050, Australia
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2018, 7(8), 192; https://doi.org/10.3390/jcm7080192
Submission received: 20 July 2018 / Revised: 30 July 2018 / Accepted: 30 July 2018 / Published: 31 July 2018

Abstract

:
Targeted molecular treatments have changed the way non-small cell lung cancer (NSCLC) is managed. Epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), v-raf murine sarcoma viral oncogene homolog B1 (BRAF), and c-ros oncogene 1 (ROS1) mutations are now used to guide specific anti-cancer therapies to improve patient outcomes. New targeted molecular treatments are constantly being developed and evaluated as a means to improve efficacy, overcome resistance, or minimise toxicity. This review article summarises the current evidence for the efficacy, resistance mechanisms, and safety of targeted molecular treatments against specific mutations in NSCLC.

1. Introduction

Worldwide, lung cancer is the most commonly diagnosed cancer and the leading cause of cancer mortality. It accounted for 1.8 million new cases in 2012 (12.9% of all cancers) and caused 1.6 million deaths (19.4% of all cancers) [1]. Non-small cell lung cancer (NSCLC) accounts for 85% of all lung cancer cases, and adenocarcinoma is the most frequent histological subtype, accounting for nearly 40% of all lung cancer cases [2]. Prior to the use of targeted therapy and immunotherapy, patients with advanced lung cancer had a poor prognosis. Platinum doublet chemotherapy, which was the standard of care for all patients with incurable locally advanced or metastatic NSCLC [3], achieved a response rate of 19%, and a median overall survival of 7.9 months [4]. Modern trials with targeted treatments have resulted in significantly better outcomes, with median overall survival now extending to around, or even beyond, two years in selected populations [5,6,7,8].
Specific mutations have been found to be prevalent in lung adenocarcinomas, some of which are predictors for response to targeted treatment. The most common mutations occur in kirsten rat sarcoma virus (KRAS, found in 24% of cases), epidermal growth factor receptor (EGFR, 17%), anaplastic lymphoma kinase (ALK, 3%), v-raf murine sarcoma viral oncogene homolog B1 (BRAF, 2%), and c-ros oncogene 1 (ROS1, 1–2%) [9,10]. We will explore the current evidence for targeted therapy for different mutations in NSCLC, with the aim of providing clinical guidance for oncologists treating NSCLC.

2. EGFR Mutation-Positive NSCLC

EGFR mutations were first described in lung adenocarcinomas in 2004 [11] and were rapidly recognised as a predictor for response to EGFR tyrosine kinase inhibitors (TKIs). The frequency of mutations in this gene varies based on phenotypic characteristics of patients. They occur more frequently in Asian non-smoking women, with an incidence up to 40% [12]. The most common EGFR mutations are exon 19 deletions and exon 21 L858R point mutations [9]. Together, these two mutations account for 90% of all EGFR mutations [13]. The remaining EGFR mutations consist of a range of rarer mutations (which can either be sensitising or non-sensitising with respect to EGFR TKIs), and include exon 18 insertions, G719X point mutations in exon 18 (1–4%), exon 20 mutations (2–5%), and complex mutations (1–2%) [14,15].
EGFR TKIs have been developed to treat EGFR mutation-positive lung cancers, and a list of these are included in Table 1.

2.1. Efficacy

Early studies investigated the efficacy of EGFR TKIs in a pre-treated population of unselected patients with advanced NSCLC. The BR.21 trial found a 30% improvement in overall survival in patients treated with erlotinib compared to placebo, with an absolute survival benefit of two months [16]. Other studies in unselected populations have not demonstrated a statistically significant overall survival benefit of using EGFR TKIs when compared to chemotherapy or placebo [17,18,19,20], in combination with chemotherapy [21,22], or as maintenance therapy after chemotherapy [23,24,25,26,27,28]. The role of erlotinib in EGFR wild-type NSCLC as maintenance therapy was most recently discounted in a phase III trial when erlotinib as maintenance treatment resulted in a median overall survival of 9.7 months compared to a median overall survival of 9.5 months when erlotinib was used on progression [29].
Although designed as a study to select patients based on phenotypic characteristics (ethnicity and smoking history), the Iressa Pan-Asia Study (IPASS) study was the first to demonstrate differential outcomes for patients treated with an EGFR TKI (gefitinib) based on the presence or absence of an activating EGFR mutation. These data were based on a subset analysis of patients, which demonstrated that the benefit of EGFR TKIs was exclusive to patients with an EGFR mutation [30]. Subsequently, trials have been performed investigating gefitinib, erlotinib, or icotinib in treatment-naive patients selected for the presence of an activating EGFR mutation. The results of these trials are summarised in Table 2. Treatment with an EGFR TKI typically resulted in superior median progression-free survival (PFS) of 9–13 months when compared to platinum doublet chemotherapy, which had median PFS in the range of 4–6 months. Furthermore, the response rates were as high as 83% in patients on an EGFR TKI, compared to 36% in patients who received chemotherapy. Due to crossover between the study arms, none of these trials demonstrated a statistically significant improvement in overall survival, which can extend up to 38 months [5,6,30,31,32,33,34,35,36,37].
The question of whether there are meaningful differences between first-generation TKIs has been addressed in three small studies. In these studies, no significant differences between erlotinib and gefitinib were observed, although there were some differences in the pattern of side effects [47,48,49]. The benefits of EGFR TKIs observed with EGFR mutation-positive cancers does not translate to patients who have high EGFR expression identified using immunohistochemistry or increased EGFR copy number detected by fluorescence in situ hybridization [31,50].
In an effort to improve further outcomes for these patients, second-generation EGFR TKIs have been developed. Afatinib and dacomitinib were both designed to bind covalently to the mutated EGFR protein. Additionally, these agents are pan-HER inhibitors and block activation of other members of the EGFR/HER family. These agents result in superior PFS compared to chemotherapy in treatment-naïve patients with EGFR-mutated tumours [7,39,40,43]. There are only limited data comparing second- and first-generation agents to each other. In a randomised phase II study, Lux Lung 7, afatinib and gefitinib were compared as first-line therapy for treatment-naïve patients. Afatinib was found to have a statistically significant PFS benefit; however, the absolute benefit was small (0.1 months) [41,42] and there seems to be little meaningful efficacy difference between the agents. The recently published overall survival data for the phase III trial of dacomitinib compared to gefitinib did show a statistically significant improvement in median overall survival (34.1 vs. 26.8 months). Despite these results, the clinical use of dacomitinib is likely to be limited by the FLAURA trial (discussed in the next paragraph), especially given the toxicity profile of dacomitinib [44].
Osimertinib, a third-generation irreversible EGFR TKI, has greater efficacy than the first- and second-generation agents. FLAURA, a randomised study comparing first-line osimertinib to erlotinib or gefitinib, showed that, despite similar response rates, patients treated with osimertinib had better PFS (18.9 months vs. 10.2 months). In patients who had stable central nervous system (CNS) metastases at time of trial enrolment, osimertinib also had a superior PFS to the first-generation EGFR TKIs (15.2 months vs. 9.6 months). Overall survival data from this study are currently immature [45].
There are no prospective studies investigating the efficacy of EGFR TKIs in patients with uncommon EGFR mutations. Observational data with small sample sizes do indicate activity of first-generation EGFR TKIs in some of the rarer EGFR mutations; however, the response rates may be lower compared to patients with common EGFR mutations [14]. In vitro data has demonstrated that cells with exon 18 mutations had better responses to second-generation EGFR TKIs such as afatinib and neratinib compared to first- or third-generation EGFR TKIs [51]. Ad hoc analyses of trial data showed a greater benefit of afatinib in patients with point mutations and duplications in exons 18–21, with a disease control rate of 84%, median PFS of 10.7 months, and median overall survival of 19 months. Meanwhile, patients who had de novo T790M mutations or exon 20 insertions had lower response rates (15% and 9%, respectively), shorter median PFS (2.9 months and 2.7 months), and shorter overall survival (14.9 months and 9.2 months) [52]. The resistance to EGFR TKIs and the poorer prognosis associated with exon 20 mutations was also seen in a retrospective analysis of 20 patients by Noronha et al. [53]. A phase II study of poziotinib in patients with EGFR exon 20 mutant advanced NSCLC is currently recruiting, with early results suggesting activity [54]. Without phase III evidence to support a different approach, EGFR TKIs are still the recommended first-line option for patients with uncommon but activating EGFR mutations.
Although several studies have been conducted in the adjuvant setting, only one trial has been completed where patient selection was prospectively based on the presence of an activating EGFR mutation. Consequently, interpretation of results is difficult. Based on the available data, EGFR TKIs may improve PFS, though the data for overall survival remains immature [55,56,57]. A phase III trial of adjuvant osimertinib in EGFR mutation-positive patients is currently recruiting, with results expected in late 2021 [58]. EGFR TKIs have yet to be implemented into routine clinical practice in this setting.

2.2. Resistance

Primary resistance, where the best response achieved is progressive disease, is a relatively rare occurrence, and is noted in 4–10% of EGFR mutation-positive NSCLC treated with an EGFR TKI [30,32,33,34,36,39,40,41,43,45]. Acquired resistance, where progressive disease develops after a period of objective response or stability, will eventually occur in all patients treated with an EGFR TKI. The most common resistance mechanisms are the T790M mutation (49–62%), c-MET amplification (5–22%), human epidermal growth factor receptor 2 (HER2) amplification (12%), epithelial–mesenchymal transition (20%), and small cell lung cancer transformation (3–14%) [59,60,61,62,63,64].
Continuation of a first- or second-generation EGFR TKI after progression on these agents has not been shown to improve outcomes [65,66]. However, osimertinib has demonstrated activity against the exon 20 T790M mutation, which occurs when threonine at position 790 is replaced by methionine [67]. This substitution leads to increased ATP affinity, cell proliferation and survival, and, ultimately, resistance to first generation TKIs [68,69]. AURA3 compared treatment with osimertinib to standard platinum doublet chemotherapy in patients with secondary T790M mutation after progression with a first-generation EGFR TKI. Osimertinib resulted in better PFS (10.1 vs. 4.4 months) and higher response rates (70% vs. 30%). A PFS benefit in patients with stable CNS metastases was also seen with osimertinib (8.5 months vs. 4.2 months) [46].

2.3. Toxicity

The adverse events of the first-generation EGFR TKIs are often Grade 1 or Grade 2 and are less likely to cause dose reductions (~20%) or drug discontinuation (<6%) compared to chemotherapy. The most common adverse events of any grade are rash or acne (66–80%), diarrhoea (25–55%) and elevated liver transaminases, particularly of alanine aminotransferase (37–55%). Most of the Grade 3 to Grade 5 adverse events occur in <6% of patients, with the exception of elevated transaminases which can occur in up to 26% of patients. The rates of pneumonitis are uncommon, in order of 1–5% [6,30,33,34,36]. There are subtle differences in the pattern of toxicity between erlotinib and gefitinib. Erlotinib is more likely to cause rash or diarrhoea, while gefitinib is more likely to cause liver function abnormalities [49]. However, these differences have little meaningful clinical impact.
Second-generation EGFR TKIs such as afatinib are more likely to result in adverse events when compared to chemotherapy or first-generation EGFR TKIs. Adverse events that are Grade 3 or higher occur in up to 49% of patients and include severe diarrhoea (up to 15%) and rash (up to 16%) [7,41]. In most studies, these have been successfully managed with dose reductions, and consequently the rate of discontinuation of drug as a result of toxicity is comparable to first-generation EGFR TKIs (6–10%) [41,43].
Meanwhile, osimertinib has been shown to have a better toxicity compared to first-generation EGFR TKIs, with a lower frequency and lower severity of adverse events of rash and transaminase elevations [45].

3. ALK-Positive NSCLC

ALK rearrangements were first identified in 2007 [70]. Although a single rearrangement with echinoderm microtubule-associated protein-like 4 (EML4) was initially identified, it has become apparent that there are several variants based on the location of the rearrangement. These variants may have prognostic significance with differences in outcome noted between them [71].
ALK rearrangements are more common in younger patients who have never smoked, or who have a light smoking history [72]. The commonly used ALK TKIs are listed in Table 1, and the pivotal randomised controlled trials for ALK TKIs are listed in Table 3.

3.1. Efficacy

Crizotinib was initially developed as a mesenchymal-to-epithelial transition (MET) inhibitor. However, during phase I trials it became apparent that it had substantial activity against ALK-rearranged tumours [73]. It was the first ALK TKI to show a clinically significant benefit in ALK-positive NSCLC, when it was evaluated in the second-line setting compared to chemotherapy. Patients treated with crizotinib had a median PFS of 7.7 months compared to 3.0 months with chemotherapy [8]. Data from this study resulted in crizotinib becoming a standard treatment in this group of patients.
Subsequently, ALK TKIs have been evaluated in the first-line setting. Crizotinib was compared to standard platinum doublet chemotherapy in the PROFILE 1014 study, with better PFS and higher response rates observed for crizotinib than for chemotherapy [74]. A study with the same design but using ceritinib resulted in similar outcomes [75]. Both crizotinib and ceritinib have shown activity in stable CNS metastases compared to chemotherapy, with median PFS of 9.0 months with crizotinib (vs. 4.0 months) and 10.7 months with ceritinib (vs. 6.7 months) [75,76].
Most recently, alectinib has been compared to crizotinib in the first-line setting in the ALEX study. In this study, alectinib had superior efficacy, with the median PFS not reached in the alectinib arm and 11.1 months in the crizotinib arm. Alectinib does have meaningful CNS activity compared to crizotinib. In patients treated with alectinib, the cumulative rate of CNS metastases was markedly reduced (9.4% vs. 41.4%), and, in patients with known CNS metastases at time of trial enrolment, the response rates were higher (59% vs. 26%) [77].
Ensartinib, a third-generation ALK TKI, had response rates of 66% and a median PFS 9.2 months in an early phase I/II trial. Recruitment is currently ongoing for a first-line phase III trial of ensartinib compared to crizotinib in ALK-positive NSCLC [78].
In parallel with trials of EGFR TKIs, these studies have all allowed crossover of patients. Consequently, it has not been possible to demonstrate improvements in overall survival.

3.2. Resistance

Acquired resistance usually occurs within the first two years of ALK TKI treatment and can occur due to acquired point mutations in ALK, or due to bypass track activation via activation of EGFR or amplification of CKIT. The most common resistance mutation is L1196M, though there are often multiple resistance mutations that occur concurrently [72].
Alectinib and ceritinib have been studied in phase III trials in the second line setting after resistance to crizotinib. They had superior efficacy when compared to chemotherapy with response rates between 35–40%, a median PFS of approximately 9 months, and evidence of efficacy in patients with known CNS metastases [79,80]. Phase II trials investigating brigatinib and lorlatinib show response rates up to 65% in patients with and without CNS metastases [81,82]. A phase II trial for entrectinib, a pan-tropomyosin receptor kinase (TRK), -ROS1, and -ALK TKI, is currently underway for mutation-positive patients with solid tumours after promising phase I data [83].
Using in vitro data on cell lines, Gainor et al. demonstrated that the activity of second-line therapy is influenced by the specific resistant mutation that occurs, as shown in Table 4 [84]. Sequential ALK TKI can also lead to the development of compound ALK mutations that may suggest resistance to specific ALK TKIs [85], thus highlighting the importance of patient selection and treatment sequencing when determining the optimal management pathway for each patient.

3.3. Toxicity

The most common toxicities of any grade due to crizotinib or ceritinib are diarrhoea (up to 85%), nausea (up to 69%), vomiting (up to 66%), constipation (up to 43%), and raised liver transaminases (up to 53%). Crizotinib is also associated with visual disorders such as visual impairment, photophobia, or blurred vision (71%), oedema (49%), and upper respiratory tract infections (32%) [74,75]. Alectinib is associated with hyperbilirubinaemia but has lower overall rates of gastrointestinal adverse events compared to the other ALK TKIs [77]. The main toxicities from lorlatinib are hypercholesterolaemia and hypertriglyceridaemia [82].

4. ROS1-Positive NSCLC

ROS1 rearrangements are more likely to be present in younger, non-smoking Asian patients [10]. There are no TKIs that have been designed to specifically target ROS1. However, in early clinical trials, it became apparent that ALK TKIs had activity in ROS1-positive patients. Crizotinib had a response rate of 71.7% and a median PFS of 15.9 months in a phase II trial of 127 patients [87]. Ceritinib had a response rate of 62% and a median PFS of 9.3 months in a phase II trial of 32 patients, though the median PFS improved to 19.3 months in patients who were treatment-naïve [88]. Finally, a phase I trial of lorlatinib included 12 ROS1-positive patients and achieved a response rate of 50% with a median PFS of 7 months [82]. Importantly, the ALK inhibitor alectinib, which has a structure that is distinct from the other agents mentioned above, is not active in ROS1-mutated tumours [89].

5. BRAF Mutation-Positive NSCLC

BRAF V600E mutations have been noted in several tumour types, most notably melanoma. They may also occur in <5% of NSCLC and are often associated with poor response to platinum-based chemotherapy. The availability of RAF inhibitors has led to their evaluation in NSCLC, although the rarity of the mutation means that the data is limited to phase II trials. Dabrafenib in monotherapy resulted in a response rate of 33%, median PFS of 5.5 months and median overall survival of 12.7 months [90]. In keeping with the experience in melanoma, the addition of the Mitogen-activated protein kinase (MEK) inhibitor trametinib resulted in better outcomes (response rates of 64% and a median PFS of 10.9 months) [91]. These agents have regulatory approval for use in NSCLC from the Food and Drug Administration (FDA) and the European Medicines Agency.

Toxicity

The most common adverse event from dabrafenib and trametinib was pyrexia, which occurred in 64% of patients. Other toxicities such as nausea, diarrhoea, fatigue, peripheral oedema, vomiting, dry skin, anorexia, and headache each occurred in 25–36% patients [91].

6. KRAS Mutation-Positive NSCLC

KRAS mutations are the most common mutation found in NSCLC. They often occur at codon 12, and can rarely occur at codon 13 and 61 [92]. They are mutually exclusive with EGFR mutations and ALK translocations in almost all cases [93]. KRAS mutations are more common in smokers, and also convey a poorer prognosis [94,95].
There are no targeted treatments that have a clinically meaningful benefit in patients with KRAS mutations. While MEK inhibitors such as selumetinib and trametinib held promise in early research, benefit could not be demonstrated in larger trials in patients with advanced NSCLC. A phase III trial of second-line selumetinib and docetaxel compared to docetaxel alone showed no difference in PFS (3.9 months vs. 2.8 months) and no difference in median overall survival (8.7 months vs. 7.9 months) [96]. A phase II trial of second-line trametinib compared to docetaxel alone showed no difference in median PFS (12 weeks vs. 11 weeks) with a response rate of 12% in both arms [97].

7. Other Mutations in NSCLC

There are other less common mutations that have been investigated as potential drug targets. Mutations in the mesenchymal-to-epithelial transition (MET) gene that cause exon 14 skipping occur in 3% of non-squamous NSCLC, and are more likely in older patients [98]. Patients with MET mutations who never received MET inhibitor therapy had a poor prognosis (median overall survival 8.1 months), which was worse if there was concurrent MET amplification (median overall survival 5.2 months). Treatment with a MET inhibitor extended the median overall survival to 24.6 months [99]. Early trials have suggested an antitumour effect of crizotinib in patients with MET exon 14-altered NSCLC, and in patients with MET amplification [100,101]. Other MET TKIs, such as capmatinib, tepotinib, salvolitinib, cabozantinib, glesatinib, and merestinib, are currently being investigated for patients with MET mutations.
RET rearrangements occur in 1–2% of patients with NSCLC [102,103]. Vandetanib, lenvatinib, and cabozantinib, which are multitargeted kinase inhibitors, have demonstrated antitumour effect in RET-positive NSCLC in phase II trials with response rates ranging from 16% to 53% [104,105,106,107]. Alectinib has also shown promising pre-clinical evidence against RET-positive NSCLC [89].
HER2 mutations occur in 1–6% of patients with NSCLC [108,109,110] and are more common in never-smokers. Gender or ethnicity did not affect incidence of the HER2 mutations [109]. Retrospective studies have shown responses to HER2-targeted therapies including trastuzumab, neratinib, afatinib, lapatinib, and trastuzumab emtansine [108,111]. Phase II trials have confirmed anti-cancer activity with trastuzumab emtansine and afatinib in patients with HER2-mutated NSCLC [112,113]. A further phase II trial investigating the benefit of afatinib is underway. In HER2-amplified NSCLC, phase II trials have shown no clinical benefit of trastuzumab monotherapy, trastuzumab with chemotherapy, trastuzumab emtansine, or pertuzumab [114,115,116,117].

8. Conclusions

Targeted molecular therapies have revolutionised the management of advanced NSCLC and have become the international standard of care for patients with driver mutations. Individualised patient care has never been so important. The optimal sequencing of TKIs to provide the best outcomes for our patients is unknown, especially in the immunotherapy era of oncology. An improved understanding of molecular resistance will guide the development of new treatments and assist with decision-making about treatment selection.

Author Contributions

K.T.B. conducted a literature review and drafted the original manuscript. M.B., S.K. and W.A.C. reviewed and edited the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ferlay, J.; Soerjomataram, I.; Dikshit, R.; Eser, S.; Mathers, C.; Rebelo, M.; Parkin, D.M.; Forman, D.; Bray, F. Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int. J. Cancer 2015, 136, E359–E386. [Google Scholar] [CrossRef] [PubMed]
  2. Dela Cruz, C.S.; Tanoue, L.T.; Matthay, R.A. Lung cancer: Epidemiology, etiology, and prevention. Clin. Chest Med. 2011, 32, 605–644. [Google Scholar] [CrossRef] [PubMed]
  3. Rajeswaran, A.; Trojan, A.; Burnand, B.; Giannelli, M. Efficacy and side effects of cisplatin- and carboplatin-based doublet chemotherapeutic regimens versus non-platinum-based doublet chemotherapeutic regimens as first line treatment of metastatic non-small cell lung carcinoma: A systematic review of randomized controlled trials. Lung Cancer 2008, 59, 1–11. [Google Scholar] [CrossRef] [PubMed]
  4. Schiller, J.H.; Harrington, D.; Belani, C.P.; Langer, C.; Sandler, A.; Krook, J.; Zhu, J.; Johnson, D.H. Comparison of four chemotherapy regimens for advanced non-small-cell lung cancer. N. Engl. J. Med. 2002, 346, 92–98. [Google Scholar] [CrossRef] [PubMed]
  5. Yoshioka, H.; Mitsudomi, T.; Morita, S.; Yatabe, Y.; Negoro, S.; Okamoto, I.; Seto, T.; Satouchi, M.; Tada, H.; Hirashima, T.; et al. Final overall survival results of WJTOG 3405, a randomized phase 3 trial comparing gefitinib (G) with cisplatin plus docetaxel (CD) as the first-line treatment for patients with non-small cell lung cancer (NSCLC) harboring mutations of the epidermal growth factor receptor (EGFR). J. Clin. Oncol. 2014, 32 (Suppl. 15), 8117. [Google Scholar] [CrossRef]
  6. Wu, Y.L.; Zhou, C.; Liam, C.K.; Wu, G.; Liu, X.; Zhong, Z.; Lu, S.; Cheng, Y.; Han, B.; Chen, L.; et al. First-line erlotinib versus gemcitabine/cisplatin in patients with advanced EGFR mutation-positive non-small-cell lung cancer: Analyses from the phase III, randomized, open-label, ENSURE study. Ann. Oncol. 2015, 26, 1883–1889. [Google Scholar] [CrossRef] [PubMed]
  7. Yang, J.C.-H.; Wu, Y.-L.; Schuler, M.; Sebastian, M.; Popat, S.; Yamamoto, N.; Zhou, C.; Hu, C.-P.; O’Byrne, K.; Feng, J.; et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): Analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol. 2015, 16, 141–151. [Google Scholar] [CrossRef]
  8. Shaw, A.T.; Kim, D.-W.; Nakagawa, K.; Seto, T.; Crino, L.; Ahn, M.-J.; De Pas, T.; Besse, B.; Solomon, B.J.; Blackhall, F.; et al. Crizotinib versus chemotherapy in advanced ALK-positive lung cancer. N. Engl. J. Med. 2013, 368, 2385–2394. [Google Scholar] [CrossRef] [PubMed]
  9. Kris, M.G.; Johnson, B.E.; Berry, L.D.; Kwiatkowski, D.J.; Iafrate, A.J.; Wistuba, I.I.; Varella-Garcia, M.; Franklin, W.A.; Aronson, S.L.; Su, P.-F.; et al. Using multiplexed assays of oncogenic drivers in lung cancers to select targeted drugs. JAMA 2014, 311, 1998–2006. [Google Scholar] [CrossRef] [PubMed]
  10. Bergethon, K.; Shaw, A.T.; Ou, S.-H.I.; Katayama, R.; Lovly, C.M.; McDonald, N.T.; Massion, P.P.; Siwak-Tapp, C.; Gonzalez, A.; Fang, R.; et al. ROS1 rearrangements define a unique molecular class of lung cancers. J. Clin. Oncol. 2012, 30, 863–870. [Google Scholar] [CrossRef] [PubMed]
  11. Lynch, T.J.; Bell, D.W.; Sordella, R.; Gurubhagavatula, S.; Okimoto, R.A.; Brannigan, B.W.; Harris, P.L.; Haserlat, S.M.; Supko, J.G.; Haluska, F.G.; et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non–small-cell lung cancer to gefitinib. N. Engl. J. Med. 2004, 350, 2129–2139. [Google Scholar] [CrossRef] [PubMed]
  12. Yatabe, Y.; Kerr, K.M.; Utomo, A.; Rajadurai, P.; Tran, V.K.; Du, X.; Chou, T.-Y.; Enriquez, M.L.D.; Lee, G.K.; Iqbal, J.; et al. EGFR mutation testing practices within the Asia Pacific region: Results of a multicenter diagnostic survey. J. Thorac. Oncol. 2015, 10, 438–445. [Google Scholar] [CrossRef] [PubMed]
  13. Li, A.R.; Chitale, D.; Riely, G.; Pao, W.; Miller, V.A.; Zakowski, M.F.; Rusch, V.; Kris, M.G.; Ladanyi, M. EGFR mutations in lung adenocarcinomas: Clinical testing experience and relationship to EGFR gene copy number and immunohistochemical expression. J. Mol. Diagn. 2008, 10, 242–248. [Google Scholar] [CrossRef] [PubMed]
  14. Beau-Faller, M.; Prim, N.; Ruppert, A.-M.; Nanni-Metéllus, I.; Lacave, R.; Lacroix, L.; Escande, F.; Lizard, S.; Pretet, J.-L.; Rouquette, I.; et al. Rare EGFR exon 18 and exon 20 mutations in non-small-cell lung cancer on 10 117 patients: A multicentre observational study by the French ERMETIC-IFCT network. Ann. Oncol. 2013, 25, 126–131. [Google Scholar] [CrossRef] [PubMed]
  15. Shi, Y.; Au, J.S.-K.; Thongprasert, S.; Srinivasan, S.; Tsai, C.-M.; Khoa, M.T.; Heeroma, K.; Itoh, Y.; Cornelio, G.; Yang, P.-C. A prospective, molecular epidemiology study of EGFR mutations in Asian patients with advanced non–small-cell lung cancer of adenocarcinoma histology (PIONEER). J. Thorac. Oncol. 2014, 9, 154–162. [Google Scholar] [CrossRef] [PubMed]
  16. Shepherd, F.A.; Pereira, J.R.; Ciuleanu, T.; Tan, E.H.; Hirsh, V.; Thongprasert, S.; Campos, D.; Maoleekoonpiroj, S.; Smylie, M.; Martins, R.; et al. Erlotinib in previously treated non-small-cell lung cancer. N. Engl. J. Med. 2005, 353, 123–132. [Google Scholar] [CrossRef] [PubMed]
  17. Kim, E.S.; Hirsh, V.; Mok, T.; Socinski, M.A.; Gervais, R.; Wu, Y.-L.; Li, L.-Y.; Watkins, C.L.; Sellers, M.V.; Lowe, E.S.; et al. Gefitinib versus docetaxel in previously treated non-small-cell lung cancer (INTEREST): A randomised phase III trial. Lancet 2008, 372, 1809–1818. [Google Scholar] [CrossRef]
  18. Ciuleanu, T.; Stelmakh, L.; Cicenas, S.; Miliauskas, S.; Grigorescu, A.C.; Hillenbach, C.; Johannsdottir, H.K.; Klughammer, B.; Gonzalez, E.E. Efficacy and safety of erlotinib versus chemotherapy in second-line treatment of patients with advanced, non-small-cell lung cancer with poor prognosis (TITAN): A randomised multicentre, open-label, phase 3 study. Lancet Oncol. 2012, 13, 300–308. [Google Scholar] [CrossRef]
  19. Miller, V.A.; Hirsh, V.; Cadranel, J.; Chen, Y.-M.; Park, K.; Kim, S.-W.; Zhou, C.; Su, W.-C.; Wang, M.; Sun, Y.; et al. Afatinib versus placebo for patients with advanced, metastatic non-small-cell lung cancer after failure of erlotinib, gefitinib, or both, and one or two lines of chemotherapy (LUX-Lung 1): A phase 2b/3 randomised trial. Lancet Oncol. 2012, 13, 528–538. [Google Scholar] [CrossRef]
  20. Ellis, P.M.; Shepherd, F.A.; Millward, M.; Perrone, F.; Seymour, L.; Liu, G.; Sun, S.; Cho, B.C.; Morabito, A.; Leighl, N.B.; et al. Dacomitinib compared with placebo in pretreated patients with advanced or metastatic non-small-cell lung cancer (NCIC CTG BR.26): A double-blind, randomised, phase 3 trial. Lancet Oncol. 2014, 15, 1379–1388. [Google Scholar] [CrossRef]
  21. Herbst, R.S.; Prager, D.; Hermann, R.; Fehrenbacher, L.; Johnson, B.E.; Sandler, A.; Kris, M.G.; Tran, H.T.; Klein, P.; Li, X.; et al. TRIBUTE: A phase III trial of erlotinib hydrochloride (OSI-774) combined with carboplatin and paclitaxel chemotherapy in advanced non-small-cell lung cancer. J. Clin. Oncol. 2005, 23, 5892–5899. [Google Scholar] [CrossRef] [PubMed]
  22. Wu, Y.-L.; Lee, J.S.; Thongprasert, S.; Yu, C.-J.; Zhang, L.; Ladrera, G.; Srimuninnimit, V.; Sriuranpong, V.; Sandoval-Tan, J.; Zhu, Y.; et al. Intercalated combination of chemotherapy and erlotinib for patients with advanced stage non-small-cell lung cancer (FASTACT-2): A randomised, double-blind trial. Lancet Oncol. 2013, 14, 777–786. [Google Scholar] [CrossRef]
  23. Takeda, K.; Hida, T.; Sato, T.; Ando, M.; Seto, T.; Satouchi, M.; Ichinose, Y.; Katakami, N.; Yamamoto, N.; Kudoh, S.; et al. Randomized phase III trial of platinum-doublet chemotherapy followed by gefitinib compared with continued platinum-doublet chemotherapy in Japanese patients with advanced non-small-cell lung cancer: Results of a west Japan thoracic oncology group trial (WJTOG0203). J. Clin. Oncol. 2010, 28, 753–760. [Google Scholar] [CrossRef] [PubMed]
  24. Cappuzzo, F.; Ciuleanu, T.; Stelmakh, L.; Cicenas, S.; Szczésna, A.; Juhász, E.; Esteban, E.; Molinier, O.; Brugger, W.; Melezínek, I.; et al. Erlotinib as maintenance treatment in advanced non-small-cell lung cancer: A multicentre, randomised, placebo-controlled phase 3 study. Lancet Oncol. 2010, 11, 521–529. [Google Scholar] [CrossRef]
  25. Coudert, B.; Ciuleanu, T.; Park, K.; Wu, Y.L.; Giaccone, G.; Brugger, W.; Gopalakrishna, P.; Cappuzzo, F. Survival benefit with erlotinib maintenance therapy in patients with advanced non-small-cell lung cancer (NSCLC) according to response to first-line chemotherapy. Ann. Oncol. 2012, 23, 388–394. [Google Scholar] [CrossRef] [PubMed]
  26. Johnson, B.E.; Kabbinavar, F.; Fehrenbacher, L.; Hainsworth, J.; Kasubhai, S.; Kressel, B.; Lin, C.-Y.; Marsland, T.; Patel, T.; Polikoff, J.; et al. ATLAS: Randomized, double-blind, placebo-controlled, phase IIIB trial comparing bevacizumab therapy with or without erlotinib, after completion of chemotherapy, with bevacizumab for first-line treatment of advanced non-small-cell lung cancer. J. Clin. Oncol. 2013, 31, 3926–3934. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, L.; Ma, S.; Song, X.; Han, B.; Cheng, Y.; Huang, C.; Yang, S.; Liu, X.; Liu, Y.; Lu, S.; et al. Gefitinib versus placebo as maintenance therapy in patients with locally advanced or metastatic non-small-cell lung cancer (INFORM; C-TONG 0804): A multicentre, double-blind randomised phase 3 trial. Lancet Oncol. 2012, 13, 466–475. [Google Scholar] [CrossRef]
  28. Zhao, H.; Fan, Y.; Ma, S.; Song, X.; Han, B.; Cheng, Y.; Huang, C.; Yang, S.; Liu, X.; Liu, Y.; et al. Final overall survival results from a phase III, randomized, placebo-controlled, parallel-group study of gefitinib versus placebo as maintenance therapy in patients with locally advanced or metastatic non-small-cell lung cancer (INFORM; C-TONG 0804). J. Thorac. Oncol. 2015, 10, 655–664. [Google Scholar] [CrossRef] [PubMed]
  29. Cicènasa, S.; Geater, S.L.; Petrov, P.; Hotko, Y.; Hooper, G.; Xia, F.; Mudie, N.; Wu, Y.-L. Maintenance erlotinib versus erlotinib at disease progression inpatients with advanced non-small-cell lung cancer who have notprogressed following platinum-based chemotherapy (IUNO study). Lung Cancer 2016, 102, 30–37. [Google Scholar] [CrossRef] [PubMed]
  30. Mok, T.S.; Wu, Y.-L.; Thongprasert, S.; Yang, C.-H.; Chu, D.-T.; Saijo, N.; Sunpaweravong, P.; Han, B.; Margono, B.; Ichinose, Y.; et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N. Engl. J. Med. 2009, 361, 947–957. [Google Scholar] [CrossRef] [PubMed]
  31. Fukuoka, M.; Wu, Y.-L.; Thongprasert, S.; Sunpaweravong, P.; Leong, S.-S.; Sriuranpong, V.; Chao, T.-Y.; Nakagawa, K.; Chu, D.-T.; Saijo, N.; et al. Biomarker analyses and final overall survival results from a phase III, randomized, open-label, first-line study of gefitinib versus carboplatin/paclitaxel in clinically selected patients with advanced non-small-cell lung cancer in Asia (IPASS). J. Clin. Oncol. 2011, 29, 2866–2874. [Google Scholar] [CrossRef] [PubMed]
  32. Mitsudomi, T.; Morita, S.; Yatabe, Y.; Negoro, S.; Okamoto, I.; Tsurutani, J.; Seto, T.; Satouchi, M.; Tada, H.; Hirashima, T.; 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, 121–128. [Google Scholar] [CrossRef]
  33. Maemondo, M.; Inoue, A.; Kobayashi, K.; Sugawara, S.; Oizumi, S.; Isobe, H.; Gemma, A.; Harada, M.; Yoshizawa, H.; Kinoshita, I.; et al. Gefitinib or chemotherapy for non-small-cell lung cancer with mutated EGFR. N. Engl. J. Med. 2010, 362, 2380–2388. [Google Scholar] [CrossRef] [PubMed]
  34. Zhou, C.; Wu, Y.-L.; Chen, G.; Feng, J.; Liu, X.-Q.; Wang, C.; Zhang, S.; Wang, J.; Zhou, S.; Ren, S.; 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. 2011, 12, 735–742. [Google Scholar] [CrossRef]
  35. Zhou, C.; Wu, Y.L.; Chen, G.; Feng, J.; Liu, X.Q.; Wang, C.; Zhang, S.; Wang, J.; Zhou, S.; Ren, S.; et al. Final overall survival results from a randomised, phase III study of erlotinib versus chemotherapy as first-line treatment of EGFR mutation-positive advanced non-small-cell lung cancer (OPTIMAL, CTONG-0802). Ann. Oncol. 2015, 26, 1877–1883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Rosell, R.; Carcereny, E.; Gervais, R.; Vergnenegre, A.; Massuti, B.; Felip, E.; Palmero, R.; Garcia-Gomez, R.; Pallares, C.; Sanchez, J.M.; 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, 239–246. [Google Scholar] [CrossRef]
  37. Shi, Y.K.; Wang, L.; Han, B.H.; Li, W.; Yu, P.; Liu, Y.P.; Ding, C.M.; Song, X.; Ma, Z.Y.; Ren, X.L.; et al. First-line icotinib versus cisplatin/pemetrexed plus pemetrexed maintenance therapy for patients with advanced EGFR mutation-positive lung adenocarcinoma (CONVINCE): A phase 3, open-label, randomized study. Ann. Oncol. 2017, 28, 2443–2450. [Google Scholar] [CrossRef] [PubMed]
  38. Inoue, A.; Kobayashi, K.; Maemondo, M.; Sugawara, S.; Oizumi, S.; Isobe, H.; Gemma, A.; Harada, M.; Yoshizawa, H.; Kinoshita, I.; et al. Updated overall survival results from a randomized phase III trial comparing gefitinib with carboplatin-paclitaxel for chemo-naive non-small cell lung cancer with sensitive EGFR gene mutations (NEJ002). Ann. Oncol. 2013, 24, 54–59. [Google Scholar] [CrossRef] [PubMed]
  39. Sequist, L.V.; Yang, J.C.-H.; Yamamoto, N.; O’Byrne, K.; Hirsh, V.; Mok, T.; Geater, S.L.; Orlov, S.; Tsai, C.-M.; Boyer, M.; 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, 3327–3334. [Google Scholar] [CrossRef] [PubMed]
  40. Wu, Y.-L.; Zhou, C.; Hu, C.-P.; Feng, J.; Lu, S.; Huang, Y.; Li, W.; Hou, M.; Shi, J.H.; Lee, K.Y.; et al. Afatinib versus cisplatin plus gemcitabine for first-line treatment of Asian patients with advanced non-small-cell lung cancer harbouring EGFR mutations (LUX-Lung 6): An open-label, randomised phase 3 trial. Lancet Oncol. 2014, 15, 213–222. [Google Scholar] [CrossRef]
  41. Park, K.; Tan, E.-H.; O’Byrne, K.; Zhang, L.; Boyer, M.; Mok, T.; Hirsh, V.; Yang, J.C.-H.; Lee, K.H.; Lu, S.; 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. 2016, 17, 577–589. [Google Scholar] [CrossRef]
  42. Paz-Ares, L.; Tan, E.H.; O’Byrne, K.; Zhang, L.; Hirsh, V.; Boyer, M.; Yang, J.C.; Mok, T.; Lee, K.H.; Lu, S.; et al. Afatinib versus gefitinib in patients with EGFR mutation-positive advanced non-small-cell lung cancer: Overall survival data from the phase IIb LUX-Lung 7 trial. Ann. Oncol. 2017, 28, 270–277. [Google Scholar] [CrossRef] [PubMed]
  43. Wu, Y.-L.; Cheng, Y.; Zhou, X.; Lee, K.H.; Nakagawa, K.; Niho, S.; Tsuji, F.; Linke, R.; Rosell, R.; Corral, J.; et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR -mutation-positive non-small-cell lung cancer (ARCHER 1050): A randomised, open-label, phase 3 trial. Lancet Oncol. 2017, 18, 1454–1466. [Google Scholar] [CrossRef]
  44. Mok, T.S.; Cheng, Y.; Zhou, X.; Lee, K.H.; Nakagawa, K.; Niho, S.; Lee, M.; Linke, R.; Rosell, R.; Corral, J.; et al. Improvement in overall survival in a randomized study that compared dacomitinib with gefitinib in patients with advanced non-small-cell lung cancer and EGFR activating mutations. J. Clin. Oncol. 2018, 36, 2244–2250. [Google Scholar] [CrossRef] [PubMed]
  45. Soria, J.-C.; Ohe, Y.; Vansteenkiste, J.; Reungwetwattana, T.; Chewaskulyong, B.; Lee, K.H.; Dechaphunkul, A.; Imamura, F.; Nogami, N.; Kurata, T.; et al. Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N. Engl. J. Med. 2018, 378, 113–125. [Google Scholar] [CrossRef] [PubMed]
  46. Mok, T.S.; Wu, Y.-L.; Ahn, M.-J.; Garassino, M.C.; Kim, H.R.; Ramalingam, S.S.; Shepherd, F.A.; He, Y.; Akamatsu, H.; Theelen, W.S.M.E.; et al. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N. Engl. J. Med. 2017, 376, 629–640. [Google Scholar] [CrossRef] [PubMed]
  47. Kim, S.T.; Uhm, J.E.; Lee, J.; Sun, J.-M.; Sohn, I.; Kim, S.W.; Jung, S.-H.; Park, Y.H.; Ahn, J.S.; Park, K.; et al. Randomized phase II study of gefitinib versus erlotinib in patients with advanced non-small cell lung cancer who failed previous chemotherapy. Lung Cancer 2012, 75, 82–88. [Google Scholar] [CrossRef] [PubMed]
  48. Yang, J.J.; Zhou, Q.; Yan, H.H.; Zhang, X.C.; Chen, H.J.; Tu, H.Y.; Wang, Z.; Xu, C.R.; Su, J.; Wang, B.C.; et al. A phase III randomised controlled trial of erlotinib vs gefitinib in advanced non-small cell lung cancer with EGFR mutations. Br. J. Cancer 2017, 116, 568–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Urata, Y.; Katakami, N.; Morita, S.; Kaji, R.; Yoshioka, H.; Seto, T.; Satouchi, M.; Iwamoto, Y.; Kanehara, M.; Fujimoto, D.; et al. Randomized phase III study comparing gefitinib with erlotinib in patients with previously treated advanced lung adenocarcinoma: WJOG 5108L. J. Clin. Oncol. 2016, 34, 3248–3257. [Google Scholar] [CrossRef] [PubMed]
  50. Hirsch, F.R.; Kabbinavar, F.; Eisen, T.; Martins, R.; Schnell, F.M.; Dziadziuszko, R.; Richardson, K.; Richardson, F.; Wacker, B.; Sternberg, D.W.; et al. A randomized, phase II, biomarker-selected study comparing erlotinib to erlotinib intercalated with chemotherapy in first-line therapy for advanced non-small-cell lung cancer. J. Clin. Oncol. 2011, 29, 3567–3573. [Google Scholar] [CrossRef] [PubMed]
  51. Kobayashi, Y.; Togashi, Y.; Yatabe, Y.; Mizuuchi, H.; Jangchul, P.; Kondo, C.; Shimoji, M.; Sato, K.; Suda, K.; Tomizawa, K.; et al. EGFR exon 18 mutations in lung cancer: Molecular predictors of augmented sensitivity to afatinib or neratinib as compared with first- or third-Generation TKIs. Clin. Cancer Res. 2015, 2015, 5305–5313. [Google Scholar] [CrossRef] [PubMed]
  52. Yang, J.C.-H.; Sequist, L.V.; Geater, S.L.; Tsai, C.-M.; Mok, T.S.K.; Schuler, M.; Yamamoto, N.; Yu, C.-J.; Ou, S.-H.I.; Zhou, C.; et al. Clinical activity of afatinib in patients with advanced non-small-cell lung cancer harbouring uncommon EGFR mutations: A combined post-hoc analysis of LUX-Lung 2, LUX-Lung 3, and LUX-Lung 6. Lancet Oncol. 2015, 16, 830–838. [Google Scholar] [CrossRef]
  53. Noronha, V.; Choughule, A.; Patil, V.M.; Joshi, A.; Kumar, R.; Philip, D.S.J.; Banavali, S.; Dutt, A.; Kumar, P. Epidermal growth factor receptor exon 20 mutation in lung cancer: Types, incidence, clinical features and impact on treatment. Onco Targets Ther. 2017, 10, 2903–2908. [Google Scholar] [CrossRef] [PubMed]
  54. Elamin, Y.; Robichaux, J.; Heymach, J. Preliminary results of a phase II study of poziotinib in EGFR exon 20 mutant advanced NSCLC. J. Thorac. Oncol. 2017, 12, S1536. [Google Scholar] [CrossRef]
  55. Goss, G.D.; O’Callaghan, C.; Lorimer, I.; Tsao, M.-S.; Masters, G.A.; Jett, J.; Edelman, M.J.; Lilenbaum, R.; Choy, H.; Khuri, F.; et al. Gefitinib versus placebo in completely resected non-small-cell lung cancer: Results of the NCIC CTG BR19 study. J. Clin. Oncol. 2013, 31, 3320–3326. [Google Scholar] [CrossRef] [PubMed]
  56. Kelly, K.; Altorki, N.K.; Eberhardt, W.E.E.; O’Brien, M.E.R.; Spigel, D.R.; Crino, L.; Tsai, C.-M.; Kim, J.-H.; Cho, E.K.; Hoffman, P.C.; et al. Adjuvant erlotinib versus placebo in patients with stage IB-IIIA non-small-cell lung cancer (RADIANT): A randomized, double-blind, phase III trial. J. Clin. Oncol. 2015, 33, 4007–4014. [Google Scholar] [CrossRef] [PubMed]
  57. Zhong, W.-Z.; Wang, Q.; Mao, W.-M.; Xu, S.-T.; Wu, L.; Shen, Y.; Liu, Y.-Y.; Chen, C.; Cheng, Y.; Xu, L.; et al. Gefitinib versus vinorelbine plus cisplatin as adjuvant treatment for stage II–IIIA (N1–N2) EGFR-mutant NSCLC (ADJUVANT/CTONG1104): A randomised, open-label, phase 3 study. Lancet Oncol. 2018, 19, 139–148. [Google Scholar] [CrossRef]
  58. Wu, Y.-L.; Herbst, R.S.; Mann, H.; Rukazenkov, Y.; Marotti, M.; Tsuboi, M. ADAURA: Phase III, double-blind, randomized study of osimertinib versus placebo in EGFR mutation-positive early stage NSCLC after complete surgical resection. Clin. Lung Cancer 2018, 19, e533–e536. [Google Scholar] [CrossRef] [PubMed]
  59. Sequist, L.V.; Waltman, B.A.; Dias-Santagata, D.; Digumarthy, S.; Turke, A.B.; Fidias, P.; Bergethon, K.; Shaw, A.T.; Gettinger, S.; Cosper, A.K.; et al. Genotypic and histological evolution of lung cancers acquring resistance to EGFR inhibitors. Sci. Transl. Med. 2011, 3, 75ra26. [Google Scholar] [CrossRef] [PubMed]
  60. Arcila, M.E.; Oxnard, G.R.; Nafa, K.; Riely, G.J.; Solomon, S.B.; Zakowski, M.F.; Kris, M.G.; Pao, W.; Miller, V.A.; Ladanyi, M. Rebiopsy of lung cancer patients with acquired resistance to EGFR inhibitors and enhanced detection of the T790M mutation using a locked nucleic acid-based assay. Clin. Cancer Res. 2011, 17, 1169–1180. [Google Scholar] [CrossRef] [PubMed]
  61. Yu, H.A.; Arcila, M.E.; Rekhtman, N.; Sima, C.S.; Zakowski, M.F.; Pao, W.; Kris, M.G.; Miller, V.A.; Ladanyi, M.; Riely, G.J. 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, 2240–2247. [Google Scholar] [CrossRef] [PubMed]
  62. Engelman, J.A.; Zejnullahu, K.; Mitsudomi, T.; Song, Y.; Hyland, C.; Park, J.O.; Lindeman, N.; Gale, C.-M.; Zhao, X.; Christensen, J.; et al. MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007, 316, 1039–1043. [Google Scholar] [CrossRef] [PubMed]
  63. Takezawa, K.; Pirazzoli, V.; Arcila, M.E.; Nebhan, C.A.; Song, X.; de Stanchina, E.; Ohashi, K.; Janjigian, Y.Y.; Spitzler, P.J.; Melnick, M.A.; et al. HER2 amplification: A potential mechanism of acquired resistance to EGFR inhibition in EGFR-mutant lung cancers that lack the second-site EGFRT790M mutation. Cancer Discov. 2012, 2, 922–933. [Google Scholar] [CrossRef] [PubMed]
  64. Zhang, Z.; Lee, J.C.; Lin, L.; Olivas, V.; Au, V.; LaFramboise, T.; Abdel-Rahman, M.; Wang, X.; Levine, A.D.; Rho, J.K.; et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat. Genet. 2012, 44, 852–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Soria, J.-C.; Wu, Y.-L.; Nakagawa, K.; Kim, S.-W.; Yang, J.-J.; Ahn, M.-J.; Wang, J.; Yang, J.C.-H.; Lu, Y.; Atagi, S.; 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. 2015, 16, 990–998. [Google Scholar] [CrossRef]
  66. Mok, T.S.K.; Kim, S.-W.; Wu, Y.-L.; Nakagawa, K.; Yang, J.-J.; Ahn, M.-J.; Wang, J.; Yang, J.C.-H.; Lu, Y.; Atagi, S.; et al. Gefitinib plus chemotherapy versus chemotherapy in epidermal growth factor receptor mutation-positive non-small-cell lung cancer resistant to first-line gefitinib (IMPRESS): Overall survival and biomarker analyses. J. Clin. Oncol. 2017, 35, 4027–4034. [Google Scholar] [CrossRef] [PubMed]
  67. Pao, W.; Miller, V.A.; Politi, K.A.; Riely, G.J.; Somwar, R.; Zakowski, M.F.; Kris, M.G.; Varmus, H. 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Song, S.; Jacobson, K.N.; McDermott, K.M.; Reddy, S.P.; Cress, A.E.; Tang, H.; Dudek, S.M.; Black, S.M.; Garcia, J.G.N.; Makino, A.; et al. ATP promotes cell survival via regulation of cytosolic [Ca2+] and Bcl-2/Bax ratio in lung cancer cells. Am. J. Physiol. Cell Physiol. 2016, 310, C99–C114. [Google Scholar] [CrossRef] [PubMed]
  69. Yun, C.-H.; Mengwasser, K.E.; Toms, A.V.; Woo, M.S.; Greulich, H.; Wong, K.-K.; Meyerson, M.; Eck, M.J. The T790M mutation in EGFR kinase causes drug resistance by increasing the affinity for ATP. Proc. Natl. Acad. Sci. USA 2008, 105, 2070–2075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Soda, M.; Choi, Y.L.; Enomoto, M.; Takada, S.; Yamashita, Y.; Ishikawa, S.; Fujiwara, S.-I.; Watanabe, H.; Kurashina, K.; Hatanaka, H.; et al. Identification of the transforming EML4–ALK fusion gene in non-small-cell lung cancer. Nature 2007, 448, 561–566. [Google Scholar] [CrossRef] [PubMed]
  71. Lin, J.J.; Zhu, V.W.; Yoda, S.; Yeap, B.Y.; Schrock, A.B.; Dagogo-Jack, I.; Jessop, N.A.; Jiang, G.Y.; Le, L.P.; Gowen, K.; et al. Impact of EML4-ALK variant on resistance mechanisms and clinical outcomes in ALK-positive lung cancer. J. Clin. Oncol. 2018, 36, 1199–1206. [Google Scholar] [CrossRef] [PubMed]
  72. Shaw, A.T.; Engelman, J.A. ALK in lung cancer: Past, present, and future. J. Clin. Oncol. 2013, 31, 1105–1111. [Google Scholar] [CrossRef] [PubMed]
  73. Kwak, E.L.; Camidge, D.R.; Clark, J.; Shapiro, G.I.; Maki, R.G.; Ratain, M.J.; Solomon, B.; Bang, Y.; Ou, S.; Salgia, R. Clinical activity observed in a phase I dose escalation trial of an oral c-met and ALK inhibitor, PF-02341066. J. Clin. Oncol. 2009, 27, 3509. [Google Scholar] [CrossRef]
  74. Solomon, B.J.; Mok, T.; Kim, D.-W.; Wu, Y.-L.; Nakagawa, K.; Mekhail, T.; Felip, E.; Cappuzzo, F.; Paolini, J.; Usari, T.; et al. First-line crizotinib versus chemotherapy in ALK-positive lung cancer. N. Engl. J. Med. 2014, 371, 2167–2177. [Google Scholar] [CrossRef] [PubMed]
  75. Soria, J.-C.; Tan, D.S.W.; Chiari, R.; Wu, Y.-L.; Paz-Ares, L.; Wolf, J.; Geater, S.L.; Orlov, S.; Cortinovis, D.; Yu, C.-J.; et al. First-line ceritinib versus platinum-based chemotherapy in advanced ALK -rearranged non-small-cell lung cancer (ASCEND-4): A randomised, open-label, phase 3 study. Lancet 2017, 389, 917–929. [Google Scholar] [CrossRef]
  76. Solomon, B.J.; Cappuzzo, F.; Felip, E.; Blackhall, F.H.; Costa, D.B.; Kim, D.-W.; Nakagawa, K.; Wu, Y.-L.; Mekhail, T.; Paolini, J.; et al. Intracranial efficacy of crizotinib versus chemotherapy in patients with advanced ALK-positive non-small-cell lung cancer: Results From PROFILE 1014. J. Clin. Oncol. 2016, 34, 2858–2865. [Google Scholar] [CrossRef] [PubMed]
  77. Peters, S.; Camidge, D.R.; Shaw, A.T.; Gadgeel, S.; Ahn, J.S.; Kim, D.-W.; Ou, S.-H.I.; Perol, M.; Dziadziuszko, R.; Rosell, R.; et al. Alectinib versus crizotinib in untreated ALK-positive non-small-cell lung cancer. N. Engl. J. Med. 2017, 377, 829–838. [Google Scholar] [CrossRef] [PubMed]
  78. Horn, L.; Infante, J.R.; Reckamp, K.L.; Blumenschein, G.R.; Leal, T.A.; Waqar, S.N.; Gitlitz, B.J.; Sanborn, R.E.; Whisenant, J.G.; Du, L.; et al. Ensartinib (X-396) in ALK-positive non-small cell lung cancer: Results from a first-in-human phase I/II, multicenter study. Clin. Cancer Res. 2018, 24, 2771–2779. [Google Scholar] [CrossRef] [PubMed]
  79. Shaw, A.T.; Kim, T.M.; Crinò, L.; Gridelli, C.; Kiura, K.; Liu, G.; Novello, S.; Bearz, A.; Gautschi, O.; Mok, T.; et al. Ceritinib versus chemotherapy in patients with ALK -rearranged non-small-cell lung cancer previously given chemotherapy and crizotinib (ASCEND-5): A randomised, controlled, open-label, phase 3 trial. Lancet Oncol. 2017, 18, 874–886. [Google Scholar] [CrossRef]
  80. Novello, S.; Mazieres, J.; Oh, I.-J.; Castro, J. d.; Migliorino, M.R.; Helland, A.; Dziadziuszko, R.; Griesinger, F.; Kotb, A.; Zeiter, A.; et al. Primary results from the phase III ALUR study of alectinib versus chemotherapy in previously treated ALK+ non-small-cell lung cancer (NSCLC). Ann. Oncol. 2017, 28 (Suppl_5). [Google Scholar] [CrossRef]
  81. Kim, D.-W.; Tiseo, M.; Ahn, M.-J.; Reckamp, K.L.; Hansen, K.H.; Kim, S.-W.; Huber, R.M.; West, H.L.; Groen, H.J.M.; Hochmair, M.J.; et al. Brigatinib in patients with crizotinib-refractory anaplastic lymphoma kinase-positive non-small cell lung cancer: A. randomized, multicenter phase II trial. J. Clin. Oncol. 2017, 35, 2490–2498. [Google Scholar] [CrossRef] [PubMed]
  82. Shaw, A.T.; Felip, E.; Bauer, T.M.; Besse, B.; Navarro, A.; Postel-Vinay, S.; Gainor, J.F.; Johnson, M.; Dietrich, J.; James, L.P.; et al. Lorlatinib in non-small-cell lung cancer with ALK or ROS1 rearrangement: An international, multicentre, open-label, single-arm first-in-man phase 1 trial. Lancet Oncol. 2017, 18, 1590–1599. [Google Scholar] [CrossRef]
  83. Drilon, A.; Siena, S.; Ou, S.-H.I.; Patel, M.; Ahn, M.J.; Lee, J.; Bauer, T.M.; Farago, A.F.; Wheler, J.J.; Liu, S.V.; et al. Safety and antitumor activity of the multitargeted pan-TRK, ROS1, and ALK inhibitor entrectinib: Combined results from two phase I trials (ALKA-372-001 and STARTRK-1). Cancer Discov. 2017, 7, 400–409. [Google Scholar] [CrossRef] [PubMed]
  84. Gainor, J.F.; Dardaei, L.; Yoda, S.; Friboulet, L.; Leshchiner, I.; Katayama, R.; Dagogo-Jack, I.; Gadgeel, S.; Schultz, K.; Singh, M.; et al. Molecular mechanisms of resistance to first- and second-generation ALK inhibitors in ALK-rearranged lung cancer. Cancer Discov. 2016, 6, 1118–1133. [Google Scholar] [CrossRef] [PubMed]
  85. Yoda, S.; Lin, J.J.; Lawrence, M.S.; Burke, B.J.; Friboulet, L.; Langenbucher, A.; Dardaei, L.; Prutisto-Chang, K.; Dagogo-Jack, I.; Timofeevski, S.; et al. Sequential ALK inhibitors can select for lorlatinb-resistance compound ALK mutations in ALK-positive lung cancer. Cancer Discov. 2018, 8, 714–729. [Google Scholar] [CrossRef] [PubMed]
  86. Hida, T.; Nokihara, H.; Kondo, M.; Kim, Y.H.; Azuma, K.; Seto, T.; Takiguchi, Y.; Nishio, M.; Yoshioka, H.; Imamura, F.; et al. Alectinib versus crizotinib in patients with ALK-positive non-small-cell lung cancer (J-ALEX): An open-label, randomised phase 3 trial. Lancet 2017, 390, 29–39. [Google Scholar] [CrossRef]
  87. Wu, Y.-L.; Yang, C.-H.; Kim, D.-W.; Lu, S.; Zhou, J.; Seto, T.; Yang, J.-J.; Yamamoto, N.; Ahn, M.-J.; Takahashi, T.; et al. Phase II study of crizotinib in East Asian patients with ROS1-positive advanced non-small cell lung cancer. J. Clin. Oncol. 2018, 36, 1405–1411. [Google Scholar] [CrossRef] [PubMed]
  88. Lim, S.M.; Kim, H.R.; Lee, J.-S.; Lee, K.H.; Lee, Y.-G.; Min, Y.J.; Cho, E.K.; Lee, S.S.; Kim, B.-S.; Choi, M.Y.; et al. Open-label, multicentre, phase II study of ceritinib in patients with non-small-cell lung cancer harboring ROS1 rearrangement. J. Clin. Oncol. 2017, 35, 2613–2618. [Google Scholar] [CrossRef] [PubMed]
  89. Kodama, T.; Tsukaguchi, T.; Satoh, Y.; Yoshida, M.; Watanabe, Y.; Kondoh, O.; Sakamoto, H. Alectinib shows potent antitumor activity against RET-rearranged non-small cell lung cancer. Mol. Cancer Ther. 2014, 13, 2910–2918. [Google Scholar] [CrossRef] [PubMed]
  90. Planchard, D.; Kim, T.M.; Mazieres, J.; Quoix, E.; Riely, G.; Barlesi, F.; Souquet, P.-J.; Smit, E.F.; Groen, H.J.M.; Kelly, R.J.; et al. Dabrafenib in patients with BRAF(V600E)-positive advanced non-small-cell lung cancer: A single-arm, multicentre, open-label, phase 2 trial. Lancet Oncol. 2016, 17, 642–650. [Google Scholar] [CrossRef]
  91. Planchard, D.; Smit, E.F.; Groen, H.J.M.; Mazieres, J.; Besse, B.; Helland, Å.; Giannone, V.; D’Amelio, A.M.; Zhang, P.; Mookerjee, B.; et al. Dabrafenib plus trametinib in patients with previously untreated BRAF V600E -mutant metastatic non-small-cell lung cancer: An open-label, phase 2 trial. Lancet Oncol. 2017, 18, 1307–1316. [Google Scholar] [CrossRef]
  92. Bos, J.L. Ras oncogenes in human cancer: A review. Cancer Res. 1989, 49, 4682–4689. [Google Scholar] [PubMed]
  93. Gainor, J.F.; Varghese, A.M.; Ou, S.-H.I.; Kabraji, S.; Awad, M.M.; Katayama, R.; Pawlak, A.; Mino-Kenudson, M.; Yeap, B.Y.; Riely, G.J.; et al. ALK rearrangements are mutually exclusive with mutations in EGFR or KRAS: An analysis of 1,683 patients with non-small cell lung cancer. Clin. Cancer Res. 2013, 19, 4273–4281. [Google Scholar] [CrossRef] [PubMed]
  94. Ahrendt, S.A.; Decker, P.A.; Alawi, E.A.; Zhu, Y.Y.R.; Sanchez-Cespedes, M.; Yang, S.C.; Haasler, G.B.; Kajdacsy-Balla, A.; Demeure, M.J.; Sidransky, D. Cigarette smoking is strongly associated with mutation of the K-ras gene in patients with primary adenocarcinoma of the lung. Cancer 2001, 92, 1525–1530. [Google Scholar] [CrossRef] [Green Version]
  95. Mascaux, C.; Iannino, N.; Martin, B.; Paesmans, M.; Berghmans, T.; Dusart, M.; Haller, A.; Lothaire, P.; Meert, A.-P.; Noel, S.; et al. The role of RAS oncogene in survival of patients with lung cancer: A systematic review of the literature with meta-analysis. Br. J. Cancer 2005, 92, 131–139. [Google Scholar] [CrossRef] [PubMed]
  96. Jänne, P.A.; van den Heuvel, M.M.; Barlesi, F.; Cobo, M.; Mazieres, J.; Crinò, L.; Orlov, S.; Blackhall, F.; Wolf, J.; Garrido, P.; et al. Selumetinib plus docetaxel compared with docetaxel alone and progression-free survival in patients with KRAS-mutant advanced non–small cell lung cancer. The SELECT-1 randomized clinical trial. JAMA 2017, 317, 1844–1853. [Google Scholar] [CrossRef] [PubMed]
  97. Blumenschein, G.R.J.; Smit, E.F.; Planchard, D.; Kim, D.-W.; Cadranel, J.; De Pas, T.; Dunphy, F.; Udud, K.; Ahn, M.-J.; Hanna, N.H.; et al. A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced non-small-cell lung cancer (NSCLC). Ann. Oncol. 2015, 26, 894–901. [Google Scholar] [CrossRef] [PubMed]
  98. Awad, M.M.; Oxnard, G.R.; Jackman, D.M.; Savukoski, D.O.; Hall, D.; Shivdasani, P.; Heng, J.C.; Dahlberg, S.E.; Janne, P.A.; Verma, S.; et al. MET exon 14 mutations in non–small-cell lung cancer are associated with advanced age and stage-dependent MET genomic amplification and c-Met overexpression. J. Clin. Oncol. 2016, 34, 721–730. [Google Scholar] [CrossRef] [PubMed]
  99. Awad, M.M.; Leonardi, G.C.; Kravets, S.; Dahlberg, S.E.; Drilon, A.E.; Noonan, S.; Camidge, D.R.; Ignatius Ou, S.-H.; Botelho, D.; Gadgeel, S.M.; et al. Impact of MET inhibitors on survival among patients (pts) with MET exon 14 mutant (METdel14) non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2017, 35, 8511. [Google Scholar] [CrossRef]
  100. Drilon, A.E.; Camidge, D.R.; Ignatius Ou, S.-H.; Clark, J.W.; Socinski, M.A.; Weiss, J.; Riely, G.J.; Winter, M.; Wang, S.C.; Monti, K.; et al. Efficacy and safety of crizotinib in patients (pts) with advanced MET exon 14-altered non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2016, 34, 108. [Google Scholar] [CrossRef]
  101. Camidge, D.R.; Ignatius Ou, S.-H.; Shapiro, G.I.; Otterson, G.A.; Villaruz, L.C.; Villalona-Calero, M.A.; Iafrate, A.J.; Varella-Garcia, M.; Dacic, S.; Cardarella, S.; et al. Efficacy and safety of crizotinib in patients with advanced c-MET-amplified non-small cell lung cancer (NSCLC). J. Clin. Oncol. 2014, 32, 8001. [Google Scholar] [CrossRef]
  102. Pan, Y.; Zhang, Y.; Li, Y.; Hu, H.; Wang, L.; Li, H.; Wang, R.; Ye, T.; Luo, X.; Zhang, Y.; et al. ALK, ROS1 and RET fusions in 1139 lung adenocarcinomas: A comprehensive study of common and fusion pattern-specific clinicopathologic, histologic and cytologic features. Lung Cancer 2014, 84, 121–126. [Google Scholar] [CrossRef] [PubMed]
  103. Lipson, D.; Capelletti, M.; Yelensky, R.; Otto, G.; Parker, A.; Jarosz, M.; Curran, J.A.; Balasubramanian, S.; Bloom, T.; Brennan, K.W.; et al. Identification of new ALK and RET gene fusions from colorectal and lung cancer biopsies. Nat. Med. 2012, 18, 382–384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Drilon, A.; Rekhtman, N.; Arcila, M.; Wang, L.; Ni, A.; Albano, M.; Van Voorthuysen, M.; Somwar, R.; Smith, R.S.; Montecalvo, J.; et al. Cabozantinib in patients with advanced RET-rearranged non-small-cell lung cancer: An open-label, single-centre, phase 2, single-arm trial. Lancet Oncol. 2016, 17, 1653–1660. [Google Scholar] [CrossRef]
  105. Lee, S.-H.; Lee, J.-K.; Ahn, M.-J.; Kim, D.-W.; Sun, J.-M.; Keam, B.; Kim, T.M.; Heo, D.S.; Ahn, J.S.; Choi, Y.-L.; et al. Vandetanib in pretreated patients with advanced non-small cell lung cancer-harboring RET rearrangement: A phase II clinical trial. Ann. Oncol. 2017, 28, 292–297. [Google Scholar] [CrossRef] [PubMed]
  106. Yoh, K.; Seto, T.; Satouchi, M.; Nishio, M.; Yamamoto, N.; Murakami, H.; Nogami, N.; Matsumoto, S.; Kohno, T.; Tsuta, K.; et al. Vandetanib in patients with previously treated RET-rearranged advanced non-small-cell lung cancer (LURET): An open-label, multicentre phase 2 trial. Lancet Respir. Med. 2016, 5, 42–50. [Google Scholar] [CrossRef]
  107. Velcheti, V.; Hida, T.; Reckamp, K.L.; Yang, J.C.; Nokihara, H.; Sachdev, P.; Feit, K.; Kubota, T.; Nakada, T.; Dutcus, C.E.; et al. Phase 2 study of lenvatinib (LN) in patients (Pts) with RET fusion-positive adenocarcinoma of the lung. Ann. Oncol. 2016, 27 (Suppl. 6), 1204PD. [Google Scholar] [CrossRef]
  108. Mazieres, J.; Peters, S.; Lepage, B.; Cortot, A.B.; Barlesi, F.; Beau-Faller, M.; Besse, B.; Blons, H.; Mansuet-Lupo, A.; Urban, T.; et al. Lung cancer that harbors an HER2 mutation: Epidemiologic characteristics and therapeutic perspectives. J. Clin. Oncol. 2013, 31, 1997–2003. [Google Scholar] [CrossRef] [PubMed]
  109. Arcila, M.E.; Chaft, J.E.; Nafa, K.; Roy-Chowdhuri, S.; Lau, C.; Zaidinski, M.; Paik, P.K.; Zakowski, M.F.; Kris, M.G.; Ladanyi, M. Prevalance, clinicopathologic associations and molecular spectrum of ERBB2 (HER2) tyrosine kinase mutations in lung adenocarcinoma. Clin. Cancer Res. 2012, 18, 4910–4918. [Google Scholar] [CrossRef] [PubMed]
  110. Shigematsu, H.; Takahashi, T.; Nomura, M.; Majmudar, K.; Suzuki, M.; Lee, H.; Wistuba, I.I.; Fong, K.M.; Toyooka, S.; Shimizu, N.; et al. Somatic mutations of the HER2 kinase domain in lung adenocarcinomas. Cancer Res. 2005, 65, 1642–1646. [Google Scholar] [CrossRef] [PubMed]
  111. Lai, W.-C.V.; Lebas, L.; Milla, J.; Barnes, T.A.; Gautschi, O.; Peters, S.; Ferrara, R.; Ni, A.; Sabari, J.K.; Clarke, S.J.; et al. Afatinib in patients with metastatic HER2-mutant lung cancers: An international multicenter study. J. Clin. Oncol. 2017, 35 (Suppl. 15), 9071. [Google Scholar] [CrossRef]
  112. De Greve, J.; Moran, T.; Graas, M.-P.; Galdermans, D.; Vuylsteke, P.; Canon, J.-L.; Chand, V.K.; Fu, Y.; Massey, D.; Vansteenkiste, J. Phase II study of afatinib, an irreversible ErbB family blocker, in demographically and genotypically defined non-small cell lung cancer (NSCLC) patients. J. Clin. Oncol. 2013, 31 (Suppl. 15), 8063. [Google Scholar] [CrossRef]
  113. Li, B.T.; Shen, R.; Buonocore, D.; Olah, Z.T.; Ni, A.; Ginsberg, M.S.; Ulaner, G.; Weber, W.; Ladanyi, M.; Won, H.H.; et al. Ado-trastuzumab emtansine in patients with HER2 mutant lung cancers: Results from a phase II basket trial. J. Clin. Oncol. 2017, 35, 8510. [Google Scholar] [CrossRef] [PubMed]
  114. Clamon, G.; Herndon, J.; Kern, J.; Govindan, R.; Garst, J.; Watson, D.; Green, M. Lack of trastuzumab activity in nonsmall cell lung carcinoma with overexpression of erb-B2: 39810: A phase II trial of Cancer and Leukemia Group, B. Cancer 2005, 103, 1670–1675. [Google Scholar] [CrossRef] [PubMed]
  115. Gatzemeier, U.; Groth, G.; Butts, C.; Van Zandwijk, N.; Shepherd, F.; Ardizzoni, A.; Barton, C.; Ghahramani, P.; Hirsh, V. Randomized phase II trial of gemcitabine-cisplatin with or without trastuzumab in HER2-positive non-small-cell lung cancer. Ann. Oncol. 2004, 15, 19–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Hotta, K.; Aoe, K.; Kozuki, T.; Ohashi, K.; Ninomiya, K.; Ichihara, E.; Kubo, T.; Ninomiya, T.; Chikamori, K.; Harada, D.; et al. A phase II study of trastuzumab emtansine in HER2-positive non–small cell lung cancer. J. Thorac. Oncol. 2018, 13, 273–279. [Google Scholar] [CrossRef] [PubMed]
  117. Herbst, R.S.; Davies, A.M.; Natale, R.B.; Dang, T.P.; Schiller, J.H.; Garland, L.L.; Miller, V.A.; Mendelson, D.; Van den Abbeele, A.D.; Melenevsky, Y.; et al. Efficacy and safety of single-agent pertuzumab, a human epidermal receptor dimerization inhibitor, in patients with non–small cell lung cancer. Clin. Cancer Res. 2007, 13, 6175–6181. [Google Scholar] [CrossRef] [PubMed]
Table 1. Tyrosine kinase inhibitors (TKIs) in non-small cell lung cancer. EGFR: epidermal growth factor receptor; ALK: anaplastic lymphoma kinase.
Table 1. Tyrosine kinase inhibitors (TKIs) in non-small cell lung cancer. EGFR: epidermal growth factor receptor; ALK: anaplastic lymphoma kinase.
First GenerationSecond GenerationThird Generation
EGFR TKIsGefinitib
Erlotinib
Icotinib
Afatinib
Dacomitinib
Neratinib
Osimertinib
Poziotinib
ALK TKIsCrizotinibCeritinib
Alectinib
Brigatinib
Lorlatinib
Entrectinib
Ensartinib
Table 2. Pivotal randomised controlled trials of Epidermal Growth Factor Receptor (EGFR) TKIs in patients with Stage IIIB/IV non-small cell lung cancer.
Table 2. Pivotal randomised controlled trials of Epidermal Growth Factor Receptor (EGFR) TKIs in patients with Stage IIIB/IV non-small cell lung cancer.
Author, Year
Trial Name
CountryPopulationIntervention
n
Control
n
Median Overall Survival (Months)
HR (95% CI)
Median Progression-Free Survival (Months)
HR (95% CI)
Response Rate
First line treatment
Mok, 2009 [30,31]
IPASS
East AsiaNon-smokers
Phase III
Gefitinib
609
Carboplatin and paclitaxel
608
18.6 vs. 17.3
HR 0.91 (0.76 to 1.10)
5.7 vs. 5.8
HR 0.75 (0.65 to 0.85)
43% vs. 32.2%
Subgroup: EGFR mutant
Subgroup: EGFR wildtype
HR 0.78 (0.50 to 1.20), NS
HR 1.38 (0.92 to 2.09), NS
HR 0.48 (0.36 to 0.64)
HR 2.85 (2.05 to 3.98)
71.2% vs. 47.3%
1.1% vs. 23.5%
Mitsudomi, 2010 [5,32]
WJTOG3405
JapanEGFR mutation +
Phase III
Gefitinib
86
Cisplatin and docetaxel
86
38.4 vs. 37.3
HR 1.25 (0.88 to 1.78)
9.2 vs. 6.3
HR 0.49 (0.34 to 0.71)
62.1% vs. 32.2%
Maemondo, 2010 [33,38]
NEJ002
JapanEGFR mutation +
Phase III
Gefitinib
115
Carboplatin & paclitaxel
115
27.7 vs. 26.6
HR 0.89 (0.63 to 1.24), NS
10.8 vs. 5.4
HR 0.32 (0.24 to 0.44)
73.7% vs. 30.7%
Zhou, 2011 [34,35]
OPTIMAL/CTONG-0802
ChinaEGFR mutation +
Phase III
Erlotinib
82
Carboplatin and gemcitabine
72
22.8 vs. 27.2
HR 1.19 (0.83 to 1.71), NS
13.1 vs. 4.6
HR 0.16 (0.10 to 0.26)
83% vs. 36%
Rosell, 2012 [36]
EURTAC
EuropeEGFR mutation +
Phase III
Erlotinib
86
Cisplatin and docetaxel or gemcitabine
87
19.3 vs. 19.5
HR 1.04 (0.65 to 1.68), NS
9.7 vs. 5.2
HR 0.37 (0.25 to 0.54)
58% vs. 15%
Wu, 2015 [6]
ENSURE
AsiaEGFR mutation +
Phase III
Erlotinib
110
Cisplatin and gemcitabine
107
26.3 vs. 25.5
HR 0.91 (0.63 to 1.31), NS
11.0 vs. 5.5
HR 0.34 (0.22 to 0.51)
62.7% vs. 33.6%
Sequist, 2013 [7,39]
LUX-Lung 3
InternationalEGFR mutation +
Phase III
Afatinib
230
Cisplatin and pemetrexed
115
28.2 vs. 28.2
HR 0.88 (0.66 to 1.17), NS
11.1 vs. 6.9
HR 0.58 (0.43 to 0.78)
56% vs. 23%
Wu, 2014 [7,40]
LUX-Lung 6
AsiaEGFR mutation +
Phase III
Afatinib
242
Cisplatin and gemcitabine
122
23.1 vs. 23.5
HR 0.93 (0.72–1.22), NS
11 vs. 5.6
HR 0.28 (0.20 to 0.39)
66.9% vs. 23%
Park, 2016 [41]
LUX-Lung 7 [42]
InternationalEGFR mutation +
Phase II
Afatinib
160
Gefitinib
159
27.9 vs. 24.5
HR 0.86 (0.66 to 1.12), NS
11.0 vs. 10.9
HR 0.73 (0.57 to 0.95)
70% vs. 56%
Wu, 2017
ARCHER 1050 [43,44]
AsiaEGFR mutation +
Phase III
Dacomitinib
227
Gefitinib
225
34.1 vs. 26.8
HR 0.76 (0.58 to 0.99)
14.7 vs. 9.2
HR 0.59 (0.47 to 0.74)
75% vs. 72%
Shi, 2017
CONVINCE [37]
ChinaEGFR mutation +
Phase III
Icotinib
148
Cisplatin and pemetrexed
137
Data immature11.2 vs. 7.9
HR 0.61 (0.43 to 0.87)
64.8% vs. 33.8%
Soria, 2018 [45]
FLAURA
InternationalEGFR mutation +
Phase III
Osimertinib
279
Gefitinib or erlotinib
277
Data immature18.9 vs. 10.2
HR 0.46 (0.37 to 0.57)
80% vs. 76% (NS)
Second line treatment
Mok, 2017 [46]
AURA3
InternationalT790M mutation +
PD after EGFR TKI
Phase III
Osimertinib
279
Platinum and pemetrexed
140
Data immature10.1 vs. 4.4
HR 0.30 (0.23 to 0.41)
71% vs. 31%
HR: hazard ratio; NS: not significant; PD: progressive disease. IPASS: Iressa Pan-Asia Study; WJTOG: West Japan Thoracic Oncology Group; NEJ: North East Japan Study Group; CTONG: Chinese Thoracic Oncology Group; EURTAC: European Randomised Trial of Tarceva vs Chemotherapy. All trials used consistent dosing of gefitinib 250 mg, erlotinib 150 mg, afatinib 40 mg daily, dacomitinib 45 mg daily, icotinib 125 mg three times a day, and osimertinib 80 mg daily.
Table 3. Pivotal randomised controlled trials of Anaplastic Lymphoma Kinase (ALK) TKIs in patients with Stage IIIB/IV non-small cell lung cancer.
Table 3. Pivotal randomised controlled trials of Anaplastic Lymphoma Kinase (ALK) TKIs in patients with Stage IIIB/IV non-small cell lung cancer.
Author, Year
Trial Name
CountryPopulationIntervention
n
Control
n
Median Overall Survival (Months)
HR (95% CI)
Median Progression Free Survival (Months)
HR (95% CI)
Response Rate
First line treatment
Solomon, 2014 [74]
PROFILE 1014
InternationalALK-positive
Phase III
Crizotinib
172
Platinum and pemetrexed
171
NR vs. NR
0.82 (0.54 to 1.26), NS
10.9 vs. 7.0
0.45 (0.35 to 0.60)
74% vs. 45%
Soria, 2017 [75]
ASCEND-4
InternationalALK-positive
Phase III
Ceritinib
189
Platinum and pemetrexed
187
NR vs. 26.2
0.73 (0.50 to 1.08), NS
16.6 vs. 8.1
0.55 (0.42 to 0.73)
72.5% vs. 26.7%
Peters, 2017 [77]
ALEX
InternationalALK-positive
Phase III
Alectinib 600 mg BD
152
Crizotinib
151
NR vs. NR
HR 0.76 (0.48 to 1.20), NS
NR vs. 11.1
0.47 (0.34 to 0.65)
82.9% vs. 75.5%
Second line treatment
Shaw, 2013 [8]
PROFILE 1007
InternationalALK-positive
PD after chemotherapy
Phase III
Crizotinib
173
Pemetrexed or docetaxel
174
Data immature
20.3 vs. 22.8
1.02 (0.68 to 1.54), NS
7.7 vs. 3.0
0.49 (0.37 to 0.64)
65% vs. 20%
Kim, 2017 [81]InternationalALK-positive
PD after crizotinib
Phase II
Brigatinib 90 mg daily
112
Brigatinib 180 mg daily
110
Not reported9.2 vs. 15.6
0.55 (0.35 to 0.86)
45% vs. 54%
Hida, 2017 [86]
J-ALEX
JapanALK-positive
First line or PD after chemotherapy
Phase III
Alectinib 300 mg BD
103
Crizotinib
104
Data immatureNR vs. 10.2
HR 0.34 (0.17 to 0.71)
92% vs. 79%
Shaw, 2017 [79]
ASCEND-5
InternationalALK-positive
PD after crizotinib
Phase III
Ceritinib
115
Pemetrexed or docetaxel
116
Data immature
1.0 (0.67 to 1.49), NS
5.4 vs. 1.6
HR 0.49 (0.36 to 0.67)
39% vs. 7%
Novello, 2017 [80]
ALUR
InternationalALK-positive
PD after crizotinib
Phase III
Alectinib 600 mg BDPemetrexed or docetaxelNot reported9.6 vs. 1.4
0.15 (0.08 to 0.29)
36.1% vs. 11.4%
HR: hazard ratio; NS: not significant; PD: progressive disease. All trials with crizotinib and ceritinib used consistent dosing at 250 mg BD and 750 mg daily, respectively.
Table 4. Spectrum of activity of different Anaplastic Lymphoma Kinase (ALK) tyrosine kinase inhibitors for different resistance mutations, as studied by Gainor et al. using in vitro data on cell lines [84].
Table 4. Spectrum of activity of different Anaplastic Lymphoma Kinase (ALK) tyrosine kinase inhibitors for different resistance mutations, as studied by Gainor et al. using in vitro data on cell lines [84].
ALK MutationCrizotinibCeritinibAlectinibBrigatinibLorlatinib
V1SSSSS
C1156YISSSS
I1171NISRSS
I1171SISISS
I1171TISSSS
F1174CISSSS
L1196MRSISS
L1198FSISSS
G1202RRIRIS
G1202delIIIIS
D1203NISSSS
E1210KSSSSS
G1269AISSNo dataS
D1203N + F1174CRRIII
D1203N + E1210KIIIIS
* R: resistant; S: sensitive; I: intermediate.

Share and Cite

MDPI and ACS Style

Bui, K.T.; Cooper, W.A.; Kao, S.; Boyer, M. Targeted Molecular Treatments in Non-Small Cell Lung Cancer: A Clinical Guide for Oncologists. J. Clin. Med. 2018, 7, 192. https://doi.org/10.3390/jcm7080192

AMA Style

Bui KT, Cooper WA, Kao S, Boyer M. Targeted Molecular Treatments in Non-Small Cell Lung Cancer: A Clinical Guide for Oncologists. Journal of Clinical Medicine. 2018; 7(8):192. https://doi.org/10.3390/jcm7080192

Chicago/Turabian Style

Bui, Kim Tam, Wendy A. Cooper, Steven Kao, and Michael Boyer. 2018. "Targeted Molecular Treatments in Non-Small Cell Lung Cancer: A Clinical Guide for Oncologists" Journal of Clinical Medicine 7, no. 8: 192. https://doi.org/10.3390/jcm7080192

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop