manuscript


doi: 10.5599/admet.5.1.352 1 

ADMET & DMPK 5(1) (2017) 1-8; doi: 10.5599/admet.5.1.352 

 
Open Access : ISSN : 1848-7718  

http://www.pub.iapchem.org/ojs/index.php/admet/index   

Mini Review 

Tyrosine kinase inhibitors: new molecules in non-small cell lung 
cancer (EGFR AND ALK) 

Fernando Franco, Míriam Méndez*, Lourdes Gutierrez, Mariano Provencio  

Puerta de Hierro Universitary Hospital, Madrid, Spain 

*Corresponding Author:  E-mail: mirimega@hotmail.com.  

Received: October 25, 2016; Revised: February 22, 2017; Published: March 25, 2017  

 

Abstract 

Lung cancer is a tumor pathology that includes a group of neoplasms with different biological characteristics, 
which determine the course of the disease and the potential treatment response depending on the subtype 
diagnosed. Non-small lung carcinoma corresponds to 85 % of all lung cancers. Adenocarcinoma is the most 
common histologic subtype of this group. In the United States approximately 130,000 new cases a year are 
diagnosed and in the world around a million. Adenocarcinoma is the most likely subtype, with possible 
molecular alterations (EGFR mutations and translocations in ALK), which are a therapeutic target. Knowledge of 
the molecular profile of the tumor and its biology has led us to identify therapeutic targets as protein tyrosine 
kinase, whose inhibition have demonstrated impact in terms of survival and quality of life for patients with 
these molecular alterations. However, the complex biology of non-small cell lung carcinoma gives the malignant 
cell the ability to generate mechanisms of resistance to these drugs and has forced us to extend the studies to 
control the disease by blocking these mechanisms. 

Keywords 

Lung cancer; EGFR inhibitors, ALK inhibitors 

 

Introduction 

Non-small lung carcinoma corresponds to 85 % of lung cancers. Adenocarcinoma is the most common 

histologic subtype. In the United States approximately 130,000 new cases are reported per year [1]. 

Patients with advanced non-small cell lung cancer (NSCLC) usually receive systemic therapy. After 

histopathological diagnosis, an extensive molecular study is required to characterize the tumor.  Regardless 

of the clinical characteristics of the patient, at least the mutational status of the epidermal growth factor 

receptor (EGFR) and ALK should be analyzed [2-4]. Activating mutations in EGFR tyrosine kinase are 

observed in approximately 15 percent of NSCLC adenocarcinoma in the United States and occur more 

frequently in nonsmokers [5]. In Asian populations, the incidence of these EGFR mutations is substantially 

higher, up to 62 percent [6].  

In unselected NSCLC populations, the ALK rearrangement is a relatively rare event. The overall incidence 

of ALK gene rearrangements has been about 4 percent. The presence of ALK gene rearrangements in 

NSCLC tumors tends to occur independently of EGFR mutations. Similar frequencies of ALK gene 

rearrangements have been reported in Asian and Western populations. 

http://www.pub.iapchem.org/ojs/index.php/admet/index


M. Mendez et al.  ADMET & DMPK 5(1) (2017) 1-8 

2  

EGFR inhibitors 

First and second generation of EGFR inhibitors 

Patients with molecular alterations constitute a subpopulation with clinical characteristics, prognosis 

and response to different treatment (chemotherapy vs tyrosine kinase inhibitors) [5,7-9]. First-generation 

EGFR TKIs (including gefitinib and erlotinib) selectively target the receptor, through a competitive, 

reversible binding at the tyrosine kinase domain. However, despite good response there is evidence that 

after a variable period of time (5-13 months), the patients develop acquired resistance to EGFR-TKI therapy 

that negatively impacts their survival. Second generation TKIs (Afatinib, Dacomitinib) differ from first 

generation EGFR-TKIs because they form an irreversible link to the EGFR kinase domain and inhibit 

receptors as HER2 and HER4 and are active agents particularly in those with exon 19 deletion. The 

increasing knowledge in tumor biology and the mechanisms, by which resistance develops, have helped to 

develop new molecules that have already demonstrated a positive impact on response rates and survival 

[9-11]. 

Third generation of EGFR inhibitors 

The T790M EGFR mutation is present in over 60 % of cases of resistance to TKIs first and second 

generation. It is the resistance mechanism of EGFR most commonly known. This acquired resistance is 

mediated by a threonine-to-methionine mutation in EGFR at position 790 (EGFR T790M), activation of a 

bypass-signalling pathway such as mesenchymal-epithelial transition (MET) pathway, human epidermal 

growth factor 2 (HER2), or mitogen-activated protein kinase 1 (MAPK1) amplification, and histologic 

transformation to small-cell lung cancer or an epithelial-to-mesenchymal transition [12-15]. Following the 

acquisition of the mutation, the median survival is less than two years, so there is a need to develop new 

drugs that will control tumor growth in patients positive EGFR T790M. 

Osimertinib (AZD9291) 

AZD9291 approved by the FDA in November 2015, is a new molecule TKI third generation, which binds 

irreversibly to the residue kinase EGFR at the site of ATP binding by covalent bond formation, in patients 

who have acquired T790M mutation. After oral administration, it is metabolized AZD9291 that has two 

pharmacologically active forms AZ5104 and AZ7550. AZ7550 showed a comparable potency and selectivity 

profile similar to osimertinib, whereas AZ5104 in addition exhibited greater potency against exon 19 

deletion and T790M mutants and wild-type EGFR. The first phase I dose escalation study was the AURA, 

made with EGFR mutated patients in progression with gefitinib or erlotinib treatment [11, 16-18]. The 

doses used were between 20 and 240 mg per day. In the expansion phase, the analysis of the T790M 

mutation patients demonstrated an objective response rate of 67 % (95 % CI 52-70 %) compared to 21 % in 

patients without the mutation. The rate of disease control in the overall analysis of all AURA studies was 

91 %. The median progression-free survival was 9.6 months versus 3 months in patients without the 

mutation. The profile of side events is favourable, since they are few and mostly grades 1 and 2. The most 

frequent were rash, diarrhea and nausea. Since no significant difference in response to higher doses was 

observed, 80 mg/day is recommended until disease progression or unacceptable toxicity [16-18]. If it is 

necessary to reduce the dose, this should be done at 40 mg per day, since tablets are 40 and 80 mg. Its 

elimination is mainly hepatic and has not been studied in patients with renal insufficiency [16]. Current 

studies of liquid biopsy are presented as an option for detection of mutations in peripheral blood. This 

probably serves not only for diagnosis but also for monitoring; however, more studies are needed in this 

line. 



ADMET & DMPK 5(1) (2017) 1-8 TKIs in non-small cell lung cancer 

doi: 10.5599/admet.5.1.352 3 

Rociletinib (CO-16086) 

Rociletinib, is a new oral selective covalent molecule EGFR inhibitor, with demonstrated activity against 

the T790M mutation, deletionn 19 and L858R [19-20]. Initial studies were performed in patients with 

acquired resistance to gefitinib and erlotinib. The subsequent inclusion of patients with mutation T790M 

has shown the particular activity of the drug in this subgroup of patients. Partial results of studies of a 

phase II study presented at ASCO 2015 reflect an objective response rate in this subgroup of 59 % (95 % CI 

45-73) and rates of disease control 85 %. However, new combined analyses of the TIGER-X and TIGER-2 

studies have shown that the actual response rate is only around 30 %. Among the most common adverse 

events are diarrhea and skin toxicity, most of them in grades 1 and 2 [21]. Cases of hyperglycemia and QT 

prolongation (> 10 %) were frequently presented, in the latter case without reaching documented 

arrhythmias. Dose reductions occurred in 51 %  of patients, most commonly due to hyperglycemia (22 %) 

and QTc prolongation (11 %). Fifty-seven percent of patients had dose interruptions, which were most 

often due to hyperglycemia (22 %), QTc prolongation (10 %), and nausea (10 %). The average survival in 

patients was 8 months with the T790M mutation in patients where the 500-650 mg dose was 

administered. Although initially seeming a promising drug, the adverse event profile and response rates 

have prompted the FDA to give a negative opinion for accelerated approval of the drug [21-22]. 

HM61713 (BI 1482694) 

HM61713 is an irreversible kinase inhibitor that binds to a cysteine residue near the EGFR kinase 

domain. Preliminary studies in cell lines, has shown activity against T790M mutations and L858R. Studies 

are currently underway with Phase I / II with a dose of 800 mg / day in patients with T790M mutation. 

Initial results show an overall response rate of 62 % in this subgroup of patients. Disease control rate was 

91 % by independent assessment. The most common treatment-related adverse events included diarrhoea 

(55 %), nausea (37 %), rash (38 %) and pruritus (36 %), the majority were mild-to-moderate [23]. It seems a 

promising agent; however further studies to confirm therapeutic activity are needed. ELUXA 1 is an 

ongoing pivotal Phase II clinical trial, designed to further investigate the efficacy and safety of BI 1482694 

in patients with NSCLC whose tumours acquired T790M-mediated resistance after first-line EGFR TKIs. 

ASP8273  

ASP8273 is another small molecule, irreversible EGFR TK inhibitor targeting EGFR mutations including 

T790M. In mouse xenograft models, ASP8273 achieved complete remission of tumors after 14 days of 

treatment. Phase I / II studies have established 400 mg / day as the optimal dose. The most common 

adverse events were gastrointestinal toxicity and thrombocytopenia. Dose-limiting toxicities were reported 

at levels higher doses (400-600 mg). Maximum tolerated dose was established as 300 mg. The objective 

response rate in patients with T790M was 25 % [24-25]. 

PF-06747775 and EGF816 

They are other new small molecules that inhibit T790M; however, they are in early stages of study and 

results are expected to verify their antitumor activity in the clinic [26-30]. 

Similarly, there are clinical phase I-II trials to evaluate the effectiveness of combinations of third 

generation TKIs with various molecules, including MET and MEK inhibitors, as well as immunotherapy, 

among others. 

 



M. Mendez et al.  ADMET & DMPK 5(1) (2017) 1-8 

4  

ALK inhibitors 

Anaplastic lymphoma kinase (ALK) is a member of the insulin receptor tyrosine kinase family (RTK). The 

echinoderm microtubule-associated protein-like 4-anaplastic lymphoma kinase (EML4-ALK) fusion 

oncogene arises from an inversion on the short arm of chromosome 2 that joins EML4 to ALK. The resulting 

chimeric protein, EML4-ALK, contains an N-terminus derived from EML4 and a C-terminus containing the 

entire intracellular tyrosine kinase domain of ALK. ALK rearrangements occur in 3–7 % of patients with 

NSCLC and are more common among patients with a non / light smoking history, adenocarcinoma 

histology, a younger age, female gender and in tumours wild type for EGFR and KRAS [31].  

Anaplastic lymphoma kinase (ALK) gene rearrangements may be detected using fluorescence in situ 

hybridization (FISH) or immunohistochemistry (IHC).  The gold standard assay for diagnosing is FISH. To be 

declared ALK positive, at least 15 % of 50 evaluated tumor cells must harbour split signals. IHC may be a 

more practical approach to detecting ALK-positive NSCLC. Moreover, the absence of ALK expression in 

normal lung tissue makes IHC less cumbersome to interpret than FISH. In 2015, an IHC test using the D5F3 

ALK antibody (Ventana) was approved after demonstration of concordance with the break apart FISH test, 

with 94 % overall agreement [32]. 

First generation of ALK inhibitors 

Crizotinib (PF-02341066, Xalkori) 

Crizotinib is the first-generation ALK inhibitor approved for ALK-positive NSCLC (4). It also has activity 

against c-MET and ROS1 tyrosine kinases. It was approved in first line and in pretreated ALK positive NSCLC 

patients [33-34]. Response rates to crizotinib are approximately 60 %, which means there are a 40 % of 

primary resistances. One reason is because some ALK translocations may not generate functional 

rearrangements in all patients [35]. There are secondary causes of resistance thought ALK kinase mutations 

and it may occur in 30 % of the cases: L1196M, F1174L, and G1202R or C1156Y mutation [36] Activation of 

other oncogenes resulting in bypass signaling cascades such as EGFR, cKIT and KRAS or copy number gains 

of the ALK fusion gene [37] and pharmacokinetic failings of the drug itself due to inadequate systemic 

levels or poor CNS penetration and about 50 % of patients develop CNS metastases during treatment with 

crizotinib [38-39] and in many cases, no definitive resistance mutation can be identified [40]. 

Second generation of ALK inhibitors 

These drugs were developed to overcome crizotinib-resistant mutations and to improve activity in CNS 

disease (Table 1). 

Ceritinib 

Ceritinib is 20-fold more potent than crizotinib in terms of ALK selectivity [41]. It strongly inhibits 

resistance mutations, and especially L1196M, G1269A, I1171T and S1206Y mutations. However, ceritinib 

was not effective against G1202R and F1174C crizotinib-resistant mutations. The updated ASCEND-1 report 

on the evaluation of the efficacy of ceritinib at a daily dose of 750 mg was recently presented. A total of 

246 patients with ALK+ NSCLC were enrolled, including 163 pretreated with an ALK inhibitor (crizotinib or 

alectinib) and 83 ALK inhibitor naïve patients. The overall rate responses (ORRs) were 56.4 %, 72.3 % and 

61.8 % and the median of progression free survival (PFS) was 18.4, 9.0 and 6.9 months in the group 

pretreated with an ALK inhibitor, ALK inhibitor-naive patients and the overall population, respectively. 124 

patients had measurable brain metastases at baseline: 28 patients pretreated with an ALK inhibitor and 8 



ADMET & DMPK 5(1) (2017) 1-8 TKIs in non-small cell lung cancer 

doi: 10.5599/admet.5.1.352 5 

patients in the ALK inhibitor naive group. The intracranial ORRs were 36 % (95 % CI 19–56 %) in patients 

pretreated with an ALK inhibitor and 63 % (95 % CI 25–92 %) in the ALK inhibitor-naive group [33,34]. The 

ASCEND-3 trial enrolled 124 patients, 40.3 % with brain metastases, 46 % of whom had no prior BRT (Brain 

Radiotherapy). The ORR was 63.7 % (95 % CI54.6–72.2 %). The intracranial ORR was 20 % (95 %CI 2.5–

55.6 %) [35]. Two randomized phase III trials are currently open to evaluate ceritinib versus first-line 

chemotherapy (ASCEND-4) and second-line chemotherapy (ASCEND-5). 

Alectinib 

Alectinib is a highly selective ALK inhibitor, in preclinical studies it showed activity against crizotinib-

resistant ALK mutations (L1196M, C1156Y and F1174L) and RET but not against InsR, IGF-R1 and ROS1. 

Japan was the first country to approve alectinib in patients with advanced ALK-rearranged NSCLC, based on 

the results of the phase I/II AF-001JP trial targeting patients with ALK+ NSCLC who had not previously been 

treated with crizotinib or other ALK inhibitors [42,43]. In the phase II trial, 46 patients were treated and 43 

of them (93.5 %) achieved an ORR (95 % CI 82.1–98.6). The 2-year PFS rate was 76 % (95 % CI 60–86 %) and 

the 2-year OS was 79 % (95 % CI 63–89 %). A phase I/II trial with 47 ALK+ NSCLC patients who progressed 

on or were intolerant to crizotinib. The ORR was 55 % and the subset analysis of 21 patients with CNS 

metastases at baseline showed a disease control rate approximating 90 %. The recommended dose for 

phase II studies was 600 mg BID [44-46]. 

Brigatinib 

Brigatinib is a novel potent oral drug that has demonstrated ability to overcome crizotinib resistance 

mutations, including G1202R and activity against ROS1 and inhibits mutant EGFR, including T790M [47-49]. 

Brigatinib showed antitumor activity in patients treated with crizotinib with an objective response rate of 

71 % and  in crizotinib-naive patients of 100 %. The median PFS was 13.4 months in the subgroup with prior 

crizotinib therapy [50]. A pivotal, randomized, phase 2 trial of brigatinib in patients with crizotinib-resistant 

ALK+ NSCLC has completed accrual. 

Entrectinib 

Entrectinib is a novel, orally available, selective tyrosine kinase inhibitor of the Trk family of tyrosine 

kinases, ROS1 and ALK. Rearrangements in NTRK1 were recently described in approximately 3 % of NSCLC 

that were negative for other oncogenic driver mutations [51]. Entrectinib has demonstrated in vitro and in 

vivo antitumour activity against various TRK, ROS1 or ALK-driven xenograft models of different human 

cancers (NPM-ALK-driven lymphoma and EML4-ALK-driven NSCLC) Entrectinib has adequate oral 

bioavailability and can efficiently cross the blood–brain barrier in mice with intracranially injected 

NCIH2228 EML4-ALK cells [52]. Entrectinib 400 mg/m2 is the dose recommended for the trials phase II 

[53]. 

Third generation of ALK inhibitors 

Lorlatinib (PF-06463922) 

Lorlatinib has been identified as an orally novel reversible available ATP-competitive selective targeting 

of a specific residue leucine at position 1198 (L1198) detected in only approximately 25 % of kinases, brain 

penetrant inhibitor of ALK (including all known mutations) and the c-Ros oncogene 1 (ROS1). In vitro and 

preclinical studies it is ∼10-fold more potent against wildtype EML4-ALK and ∼40-fold more potent against 

EML4-ALK L1196M compared with crizotinib [54]. Importantly, lorlatinib has demonstrated activity in 

patients whose tumors harbor the highly resistant mutation ALK G1202R. This mutation confers resistance 



M. Mendez et al.  ADMET & DMPK 5(1) (2017) 1-8 

6  

to other next-generation ALK inhibitors. 

Other drugs for crizotinib resistant tumor 

ASP3026 

This is an ALK inhibitor active against crizotinib-resistant ALK-positive advanced tumors [55]. 

X-376 and X-396 (Xcovery) 

These are more potent inhibitors of ALK but less potent inhibitors of MET compared to crizotinib.  

Moreover, X-396 could potently inhibit ALK with two point mutations (L1196M and C1156Y) associated 

with acquired resistance to crizotinib and there is preclinical data about CNS penetration [56]. 

CEP-28122 

CEP-29122 is a potent and selective ALK inhibitor which showed selectivity in preclinical studies against 

ALK compared to various types of tyrosine kinases, including InsR, IGF-R1 and c-MET. CEP- 37440 is an ALK 

inhibitor and focal adhesion kinase (FAK) [57-58]. 

HSP90 inhibitors 

Another possibility to overcome resistance to ALK inhibitors includes the combination with HSP90 

inhibitors such as retaspimycin hydrochloride (IPI-504) and ganetespib (STA-9090) that have demonstrated 

clinical activity in ALK+ NSCLC patients [60-62]. Several clinical trials are evaluating HSP90 inhibitors in 

combination with ALK inhibitors. Crizotinib is combined with ganetespib in crizotinib-naive patients and in 

crizotinib-resistant disease. In other studies crizotinib is combined with onalespib (AT13387), while ceritinib 

is combined with AUY922. 

Table 1. Main molecular characteristics of second and third-generation ALK inhibitors. 

Drug 
Targets other 

than ALK 

L1196M 
resistance 
mutation 

C1156Y 
resistance 
mutation 

G1202R 
resistance 
mutation 

Ceritinib IGF-R1, InsR, 
ROS1 

Sens. Res. Res. 

Alectinib LTK, GAK Sens. Sens. Res. 

Brigatinib ROS1, EGFR Sens. Sens. Sens. 

Entrectinib TrkA, TrkB, 
TrkC, ROS1 

Sens. Sens. ND 

Lorlatinib ROS1 Sens. ND Sens. 

TSR-011 TrkA, TrkB, 
TrkC 

Sens. ND ND 

ASP3026 ROS1, ACK Sens. ND ND 

X-396 MET Sens. Sens. ND 

Conclusions  

Current knowledge of the biology of NSCLC, allows the development of new molecules that are able to 

control the disease and have an impact on disease control, survival and quality of life. The new third-

generation EGFR TKIs demonstrate efficacy in patients with EGFR resistance mutations patients while the 

new ALK inhibitors show activity in patients with ALK rearrangements and mutations. These benefits alone 

have already allowed the registration of some of them. Results from other studies are pending, as well as 

those related to combinations with other drugs. 

 



ADMET & DMPK 5(1) (2017) 1-8 TKIs in non-small cell lung cancer 

doi: 10.5599/admet.5.1.352 7 

References 

[1] R. Siegel, J. Ma, Z. Zou, A. Jemal. CA Cancer J. Clin. 64(1) (2014) 9-29. 

[2] N. Lindeman, P. Cagle, M.B. Beasley et al. Arch. Pathol. Lab. Med. 137(6) (2013) 828–860. 

[3] T.J. Lynch, D.W. Bell, R. Sordella, et al. N. Engl. J. Med. 350(21) (2004) 2129-2139. 

[4] E.L. Kwak, Y.J. Bang, D.R. Camidge, et al. N. Engl. J. Med. 363(18) (2010) 1693-1703. 

[5] R. Rosell, E. Carcereny, R. Gervais, et al. Lancet Oncol. 13(3) (2012) 239–246. 

[6] F.-Y. Niu, Y.-L.Wu. Exp. Hematol. Oncol. 3(1) (2014) 2. 

[7] N.B. Leighl, N. Rekhtman, W.A. Biermann, et al. J. Clin. Oncol. 32(32) (2014) 3673. 

[8] A. Chi, S. Remick, W. Tse. Biomark. Res. 1(1) (2013) 2. 

[9] C.K. Lee, Y.-L. Wu, P.N. Ding, et al.  J. Clin. Oncol. 33(17) (2015) 1958–1965. 

[10] A.O. Walter, R.T. Sjin, H.J. Haringsma, et al. Cancer Discov. 3(12) (2013) 1404–1415. 

[11] D.A. Cross, S.E. Ashton, S. Ghiorghiu, et al. Cancer Discov. 4(9) (2014) 1046–1061. 

[12] S. Kobayashi, T.J. Boggon, T. Dayaram, et al. N. Engl. J. Med. 352 (2005) 786–792. 

[13] J.A. Engelman, K. Zejnullahu, T. Mitsudomi, et al. Science 316 (2007) 1039–1043. 

[14] K. Takezawa, V. Pirazzoli, M.E. Arcila, et al. Cancer Discov. 2 (2012) 922–933. 

[15] T.M. Kim, A. Song, D.-W. Kim, et al. J. Thorac. Oncol. 10 (2015) 1736–1744. 

[16] P.A. Jänne, J.C. Yang, D.W. Kim, et al. N. Engl. J. Med. 372(18) (2015) 1689. 

[17] S.S. Ramalingam, J. Yang, C. Lee, et al. J. Thoracic. Oncol. 10(9_suppl 2) (2015) S319. 

[18] C.A. Eberlein, D. Stetson, A.A. Markovets, et al. Cancer Res. 75(12) (2015) 2489–500. 

[19] A.O. Walter, R.T. Sjin, H.J. Haringsma, et al. Cancer Discov. 3 (2013) 1404-1415. 

[20] L.V. Sequist, J.C. Soria, J.W. Goldman, et al. N. Engl. J. Med. 372(18) (2015) 1700–1709. 

[21] J.C. Soria, D.R. Camidge. N. Engl. J. Med. 374 (2016) 2296-2297 

[22] FDA Briefing Document Oncologic Drugs Advisory Committee Meeting. NDA 208542 Rociletinib. April 
12, 2016  

[23] K.-O. Lee, M.Y. Cha, M. Kim, et al. Cancer Res. 74(19) (2014) LB-100. 

[24] H. Sakagami, S. Konagai, H. Yamamoto, et al. Cancer Res. 74(19 Supplement) (2014) 1728. 

[25] Y. Jia, J. Juarez, M. Manuia, G. Lelais, et al. Cancer Res. 74(19 Supplement) (2014) 1734. 

[26] S. Kasibhatla, J. Li, C. Tompkins, et al. Cancer Res. 74(19 Supplement) (2014) 1733. 

[27] Y. Goto, H. Nokihara, H. Murakami, et al. ASCO Meet Abstr. 33(15_suppl) (2015) 8014. 

[28] G. Lelais, R. Epple, P.-Y. Michellys, et al. Cancer Res. 75(15 Supplement) (2015) 2585. 

[29] Study For Patients with NSCLC EGFR Mutations (Del 19 or L858R +/- T790M). ClinicalTrials.gov 
Identifier: NCT02349633. 

[30] A. Ullrich, J. Schlessinger. Cell 61(2) (1990) 203–212.  

[31] D. Camidge,  S. Kono, A. Flacco, et al. Clin. Cancer Res. 16(22) (2010) 5581–5590. 

[32] J.J. Cui, M. Tran-Dubé, H. Shen, et al. J. Med. Chem. 54(18) (2011) 6342–6363. 

[33] A.T. Shaw, S.H. Ou, Y.J. Bang, et al. N. Engl. J. Med. 371(21) (2014) 1963–1971. 

[34] B.J. Solomon, T. Mok, D.W. Kim, et al. N. Engl. J. Med. 371(23) (2014) 2167–2177. 

[35] E. Felip, S. Orlov, K. Park, et al. J. Clin. Oncol. 33 (2015) 8060. 

[36] J.M. Heuckmann, H. Balke-Want, F. Malchers, et al. Clin. Cancer Res. 18(17) (2012) 4682–4690. 

[37] Y.L. Choi, M. Soda, Y. Yamashita, et al. N. Engl. J. Med. 363(18) (2010) 1734–1739. 

[38] R. Katayama, A.T. Shaw, T.M. Khan et al. Sci. Transl. Med. 4(120) (2012) 120ra17. 

[39] J. Gainor, S.H. Ou, J. Logan, L.F. Borges, A.T. Sahw. J. Thorac. Oncol. 8(12) (2013) 1570–1573. 

[40] D. Costa, A.T. Shaw, S.H. Ou, et al. J. Clin. Oncol. 33(17) (2015) 1881–88. 

[41] R. Doebele, A. Pilling, D. Aisner, et al. Clin. Cancer Res. 18(5) (2012) 1472–1482. 



M. Mendez et al.  ADMET & DMPK 5(1) (2017) 1-8 

8  

[42] L. Friboulet, N. Li, R. Katayama, et al. Cancer Discov. 4(6) (2014) 662–673. 

[43] K. Kinoshita, K. Asoh, N. Furuchi, et al Bioorg. Med. Chem. 20(3) (2012) 1271–1280. 

[44] T. Seto, K. Kiura, M. Nishio, et al. Lancet Oncol. 14(7) (2013) 590-598. 

[45] T. Tamura, T. Seto, K. Nakagawa, et al. Radiat. Oncol. 90 (2014) S6. 

[46] S. Gadgeel, L. Gandhi, G. Riely, et al. Lancet Oncol. 15 (2014) 1119–1128. 

[47] J. Wu, J. Savooji, D. Liu. J. Hematol. Oncol. 9 (2016) 9. 

[48] R. Squillace, R. Anjum, D. Miller, et al. Cancer Res. 73(8 Suppl) (2013) Abstract #5655. 

[49] V. Rivera, F. Wang, R. Anjum, et al. Cancer Res. 72(8 Suppl) (2012) Abstract #1794. 

[50] D. Camidge, L. Bazhenova, R. Salgia, et al. J. Clin. Oncol. 33 (2015) 8062. 

[51] A. Vaishnavi, M. Capelletti, A.T. Le, et al. Nat. Med. 19(11) (2013) 1469–1472. 

[52] E. Ardini, M. Menichincheri, C. De Ponti, et al. Mol. Cancer Ther. 8(12 Suppl) (2009) A243. 

[53] M. Patel, T. Bauer, S. Liu, et al. J. Clin. Oncol. 33 (2015) 2596. 

[54] T. Johnson, P. Richardson, S. Bailey, et al. J. Med. Chem. 57(11) (2014) 4720–4744. 

[55] T. Li, P. LoRusso, M. Maitland, et al. J. Hematol. Oncol. 9 (2016) 23. 

[56] C. Lovly, J. Heuckmann, E. de Stanchina, et al. Cancer Res. 71(14) (2011) 4920–4931. 

[57] M. Cheng, M. Quail, D. Girgrich, et al. Mol. Cancer Ther. 11 (2012) 670–679. 

[58] I. Sullivan, D. Planchard. Ther. Adv. Med. Oncol.  8(1) (2016) 32–47. 

[59] L. Sequist, S. Gettinger, N. Sanzer, et al. J. Clin. Oncol. 28(33) (2010) 4953–4960. 

[60] E. Normant, G. Paez, K. West, et al. Oncogene 30(22) (2011) 2581–2586. 

[61] M. Socinski, J. Goldman, I. El-Harary, et al. Clin. Cancer Res. 19(11) (2013) 3068–3077. 

 

 

 

©2017 by the authors; licensee IAPC, Zagreb, Croatia. This article is an open-access article distributed under the terms and 
conditions of the Creative Commons Attribution license (http://creativecommons.org/licenses/by/3.0/) . 

http://creativecommons.org/licenses/by/3.0/