Checkpoint inhibitors after chemoradiation: is it ready for prime time?
Editorial

Checkpoint inhibitors after chemoradiation: is it ready for prime time?

Carlos Rojas1, Vamsidhar Velcheti2

1Clınica Alemana de Santiago, Santiago, Chile; 2Department of Hematology and Oncology, Tausig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA

Correspondence to: Vamsidhar Velcheti, MD, FACP. Department of Hematology and Oncology, Tausig Cancer Institute, Cleveland Clinic, 9500 Euclid ave, Cleveland, OH 44124, USA. Email: velchev@ccf.org.

Comment on: 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.


Submitted Dec 04, 2017. Accepted for publication Dec 14, 2017.

doi: 10.21037/tcr.2017.12.22


Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death in the United States (1). About a third of the patients diagnosed with NSCLC are diagnosed at a locally advanced stage (stage III). Despite concurrent chemoradiation, the outcome for patients diagnosed with locally advanced NSCLC is very poor with a 5-year survival of <20% (2-4). Recently a new class of drugs blocking the immune checkpoint pathways have revolutionized the treatment paradigm for many solid tumors (5). Immune checkpoint pathways serve a critical role in maintaining immune-homeostasis and inducing immune tolerance to self. Immune checkpoint pathways, particularly the programmed death-1 (PD-1) axis is often co-opted by cancer cells to evade the anti-tumor immune response (6-8). Several such drugs targeting the PD-1 axis are approved by the United States Food and Drug Administration (US-FDA) for treating patients with metastatic NSCLC. However, the response rates are modest and are lower than 50% even in patients expressing high levels of programmed death ligand 1 (PD-L1) (>50% TPS) (9-11). Several pre-clinical and clinical studies demonstrate the possible synergistic effect of radiation and immunotherapy (12-14). Tumor antigen presentation, immune recognition, and activation are the key steps involved in generating an effective anti-tumor immune response (5). Radiation can induce damage-associated molecular patterns (DAMPs), the release of tumor-specific antigens and enhance antigen presentation thus augmenting an anti-tumor immune response (in-situ vaccination) (12). In a large phase 1 trial of pembrolizumab (PD-1 inhibitor) patients with metastatic NSCLC patients (KEYNOTE-1) who had prior radiation had nearly two-fold improvement in both progression-free survival (PFS) and overall survival (OS) [PFS: hazard ratio (HR), 0.56; 95% CI, 0.34–0.91, P=0.019; mPFS 4.4 vs. 2.1 months] and (OS: HR, 0.58, 95% CI, 0.36–0.94, P=0.026; mOS 10.7 vs. 5.3 months) (14).

Durvalumab is a highly selective IgG1 monoclonal antibody with high affinity to PD-L1 which is a ligand of PD-1. Several early phase trials of durvalumab as a single agent and in combination demonstrate encouraging anti-tumor activity in metastatic NSCLC and larger confirmatory trials are ongoing (15,16).

Dr. Antonia et al. recently reported the interim results of the phase III double-blind randomized (PACIFIC) study evaluating the role of consolidation durvalumab for patients with locally advanced and unresectable NSCLC after definitive chemoradiation (17). This study included patients with stage IIIA and IIIB NSCLC patients who completed platinum-based chemotherapy with concurrent radiotherapy (54–66 Gy) and patients who did not progress after chemoradiation were randomized (2:1) to consolidation durvalumab (10 mg/kg every 2 weeks) or placebo for 1 year. The study had two co-primary end points of PFS and OS. At the time of the preplanned interim analysis, OS data of the study were yet to mature however, PFS in the durvalumab group was 16.8 months compared to 5.6 months in the placebo group. The HR for disease progression or death was 0.52 (95% CI, 0.42–0.65; P<0.001). The 1-year PFS rate was 55.9% (95% CI, 51.0–60.4) for patients treated with durvalumab compared to 35.3% (95% CI, 29.0–41.7) for patients receiving placebo. Patients in the durvalumab arm also had significantly higher objective response rate (ORR) than in the placebo arm (28.4% vs. 16.0%; P<0.001). The clinical benefit was seen across all patients independent of pre-specified sub-groups including in smoking status, EGFR status and pre-chemoradiation PD-L1 status (<25% and ≥25% tumor cell expression). Treatment related adverse events in the durvalumab compared to the placebo were 67.8% vs. 53.4%, respectively. All grade Immune-related adverse events were seen in 24.2% of patients treated with durvalumab with 3.4% of them were grade 3 or 4. The most common adverse event resulting in treatment discontinuation was pneumonitis and was seen in 6.3% of patients in the durvalumab arm compared to 4.3% in the placebo arm. Grade 3 or 4 pneumonitis was seen in 3.4% vs. 2.6%, respectively.

Overall as expected treatment with durvalumab maintenance resulted in an increase in immune-related adverse events however the rates of grade 3/4 adverse events were modest and manageable. Updates on the OS from the PACIFIC study are highly anticipated, however, the magnitude of the PFS benefit (16.8 months for the durvalumab vs. 5.6 months in the control arm) is quite significant. The mPFS in the control arm was 5.6 months and was measured from the time of randomization after chemoradiation. This is comparable to other concurrent chemoradiation trials (2).

The benefit was seen in both patients with squamous and non-squamous NSCLC, and was independent of PD-L1 status. These data thus support the use of durvalumab consolidation therapy in all patients with unresectable stage III NSCLC. The National Comprehensive Cancer Network (NCCN) update (version 9) incorporated durvalumab consolidation treatment for stage III NSCLC following concurrent chemoradiation (level 2A evidence) (18).

Treatment with chemoradiation could result in improved immunogenicity of the tumor resulting from improved antigen presentation and amplification of tumor directed immune response by “in-situ vaccination” (12). The data from the PACIFIC trial further support the idea of combination/sequencing of immune-checkpoint inhibitors with chemotherapy and radiation with the intent to “immune-prime” tumors. There is increasing clinical evidence for an immune priming role of radiation. Recent post-hoc analysis of a large phase 1 trial of pembrolizumab in patients with stage IV NSCLC (KEYNOTE-1) there was a nearly doubling of OS in patients who had previous radiation (14). The optimal dose, radiation technique and sequencing of radiation with immunotherapy is still unclear and further evaluation in prospective trials is warranted. Several prospective clinical trials are currently ongoing to explore the additive benefit of radiation to immune-checkpoint therapy in metastatic NSCLC (NCT02444741, NCT03217071, NCT02658097, NCT03307759, NCT02492568, NCT02407171, and NCT02858869) (19).

Is durvalumab maintenance therapy ready for prime time on stage III NSCLC after chemoradiation? For patients with locally advanced NSCLC based on the PACIFIC study durvalumab is the new standard of care following chemoradiation for un-resectable NSCLC. OS data has not been presented yet however, with nearly three-fold improvement in the PFS and the durability of the responses seen in patients receiving durvalumab it is likely that this will translate to an OS benefit.

The role of surgery in stage IIIA NSCLC has been widely debated and the patterns of practice vary across institutions. The only randomized phase III study (Intergroup 0139 trial) that evaluated induction chemoradiation followed by surgery versus definitive chemoradiation demonstrated no survival advantage for doing surgery in resectable stage IIIA NSCLC (20). However, based on post-hoc analysis from the intergroup 0129 trial and several retrospective analyses patients with limited/non-bulky mediastinal (N2) and candidates for lobectomy could benefit from a trimodality approach (21). In some institutions with high surgical volumes and low operative mortality trimodality approach with induction chemotherapy followed by surgery has come to be the standard approach in managing “resectable” stage IIIA NSCLC. The PACIFIC trial does not address the role of immune-checkpoint inhibitors in the surgically resectable stage IIIA NSCLC patients. It could be plausible that patients undergoing chemoradiation followed by surgery could also benefit from maintenance durvalumab. However, considering that this patient population was not studied in the PACIFIC trial it may not be considered as a standard of care at this time. Considering the PACIFIC data, it would be even more important to be highly selective in offering a trimodality approach to patients with “resectable” stage IIIA NSCLC. Several trials incorporating checkpoint inhibitors in the management of early stage and locally advanced NSCLC are ongoing. Future trials particularly in locally advanced stage NSCLC should incorporate better patient selection strategies using biomarkers for selecting patients who could benefit most from immunotherapy. Identifying patients who could potentially benefit from a more aggressive surgical approach incorporating immunotherapy could lead to an improved chance of cure for these patients.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned and reviewed by the Section Editor Peng Zhang, MD, PhD (Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China).

Conflicts of Interest: V Velcheti: Consultant/Advisory Role: BMS, Genentech, Astrazenca, Celgene, Amgen, Foundation Medicine. C Rojas has 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. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin 2015;65:5-29. [Crossref] [PubMed]
  2. Bradley JD, Paulus R, Komaki R, et al. Standard-dose versus high-dose conformal radiotherapy with concurrent and consolidation carboplatin plus paclitaxel with or without cetuximab for patients with stage IIIA or IIIB non-small-cell lung cancer (RTOG 0617): a randomised, two-by-two factorial phase 3 study. Lancet Oncol 2015;16:187-99. [Crossref] [PubMed]
  3. Aupérin A, Le Pechoux C, Rolland E, et al. Meta-analysis of concomitant versus sequential radiochemotherapy in locally advanced non-small-cell lung cancer. J Clin Oncol 2010;28:2181-90. [Crossref] [PubMed]
  4. Bezjak A, Temin S, Franklin G, et al. Definitive and adjuvant radiotherapy in locally advanced non-small-cell lung cancer: American Society of Clinical Oncology Clinical Practice Guideline Endorsement of the American Society for Radiation Oncology Evidence-Based Clinical Practice Guideline. J Clin Oncol 2015;33:2100-5. [Crossref] [PubMed]
  5. Velcheti V, Schalper K. Basic Overview of Current Immunotherapy Approaches in Cancer. Am Soc Clin Oncol Educ Book 2016;35:298-308. [Crossref] [PubMed]
  6. Schalper KA, Brown J, Carvajal-Hausdorf D, et al. Objective measurement and clinical significance of TILs in non-small cell lung cancer. J Natl Cancer Inst 2015;107:dju435 [Crossref] [PubMed]
  7. Schalper KA, Carvajal-Hausdorf D, McLaughlin J, et al. Clinical significance of PD-L1 protein expression on tumor-associated macrophages in lung cancer. J Immunother Cancer 2015;3:415. [Crossref]
  8. Velcheti V, Schalper KA, Carvajal DE, et al. Programmed death ligand-1 expression in non-small cell lung cancer. Lab Invest 2014;94:107-16. [Crossref] [PubMed]
  9. Reck M, Rodriguez-Abreu D, Robinson AG, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med 2016;375:1823-33. [Crossref] [PubMed]
  10. Herbst RS, Baas P, Kim DW, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016;387:1540-50. [Crossref] [PubMed]
  11. Khunger M, Hernandez AV, Pasupuleti V, et al. Programmed cell death 1 (PD-1) Ligand (PD-L1) expression in solid tumors as a predictive biomarker of benefit from PD-1/PD-L1 axis inhibitors: a systematic review and meta-analysis. JCO Precis Oncol 2017;1:1-15.
  12. Formenti SC, Demaria S. Radiation therapy to convert the tumor into an in situ vaccine. Int J Radiat Oncol Biol Phys 2012;84:879-80. [Crossref] [PubMed]
  13. Demaria S, Golden EB, Formenti SC. Role of local radiation therapy in cancer immunotherapy. JAMA Oncol 2015;1:1325-32. [Crossref] [PubMed]
  14. Shaverdian N, Lisberg AE, Bornazyan K, et al. Previous radiotherapy and the clinical activity and toxicity of pembrolizumab in the treatment of non-small-cell lung cancer: a secondary analysis of the KEYNOTE-001 phase 1 trial. Lancet Oncol 2017;18:895-903. [Crossref] [PubMed]
  15. Stewart R, Morrow M, Hammond SA, et al. Identification and Characterization of MEDI4736, an Antagonistic Anti-PD-L1 Monoclonal Antibody. Cancer Immunol Res 2015;3:1052-62. [Crossref] [PubMed]
  16. Mezquita L, Planchard D. Durvalumab in non-small-cell lung cancer patients: current developments. Future Oncol 2018;14:205-22. [Crossref] [PubMed]
  17. 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]
  18. Ettinger DS, Wood DE, Aisner DL, et al. Non-Small Cell Lung Cancer, Version 9.2017, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2017;15:504-35. [Crossref] [PubMed]
  19. Available online: https://clinicaltrials.gov/
  20. 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]
  21. Francolini G, Ferrari K, Scotti V. Neoadjuvant approach for nonsmall cell lung cancer: overview of the current issues. Curr Opin Oncol 2017;29:123-8. [PubMed]
Cite this article as: Rojas C, Velcheti V. Checkpoint inhibitors after chemoradiation: is it ready for prime time? Transl Cancer Res 2018;7(Suppl 1):S41-S43. doi: 10.21037/tcr.2017.12.22

Download Citation