Induction immune-checkpoint inhibitors for resectable oncogene-mutant NSCLC: A multicenter pooled analysis

Chao Zhang, Hua-Fei Chen, Shi Yan, Lin Wu, Li-Xu Yan, Xiao-Long Yan, Dong-Sheng Yue, Chun-Wei Xu, Min Zheng, Ji-Sheng Li, Si-Yang Liu, Ling-Ling Yang, Ben-Yuan Jiang, Qiu-Xiang Ou, Zhen-Bin Qiu, Yang Shao, Yi-Long Wu, Wen-Zhao Zhong, Chao Zhang, Hua-Fei Chen, Shi Yan, Lin Wu, Li-Xu Yan, Xiao-Long Yan, Dong-Sheng Yue, Chun-Wei Xu, Min Zheng, Ji-Sheng Li, Si-Yang Liu, Ling-Ling Yang, Ben-Yuan Jiang, Qiu-Xiang Ou, Zhen-Bin Qiu, Yang Shao, Yi-Long Wu, Wen-Zhao Zhong

Abstract

Despite limited efficacy of immunotherapy for advanced non-small-cell lung cancer (NSCLC) with driver mutations, whether neoadjuvant immunotherapy could be clinically valuable in those patients warrants further investigation. We utilized 40 oncogene-mutant NSCLC treated with induction immunotherapy from a large consecutive multicenter cohort. Overall response rate was 62.5% while 2 patients had disease progression. Of 39 patients that received surgery, R0 resection rate was 97.4%. The major pathological response (MPR) rate was 37.5% and the pathological complete response (pCR) rate was 12.5%. Pre-treatment PD-L1 expression was not a predictive biomarker in these patients. Median disease-free survival for all oncogenic mutation and EGFR mutation was 28.5 months. Indirect comparison through integrating CTONG1103 cohort showed neoadjuvant immunotherapy plus chemotherapy yielded the most superior efficacy among erlotinib and chemotherapy for resectable EGFR-mutant NSCLC. No MPR patients were identified with neoadjuvant immunotherapy plus chemotherapy for uncommon EGFR insertion or point mutations. Our results indicated the potential clinical feasibility of neoadjuvant immunotherapy for resectable localized oncogene-mutant NSCLC especially for EGFR-mutant NSCLC.

Conflict of interest statement

W.-Z.Z. received speech honoraria from AstraZeneca, Roche, Eli Lilly, and Pfizer. Y.-L.W. received research funding from Roche and speech honoraria from AstraZeneca, Roche, Eli Lilly, Pfizer and Sanofi, and he was a research consultant for AstraZeneca. L.W. and D.S.Y. received speech honoraria from AstraZeneca and Roche. L.-L.Y., Q.-X.O., and Y.S. are employees of Nanjing Geneseeq Technology Inc. The remaining authors declare no conflict of interest.

© 2022. The Author(s).

Figures

Fig. 1. Overview of clinicopathological characteristics and…
Fig. 1. Overview of clinicopathological characteristics and clinical outcome of oncogene-mutant NSCLC treated with neoadjuvant immunotherapy.
a Overview of clinicopathological charteristics and clinical outcome of oncogene-mutant NSCLC treated with neoadjuvant immunotherapy. Clinicopathological charateristics including smoking status, therpeutic regimens, PD-L1 expression, treatment duration and preoperative radiologic response (according to Response Evaluation Criteria in Solid Tumors [RECIST]) were annotated for each patients. The horizontal dash line indicates the threshold for a major pathological response (90% regression). Pathological regression was presented in colored bar plot regarding different oncogene mutation. b Correlation between radiological shrinkage and pathological regression. Spearman correlation was used to measure the relevance. c Association between PD-L1 expression and MPR status. Centre line represents the median value of PD-L1 expression. Error bars represent the upper and lower quartiles of PD-L1 expression and whiskers defines the minimal and maximun value. PD Progression disease, SD Stable disease, PR Partial response, MPR Major pathological response, pCR Pathological complete response. d Disease-free survival (DFS) of all patients, EGFR-mutant patients and KRAS-mutant patients treated with neoadjuvant immunotherapy through Kaplan-Meier survival analysis.
Fig. 2. Efficacy of neoadjuvant treatments for…
Fig. 2. Efficacy of neoadjuvant treatments for EGFR-mutant NSCLC.
Comparison of different treatment modalities for resectable EGFR-mutant NSCLC in regard to radiological response and pathological assessment by integrating chemotherapy and EGFR-TKI cohorts from CTONG1103 study. ADC Adenocarcinoma, SQC Squamous cell carcinoma, SD Stable disease, PR Partial response, PD Progression disease, CR Compelete response, MPR Major pathological response, pCR P athological complete response.
Fig. 3. Overview of infiltrating immune contexture…
Fig. 3. Overview of infiltrating immune contexture and genomic profile of resected PL and DLNs after neoadjuvant immunotherapy.
a Non-supervised clustering of infiltrating immune microenviornment among PL and DLNs through ImmuCellAI. Pie chart indicates the proportion of samples which are antipated as responsive to immunotherapy. R Responder, NR Non-responder. b Comparison of specific immune cell subtypes among different patients. Kruskal-Wallis test was used to testify the difference among patients. Centre dots represent the median value of specific immune cell infiltration. Error bars represent the upper and lower quartiles and whiskers defines the range within 1.5IQR. c Radiological evaluation before and after neoadjuvant immunotherapy for PT4. d Genomic evolutionary trajectory between PL and DLNs of PT4. e Indel- and SNV-induced neoantigens burden among PL and DLNs through NeoPredPipe.
Fig. 4. Immune phenotypes and enriched functional…
Fig. 4. Immune phenotypes and enriched functional pathway among PLs and DLNs.
a Expression profile of functional genesets including antigen presenting cell abundece, T/NK cell abundence, IFN activity and T cell exhasution were presented through z-score. b Radar plots of comprehensive immune phenotypes including PD-L1 expression in PL, T-cell inflamed GEP and suppressor cell score in PL and DLNs were scaled into 0–100%. T_PD-L1 represents expression of PD-L1 in tumor; T_GEP represents gene-expression profile of infiltrating lymphocytes in tumor; T_SC represents suppressor cell in tumor; LN_GEP represents gene-expression profile of infiltrating lymphocytes in draining lymph nodes; LN_SC represents suppressor cell in draining lymph nodes. c Gene set enrichment analysis (GSEA) of PL and DLNs upon responders and non-responders.

References

    1. Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science (N.Y., NY) 2018;359:1350–1355. doi: 10.1126/science.aar4060.
    1. Hargadon KM, Johnson CE, Williams CJ. Immune checkpoint blockade therapy for cancer: An overview of FDA-approved immune checkpoint inhibitors. Int. Immunopharmacol. 2018;62:29–39. doi: 10.1016/j.intimp.2018.06.001.
    1. Postow MA, Callahan MK, Wolchok JD. Immune checkpoint blockade in cancer therapy. J. Clin. Oncol.: Off. J. Am. Soc. Clin. Oncol. 2015;33:1974–1982. doi: 10.1200/JCO.2014.59.4358.
    1. Garassino MC, et al. Durvalumab as third-line or later treatment for advanced non-small-cell lung cancer (ATLANTIC): An open-label, single-arm, phase 2 study. Lancet Oncol. 2018;19:521–536. doi: 10.1016/S1470-2045(18)30144-X.
    1. Mazieres J, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann. Oncol.: Off. J. Eur. Soc. Med. Oncol. 2019;30:1321–1328. doi: 10.1093/annonc/mdz167.
    1. Lee CK, et al. Checkpoint inhibitors in metastatic EGFR-mutated non-small cell lung cancer-a meta-analysis. J. Thorac. Oncol.: Off. Publ. Int. Assoc. Study Lung Cancer. 2017;12:403–407. doi: 10.1016/j.jtho.2016.10.007.
    1. Ng TL, et al. Predictive value of oncogenic driver subtype, programmed death-1 ligand (PD-L1) score, and smoking status on the efficacy of PD-1/PD-L1 inhibitors in patients with oncogene-driven non-small cell lung cancer. Cancer. 2019;125:1038–1049. doi: 10.1002/cncr.31871.
    1. Gainor JF, et al. EGFR Mutations and ALK Rearrangements Are Associated with Low Response Rates to PD-1 Pathway Blockade in Non-Small Cell Lung Cancer: A Retrospective Analysis. Clin. Cancer Res.: Off. J. Am. Assoc. Cancer Res. 2016;22:4585–4593. doi: 10.1158/1078-0432.CCR-15-3101.
    1. Lin A, et al. Role of the dynamic tumor microenvironment in controversies regarding immune checkpoint inhibitors for the treatment of non-small cell lung cancer (NSCLC) with EGFR mutations. Mol. Cancer. 2019;18:139. doi: 10.1186/s12943-019-1062-7.
    1. Socinski, M. A. et al. Abstract CT216: IMpower150 final analysis: Efficacy of atezolizumab (atezo) + bevacizumab (bev) and chemotherapy in first-line (1L) metastatic nonsquamous (nsq) non-small cell lung cancer (NSCLC) across key subgroups. 80, CT216-CT216 (2020).
    1. Nagai K, et al. A randomized trial comparing induction chemotherapy followed by surgery with surgery alone for patients with stage IIIA N2 non-small cell lung cancer (JCOG 9209) J. Thorac. Cardiovascular Surg. 2003;125:254–260. doi: 10.1067/mtc.2003.15.
    1. Blumenthal GM, et al. Current Status and Future Perspectives on Neoadjuvant Therapy in Lung Cancer. J. Thorac. Oncol.: Off. Publ. Int. Assoc. Study Lung Cancer. 2018;13:1818–1831. doi: 10.1016/j.jtho.2018.09.017.
    1. Detterbeck FC, et al. Neoadjuvant chemotherapy with gemcitabine-containing regimens in patients with early-stage non-small cell lung cancer. J. Thorac. Oncol.: Off. Publ. Int. Assoc. Study Lung Cancer. 2008;3:37–45. doi: 10.1097/JTO.0b013e31815e5d9a.
    1. Gao S, et al. Neoadjuvant PD-1 inhibitor (Sintilimab) in NSCLC. J. Thorac. Oncol.: Off. Publ. Int. Assoc. Study Lung Cancer. 2020;15:816–826. doi: 10.1016/j.jtho.2020.01.017.
    1. Forde PM, et al. Neoadjuvant PD-1 Blockade in Resectable Lung Cancer. N. Engl. J. Med. 2018;378:1976–1986. doi: 10.1056/NEJMoa1716078.
    1. Shu, C. A. et al. Neoadjuvant atezolizumab and chemotherapy in patients with resectable non-small-cell lung cancer: an open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol.21, 786–795 (2020).
    1. Zhong, W. Z. 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. Clinical Oncol.: Off. J. American Soc. Clinical Oncol. 2019:Jco1900075.
    1. Jiang SX, et al. EGFR genetic heterogeneity of nonsmall cell lung cancers contributing to acquired gefitinib resistance. Int. J. Cancer. 2008;123:2480–2486. doi: 10.1002/ijc.23868.
    1. Dong ZY, et al. Mixed responses to systemic therapy revealed potential genetic heterogeneity and poor survival in patients with non-small cell lung cancer. Oncologist. 2017;22:61–69. doi: 10.1634/theoncologist.2016-0150.
    1. Miao YR, et al. ImmuCellAI: A unique method for comprehensive T-Cell subsets abundance prediction and its application in cancer immunotherapy. Adv. Sci. (Weinh., Baden.-Wurtt., Ger.) 2020;7:1902880.
    1. Inoue Y, et al. Evaluation of Programmed Death Ligand 1 (PD-L1) gene amplification and response to nivolumab monotherapy in non-small cell lung cancer. JAMA Netw. open. 2020;3:e2011818. doi: 10.1001/jamanetworkopen.2020.11818.
    1. Kato S, et al. Hyperprogressors after Immunotherapy: Analysis of genomic alterations associated with accelerated growth rate. Clin. Cancer Res.: Off. J. Am. Assoc. Cancer Res. 2017;23:4242–4250. doi: 10.1158/1078-0432.CCR-16-3133.
    1. Schenck RO, et al. NeoPredPipe: High-throughput neoantigen prediction and recognition potential pipeline. BMC Bioinforma. 2019;20:264. doi: 10.1186/s12859-019-2876-4.
    1. Ayers M, et al. IFN-γ-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Investig. 2017;127:2930–2940. doi: 10.1172/JCI91190.
    1. Charoentong P, et al. Pan-cancer Immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep. 2017;18:248–262. doi: 10.1016/j.celrep.2016.12.019.
    1. Forde, P. M. et al. Neoadjuvant Nivolumab plus Chemotherapy in Resectable Lung Cancer. New England J. Med.386,1973–1985 (2022).
    1. Spicer, J. Surgical outcomes from the phase 3 CheckMate 816 trial: Nivolumab (NIVO) + platinum-doublet chemotherapy (chemo) vs chemo alone as neoadjuvant treatment for patients with resectable non-small cell lung cancer (NSCLC). In: Jonathan Spicer CWFTGBSK-NCMLEEVNGSLMPTMMMAEF, ed.ASCO Annual Meeting: American Soc. Clinical Oncol.; 2021 Available at .
    1. Uprety D, et al. Neoadjuvant Immunotherapy for NSCLC: Current Concepts and Future Approaches. J. Thorac. Oncol.: Off. Publ. Int. Assoc. Study Lung Cancer. 2020;15:1281–1297. doi: 10.1016/j.jtho.2020.05.020.
    1. Zhou, N. et al. Impact of genomic aberrations and additional therapies on survival outcomes of patients with operable non-small cell lung cancer (NSCLC) from the NEOSTAR study. J. Clinical Oncol.39, 8542–8542 (2021).
    1. Provencio M, et al. Neoadjuvant chemotherapy and nivolumab in resectable non-small-cell lung cancer (NADIM): An open-label, multicentre, single-arm, phase 2 trial. Lancet Oncol. 2020;21:1413–1422. doi: 10.1016/S1470-2045(20)30453-8.
    1. Skoulidis F, Heymach JV. Co-occurring genomic alterations in non-small-cell lung cancer biology and therapy. Nat. Rev. Cancer. 2019;19:495–509. doi: 10.1038/s41568-019-0179-8.
    1. Herbst RS, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet (Lond., Engl.) 2016;387:1540–1550. doi: 10.1016/S0140-6736(15)01281-7.
    1. Borghaei H, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N. Engl. J. Med. 2015;373:1627–1639. doi: 10.1056/NEJMoa1507643.
    1. Brahmer J, et al. Nivolumab versus docetaxel in advanced squamous-cell non-small-cell lung cancer. N. Engl. J. Med. 2015;373:123–135. doi: 10.1056/NEJMoa1504627.
    1. Rothschild, S. I. et al. SAKK 16/14: Durvalumab in Addition to Neoadjuvant Chemotherapy in Patients With Stage IIIA(N2) Non-Small-Cell Lung Cancer-A Multicenter Single-Arm Phase II Trial. J. Clinical Oncol.: Off. J. American Soc. Clinical Oncol. 2021:Jco2100276.
    1. Travis WD, et al. IASLC multidisciplinary recommendations for pathologic assessment of lung cancer resection specimens after neoadjuvant therapy. J. Thorac. Oncol.: Off. Publ. Int. Assoc. Study Lung Cancer. 2020;15:709–740. doi: 10.1016/j.jtho.2020.01.005.
    1. Li H, et al. The Sequence Alignment/Map format and SAMtools. Bioinforma. (Oxf., Engl.) 2009;25:2078–2079. doi: 10.1093/bioinformatics/btp352.
    1. Liu, M. et al. MesKit: A tool kit for dissecting cancer evolution of multi-region tumor biopsies through somatic alterations. GigaScience10, giab036 (2021).

Source: PubMed

3
Suscribir