Neoadjuvant therapy in localized non-small cell lung cancer: can we do better than chemotherapy?
Editorial Commentary

Neoadjuvant therapy in localized non-small cell lung cancer: can we do better than chemotherapy?

Brittney Chau, Ashwin Shinde, Arya Amini

Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA, USA

Correspondence to: Arya Amini, MD. Department of Radiation Oncology, City of Hope National Medical Center, 1500 East Duarte Road, Duarte CA 91010, USA. Email: aamini@coh.org.

Comment on: Zhong WZ, Chen KN, Chen C, et al. Erlotinib Versus Gemcitabine Plus Cisplatin as Neoadjuvant Treatment of Stage IIIA-N2 EGFR-Mutant Non-Small-Cell Lung Cancer (EMERGING-CTONG 1103): A Randomized Phase II Study. J Clin Oncol 2019;37:2235-45.


Submitted Aug 16, 2019. Accepted for publication Aug 29, 2019.

doi: 10.21037/tcr.2019.08.43


The oncologic importance of an epidermal growth factor receptor (EGFR) mutation in the era of tyrosine-kinase inhibitors (TKIs) for patients with metastatic non-small cell lung cancer (NSCLC) cannot be overstated. The multiple positive trials evaluating gefitinib, erlotinib, and more recently osimeritinib as first line treatment of EGFR mutated NSCLC have revolutionized NSCLC management, with significantly improved oncologic and toxicity outcomes in this subset of patients (1-3). However, use of EGFR-TKIs in the management of non-metastatic NSCLC has not been proven as of yet. Currently, patients with non-metastatic disease do not have any indications for EGFR-TKI outside of a clinical trial setting, with traditional chemotherapy given adjuvantly and neoadjuvantly around surgical resection, and concurrently with radiation therapy (4).

Zhong et al. recently published anticipated results from the EMERGING-CTONG 1,103 multicenter, randomized phase II trial, which explored the use of erlotinib (TKI) vs. gemcitabine plus cisplatin (GC) chemotherapy in the neoadjuvant/adjuvant setting for stage IIIA–N2 resectable NSCLC (5). Patients were randomly assigned to receive 6 weeks of neoadjuvant TKI, given daily, or GC, given every 3 weeks. Following surgery, patients received additional TKI therapy for up to 12 months or GC for an additional 2 cycles. Patients were deemed to have resectable N2 disease if the short-axis diameter was less than 3 cm and if there was no evidence of bulky or fixed lymph nodes. The primary endpoint was objective response rate (ORR), defined as either complete or partial response per the Response Evaluation Criteria in Solid Tumors (RECIST) criteria. Secondary end points included pathologic complete response, rates of pathological nodal downstaging (from N2 to N1 or N0), progression-free survival (PFS), overall survival (OS), safety, and tolerability.

Baseline characteristics were not evaluated for potentially statistically significant differences. There were numerically higher rates of T-classification ≥2 (85.7% vs. 64.8%), but lower rates of multi-station N2 disease (45.7% vs. 54.1%) in the GC arm. In total, 31/37 (83.8%) TKI and 24/35 (70.6%) GC patients were able to undergo surgery. No patients had a pathologic complete response. Rates of R0 resection were 73% and 63% for the TKI and GC arms respectively (P=NS). Lymph node downstaging occurred in 10.8% and 2.9% for the TKI and GC cohorts respectively (P=NS).

Out of 72 patients evaluated, the study found no significant difference in the primary endpoint of ORR (54.1% in TKI group vs. 34.3% in GC group, P=0.092). However, there was a significant increase in PFS in patients treated with TKI versus patients treated with GC chemotherapy (median PFS 21.5 vs. 11.4 months, P<0.001). This may have been driven by the planned difference in length of adjuvant therapy, as most patients in both arms received adjuvant therapy (75.7% for TKI, 62.9% for GC), but median duration of adjuvant TKI was 12 months, while GC was given for an additional 2 cycles. OS was similar between the two arms (45.8 months for TKI vs. 39.2 for GC, P=0.42). As expected, neoadjuvant TKI was better tolerated than GC (Grade 3+ toxicities 0% vs. 29.4%). In general, adverse events correlated with the known toxicity profiles of the two regimens, with increased skin toxicity associated with Erlotinib and more hematologic and gastrointestinal toxicities associated with GC.

The results of this trial are certainly interesting but require framing in an appropriate context to optimize patient management. The lack of evaluation of potential statistically significant differences between the two groups at baseline may have led to confounding variables affecting some of the outcomes. The lack of a statistically significant benefit in ORR is likely driven by lack of sample size (rather than a true lack of benefit) in this phase II trial without adjustment to the alpha threshold of statistical significance, especially given the 20% absolute increase in ORR seen in this cohort, which is clinically meaningful. The discordant results of no difference in ORR at time of surgery but a significant PFS benefit is likely driven by the extended duration of adjuvant TKI, and additional follow-up is required to determine if this will manifest to an OS benefit with additional follow-up. This trial provides promising evidence that treating stage IIIA–N2 patients with erlotinib and surgical resection shows promise as a neoadjuvant and adjuvant therapy strategy in patients with EGFR mutated NSCLC.

Stage IIIA–N2 NSCLC patients have a multitude of treatment options, including definitive chemoradiation with consolidative durvalumab, induction chemoradiation followed by surgery, or induction chemotherapy followed by surgery and adjuvant radiation therapy when indicated (4,6-8). The ability to potentially use an oral medication which is a less toxic and more convenient form of systemic therapy that can have an effective ORR to improve surgical outcomes is highly desirable. However, a pathological nodal downstaging of 10% with erlotinib as seen in the study is less than ideal, given its importance in predicting rates of recurrence and OS (9-13). Additionally, whether postoperative radiation was utilized was not reported and may be an important factor in differentiating outcomes in one study (9). Regardless of these limitations, the study adds to the growing body of evidence demonstrating the potential benefit of routine testing of EGFR and other targetable mutations even in the non-metastatic NSCLC patient population (14). As the authors suggest, future studies are drastically needed to evaluate the role of targeted agents such as EGFR inhibitors, anaplastic lymphoma kinase (ALK) inhibitors, and programmed death-ligand 1 (PD-L1) inhibitors for example, in the setting of non-metastatic NSCLC given how impactful they have been in the metastatic cohort.

Another area of strong interest is the utilization of erlotinib or other TKIs concurrently with chemoradiation in stage IIIA-N2 disease. Several studies support the use of erlotinib as a concurrent therapy to chemoradiation in patients with locally advanced NSCLC. They found that adding erlotinib to chemoradiation or radiation therapy alone has a favorable safety profile and correlates with promising progression-free and overall survival rates (15,16). However, randomized phase II/III data is required in this clinical scenario as well in order to drive a paradigm shift in treatment and confirm the role of TKIs in the non-metastatic scenario. Lastly, there is some early evidence suggesting a positive role for TKIs such as gefinitib in early stage NSCLC (17).

Several ongoing clinical trials are testing the efficacy, safety, and timing for use of TKIs with chemotherapy, radiation, or both in NSCLC (Clinicaltrials.gov ID#: NCT00888511, NCT01573702, NCT02859077, NCT02759835, NCT01573702, NCT02759835). These clinical trials are paving the way for the use of erlotinib and other TKIs in treatment for non-metastatic NSCLC. The EMERGING-CTONG 1103 is the start of multiple positive studies liking to come forward in the next decade and offer more tailored therapy for EGFR positive NSCLC patients.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned and reviewed by the Section Editor Dr. Wei Xu (Division of Respiratory Disease, Department of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China).

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/tcr.2019.08.43). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  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;362:2380-8. [Crossref] [PubMed]
  2. Soria JC, Ohe Y, Vansteenkiste J, et al. Osimertinib in Untreated EGFR-Mutated Advanced Non-Small-Cell Lung Cancer. N Engl J Med 2018;378:113-25. [Crossref] [PubMed]
  3. Rosell R, Carcereny E, Gervais R, et al. Erlotinib versus standard chemotherapy as first-line treatment for European patients with advanced EGFR mutation-positive non-small-cell lung cancer (EURTAC): a multicentre, open-label, randomised phase 3 trial. Lancet Oncol 2012;13:239-46. [Crossref] [PubMed]
  4. National Compressive Cancer Network Clinical Practice Guidelines in Oncology: Non-Small Cell Lung Cancer (2018). Available line: https://www.nccn.org/professionals/physician_gls/pdf/nscl.pdf
  5. Zhong WZ, Chen KN, Chen C, et al. Erlotinib Versus Gemcitabine Plus Cisplatin as Neoadjuvant Treatment of Stage IIIA-N2 EGFR-Mutant Non-Small-Cell Lung Cancer (EMERGING-CTONG 1103): A Randomized Phase II Study. J Clin Oncol 2019;37:2235-45. [Crossref] [PubMed]
  6. Antonia SJ, Villegas A, Daniel D, et al. Durvalumab after Chemoradiotherapy in Stage III Non-Small-Cell Lung Cancer. N Engl J Med 2017;377:1919-29. [Crossref] [PubMed]
  7. Albain KS, Swann RS, Rusch VW, et al. Radiotherapy plus chemotherapy with or without surgical resection for stage III non-small-cell lung cancer: a phase III randomised controlled trial. Lancet 2009;374:379-86. [Crossref] [PubMed]
  8. Lewis J, Gillaspie EA, Osmundson EC, et al. Before or After: Evolving Neoadjuvant Approaches to Locally Advanced Non-Small Cell Lung Cancer. Front Oncol 2018;8:5. [Crossref] [PubMed]
  9. Shinde A, Horne ZD, Li R, et al. Optimal adjuvant therapy in clinically N2 non-small cell lung cancer patients undergoing neoadjuvant chemotherapy and surgery: The importance of pathological response and lymph node ratio. Lung Cancer 2019;133:136-43. [Crossref] [PubMed]
  10. Betticher DC, Hsu Schmitz SF, Tötsch M, et al. Prognostic factors affecting long-term outcomes in patients with resected stage IIIA pN2 non-small-cell lung cancer: 5-year follow-up of a phase II study. Br J Cancer 2006;94:1099-106. [Crossref] [PubMed]
  11. Amini A, Lou F, Correa AM, et al. Predictors for locoregional recurrence for clinical stage III-N2 non-small cell lung cancer with nodal downstaging after induction chemotherapy and surgery. Ann Surg Oncol 2013;20:1934-40. [Crossref] [PubMed]
  12. Amini A, Correa AM, Komaki R, et al. The role of consolidation therapy for stage III non-small cell lung cancer with persistent N2 disease after induction chemotherapy. Ann Thorac Surg 2012;94:914-20. [Crossref] [PubMed]
  13. Haager B, Wiesemann S, Passlick B, et al. Prognostic value of lymph node ratio after induction therapy in stage IIIA/N2 non-small cell lung cancer: a monocentric clinical study. J Thorac Dis 2018;10:3225-31. [Crossref] [PubMed]
  14. Lindeman NI, Cagle PT, Beasley MB, et al. Molecular testing guideline for selection of lung cancer patients for EGFR and ALK tyrosine kinase inhibitors: guideline from the College of American Pathologists, International Association for the Study of Lung Cancer, and Association for Molecular Pathology. J Thorac Oncol 2013;8:823-59. [Crossref] [PubMed]
  15. Ramella S, Alberti AM, Cammilluzzi E, et al. Erlotinib and concurrent chemoradiation in pretreated NSCLC patients: radiobiological basis and clinical results. Biomed Res Int 2013;2013:403869. [Crossref] [PubMed]
  16. Wang J, Xia TY, Wang YJ, et al. Prospective study of epidermal growth factor receptor tyrosine kinase inhibitors concurrent with individualized radiotherapy for patients with locally advanced or metastatic non-small-cell lung cancer. Int J Radiat Oncol Biol Phys 2011;81:e59-65. [Crossref] [PubMed]
  17. Zhong WZ, Wang Q, Mao WM, et al. Gefitinib versus vinorelbine plus cisplatin as adjuvant treatment for stage II-IIIA (N1-N2) EGFR-mutant NSCLC (ADJUVANT/CTONG1104): a randomised, open-label, phase 3 study. Lancet Oncol 2018;19:139-48. [Crossref] [PubMed]
Cite this article as: Chau B, Shinde A, Amini A. Neoadjuvant therapy in localized non-small cell lung cancer: can we do better than chemotherapy? Transl Cancer Res 2019;8(Suppl 6):S633-S635. doi: 10.21037/tcr.2019.08.43

Download Citation