Real world data are needed to establish safety of adjuvant immune checkpoint inhibitors after pneumonectomy
Editorial Commentary

Real world data are needed to establish safety of adjuvant immune checkpoint inhibitors after pneumonectomy

Ikuo Sekine

Department of Medical Oncology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan

Correspondence to: Ikuo Sekine, MD, PhD. Department of Medical Oncology, Institute of Medicine, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki 305-8575, Japan. Email: isekine@md.tsukuba.ac.jp.

Comment on: Lee JM, Vallières E, Ding B, et al. Safety of adjuvant atezolizumab after pneumonectomy/bilobectomy in stage II-IIIA non-small cell lung cancer in the randomized phase III IMpower010 trial. J Thorac Cardiovasc Surg 2023;166:655-66.e7.


Keywords: Atezolizumab; lobectomy; non-small cell lung cancer (NSCLC)


Received: 21 August 2023; Accepted: 08 October 2023; Published online: 16 October 2023.

doi: 10.21037/actr-23-16


For stage II–IIIA non-small cell lung cancer (NSCLC), surgical resection of the tumor is the mainstay treatment, but the 5-year recurrence rate is as high as 50% to 78%, and the overall survival rate at 5 years remains 48% to 64% (1,2). Conventional adjuvant chemotherapy improves 5-year survival by only 5% at best (3). Atezolizumab, anti-programmed death ligand 1 (PD-L1) monoclonal antibody, has demonstrated significant therapeutic benefit in combination with platinum-based chemotherapy in first-line treatment and as monotherapy in second-line treatment for patients with advanced NSCLC. Thus, it has been anticipated that this agent could be used as adjuvant therapy after surgical resection in patients with resectable NSCLC. The IMpower010 trial is a randomized, open-label phase III trial of adjuvant atezolizumab or best supportive care after adjuvant platinum-based chemotherapy in patients with pathologically stage IB–IIIA NSCLC that has been completely resected. Pre-planned interim analyses of this trial showed better disease-free survival [hazard ratio (HR) 0.66, 95% confidence interval (CI): 0.50–0.88, P=0.0039] at a median follow-up of 32.2 months (4) and a trend toward better overall survival (HR 0.71, 95% CI: 0.49–1.03) at a median follow-up of 45.3 months in the atezolizumab group in stage II–IIIA patients with programmed death-ligand 1 (PD-L1) expression in more than 1% of tumor cells (5). Toxicity of adjuvant atezolizumab was acceptable. Grade 3–4 and serious adverse events (AEs) were observed in 22% and 18% of patients in the atezolizumab group and in 12% and 8% for the best supportive care group, respectively (4). Immune-related grade 3–4 AEs developed in 8%, and the use of systemic corticosteroids was required in 12% of patients in the atezolizumab group. Treatment-related death was noted in 1.6% (atezolizumab-related death 0.8%) and 0.6% of patients in the atezolizumab and best supportive care groups, respectively (4). Although these initial data appear very promising, special considerations must be given to the postoperative drug treatment of patients with resectable stage disease. This is because such patients include those already cured by surgical resection alone. Therefore, safety considerations are very important when using immune checkpoint inhibitors (ICIs) in the postoperative period.

Dr. Lee and colleagues published a paper entitled “Safety of adjuvant atezolizumab after pneumonectomy/bilobectomy in stage II-IIIA non-small cell lung cancer in the randomized phase III IMpower010 trial” (6). Of 433 patients with stage II–IIIA NSCLC in the adjuvant atezolizumab group, 100 (23%) received pneumonectomy or bilobectomy and 332 (77%) received lobectomy or sleeve lobectomy. There was no difference in the frequency of grade 3–4 any AEs (21% vs. 23%), grade 3–4 pneumonitis (0% vs. 1.2%), atezolizumab discontinuation (32% vs. 35%), hospitalization for AEs (13% vs. 17%), or treatment-related death (1.0% vs. 1.2%) between the pneumonectomy or bilobectomy group and the lobectomy or sleeve lobectomy group, respectively. These exploratory findings may support the adjuvant use of atezolizumab in patients undergoing pneumonectomy. However, these results should be evaluated with caution because pneumonectomy, surgical removal of an entire lung, has traditionally been a great concern for its mortality and morbidity. After a literature review of more than 300 pneumonectomy cases published since 1990, contemporary 30-day mortality rates ranging from 3% to 12% and 90-day or in-hospital mortality rates ranging from 2% to 12% (Table 1) (7-15). The most common cause of 30-day death was acute respiratory distress syndrome (ARDS) in 51–54% of cases, followed by bronchopulmonary fistula in 11–23% and cardiac in 8–14% (10,13). These studies also identified risk factors for mortality. For example, Yu et al. retrieved 10,337 pneumonectomy cases from the Surveillance, Epidemiology, and End Results (SEER) cancer registry, and reported that old age, male gender, right-sided pneumonectomy, and squamous cell carcinoma histology were significant risk factors for death within 90 days after pneumonectomy (14). Major postoperative complications occurred in 20–26% of patients, with ARDS in 3–16%, pneumonia in 4–10%, bronchopulmonary fistula in 2–4%, and heart failure in 1–7% (9,10,15). Furthermore, pneumonectomy is associated with a variety of significant anatomic changes, which can lead to late complications. Postpneumonectomy syndrome characterized by gradually increasing shortness of breath, audible stridor, and recurrent airway infections 1 month to several years (median, 2 years) after pneumonectomy. This syndrome occurs in about 2% of pneumonectomies and results from extrinsic compression of the distal trachea or main bronchus by the vertebral body or aorta due to excessive movement and rotation of the mediastinum into the pneumonectomy space and hyperextension of the remaining lung and airway (16,17). Empyema in the postpneumonectomy space occurs at an incidence of 4% to 16% after pneumonectomy. Although most postpneumonectomy empyema has been thought to develop within 3 months of pneumonectomy, large case series studies found that the median interval between pneumonectomy and postpneumonectomy empyema was 3.4–4.3 months, ranging from 1–7 to 15.6–19.7 years (18,19).

Table 1

Literature review of more than 300 pneumonectomy cases published since 1990

Author Published year Mortality rate of pneumonectomy Risk factors for mortality from pneumonectomy
No. of patients 30-day (%) In-hospital or 90-day (%)
Romano (7) 1992 1,529 10.1 11.6 Age >60 years, chronic heart disease
Wada (8) 1998 586 3.2 NA NA
Harpole (9) 1999 567 11.5 NA NA
Bernard (10) 2001 639 NA 7.0 Lower preoperative Hb, hematologic disease
Watanabe (11) 2004 355 3.1 5.9 NA
Safi (12) 2015 774 NA 6.8 Current alcohol use, preoperative WBC >13,000/μL, CAD
Gu (13) 2017 406 3.2 6.2 NA
Yu (14) 2021 10,337 4.2 10.3 Age >40 years, male, right-sided pneumonectomy, SCC
Wang (15) 2022 1,052 NA 1.8 NA

NA, not available; Hb, hemoglobin; WBC, white blood cell count; CAD, coronary artery disease; SCC, squamous cell carcinoma.

Thus, in general, pneumonectomy has more postoperative complications than lobectomy, which raises some concerns about the interpretation of the results of the IMpower010 trial. The most serious concern is that these results were obtained from highly selected patients. The standard number of cycles of cisplatin-based chemotherapy in postoperative adjuvant therapy is four, but only half of patients actually complete four cycles of chemotherapy after surgical resection (20,21). In the IMpower010 trial, 264 of the 1,269 patients (21%) who started cisplatin-based chemotherapy dropped out of the study and were excluded from proceeding to atezolizumab treatment (4). As a result, patients in good enough general condition to receive four cycles of chemotherapy accounted for nearly 90% of those who received adjuvant atezolizumab therapy (6). The interval between surgery and the start of atezolizumab administration is also important. Most complications associated with pneumonectomy occur within the first 3 months after surgery. Therefore, careful monitoring of the postoperative clinical course during this period or longer can help select patients suitable for adjuvant therapy. The median time from surgery to atezolizumab adjuvant therapy in the IMpower010 trial was 5.1 months (6), which appears to have been a sufficient observation period to select appropriate patients. Another concern is the relatively short median observation period of 32.2 months (interquartile range, 27.4–38.3 months) for the evaluation of any difference in safety by surgical technique in IMpower010 trial (6). This is because a case was reported in which an immune-related AE occurred 58 months after the start of ICI administration (22), and in another case, an immune-related AE developed 28 months after discontinuation of ICI administration (23). Finally, the relatively small number of patients [100] makes it difficult to evaluate highly lethal immune-related AEs including myocarditis and toxic epidermal necrolysis, because the frequency of such immune-related AEs is less than 1% (24). Thus, to support use of adjuvant ICIs in patients undergoing pneumonectomy, real world data based on a sufficient number of patients with sufficiently long observation period are needed.


Acknowledgments

Funding: None.


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, AME Clinical Trials Review. The article has undergone external peer review.

Peer Review File: Available at https://actr.amegroups.com/article/view/10.21037/actr-23-16/prf

Conflicts of Interest: The author has completed the ICMJE uniform disclosure form (available at https://actr.amegroups.com/article/view/10.21037/actr-23-16/coif). The author has no conflicts of interest to declare.

Ethical Statement: The author is 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. Okami J, Shintani Y, Okumura M, et al. Demographics, Safety and Quality, and Prognostic Information in Both the Seventh and Eighth Editions of the TNM Classification in 18,973 Surgical Cases of the Japanese Joint Committee of Lung Cancer Registry Database in 2010. J Thorac Oncol 2019;14:212-22.
  2. West H, Hu X, Zhang S, et al. Treatment Patterns and Outcomes in Resected Early-stage Non-small Cell Lung Cancer: An Analysis of the SEER-Medicare Data. Clin Lung Cancer 2023;24:260-8. [Crossref] [PubMed]
  3. Arriagada R, Auperin A, Burdett S, et al. Adjuvant chemotherapy, with or without postoperative radiotherapy, in operable non-small-cell lung cancer: two meta-analyses of individual patient data. Lancet 2010;375:1267-77. [Crossref] [PubMed]
  4. Felip E, Altorki N, Zhou C, et al. Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase 3 trial. Lancet 2021;398:1344-57. [Crossref] [PubMed]
  5. Felip E, Altorki N, Zhou C, et al. Overall survival with adjuvant atezolizumab after chemotherapy in resected stage II-IIIA non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label, phase III trial. Ann Oncol 2023;34:907-19. [Crossref] [PubMed]
  6. Lee JM, Vallières E, Ding B, et al. Safety of adjuvant atezolizumab after pneumonectomy/bilobectomy in stage II-IIIA non-small cell lung cancer in the randomized phase III IMpower010 trial. J Thorac Cardiovasc Surg 2023;166:655-66.e7. [Crossref] [PubMed]
  7. Romano PS, Mark DH. Patient and hospital characteristics related to in-hospital mortality after lung cancer resection. Chest 1992;101:1332-7. [Crossref] [PubMed]
  8. Wada H, Nakamura T, Nakamoto K, et al. Thirty-day operative mortality for thoracotomy in lung cancer. J Thorac Cardiovasc Surg 1998;115:70-3. [Crossref] [PubMed]
  9. Harpole DH Jr, DeCamp MM Jr, Daley J, et al. Prognostic models of thirty-day mortality and morbidity after major pulmonary resection. J Thorac Cardiovasc Surg 1999;117:969-79. [Crossref] [PubMed]
  10. Bernard A, Deschamps C, Allen MS, et al. Pneumonectomy for malignant disease: factors affecting early morbidity and mortality. J Thorac Cardiovasc Surg 2001;121:1076-82. [Crossref] [PubMed]
  11. Watanabe S, Asamura H, Suzuki K, et al. Recent results of postoperative mortality for surgical resections in lung cancer. Ann Thorac Surg 2004;78:999-1002; discussion 1002-3. [Crossref] [PubMed]
  12. Safi S, Benner A, Walloschek J, et al. Development and validation of a risk score for predicting death after pneumonectomy. PLoS One 2015;10:e0121295. [Crossref] [PubMed]
  13. Gu C, Wang R, Pan X, et al. Comprehensive study of prognostic risk factors of patients underwent pneumonectomy. J Cancer 2017;8:2097-103. [Crossref] [PubMed]
  14. Yu X, Gao S, Xue Q, et al. Development of a nomogram for predicting the operative mortality of patients who underwent pneumonectomy for lung cancer: a population-based analysis. Transl Lung Cancer Res 2021;10:381-91. [Crossref] [PubMed]
  15. Wang C, Wang S, Li Z, et al. A Multiple-Center Nomogram to Predict Pneumonectomy Complication Risk for Non-Small Cell Lung Cancer Patients. Ann Surg Oncol 2022;29:561-9. [Crossref] [PubMed]
  16. Shen KR, Wain JC, Wright CD, et al. Postpneumonectomy syndrome: surgical management and long-term results. J Thorac Cardiovasc Surg 2008;135:1210-6; discussion 1216-9. [Crossref] [PubMed]
  17. Soll C, Hahnloser D, Frauenfelder T, et al. The postpneumonectomy syndrome: clinical presentation and treatment. Eur J Cardiothorac Surg 2009;35:319-24. [Crossref] [PubMed]
  18. Zaheer S, Allen MS, Cassivi SD, et al. Postpneumonectomy empyema: results after the Clagett procedure. Ann Thorac Surg 2006;82:279-86; discussion 286-7. [Crossref] [PubMed]
  19. Schneiter D, Grodzki T, Lardinois D, et al. Accelerated treatment of postpneumonectomy empyema: a binational long-term study. J Thorac Cardiovasc Surg 2008;136:179-85. [Crossref] [PubMed]
  20. Winton T, Livingston R, Johnson D, et al. Vinorelbine plus cisplatin vs. observation in resected non-small-cell lung cancer. N Engl J Med 2005;352:2589-97. [Crossref] [PubMed]
  21. Douillard JY, Rosell R, De Lena M, et al. Adjuvant vinorelbine plus cisplatin versus observation in patients with completely resected stage IB-IIIA non-small-cell lung cancer (Adjuvant Navelbine International Trialist Association [ANITA]): a randomised controlled trial. Lancet Oncol 2006;7:719-27. [Crossref] [PubMed]
  22. Owen CN, Bai X, Quah T, et al. Delayed immune-related adverse events with anti-PD-1-based immunotherapy in melanoma. Ann Oncol 2021;32:917-25. [Crossref] [PubMed]
  23. Couey MA, Bell RB, Patel AA, et al. Delayed immune-related events (DIRE) after discontinuation of immunotherapy: diagnostic hazard of autoimmunity at a distance. J Immunother Cancer 2019;7:165. [Crossref] [PubMed]
  24. Raschi E, Gatti M, Gelsomino F, et al. Lessons to be Learnt from Real-World Studies on Immune-Related Adverse Events with Checkpoint Inhibitors: A Clinical Perspective from Pharmacovigilance. Target Oncol 2020;15:449-66. [Crossref] [PubMed]
doi: 10.21037/actr-23-16
Cite this article as: Sekine I. Real world data are needed to establish safety of adjuvant immune checkpoint inhibitors after pneumonectomy. AME Clin Trials Rev 2023;1:7.

Download Citation