Dual blockade of EGFR and VEGFR pathways: Results from a pilot study evaluating apatinib plus gefitinib as a first-line treatment for advanced EGFR-mutant non-small cell lung cancer

Zhonghan Zhang, Yang Zhang, Fan Luo, Yuxiang Ma, Wenfeng Fang, Jing Zhan, Su Li, Yunpeng Yang, Yuanyuan Zhao, Shaodong Hong, Ting Zhou, Yaxiong Zhang, Shen Zhao, Yan Huang, Hongyun Zhao, Li Zhang, Zhonghan Zhang, Yang Zhang, Fan Luo, Yuxiang Ma, Wenfeng Fang, Jing Zhan, Su Li, Yunpeng Yang, Yuanyuan Zhao, Shaodong Hong, Ting Zhou, Yaxiong Zhang, Shen Zhao, Yan Huang, Hongyun Zhao, Li Zhang

Abstract

Background: Dual blockade of both EGFR and VEGFR pathways in EGFR-mutant NSCLC have shown enhanced antitumor efficacy versus EGFR-TKIs alone. Apatinib is an orally effective VEGFR-2 tyrosine kinase inhibitor (TKI). This pilot study aims to evaluate the tolerability, pharmacokinetic profile, and antitumor activity of apatinib plus gefitinib as a therapy for EGFR-mutant advanced NSCLC.

Methods: Advanced non-squamous NSCLC participants harbored with the EGFR 19 deletion or the 21 L858R point mutation were included. There were two cohorts: Cohort A: apatinib 500 mg + gefitinib 250 mg. Cohort B: apatinib 250 mg + gefitinib 250 mg. The primary endpoint was safety profile. Other endpoints consisted of PK analysis, objective response rate (ORR), and progression-free survival (PFS). Exploratory analysis was conducted using next-generation sequencing of plasma circulating-tumor DNA.

Results: Between July 2016 and April 2017, 13 of NSCLC patients were recruited. Six patients were pooled in Cohort A, while seven patients were in Cohort B. Adverse events (AEs) were tolerable (mostly grade 1-2) and the treatment-related AEs were similar in both cohorts: rash (100% vs 71.4%), diarrhea (66.7% vs 71.4%), hypertension (66.7% vs 71.4%), proteinuria (66.7% vs 42.9%), and hand-foot skin reaction (33.3% vs 28.6%). The area under plasma concentration-time curve for the steady state of apatinib was 2864.73 ± 2605.54 ng mL-1 h-1 in Cohort A and 2445.09 ± 1550.89 ng mL-1 h-1 in Cohort B. Of the 11 patients evaluable for efficacy, Cohort A achieved an ORR of 80.0% and reached a median PFS of 19.2 months, while it was 83.3% and 13.4 months in Cohort B. Patients without a concomitant mutation at baseline had a prolonged PFS tendency (20.99 months v 13.21 months, P = .0624). The EGFR-T790M mutation remained the dominant resistance mechanism.

Conclusion: Apatinib (500 mg) plus gefitinib (250 mg) showed a tolerable safety profile and encouraging antitumor activity for advanced EGFR-mutant NSCLC in the first-line setting. Phase III trials of apatinib (500 mg) plus gefitinib (250 mg) are warranted.

Trial registration: Clinicaltrials.gov, NCT02824458, date of registration June 23, 2016.

Keywords: Apatinib; EGFR-TKIs; Gefitinib; NSCLC; antiangiogenic therapy.

Conflict of interest statement

The authors declare no conflict of interest.

© 2020 The Authors. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

Figures

FIGURE 1
FIGURE 1
Trial profile
FIGURE 2
FIGURE 2
The concentration time curve of apatinib and gefitinib. A‐D, Mean Apatinib plasma concentration–time profile on Day 1 (single dose) and Day 15 (continuous dose). Patients were administered with 500 mg or 250 mg Apatinib plus 250 mg Gefitinib once a day; PK evaluation was performed on Cycle 1, Day 1 and Day 15. E and F, Mean Gefitinib plasma concentration–time profile on Day 1 (single dose) and Day 15 (continuous dose). Patients were administered with 250 mg Gefitinib 250 mg and 500 mg or 250 mg Apatinib once a day; PK evaluation was performed on Cycle 1, Day 1, and Day 15. *Two patients did not have PK data on Cycle 1, Day 15 (one patient had SAE and one was no longer willing to participate and was lost to follow‐up after C1D14)
FIGURE 3
FIGURE 3
Objective response rate (n = 11). A, Waterfall plot: A maximum tumor change from baseline by best overall response (intention‐to‐treat population). Best change of target‐lesion compared to baseline at best overall response of Apatinib combined with Gefitinib treatment. Waterfall plots for best change of target‐lesion for all patients. The colored bars represent the different doses of Apatinib. The dashed lines are at 20% and −30% represents the boundary for determination of PD and PR, respectively. Asterisks represent PD. B, Swimmer plot: Time to treatment failure from enrollment to PD during treatment with apatinib and gefitinib. Each bar represents one patient's duration exposure of apatinib plus gefitinib, and each color presents the best response of one patient. ∆ represent PD
FIGURE 4
FIGURE 4
Plasma circulating‐tumor DNA (ct‐DNA) sequencing description summary and clinical outcome exploratory analysis results of 11 patients. A, Mutation plots of sequencing profile at baseline, best of response, and after PD samples, were sorted by the apatinib dosage (500 or 250 mg). Each column represents a distinct patient. BOR, smoking status, and sex groups are shown at the top. B, Kaplan‐Meier curves of progression‐free survival (PFS) in patients whose ct‐DNA had concomitant mutations compared with those without concomitant mutations at baseline. (C) Kaplan‐Meier curves of progression‐free survival (PFS) in 8 patients who received Osimertinib after PD in the second‐line. D, Pie chart depicting the T790M resistant distribution. E, Scatter plot of EGFR‐T790M VAFs (%). The red dashed line represents the median (0 at baseline, 0.6375 at PR/SD, and 4.145 at PD). F, Percentage changes in SLD of target lesions correlated with T790M VAFs. Abbreviations: del, deletion; EGFR, epidermal growth factor receptor; HR, hazard ratio; NR, not reached; PD, progressive disease; PR, partial response; SD, stable disease; PD, progression disease and VAF, variant allele frequency; SLD, sum of the longest diameters

References

    1. Chen W, Zheng R, Baade PD, et al. Cancer statistics in China, 2015. CA Cancer J Clin. 2016;66:115‐132.
    1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69:7‐30.
    1. Georgoulias V, Vlachonikolis IG. Chemotherapy for non‐small‐cell lung cancer. The Lancet. 2003;358:1271‐1272.
    1. Shigematsu H, Lin L, Takahashi T, et al. Clinical and biological features associated with epidermal growth factor receptor gene mutations in lung cancers. J Natl Cancer Inst. 2005;97:339‐346.
    1. Ning Y, Mei G, Song Y, Li M, Pu X, Clinical signficance of EGFR,KRAS,BRAF and PIK3CA gene mutations in 430 Chinese patients with non‐small cell lung cancer. Chin J Cancer Biother 2015. 10.3872/j.issn.1007-385X.2015.06.010. Details can be found at:
    1. Shi Y, Au SK, Thongprasert S, et al. 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.
    1. Sholl LM, Aisner DL, Varella‐Garcia M, et al. Multi‐institutional oncogenic driver mutation analysis in lung adenocarcinoma: the lung cancer mutation consortium experience. J Thorac Oncol. 2015;10:768‐777.
    1. Inoue A, Kobayashi K, Maemondo M, et al. Updated overall survival results from a randomized phase III trial comparing gefitinib with carboplatin‐paclitaxel for chemo‐na?ve non‐small cell lung cancer with sensitive EGFR gene mutations (NEJ002). J Annals of Oncology. 2013;24:54‐59.
    1. Maemondo M, Inoue A, Kobayashi K, et al. Gefitinib or chemotherapy for non‐small‐cell lung cancer with mutated EGFR. N Engl J Med. 2010;362:2380‐2388.
    1. Masahiro F, Yi‐Long W, Sumitra T, 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.
    1. Mitsudomi T, Morita S, Yatabe Y, 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.
    1. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin‐paclitaxel in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947‐957.
    1. Paeng JY, Hong J, Kim CS, Kim MJ. First‐SIGNAL: first‐line single‐agent iressa versus gemcitabine and cisplatin trial in never‐smokers with adenocarcinoma of the lung. J Clin Oncol. 2012;30:1122‐1128.
    1. Kerbel RS. Antiangiogenic therapy: a universal chemosensitization strategy for cancer. J Sci. 2006;312:1171‐1175.
    1. Pietras K, Hanahan D. A multitargeted, metronomic, and maximum‐tolerated dose “chemo‐switch” regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of cancer. J Clin Oncol. 2005;23:939‐952.
    1. Seto T, Kato T, Nishio M, et al. Erlotinib alone or with bevacizumab as first‐line therapy in patients with advanced non‐squamous non‐small‐cell lung cancer harbouring EGFR mutations (JO25567): an open‐label, randomised, multicentre, phase 2 study. Lancet Oncol. 2014;15:1236‐1244.
    1. Saito H, Fukuhara T, Furuya N, et al. Erlotinib plus bevacizumab versus erlotinib alone in patients with EGFR‐positive advanced non‐squamous non‐small‐cell lung cancer (NEJ026): interim analysis of an open‐label, randomised, multicentre, phase 3 trial. Lancet Oncol. 2019;20:625‐635.
    1. Zhao H, Zhang Z, Luo F, et al. A multicenter, randomized, double‐blind, placebo‐controlled phase III study of apatinib or placebo plus gefitinib as first‐line treatment in patients with EGFR‐mutant advanced non‐small cell lung cancer (NSCLC). J Clin Oncol. 2018;36:TPS9116‐TPS9116.
    1. Zhao H, Zhang Z, Li S, et al. P1.01‐109 phase I study of apatinib plus gefitinib as first‐line therapy in patients with egfr mutant advanced non‐small cell lung cancer. J Thorac Oncol. 2018;13:S506.
    1. Reck M, Garon EB, Paz‐Ares L, et al. Randomized, double‐blind phase Ib/III study of erlotinib with ramucirumab or placebo in previously untreated EGFR‐mutant metastatic non‐small‐cell lung cancer (RELAY): phase Ib results. Clin Lung Cancer. 2018;19:213‐220. e214.
    1. Rosell R, Dafni U, Felip E, et al. Erlotinib and bevacizumab in patients with advanced non‐small‐cell lung cancer and activating EGFR mutations (BELIEF): an international, multicentre, single‐arm, phase 2 trial. Lancet Respir Med. 2017;5:435‐444.
    1. Scott AJ, Messersmith WA, Jimeno A. Apatinib: a promising oral antiangiogenic agent in the treatment of multiple solid tumors. Drugs Today. 2015;51:223‐229.
    1. Tian S, Quan H, Xie C, et al. YN968D1 is a novel and selective inhibitor of vascular endothelial growth factor receptor‐2 tyrosine kinase with potent activity in vitro and in vivo. Cancer Sci. 2011;102:1374‐1380.
    1. Yan‐Jun M, Yong‐Ju L, Hong‐Bing H, et al. Apatinib (YN968D1) reverses multidrug resistance by inhibiting the efflux function of multiple ATP‐binding cassette transporters. J Cancer Research. 2010;70:7981‐7991.
    1. Li Z, Shi M, Cheng H, et al. A phase II, multicenter, placebo‐controlled trial of apatinib in patients with advanced nonsquamous non‐small cell lung cancer (NSCLC) after two previous treatment regimens. J Clin Oncol. 2012;30.
    1. Zhang Z, Luo F, Zhang Y, et al. The ACTIVE study protocol: apatinib or placebo plus gefitinib as first‐line treatment for patients with EGFR‐mutant advanced non‐small cell lung cancer (CTONG1706). Cancer Commun. 2019;39:69.
    1. David J, William P, Riely GJ, et al. Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non‐small‐cell lung cancer. J Clin Oncol. 2010;28:357‐360.
    1. Lu S, Zhou J, Niu X, Chen M, Hua Y, Su W. P3.01‐070 A phase II study of fruquintinib in combination with gefitinib in stage IIIb/IV NSCLC patients harboring EGFR activating mutations. J Thorac Oncol. 2017;12:S2228.
    1. Haijun Z. Apatinib for molecular targeted therapy in tumor. Drug Design Dev. 2015;9:6075‐6081.
    1. Li F, Zhu T, Cao B, Wang J, Liang L. Apatinib enhances antitumour activity of EGFR‐TKIs in non‐small cell lung cancer with EGFR‐TKI resistance. Eur J Cancer. 2017;84:184‐192.
    1. Nakagawa K, Tamura T, Negoro S, et al. Phase I pharmacokinetic trial of the selective oral epidermal growth factor receptor tyrosine kinase inhibitor gefitinib (‘Iressa’, ZD1839) in Japanese patients with solid malignant tumors. J Annals of Oncology. 2003;14:922‐930.
    1. Herbst RS, Anne‐Marie M, Rothenberg ML, et al. Selective oral epidermal growth factor receptor tyrosine kinase inhibitor ZD1839 is generally well‐tolerated and has activity in non‐small‐cell lung cancer and other solid tumors: results of a phase I trial. J Clin Oncol. 2002;20:3815‐3825.
    1. Ranson M, Hammond LA, Ferry D, et al. ZD1839, a selective oral epidermal growth factor receptor‐tyrosine kinase inhibitor, is well tolerated and active in patients with solid, malignant tumors: results of a phase I trial. J Clin Oncol. 2002;20:2240.
    1. Cohen MH, Williams GA, Sridhara R, et al. United States Food and Drug Administration Drug approval summary: gefitinib (ZD1839; Iressa) tablets. Clin Cancer Res. 2004;10:1212‐1218.
    1. Cohen MH, Williams GA, Sridhara R, Chen G, Pazdur R. FDA drug approval summary: gefitinib (ZD1839) (Iressa) tablets. Oncologist. 2003;8:303‐306.
    1. Liu X, Zhang Y, Chen Q, et al. Pharmacokinetic drug interactions of apatinib with rifampin and itraconazole. J Clin Pharmacol. 2018;58:347‐356.
    1. Blakely CM, Watkins TBK, Wu W, et al. Evolution and clinical impact of co‐occurring genetic alterations in advanced‐stage EGFR‐mutant lung cancers. Nat Genet. 2017;49:1693‐1704.
    1. Hong S, Gao F, Fu S, et al. Concomitant genetic alterations with response to treatment and epidermal growth factor receptor tyrosine kinase inhibitors in patients with EGFR‐mutant advanced non‐small cell lung cancer. JAMA Oncol. 2018;4:739‐742.
    1. Sequist LV, Waltman BA, Dora DS, et al. Genotypic and histological evolution of lung cancers acquiring resistance to EGFR inhibitors. J Sci Transl Med. 2011;3:75ra26.
    1. Yu HA, Arcila ME, Natasha R, et al. Analysis of tumor specimens at the time of acquired resistance to EGFR‐TKI therapy in 155 patients with EGFR‐mutant lung cancers. J Clin Cancer Res. 2013;19:2240‐2247.
    1. Mok TS, Wu Y‐L, Ahn M‐J, et al. Osimertinib or platinum–pemetrexed in EGFR T790M–positive lung cancer. N Engl J Med. 2017;376:629‐640.
    1. Zhou Q, Wu YL, Cheng Y, et al. CTONG 1509: phase III study of bevacizumab with or without erlotinib in untreated Chinese patients with advanced EGFR‐mutated NSCLC. Ann Oncol. 2019;30:v603.
    1. Nakagawa K, Garon EB, Seto T, et al. Ramucirumab plus erlotinib in patients with untreated, EGFR‐mutated, advanced non‐small‐cell lung cancer (RELAY): a randomised, double‐blind, placebo‐controlled, phase 3 trial. Lancet Oncol. 2019;20:1655‐1669.

Source: PubMed

3
Sottoscrivi