Elsevier

Lung Cancer

Volume 108, June 2017, Pages 109-114
Lung Cancer

Patterns of initial and intracranial failure in metastatic EGFR-mutant non-small cell lung cancer treated with erlotinib

https://doi.org/10.1016/j.lungcan.2017.03.010Get rights and content

Highlights

  • We analyzed failure patterns in EGFR-mutant NSCLC patients treated with erlotinib.

  • Main pattern of progression was in initial sites of disease.

  • Risk of developing brain metastasis was also substantial.

  • This data supports earlier incorporation of local or brain-directed therapy.

Abstract

Objectives

Patients with metastatic EGFR-mutant (mEGFRmt) NSCLC have favorable survival when treated with erlotinib. We hypothesized that treatment failure in most patients is limited to initial sites of disease, in which case incorporating local therapy such as radiation might further delay progression. We therefore analyzed patterns and predictors of failure in a large cohort of such patients.

Materials and methods

We reviewed 189 patients treated with erlotinib for mEGFRmt NSCLC. We classified first pattern of failure as involving initial sites only (ISF), new sites only (NSF), or the combination (CSF), and used competing-risks regression to identify factors associated with ISF, progression and overall survival (OS). We also separately analyzed intracranial and intrathoracic failure.

Results

Of 171 patients who progressed, 103 (60.2%) had ISF, 30 (17.5%) had NSF, and 38 (22.2%) had CSF. Younger age and lack of initial CNS involvement independently correlated with ISF, with a trend for higher T and N stage. Higher T and N stage was also a significant predictor of progression. Factors predicting shorter OS were female gender, weight loss, initial intracranial involvement, and ≥4 extracranial metastases. Intrathoracic progression was a component of first failure in 61%, and three-year cumulative incidence of brain metastasis was 30%.

Conclusion

The main pattern of progression in mEGFRmt NSCLC on erlotinib is in the initial sites of disease. Younger patients and those without brain involvement are particularly likely to develop ISF. This suggests a role for incorporating local therapy into treatment of selected patients with mEGFRmt NSCLC.

Introduction

Given the effectiveness of the tyrosine kinase inhibitors (TKIs) such as erlotinib and gefitinib, patients with metastatic non-small cell lung cancer (NSCLC) harboring activating mutations in the epidermal growth factor receptor (EGFRmt) have a remarkably long natural history [1], [2], [3], [4]. Though such patients eventually still experience disease progression, their relatively long survival raises the question of whether local therapy such as radiation might prolong progression-free or even overall survival in some of these patients. A benefit for local therapy seems most likely for patients with oligometastatic disease (often defined as five or fewer total lesions) for whom local therapy to all sites is often feasible, and whose prognosis has been theorized to be better than those with florid metastatic disease.

While interest in local ablative therapy for oligometastatic disease has increased tremendously in recent years, the paucity of randomized evidence underscores the need for caution and for prospective trials, as highlighted in several recent reviews [5], [6], [7]. In the case of oligometastatic NSCLC, multiple studies are underway to evaluate this question in molecularly unselected patients: the SABR-COMET trial will test stereotactic ablative radiotherapy (SABR) in treating oligometastatic recurrence of an otherwise controlled solid primary tumor, though entry criteria are not limited specifically to lung primary tumors; and another phase II study at MD Anderson Cancer Center will assess local consolidative therapy (either surgery or radiotherapy) following chemotherapy for patients with NSCLC with one to three metastases, as well as several other similar studies [8], [9], [10], [11]. Finally, the randomized phase III SARON trial will evaluate the addition of definitive radiotherapy to standard chemotherapy in oligometastatic NSCLC patients limited to 3 metastases [12].

However, such studies of local therapy in oligometastatic disease include a variety of histologies, or do not utilize molecular selection to enhance the underlying probability that patients will have prolonged natural history and a tendency to fail in initially involved sites. Molecularly-selected EGFRmt patients treated with erlotinib represent a particularly promising population for local therapy, but we currently lack a thorough understanding of the pattern of disease progression in these patients. Knowing whether these patients first progress only in their initial sites of disease or in new sites, and identifying predictive factors for either pattern, would not only aid in understanding what sort of surveillance such patients might require, but also in identifying potential subsets that might benefit from early incorporation of local therapy. Though several small reports of oligometastatic progression of oncogene-addicted NSCLC treated with local therapy have shown promising results [13], [14], [15], only two studies to our knowledge have evaluated the pattern of failure in metastatic patients treated with TKIs—one from a group of molecularly unselected patients and one from a relatively small set of EGFRmt patients treated with TKI [16], [17]. In both, half or more of the patients had progression limited to their initial sites of disease. Using a larger cohort of molecularly-selected EGFRmt patients treated with erlotinib, we sought to further characterize the frequency with which such patients progress in their initial sites of disease, and identify potential subsets of patients most likely to exhibit this pattern and therefore, potentially benefit from early local therapy. As disease progression in the CNS (typically, in the brain) is also a common pattern of failure in NSCLC with different clinical and treatment paradigms, we also performed a separate, novel analysis of CNS failure and its predictors.

Section snippets

Patients

All new patients seen at our institution from 2004 to 2013 were reviewed to identify eligible patients. Eligibility criteria included initial presentation with biopsy-confirmed NSCLC with at least one metastatic site according to the American Joint Committee on Cancer 7th edition, and with pathological molecular testing to confirm the presence of a sensitizing EGFR mutation. We collected baseline variables such as age at diagnosis, sex, stage at diagnosis, smoking history, baseline performance

Patients

Between 2004 and 2013, 189 patients with metastatic EGFRmt NSCLC met the eligibility criteria and were analyzed. Clinical and baseline characteristics are shown in Table 1. Median age for all patients was 60 years. Two-thirds of patients were non-smokers, 70% were female, and ≥80% had intrathoracic stage III disease (i.e. T4 or N2-3 disease). The predominant EGFR mutation was an in-frame deletion of exon 19, but a third of patients had an exon 21 L858R deletion, and a small minority had other

Discussion

To our knowledge, this is the largest comprehensive analysis of prognostic factors and patterns of disease progression in metastatic EGFRmt NSCLC patients treated with erlotinib. Though smaller institutional retrospective reports of EGFRmt patients treated with erlotinib or gefitinib have suggested improved OS in those with exon 19 deletion versus exon 21 L858R mutation [21], [22], this is not confirmed in our dataset of initially-metastatic patients. Instead, we find worse OS in female

Conclusions

Our data indicates that over half of patients with metastatic EGFRmt NSCLC treated with erlotinib are likely to fail at their initial sites of disease, and that younger age and lack of initial CNS involvement increase the likelihood of ISF. Furthermore, we indicate that patients without initial CNS disease are at significant risk to develop brain metastasis, and that EGFR exon 21 mutants are nearly twice as likely as exon 19 mutants to develop BM. The significant rates of CNS failure suggest

Sources of support

This research was supported by NIH Core GrantP30 CA008748.

References (32)

  • T.S. Mok et al.

    Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma

    N. Engl. J. Med.

    (2009)
  • D.A. Palma et al.

    The oligometastatic state − separating truth from wishful thinking

    Nat. Rev. Clin. Oncol.

    (2014)
  • C.G. Rusthoven et al.

    Radiation therapy for oligometastatic non-small cell lung cancer: theory and practice

    Cancer J.

    (2015)
  • D.A. Palma et al.

    Stereotactic ablative radiotherapy for comprehensive treatment of oligometastatic tumors (SABR-COMET): study protocol for a randomized phase II trial

    BMC Cancer

    (2012)
  • A Randomized Phase II Study Assessing the Efficacy of Local Consolidative Therapy for Non-Small Cell Lung Cancer...
  • Stereotactic Body Radiation Therapy (SBRT) in Metastatic Non-small Cell Lung Cancer. Available at...
  • Cited by (39)

    • Nazartinib for treatment-naive EGFR-mutant non−small cell lung cancer: Results of a phase 2, single-arm, open-label study

      2022, European Journal of Cancer
      Citation Excerpt :

      After a median follow-up time of 25 months, the median OS was not reached, with 56% of patients alive at 33 months. CNS recurrence is a frequent complication in patients with EGFR-mutant NSCLC, with a 2-year estimated risk of CNS progression of ∼15%–20% while on treatment with EGFR-TKIs [16–19]. Since the presence of brain metastases is associated with a poor prognosis and impaired quality of life, control of CNS progression is an important consideration for these patients [17,20,21].

    • EGFR mutation is not a prognostic factor for CNS metastasis in curatively resected lung adenocarcinoma patients

      2022, Lung Cancer
      Citation Excerpt :

      CNS metastasis is an important cause of treatment failure and death in NSCLC patients. In patients with advanced NSCLC receiving first-generation EGFR-TKI treatment, CNS metastasis was the most common disease progression mode[10–12]. This was mainly caused by the poor blood–brain barrier permeability of first-generation EGFR-TKIs.

    View all citing articles on Scopus

    This work has been presented at ASTRO’s 56th annual meeting, as well as the 2014 Chicago Multidisciplinary Symposium in Thoracic Oncology.

    View full text