Skip to main content
ESMO Open logoLink to ESMO Open
. 2016 Jul 21;1(4):e000063. doi: 10.1136/esmoopen-2016-000063

Phase II study of erlotinib plus tivantinib (ARQ 197) in patients with locally advanced or metastatic EGFR mutation-positive non-small-cell lung cancer just after progression on EGFR-TKI, gefitinib or erlotinib

Koichi Azuma 1, Tomonori Hirashima 2, Nobuyuki Yamamoto 3, Isamu Okamoto 4,5, Toshiaki Takahashi 6, Makoto Nishio 7, Taizo Hirata 8, Kaoru Kubota 9, Kazuo Kasahara 10, Toyoaki Hida 11, Hiroshige Yoshioka 12, Kaoru Nakanishi 13, Shiro Akinaga 13, Kazuto Nishio 14, Tetsuya Mitsudomi 15, Kazuhiko Nakagawa 4
PMCID: PMC5070235  PMID: 27843623

Abstract

Background

Patients with epidermal growth factor receptor (EGFR) activation mutation-positive non-small-cell lung cancer (NSCLC) respond well to EGFR tyrosine kinase inhibitors (EGFR-TKIs), but eventually become resistant in most cases. The hepatocyte growth factor/c-Met (HGF/c-Met) pathway is reported as a poor prognostic factor in various cancers. As c-Met is involved in EGFR-TKI resistance, a c-Met inhibitor and EGFR-TKI combination may reverse the resistance. This study evaluated the efficacy and safety of a c-Met selective inhibitor, tivantinib (ARQ 197), in combination with erlotinib, in Japanese EGFR mutation-positive patients with NSCLC who progressed while on EGFR-TKIs.

Methods

This study enrolled 45 patients with NSCLC with acquired resistance to EGFR-TKIs, who were orally administered a daily combination of tivantinib/erlotinib. The primary end point was the overall response rate (ORR) and secondary end points included disease control rate, progression-free survival (PFS) and overall survival (OS). The patients underwent a mandatory second biopsy just after progression on EGFR-TKIs. The predictive biomarkers were extensively analysed using tumour and blood samples.

Results

The ORR was 6.7% (95% CI 1.4% to 18.3%), and the lower limit of 95% CI did not exceed the target of 5%. The median PFS (mPFS) and median OS (mOS) were 2.7 months (95% CI 1.4 to 4.2) and 18.0 months (95% CI 13.4 to 22.2), respectively. Both were longer in c-Met high patients (c-Met high vs low: mPFS 4.1 vs 1.4 months; mOS 20.7 vs 13.9 months). Partial response was observed in three patients, all of whom were c-Met and HGF high. The common adverse events and their frequencies were similar to those known to occur with tivantinib or erlotinib alone.

Conclusions

Although this study did not prove clinical benefit of tivantinib in patients with acquired resistance to EGFR-TKIs, activated HGF/c-Met signalling, a poor prognostic factor, may define a patient subset associated with longer survival by the tivantinib/erlotinib combination.

Trial registration number

NCT01580735.

Keywords: Tivantinib, Non-small cell lung cancer, c-Met inhibitor, EGFR mutation positive, EGFR-TKI resistance


Key questions.

What is already known about this subject?

  • Patients with epidermal growth factor receptor (EGFR) activation mutation-positive non-small-cell lung cancer (NSCLC) respond well to EGFR tyrosine kinase inhibitors (EGFR-TKIs), but eventually become resistant in most cases.

  • The hepatocyte growth factor/c-Met (HGF/c-Met) pathway is reported as a poor prognostic factor in various cancers.

  • As c-Met is involved in EGFR-TKI resistance, a c-Met inhibitor and EGFR-TKI combination may reverse the resistance.

What does this study add?

  • This is the first study to evaluate the efficacy and safety of a c-Met selective inhibitor, tivantinib (ARQ 197), in combination with erlotinib, in Japanese EGFR mutation-positive patients with NSCLC who progressed while on EGFR-TKIs, and to necessitate a second biopsy just after progression on EGFR-TKIs.

  • The primary end point (objective response rate) did not achieve the target level (ie, the lower limit of 95% CI exceeding the 5% threshold).

  • Partial response was observed in three patients, all of whom were c-Met and HGF high expression by immunohistochemistry.

  • Median progression-free survival (mPFS) and median overall survival (mOS) of c-Met high patients were longer than those of c-Met low patients, and, similarly, mPFS and mOS of HGF high patients were longer than those of HGF low patients.

How might this impact on clinical practice?

  • Activated HGF/c-Met signalling, a poor prognostic factor, may define a patient subset associated with longer survival using the tivantinib/erlotinib combination.

  • Taken together with the results of the previous phase III studies (the MARQUEE study and the ATTENTION study), activated HGF/c-Met signalling could be an independent predictive biomarker for selecting patients with NSCLC who may respond to tivantinib and, furthermore, tivantinib might have some potential as a single agent particularly for NSCLC with activated HGF/c-Met signalling, regardless of EGFR activation or inhibition.

Introduction

In Asia, lung cancer is the most frequent malignant tumour in males and the second most frequent in females.1 About 85% of lung cancer is classified into non-small-cell lung cancer (NSCLC), and activating epidermal growth factor receptor (EGFR) mutation is present in 32% of Asian and 7% of non-Asian patients with NSCLC.2 EGFR tyrosine kinase inhibitors (EGFR-TKIs) showed significant clinical benefit as a first-line treatment in patients with advanced or metastatic NSCLC with activating EGFR mutation.3 However, most of those responders eventually become resistant. Secondary mutation of EGFR (T790M), conversion to small-cell lung cancer and activated hepatocyte growth factor/c-Met (HGF/c-Met) signalling have been reported as the mechanisms of acquired EGFR-TKI resistance.4–6 A non-clinical study reported that acquired EGFR-TKI resistance was reversed by the combination of a c-Met inhibitor and gefitinib in an EGFR-TKI-resistant lung cancer cell line with c-Met amplification.7

Activation of HGF/c-Met signalling due to overexpression of HGF/c-Met is reported to be involved in tumour infiltration and metastasis, and is identified as a poor prognosis factor in NSCLC.8–11 Tivantinib (ARQ 197) is an oral, non-ATP-competitive, low-molecular weight selective c-Met inhibitor. The primary metabolic enzyme of tivantinib, CYP2C19, is known for the gene polymorphism associated with loss of function. The frequency of homozygotes with CYP2C19 loss-of-function polymorphism (poor metabolisers (PMs)) is about 3% in Caucasians and 15–20% in Asians.12 A previous Japanese phase I study showed a recommended tivantinib dose of 240 mg twice daily in PMs, and 360 mg twice daily in the other patients (extensive metabolisers (EMs)) with or without erlotinib, an EGFR-TKI, in patients with NSCLC (ARQ 197–0701, ARQ 197–003 and ARQ 197–005 studies).13 14

The clinical efficacy of the tivantinib/erlotinib combination in EGFR-TKI-naive NSCLC has been evaluated by comparing it with the placebo/erlotinib combination in three randomised phase II/III trials: ARQ 197–209 study (n=167 from the USA/European Union (EU)), MARQUEE study (n=1048 from the USA/EU) and ATTENTION study (n=307 from Asia, only EGFR mutation-negative patients were enrolled). The primary end point of the ARQ 197–209 study was progression-free survival (PFS) and that for the other two studies was overall survival (OS).15–17 These studies showed an extension of PFS, with the p value in ARQ 197–209, MARQUEE and ATTENTION studies as 0.038 (HR 0.68), 0.001 (HR 0.74) and 0.019 (HR 0.719), respectively.15–17 The MARQUEE study also showed an extension of OS in high c-Met patients (HR 0.70; p=0.03). However, the percentage of EGFR mutation-positive patients in the ARQ 197–209, MARQUEE and ATTENTION studies was merely 10.2%, 10.4% and 0%, respectively. Therefore, clinical profiles including efficacy and safety of the tivantinib/erlotinib combination in EGFR mutation-positive patients have hardly been examined yet.

This is the first phase II study to evaluate the efficacy of the tivantinib/erlotinib combination in EGFR mutation-positive patients who are resistant to previous EGFR-TKI treatment. Tumour biopsy just after progression on EGFR-TKIs was mandatory for study entry to explore predictive biomarkers of efficacy of the tivantinib/erlotinib combination.

Patients and methods

Study design

This study was a phase II, single-arm, open-label, 10-centre study with a target sample size of 40 (ARQ 197–007 study; NCT01580735). Patients with EGFR mutation-positive advanced or metastatic NSCLC, just after gefitinib or erlotinib treatment, were enrolled to receive the tivantinib/erlotinib combination. Only gefitinib and erlotinib were approved as EGFR-TKIs at the time of this study. Prior platinum-based regimen was allowed. Tivantinib was provided by Kyowa Hakko Kirin Co, Ltd. Tivantinib was administered at 360 mg twice daily to CYP2C19 EMs and 240 mg twice daily to PMs, during or immediately after meals. Erlotinib 150 mg four times a day was given on an empty stomach, ≥1 hour before or ≥2 hours after meals, regardless of CYP2C19 polymorphism. Treatments were continued until patients met the discontinuation criteria including disease progression (PD) and >14 days of drug interruption.

The primary end point was objective response rate (ORR), and the secondary end points included disease control rate (DCR), PFS, OS and safety. Tumour response was evaluated by an independent review committee.

Predictive biomarkers of antitumour activity were exploratory end points. Tissue samples with confirmed tumour cells were collected in the period between progression on EGFR-TKIs and study registration. The pretreatment tumour tissues were assayed for c-Met by immunohistochemistry (IHC) and fluorescence in situ hybridisation (FISH), HGF by IHC (SRL, Inc, Tokyo) and extensive lung cancer gene mutation analysis LungCarta (MassARRAY, Agena Bioscience, California, USA). BioPlex test (BioRad, California, USA) and soluble c-Met concentration analysis (Immuno-Biological Laboratories Co, Ltd, Gumma, Japan) were performed on blood samples at screening and 2 weeks after the start of the tivantinib/erlotinib combination. Details of the analyses are described in online supplementary Data.

supplementary data

esmoopen-2016-000063supp.pdf (210.6KB, pdf)

This study was conducted in compliance with the Declaration of Helsinki and Good Clinical Practice (GCP). The Institutional Review Boards in all hospitals approved this study, and all patients gave written consent to participate in the study.

Inclusion/exclusion criteria: The main inclusion criteria included: age ≥20 years; Eastern Cooperative Oncology Group (ECOG) performance status ≤1; stage IIIB or IV NSCLC at the time of registration; EGFR mutation-positive (exon 19 deletion mutation and/or exon 21 L858R point mutation) before informed consent; history of receiving at least one prior regimen of systemic chemotherapy; history of only one regimen of either gefitinib or erlotinib monotherapy immediately before this study and disease progression following the prior EGFR-TKI monotherapy.

Patient evaluation: The baseline evaluation included vital signs, haematological tests, blood biochemistry tests, ECG, CYP2C19 polymorphism analysis and tumour measurement. Vital signs and haematological/blood biochemistry tests were performed every week in the first 4 weeks and every 2 weeks thereafter. ECG and tumour measurement were performed every 6 weeks. Tumour measurement was evaluated based on Response Evaluation Criteria in Solid Tumor (RECIST) V.1.1. Adverse events (AEs) were evaluated based on Common Terminology Criteria for Adverse Events (CTCAE) V.4.0.

Statistical analysis

The statistical analysis in this study was prospectively defined in the protocol. ORR, the primary end point, with its 95% CI, was estimated. ORR in the tivantinib/erlotinib combination was assumed to be 20% based on the 16.7% ORR reported in the phase II ARQ 197–209 study.15 The target level of ORR was the lower limit of the 95% CI exceeding 5% threshold of ORR, based on the response rate of docetaxel, which is commonly used after EGFR-TKI failure.18–20 Under these assumptions, efficacy could be evaluated in 40 patients at a power of 80%.

PFS and OS, the secondary end points, were estimated by the Kaplan-Meier method. The patients who received poststudy treatment before PD or death confirmation and those with no PD or death confirmation were censored for PFS on the day of non-PD confirmation, and data of patients whose deaths had not been confirmed were censored for OS on the day of the most recent survival confirmation.

Results

Forty-five patients were registered between June 2012 and February 2013. Data were cut-off in September 2015. The patient characteristics are shown in table 1. The proportion of females, adenocarcinoma and non-smokers was high, and amplified c-Met (FISH) was low (6.7%).

Table 1.

Patient characteristics

Overall EM PM
N=45 N=36 N=9
Gender
 Female 28 (62.2%) 19 (52.8%) 9 (100.0%)
 Male 17 (37.8%) 17 (47.2%) 0
Age (years)
 Mean (minimum–maximum) 65.2 (35–85) 65.2 (41–85) 65.1 (35–79)
CYP2C19 phenotype
 EM 36 (80.0%) 36 (100.0%) 0
 PM 9 (20.0%) 0 9 (100.0%)
EGFR mutation status (possibly duplicated)
 Exon19 deletions 23 21 2
 L858R 22 15 7
Tumour histology
 Adenocarcinoma 44 (97.8%) 35 (97.2%) 9 (100.0%)
 Large cell carcinoma 1 (2.2%) 1 (2.8%) 0
Number of prior chemotherapies
 1 33 (73.3%) 27 (75.0%) 6 (66.7%)
 2 10 (22.2%) 8 (22.2%) 2 (22.2%)
 3 2 (4.4%) 1 (2.8%) 1 (11.1%)
Smoking history
 Current 0 0 0
 Previous 19 (42.2%) 17 (47.2%) 2 (22.2%)
 Never 26 (57.8%) 19 (52.8%) 7 (77.8%)
Prior surgeries
 Yes 8 (17.8%) 5 (13.9%) 3 (33.3%)
 No 37 (82.2%) 31 (86.1%) 6 (66.7%)
Prior radiotherapies
 Yes 19 (42.2%) 16 (44.4%) 3 (33.3%)
 No 26 (57.8%) 20 (55.6%) 6 (66.7%)
ECOG PS (baseline)
 0 22 (48.9%) 16 (44.4%) 6 (66.7%)
 1 23 (51.1%) 20 (55.6%) 3 (33.3%)
Recent prior chemotherapy regimen (except for maintenance/adjuvant)
 ERL 9 (20.0%) 9 (25.0%) 0
 GEF 36 (80.0%) 27 (75.0%) 9 (100.0%)
Best overall response to recent prior chemotherapy (except for maintenance/adjuvant)
 CR 1 (2.2%) 1 (2.8%) 0
 PR 32 (71.1%) 25 (69.4%) 7 (77.8%)
 SD 11 (24.4%) 9 (25.0%) 2 (22.2%)
 NE 1 (2.2%) 1 (2.8%) 0
Met status
 High 22 (48.9%) 17 (47.2%) 5 (55.6%)
 Low 22 (48.9%) 18 (50.0%) 4 (44.4%)
 Unknown 1 (2.2%) 1 (2.8%) 0
Met (FISH) status
 Amplified 3 (6.7%) 3 (8.3%) 0
 Normal 25 (55.6%) 22 (61.1%) 3 (33.3%)
 Unknown 17 (37.8%) 11 (30.6%) 6 (66.7%)
HGF status
 High 32 (71.1%) 26 (72.2%) 6 (66.7%)
 Low 13 (28.9%) 10 (27.8%) 3 (33.3%)

Met status:

 High: 50% or more tumour cells with moderate or strong (2+ or 3+) staining intensity by IHC.

 Low: other than ‘Met status high’ or ‘unknown’.

 Unknown: ‘missing data’ or reported as ‘reference value’.

Met (FISH) status:

 High: defined as gene copy number ≥4.

 Low: other than ‘Met (FISH) status high’ or ‘unknown’.

 Unknown: ‘missing data’ or reported as ‘reference value’.

HGF status:

 High: H-score of ≥200 by IHC.

 Low: H-score of <200 by IHC.

CR, complete response; ECOG, Eastern Cooperative Oncology Group; EGFR, epidermal growth factor receptor; EM, extensive metabolisers; ERL, erlotinib; FISH, fluorescence in situ hybridisation; GEF, gefitinib; HGF, hepatocyte growth factor; IHC, immunohistochemistry; NE, not evaluable; PM, poor metabolisers; PR, partial response; PS, performance status; SD, stable disease.

All registered patients were included in the efficacy analysis. As shown in table 2, none of the 45 patients achieved complete response as the best overall response, while 3 patients achieved partial response (PR). ORR was 6.7% (95% CI 1.4% to 18.3%), which did not achieve the target level (ie, the lower limit of 95% CI exceeding the 5% threshold). Twenty-two patients showed stable disease (SD), and DCR was 48.9% (95% CI 33.7% to 64.2%). Median PFS (mPFS) and median OS (mOS) were 2.7 months (95% CI 1.4 to 4.2) and 18.0 months (95% CI 13.4 to 22.2), respectively.

Table 2.

Tumour response

Overall
N 45
Best overall response
 CR 0
 PR 3
 SD 19
 Non-CR/non-PD 0
 PD 21
 NE 2
Response 3
ORR (%) (95% CI) 6.7 (1.4 to 18.3)
Disease control 22
DCR (%) (95% CI) 48.9 (33.7 to 64.2)

CR, complete response; DCR, disease control rate; disease control, CR/PR/SD, DCR: disease control rate; NE, not evaluable; ORR, objective response rate; PD, progressive disease; PR, partial response; response, CR/PR, ORR: objective response rate; SD, stable disease.

All the registered patients were included in the safety analysis. Drug-related AEs occurred in 41 of 45 patients (91.1%). Table 3 shows drug-related AEs that occurred at a frequency of ≥5%. The most frequent drug-related AEs were dermatitis acneiform, decreased appetite and stomatitis, and those events were all grade ≤2 except for a grade 3 decreased appetite (2.2%). Anaemia in eight patients (17.8%), and decreased neutrophil count and white cell count in five patients each (11.1%) were observed as haematotoxicity, typical AE of tivantinib,13–17 and approximately half of these haematotoxicities were grade ≥3. All grade ≥3 haematotoxicities, except one case of anaemia, occurred within a month of starting the tivantinib/erlotinib combination. These haematotoxicities resolved in all the patients following the study-treatment interruption and granulocyte colony-stimulating factor (G-CSF) therapy. Interstitial lung disease (ILD), possibly related to the study drugs, occurred in two patients (4.4%). One patient developed grade 3 ILD 92 days after starting the combination. The event resolved with steroid pulse therapy. The other patient developed concurrent lung infection and ILD 124 days after starting the combination. Although these events improved initially with study-treatment interruption, and antimicrobial agent and steroid pulse therapy, the patient died of ILD 137 days after starting the combination.

Table 3.

Summary of drug-related treatment emergent AEs stratified by worst grade (MedDRA/J)

N (Per cent) Grade
Grade ≥3
AE PT 1 2 3 4 5 n Per cent
At least one TEAE 41 91.1 14 15 9 2 1 12 26.7
Dermatitis acneiform 24 53.3 15 9 0 0 0 0 0
Decreased appetite 14 31.1 8 5 1 0 0 1 2.2
Stomatitis 13 28.9 11 2 0 0 0 0 0
Diarrhoea 12 26.7 9 3 0 0 0 0 0
Dry skin 10 22.2 9 1 0 0 0 0 0
Anaemia 8 17.8 0 4 4 0 0 4 8.9
Malaise 8 17.8 4 3 1 0 0 1 2.2
Paronychia 7 15.6 5 2 0 0 0 0 0
Fatigue 6 13.3 4 1 1 0 0 1 2.2
Alanine aminotransferase increased 6 13.3 6 0 0 0 0 0 0
Weight decreased 6 13.3 1 5 0 0 0 0 0
Dysgeusia 6 13.3 4 2 0 0 0 0 0
Nausea 5 11.1 5 0 0 0 0 0 0
Neutrophil count decreased 5 11.1 0 2 1 2 0 3 6.7
White cell count decreased 5 11.1 0 2 2 1 0 3 6.7
Pruritus 5 11.1 5 0 0 0 0 0 0
Aspartate aminotransferase increased 4 8.9 4 0 0 0 0 0 0
Hypertension 4 8.9 0 4 0 0 0 0 0
Abdominal pain 3 6.7 3 0 0 0 0 0 0
Vomiting 3 6.7 3 0 0 0 0 0 0
Nasopharyngitis 3 6.7 2 1 0 0 0 0 0
Blood bilirubin increased 3 6.7 1 2 0 0 0 0 0
Lymphocyte count decreased 3 6.7 0 1 2 0 0 2 4.4

AE, adverse event; MedDRA/J, Medical Dictionary for Regulatory Activities/Japanese version; PT, preferred term; TEAE, treatment emergent AE.

This study also investigates the correlation between c-Met status and antitumour activities. Immunostaining showed the same incidence (48.9%) of high and low expression of c-Met (table 1). All three patients who achieved PR were c-Met high. Of the 22 c-Met high patients, ORR and DCR were 13.6% (95% CI 2.9% to 34.9%) and 54.5% (95% CI 32.2% to 75.6%), respectively. As shown in figure 1A, B, mPFS and mOS were longer in c-Met high patients; mPFS was 4.1 months (95% CI 1.4 to 7.0) in c-Met high and 1.4 months (95% CI 1.4 to 4.2) in c-Met low, while mOS was 20.7 months (95% CI 13.7 to 33.1) in c-Met high and 13.9 months (95% CI 8.2 to 27.3) in c-Met low patients.

Figure 1.

Figure 1

Kaplan-Meier-estimated PFS and OS are presented, with PFS in c-Met high and low patients shown in (A), OS in c-Met high and low patients in (B), PFS in HGF high and low patients in (C), and OS in HGF high and low patients in (D). HGF, hepatocyte growth factor; OS, overall survival; PFS, progression-free survival.

The correlation between expression level of HGF, the only known c-Met ligand, and antitumour activities, was also evaluated. As shown in table 1, there were 32 HGF high patients and 13 HGF low patients. All three patients who achieved PR were HGF high. Of the 32 HGF high patients, ORR and DCR were 9.4% (95% CI 2.0% to 25.0) and 56.3% (95% CI 37.7% to 73.6%), respectively. As shown in figure 1C, D, mPFS and mOS were longer in HGF high; mPFS was 2.8 months (95% CI 1.4 to 4.2) in HGF high and 1.4 months (95% CI 0.7 to 5.5) in HGF low, and while mOS was 18.2 months (95% CI 13.6 to 27.3) in HGF high and 12.4 months (95% CI 2.8 to 28.8) in HGF low patients.

To explore predictive biomarkers of tivantinib, an extensive lung cancer gene mutation analysis LungCarta of 26 genes known to contribute to tumour progression was performed in tumour samples collected from all patients after confirmation of PD on the previous EGFR-TKI treatment (see online supplementary table S1). At least one gene mutation was found in 43 of 45 patients. Of these 43 patients, 41 had exon 19 deletion mutation and/or exon 21 L858R point mutation, as expected from the target patient population for this study. T790M mutation was found in half of the patients, and this result was consistent with the known proportion of EGFR-TKI-resistant mutations.4–6 Other than these mutations, mutation of STK11, a tumour suppressor and an upregulator of AMP-activated proteinkinase, was found in seven patients,21 TP53 (another tumour suppressor) in four patients,21 and of PIK3CA and MET (N375S) in one patient each. However, no apparent correlation was observed between the presence of these mutations and antitumour activities of the tivantinib/erlotinib combination.

supplementary table

esmoopen-2016-000063supp_table1.pdf (48.7KB, pdf)

Soluble c-Met concentrations seemed to associate with longer PFS and OS (see online supplementary figure S1), but did not vary much among patients as a predictive biomarker (see online supplementary table S2). On the other hand, there was no particular trend between the efficacy and concentration of nine types of protein (angiopoietin-2, follistatin, G-CSF, HGF, interleukin-8, leptin, platelet derived growth factor-BB, platelet endothelial cell adhesion molecule-1 and vascular endothelial growth factor), which were measured using the human angiogenesis panel BioPlex (see online supplementary table S3).

supplementary table

esmoopen-2016-000063supp_table2.pdf (79.6KB, pdf)

supplementary table

esmoopen-2016-000063supp_table3.pdf (82.8KB, pdf)

supplementary figure

Kaplan-Meier-estimated PFS and OS presented, with PFS in soluble c-Met high and low patients shown in A, OS in soluble c-Met high and low patients in B. NA: not available.

esmoopen-2016-000063supp_figure.pdf (75.9KB, pdf)

Discussion

This study was the first clinical trial of tivantinib in EGFR mutation-positive patients with NSCLC resistant to EGFR-TKIs. Although the primary end point was not met for the target level (ie, lower limit of ORR 95% CI did not exceed 5%), preplanned biomarker tests revealed several important findings.

Our biomarker analysis demonstrated that PFS and OS were longer in c-Met high and HGF high patients (figure 1), and all three PR patients were diagnosed as both c-Met and HGF high. This indicates that the tivantinib/erlotinib combination presents superior efficacy in EGFR-resistant patients with high expression of c-Met high or HGF high, both reported as poor prognostic factors.8–11 Interestingly, similar superior efficacy in a poor prognosis population (ie, c-Met high and/or HGF high) was observed in previous phase III studies testing the tivantinib/erlotinib combination, even though those studies enrolled patients with NSCLC with backgrounds different from this study. The MARQUEE study enrolled EGFR-TKI-naive Caucasian patients including about 10% EGFR mutation-positive patients, and resulted in longer OS in c-Met high; HR was 0.70 (95% CI 0.49 to 1.01, vs placebo) in c-Met high, and 0.90 (95% CI 0.64 to 1.26, vs placebo) in c-Met low.16 Similarly, the ATTENTION study that enrolled EGFR-TKI-naive Asian patients, all of whom were EGFR mutation-negative, showed longer OS in HGF high patients; HR was 0.541 (95% CI 0.303 to 0.964, vs placebo) in HGF high, and 0.949 (95% CI 0.523 to 1.720, vs placebo) in HGF low.17 Considered together, these data suggest that activated HGF/c-Met signalling could be an independent predictive biomarker for selecting patients with NSCLC who may respond to tivantinib and, furthermore, tivantinib might have some potential as a single agent particularly for NSCLC with activated HGF/c-Met signalling, regardless of EGFR activation or inhibition. In fact, PR was reported in 2 of 25 patients with NSCLC in a phase I study on the safety of tivantinib as a single agent,13 and this may encourage further studies to evaluate the efficacy of tivantinib as a single agent.

Tumour samples of all patients just after progression following EGFR-TKIs were collected to perform an extensive lung cancer gene mutation analysis LungCarta on 26 genes known to contribute to tumour progression. Exon 19 deletion mutation, exon 21 L858R point mutation and T790M mutation comprised almost all the other mutations found in this study. Thus, no mutation that could possibly be used as a predictive biomarker for the tivantinib/erlotinib combination was suggested in this study. The frequency of c-Met exon 14 skipping, which has been reported in recent studies,22 was not covered by the LungCarta panel we used.

Regarding c-Met copy number, Engelman et al7 reported the involvement of amplified c-Met for the resistance mechanism to EGFR-TKIs in EGFR mutation-positive NSCLC. In this study, only three patients (6.7%) had amplified c-Met (FISH), and the best overall response was one SD and two PD. Owing to the small sample size of this population, tivantinib did not show a clear reversal of acquired EGFR-TKI resistance related to the amplified c-Met.

The common AEs and their frequencies reported in this study were similar to those known for tivantinib and erlotinib monotherapy.13–17 23 Therefore, the tivantinib/erlotinib combination may be expected to be generally tolerable for EGFR mutation-positive patients with NSCLC previously treated with EGFR-TKIs. The incidence of ILD in this study was 4.4% (2/45 patients), which was comparable to that (4.3%; 429/9909) reported in patients treated with erlotinib alone in the phase IV POLARSTAR study (postmarketing surveillance conducted in Japan).24 However, the risk of ILD with the tivantinib/erlotinib combination was not completely ruled out because the sample size in this study was small. The Asian phase III ATTENTION study implied an increased risk of ILD in patients with NSCLC treated with the tivantinib/erlotinib combination.17 Further safety evaluation and selection of patients likely to respond is necessary to develop the tivantinib/erlotinib combination for patients with NSCLC resistant to EGFR-TKIs.

Although this study did not prove clinical benefit of tivantinib in patients with acquired resistance to EGFR-TKIs, activated HGF/c-Met signalling, which is reported as a poor prognostic factor in NSCLC,8–11 may define a patient subset associated with longer survival by treatment using the tivantinib/erlotinib combination. It will be interesting to evaluate the efficacy of tivantinib alone in patients with activated HGF/c-Met signalling in the future.

Acknowledgments

The authors thank the patients, their families, caregivers and all the personnel who contributed to patient care and data collection in this study. They also express their gratitude to Dr Kazuko Sakai for genomic analysis and expert advice. The authors would like to thank two important groups of people, the members of the Safety Review Committee (Dr Kazuo Tamura, Dr Masahiro Fukuoka and Dr Akihiko Gemma), and the members of the Independent Review Committee (Dr Ukihide Tateishi, Dr Takashi Terauchi, Dr Toshimi Takano and Dr Yuji Miura).

Footnotes

Funding: This work was supported by Kyowa Hakko Kirin, Co, Ltd (Tokyo, Japan).

Competing interests: TomH received research funding from AstraZeneca and Kyowa Hakko Kirin (KHK). MN received research funding and honoraria from Chugai pharmaceutical company (Chugai), Novartis Pharmaceuticals (Novartis) and Pfizer Inc (Pfizer). ToyH received research grants from AstraZeneca, Novartis and KHK. KaoN and SA are employees of KHK and hold stock in the company. TM is a member of the advisory board for F Hoffmann-La Roche (Roche), Chugai, Novartis and KHK, and received honoraria from Chugai and KHK, and a research grant from Chugai. KazutN received honoraria and research funding from Chugai, Pfizer and KHK.

Patient consent: Obtained.

Ethics approval: The authors obtained institutional review board approvals in all hospitals.

Provenance and peer review: Not commissioned; internally peer reviewed.

References

  • 1.Ferlay J, Soerjomataram I, Dikshit R et al. . Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015;136:E359–386. 10.1002/ijc.29210 [DOI] [PubMed] [Google Scholar]
  • 2.Mitsudomi T, Yatabe Y. Mutations of the epidermal growth factor receptor gene and related genes as determinants of epidermal growth factor receptor tyrosine kinase inhibitors sensitivity in lung cancer. Cancer Sci 2007;98:1817–24. 10.1111/j.1349-7006.2007.00607.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Masters GA, Temin S, Azzoli CG et al. . Systemic therapy for stage IV non-small-cell lung cancer: American Society of Clinical Oncology clinical practice guideline update. J Clin Oncol 2015;20:3488–515. 10.1200/JCO.2015.62.1342 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Sequist LV, Waltman BA, Dias-Santagata D et al. . Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. Sci Transl Med 2011;23:75ra26 10.1126/scitranslmed.3002003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Yu HA, Arcila ME, Rekhtman N et al. . Analysis of tumor specimens at the time of acquired resistance to EGFR-TKI therapy in 155 patients with EGFR-mutant lung cancers. Clin Cancer Res 2013;15:2240–7. 10.1158/1078-0432.CCR-12-2246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Yano S, Yamada T, Takeuchi S et al. . Hepatocyte growth factor expression in EGFR mutant lung cancer with intrinsic and acquired resistance to tyrosine kinase inhibitors in a Japanese cohort. J Thorac Oncol 2011;6:2011–17. 10.1097/JTO.0b013e31823ab0dd [DOI] [PubMed] [Google Scholar]
  • 7.Engelman JA, Zejnullahu K, Mitsudomi T et al. . MET amplification leads to gefitinib resistance in lung cancer by activating ERBB3 signaling. Science 2007;316:1039–43. 10.1126/science.1141478 [DOI] [PubMed] [Google Scholar]
  • 8.Tanaka A, Sueoka-Aragane N, Nakamura T et al. . Co-existence of positive MET FISH status with EGFR mutations signifies poor prognosis in lung adenocarcinoma patients. Lung Cancer 2012;75:89–94. 10.1016/j.lungcan.2011.06.004 [DOI] [PubMed] [Google Scholar]
  • 9.Finocchiaro G, Toschi L, Gianoncelli L et al. . Prognostic and predictive value of MET deregulation in non-small cell lung cancer. Ann Transl Med 2015;3:83 10.3978/j.issn.2305-5839.2015.03.43 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Sun W, Song L, Ai T et al. . Prognostic value of MET, cyclin D1 and MET gene copy number in non-small cell lung cancer. J Biomed Res 2013;27:220–30. 10.7555/JBR.27.20130004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Noro R, Seike M, Zou F et al. . MET FISH-positive status predicts short progression-free survival and overall survival after gefitinib treatment in lung adenocarcinoma with EGFR mutation. BMC Cancer 2015;6:31 10.1186/s12885-015-1019-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kubota T, Chiba K, Iga T. Frequency distribution of CYP2C19, CYP2D6, and CYP2C9 mutant-alleles in several different populations. Xenobio Metab Dispos 2001;16:69–74. [Google Scholar]
  • 13.Yamamoto N, Murakami H, Nishina T et al. . The effect of CYP2C19 polymorphism on the safety, tolerability, and pharmacokinetics of tivantinib (ARQ 197): results from a phase I trial in advanced solid tumors. Ann Oncol 2013;24:1653–9. 10.1093/annonc/mdt014 [DOI] [PubMed] [Google Scholar]
  • 14.Yamamoto N, Murakami H, Hayashi H et al. . CYP2C19 genotype-based phase I studies of a c-Met inhibitor tivantinib in combination with erlotinib, in advanced/metastatic non-small cell lung cancer. Br J Cancer 2013;109:2803–9. 10.1038/bjc.2013.588 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Sequist LV, von Pawel J, Garmey EG et al. . Randomized phase II study of erlotinib plus tivantinib versus erlotinib plus placebo in previously treated non-small-cell lung cancer. J Clin Oncol 2011;29:3307–15. 10.1200/JCO.2010.34.0570 [DOI] [PubMed] [Google Scholar]
  • 16.Scagliotti G, von Pawel J, Novello S et al. . Phase III multinational, randomized, double-blind, placebo-controlled study of tivantinib (ARQ 197) plus erlotinib versus erlotinib alone in previously treated patients with locally advanced or metastatic nonsquamous non-small-cell lung cancer. J Clin Oncol 2015;20:2667–74. 10.1200/JCO.2014.60.7317 [DOI] [PubMed] [Google Scholar]
  • 17.Yoshioka H, Azuma K, Yamamoto N et al. . A randomized, double-blind, placebo-controlled, phase III trial of erlotinib with or without a c-Met inhibitor tivantinib (ARQ 197) in Asian patients with previously treated stage IIIB/IV nonsquamous nonsmall-cell lung cancer harboring wild-type epidermal growth factor receptor (ATTENTION study). Ann Oncol 2015;26:2066–72. 10.1093/annonc/mdv288 [DOI] [PubMed] [Google Scholar]
  • 18.Hanna N, Shepherd FA, Fossella FV et al. . Randomized phase III trial of pemetrexed versus docetaxel in patients with non–small-cell lung cancer previously treated with chemotherapy. J Clin Oncol 2004;22:1589–97. 10.1200/JCO.2004.08.163 [DOI] [PubMed] [Google Scholar]
  • 19.Fossella FV, DeVore R, Kerr RN et al. . Randomized phase III trial of docetaxel versus vinorelbine or ifosfamide in patients with advanced non-small-cell lung cancer previously treated with platinum-containing chemotherapy regimens. J Clin Oncol 2000;18:2354–62. [DOI] [PubMed] [Google Scholar]
  • 20.Ding L, Getz G, Wheeler DA et al. . Somatic mutations affect key pathways in lung adenocarcinoma. Nature 2008;455:1069–75. 10.1038/nature07423 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Shepherd FA, Dancey J, Ramlau R et al. . Prospective randomized trial of docetaxel versus best supportive care in patients with non-small-cell lung cancer previously treated with platinum-based chemotherapy. J Clin Oncol 2000;18:2095–103. [DOI] [PubMed] [Google Scholar]
  • 22.Awad MM, Oxnard GR, Jackman DM 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;34:721–30. 10.1200/JCO.2015.63.4600 [DOI] [PubMed] [Google Scholar]
  • 23.Kubota K, Nishiwaki Y, Tamura T et al. . Efficacy and safety of erlotinib monotherapy for Japanese patients with advanced non-small cell lung cancer: a phase II study. J Thorac Oncol 2008;3:1439–45. 10.1097/JTO.0b013e31818d6702 [DOI] [PubMed] [Google Scholar]
  • 24.Gemma A, Kudoh S, Ando M et al. . Final safety and efficacy of erlotinib in the phase 4 POLARSTAR surveillance study of 10 708 Japanese patients with non-small-cell lung cancer. Cancer Sci 2014;105:1584–90. 10.1111/cas.12550 [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

supplementary data

esmoopen-2016-000063supp.pdf (210.6KB, pdf)

supplementary table

esmoopen-2016-000063supp_table1.pdf (48.7KB, pdf)

supplementary table

esmoopen-2016-000063supp_table2.pdf (79.6KB, pdf)

supplementary table

esmoopen-2016-000063supp_table3.pdf (82.8KB, pdf)

supplementary figure

Kaplan-Meier-estimated PFS and OS presented, with PFS in soluble c-Met high and low patients shown in A, OS in soluble c-Met high and low patients in B. NA: not available.

esmoopen-2016-000063supp_figure.pdf (75.9KB, pdf)

RESOURCES