EGFR inhibitor

Progression of Central Nervous System Metastases in Advanced Nonsmall Cell Lung Cancer Patients Effectively Treated with First-Generation Epidermal Growth Factor Receptor-Tyrosine Kinase Inhibitor

Abstract
Central nervous system (CNS) progression is frequently detected in patients with favorable initial responses to first-generation epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs), but data are incom- plete with respect to clinical features and prognostic factors of CNS failure in this population. We retrospectively evaluated 420 advanced nonsmall cell lung cancer (NSCLC) patients treated with first-generation EGFR-TKI for over 3 months. We analyzed CNS progression of these patients, defining as newly developed CNS metastases or progression of preexisting brain lesions after EGFR-TKI treatment. Of the 420 patients, 99 (23.6%) with CNS progression after EGFR-TKI initiation were identified. The median time to CNS progression was 12 months (95% confidence interval [CI] 10.5–13.5). Patients with L858R mutation were more likely to experience CNS failure than those with exon 19 deletion ( p = 0.008). For patients with previous brain metastases (BM), the median time to CNS progression of patients with EGFR-TKI and local CNS therapy was significantly longer than those treated with EGFR-TKI alone (14.0 months vs. 11.2 months; p = 0.016). The median survival time after CNS progression was 11.2 months (95% CI 8.8–13.5). L858R mutation, multiple BM progression and CNS with other site failures were negative prognostic factor, while localized CNS therapy was the only significant fa- vorable prognostic factor for overall survival after CNS progression. For advanced NSCLC patients treated successfully with first-generation EGFR-TKI, careful monitoring for CNS progression should be considered, especially for those with L858R mutation. Localized CNS therapy should be considered for CNS progression.

Introduction
eNTRAL NeRvoUs sYsTeM (CNS) tumor metastases are frequent and serious complications of nonsmall cell lung cancer (NSCLC) that can decrease life quality and expec- tancy. Recently, with advances in neuroimaging techniques and prolonged survival resulting from new treatments, CNS metastases of NSCLC are increasingly diagnosed. Whole brain radiotherapy (WBRT), alone or combined with surgery and stereotactic radiosurgery (SRS), has been the standard treatment for patients with brain metastases (BM). However, the overall survival (OS) is only 4–5 months for unselected NSCLC patients treated with WBRT.The first-generation tyrosine kinase inhibitors (TKIs) of the epidermal growth factor receptor (EGFR), such as gefitinb, erlotinib, and icotinib have offered therapeutic improvement for patients with NSCLC harboring EGFR mutations. Patients with NSCLC that harbor EGFR mutation can achieve better disease control and prolonged survival with EGFR-TKI. However, the prolonged survival of these patients with EGFR-TKIs may result in increased develop- ment of CNS metastases. EGFR-TKIs have distinct thera- peutic benefits against BM for EGFR-mutant patients, offering an intracranial overall response rate of 85%, a disease control rate (DCR) of 94.6%, a median progression- free survival of 12.3 months, and a median survival time (mOS) of 16.2 months.2 Thus, it may be reasonable to consider EGFR-TKIs for first-line treatment of patients with BM harboring EGFR mutations, which delay brain irradia- tion until CNS imaging or symptom progression, thereby avoiding WBRT-related neurocognitive side-effects.CNS disease progression, either new lesions or the increase in preexisting brain lesions, frequently has been detected in patients with initially good responses to EGFR-TKIs.3–5 Data are incomplete regarding clinical features and prognostic factors of CNS failure for these patients, and patients with CNS metastases are under-represented in clinical trials of systemic administration. Thus, we performed a retrospective study to examine a large cohort of patients who had initially good responses to EGFR-TKI to assess the clinical features of CNS progression and clinical factors related to prognosis.

A total of 420 NSCLC patients treated with first or second- line EGFR-TKI for over 3 months with good responses were reviewed at the Cancer center of West China Hospital, Si- chuan University between June 2010 and June 2014. All patients received an initial gadolinium-enhanced brain mag- netic resonance imaging (MRI) at the time of initial diagno- sis of NSCLC. A subsequent brain MRI was performed if symptoms or signs were present to indicate CNS involve- ment. Medical records and imaging data for patients were collected to identify patients who developed CNS metastasis during their disease course, which was defined as the presence of one or more intra-axial enhancing lesions on MRI. Patients with leptomeningeal metastases (LM) were also included. CNS progression was defined as newly developed CNS me- tastases or progression of preexisting brain lesions after the response to first-generation EGFR-TKI. We evaluated the treatment effects according to RECIST 1.1. This study was approved by the Institutional Review Board, and informed consent was obtained from each pa- tient for the use of tissue in molecular analysis.EGFR mutations were identified by real-time Amplification Refractory Mutation System (ARMS) quantitative polymerase chain reaction analysis using AmoyDxTM EGFR Mutation Detection Kit (Amoy Diagnostics Co., LTD, China) and EGFR Scorpion ARMS Kit (DxS Ltd., Manchester, United Kingdom).The crude incidence of CNS progression was calculated as the proportion of patients with newly developed BM/LM or progression of preexisting CNS involvement to the number of patients treated with EGFR-TKI. Time to CNS progression was calculated from first-day of treatment with EGFR-TKI to the date of progression. OS after CNS pro- gression was calculated from the time of CNS progression diagnosis until death from any cause. Patients alive at the time of statistical analysis were censored at last follow-up. The Pearson chi-square test or the Fisher exact test was used to compare the frequency of CNS failure according to the risk factors. The Kaplan–Meier method was used to perform survival analysis, and survival curves were com- pared using the log-rank test. Multivariable predictors of survival were determined using Cox regression analysis. Two-sided values of p < 0.05 were considered statistically significant. All statistical analyses were performed with SPSS 20.0 software. Results Of the 420 patients, 99 who developed CNS metastases at any point in their disease course were identified, including 42 patients with parenchymal BM before EGFR-TKI treat- ment and 57 patients with no history of BM before the initiation of EGFR-TKI but who developed CNS metastasis after treatment. Therefore, 99 patients had CNS progression after initiation of treatment with EGFR-TKI and their characteristics are presented in Table 1. Isolated CNS fail- ure, without systemic progression, was observed in 53 pa- tients (53.5%). Most patients (79.8%) had numerous brain lesions. Patients (56/197) with L858R mutations were more likely to experience CNS failure, compared with 17.2% of patients (35/203) with exon 19 deletions ( p = 0.008). The median follow-up time for all patients was 32.0 months (range, 6.5–101.0 months). The crude incidence of CNS progression in the cohort was 23.6%. The median time to CNS progression was 12 months (95% confidence interval [CI] 10.5–13.5). Of the 42 patients with parenchymal BM before EGFR-TKI treatment, 24 (57.1%) had received lo- calized CNS therapy before or concomitant with EGFR- TKI, including 10 patients treated with SRS and 14 with WBRT. The median time to CNS progression of patients with local CNS therapy was significantly longer than those without local CNS therapy (14.0 months vs. 11.2 months; p = 0.016; Fig. 1).After CNS progression, 48 patients received localized CNS therapy. Among them, 36 patients received WBRT; 9 patients were treated with SRS; and 1 patient received SRS and then WBRT; 1 patient received SRS followed by BM resection; and 1 patient received only resection of a solitary BM. Of the 53 patients with isolated CNS failure, 33 continued with gefitinib, 9 were treated with erlotinib, and 11 received chemotherapy. Of the 46 patients who had CNS and other site failures, 28 patients received chemotherapy and 4 pa- tients were treated with erlotinib. The other 14 patients had no further systemic therapy. At the final analysis (March 30, 2016), 26 patients were alive. The median survival time after CNS progression was 11.2 months (95% CI 8.8–13.5) (Fig. 2). In univariate analysis, the patients with exon 19 deletions had a longer OS than those with L858R mutations (22.8 vs. 9.8 months, vs. 8.5 months, p = 0.002, Fig. 3b). The OS of patients who had single brain metastasis was greater than that of patients with multiple BM (16.0 vs. 10.0 months, p = 0.011, Fig. 3c). Patients who received localized CNS therapy had increased survival, with a median survival of 13.6 months versus 8.0 months for patients without localized CNS therapy ( p = 0.001, Fig. 3d). In multivariate analysis, the addition of localized CNS therapy was the only favorable independent predictor of better OS for these patients, with a hazards ratio (HR) of 0.33 (95% CI, 0.19–0.58, p = 0.002). Multiple BM (HR = 4.52; 95% CI, 1.35–15.16; p = 0.015) and L858R mutations (HR = 1.46; 95% CI, 1.01–2.12; p = 0.047) were identified as adverse prognostic factors for CNS progression (Table 2). Discussion Here, we describe clinical features and prognostic factors of CNS progression in a cohort of patients who initially had good responses to first-generation EGFR-TKI but experi- enced CNS failure. To our knowledge, this is the largest study of this patient population and the first study to report predictors of survival after CNS progression in these pa- tients. Our data will offer clinicians more information re- garding CNS failure for patients who initially benefited from first-generation EGFR-TKI treatment.Our analysis revealed that the crude incidence of CNS progression in patients who were effectively treated with first-generation EGFR-TKI was 23.6% after a median follow-up time of 32.0 months. Previous reports in patients who experienced CNS failure after clinical benefit from EGFR- TKIs have suggested a 26% to 28% crude incidence of overall CNS failure after a median follow-up of 21.4–42.2 months [4–5]. Our results therefore suggest a similar risk of CNS progression in this patient cohort. Approximately half of the patients had isolated CNS failure without systemic progres- sion and most patients (79.8%) had numerous brain lesions. LM were observed in 29 patients as a relatively late event (median time 16.5 months) after EGFR-TKI treatment. Lee et al. reported that the median time to LM progression was 21.4 months and EGFR-TKIs did not affect the incidence or timing of LM development.6 Our study had too few patients with LM to confirm this (data not shown). Our study showed that for patients with previous BM, the median time to CNS progression of patients with EGFR- TKI and local CNS therapy was significantly longer than those treated with EGFR-TKI alone (14.0 months vs. 11.2 months; p = 0.016). Zeng et al. also reported the median time to progression of BM was longer in gefitinib-WBRT group than gefitinib group.7 The attribution of the enhanced ef- fectiveness might be synergistic effects of EGFR-TKIs and local CNS therapy. An interesting finding in our study was that patients with L858R mutations were more likely to experience CNS failure than those with exon 19 deletions (28.4% vs. 17.2%; p = 0.008). The patients with L858R mu- tations had a poor OS after CNS failure than those with exon 19 deletion (9.8 months vs. 22.8 months; p = 0.020). Several studies have indicated that advanced NSCLC patients with exon 19 deletion had a longer OS and progression-free survival (PFS) following treatment with gefitinib or erlotinib compared with those harboring L858R mutation.8–10 Up to present, there are no studies comparing the clinical differences between exon 19 deletion and L858R mutation in advanced NSCLC patients with CNS progression who had initially good responses to EGFR-TKI. We hypothesized that the lower risk of CNS progression in patients with exon 19 deletion after EGFR-TKI treatment contributed to PFS and OS advantage. Although we make note of this finding, given the small number of patients in our study and its retrospective nature, further prospective studies with greater numbers of patients with adequate follow- up are needed to define the predictive and prognostic roles of different EGFR mutations on the outcome of CNS progression after EGFR-TKIs treatment. Understanding mechanism of CNS progression in patients effectively treated with EGFR-TKIs may help with treat- ment strategies. Two possible reasons for CNS progression may exist. First, EGFR-TKIs may incompletely penetrate the blood-brain barrier. McKillop et al. confirmed in a preclinical study that after oral administration of [14C]- gefitinib to albino and pigmented rats, radioactivity had limited distribution in the CNS.11 Jackman et al. showed that greater than standard doses of gefitinib were insufficient for tumor growth inhibition in the cerebrospinal fluid (CSF) of one patient.12 Some clinical studies suggest high dose pulsatile EGFR-TKIs are successful after CNS failure with standard dose EGFR-TKI.13,14 However, inadequate drug delivery to the CSF might not be exclusively responsible for CNS failures with EGFR-TKI therapy. CNS metastases might acquire drug resistance during EGFR-TKI therapy, or they may be heterogeneous with the primary tumor in terms of their EGFR mutation status. Heon et al. reported the presence of a sensitizing exon 19 mutation concurrent with a T790M mutation in a brain specimen of a patient who expe- rienced CNS failure with gefitinib therapy after initially re- sponding.5 Ruppert et al. observed heterogeneity regarding EGFR mutation status of cranial and extracranial disease after treatment with gefitinib.15 Thus, both the unique microenvi- ronment of the CNS and the intrinsic characteristics of met- astatic CNS clones may contribute to different patterns of resistance. Studies are needed to characterize the molecular evolution of the primary tumor and CNS metastases in pa- tients with EGFR mutations treated with EGFR-TKIs. After CNS failure of standard EGFR-TKI therapy, dif- ferent treatment patterns have been suggested. Some authors suggested using another TKI.16–18 Osimertinib is an oral EGFR-TKI, which is selective for T790M resistance muta- tions with activity in the pulmonary lesions and CNS me- tastases. Since osimertinib demonstrated greater penetration of blood-brain barrier than other EGFR-TKIs, and showed specific effects on T790M-positive lesions, osimertinib had significantly great efficacy in patients with advanced NSCLC with CNS metastases in whom disease had progressed during first-line EGFR-TKI therapy.We also found that localized CNS therapy was the favor- able independent predictor of OS for patients with CNS progression. Patients who received localized CNS therapy lived longer than those without localized CNS therapy. Re- cently, patients harboring EGFR mutation with cerebral me- tastases were recommended to receive EGFR-TKI first, in hopes of delaying or obviating the need for WBRT and the risks of neurocognitive side-effects from brain radiation.21,22 Delaying WBRT for patients without obvious neurological signs or symptoms is an emerging concept. In our study, some patients treated with EGFR-TKIs eventually experienced CNS progression, and traditional methods such as WBRT or SRS were beneficial.In vitro studies documented radiosensitivity of lung cancer cells with mutant EGFR.23 Various reports indicate a favor- able clinical effect with WBRT and EGFR-TKI therapy, which offers an excellent intracranial DCR.24–27 For EGFR- mutant patients with BM at initial staging, optimal manage- ment strategy with local radiation therapy and TKI should be evaluated in prospective clinical trials. The ongoing pro- spective TRACTS trial (clinicaltrials.gov/NCT01763385), to compare concurrent WBRT and erlotinib to erlotinib alone with WBRT at time of progression, may help us understand this issue. Conclusions The CNS was frequently a failure site for NSCLC patients treated otherwise successfully with EGFR-TKI. L858R mutations might be associated with higher risk of CNS progression and represent a negative prognostic factor, compared with exon 19 deletions. Careful monitoring for CNS failure and localized CNS therapy should be consid- ered for CNS progression. Further prospective studies are needed to define the predictive and prognostic roles of dif- ferent EGFR mutations on the outcome of CNS EGFR inhibitor progression after EGFR-TKIs treatment.