Toripalimab plus chemotherapy as second-line treatment in previously EGFR-TKI treated patients with EGFR-mutant-advanced NSCLC: a multicenter phase-II trial

Tao Jiang, Pingyang Wang, Jie Zhang, Yanqiu Zhao, Jianying Zhou, Yun Fan, Yongqian Shu, Xiaoqing Liu, Helong Zhang, Jianxing He, Guanghui Gao, Xiaoqian Mu, Zhang Bao, Yanjun Xu, Renhua Guo, Hong Wang, Lin Deng, Ningqiang Ma, Yalei Zhang, Hui Feng, Sheng Yao, Jiarui Wu, Luonan Chen, Caicun Zhou, Shengxiang Ren, Tao Jiang, Pingyang Wang, Jie Zhang, Yanqiu Zhao, Jianying Zhou, Yun Fan, Yongqian Shu, Xiaoqing Liu, Helong Zhang, Jianxing He, Guanghui Gao, Xiaoqian Mu, Zhang Bao, Yanjun Xu, Renhua Guo, Hong Wang, Lin Deng, Ningqiang Ma, Yalei Zhang, Hui Feng, Sheng Yao, Jiarui Wu, Luonan Chen, Caicun Zhou, Shengxiang Ren

Abstract

This multicenter phase-II trial aimed to investigate the efficacy, safety, and predictive biomarkers of toripalimab plus chemotherapy as second-line treatment in patients with EGFR-mutant-advanced NSCLC. Patients who failed from first-line EGFR-TKIs and did not harbor T790M mutation were enrolled. Toripalimab plus carboplatin and pemetrexed were administrated every three weeks for up to six cycles, followed by the maintenance of toripalimab and pemetrexed. The primary endpoint was objective-response rate (ORR). Integrated biomarker analysis of PD-L1 expression, tumor mutational burden (TMB), CD8 + tumor-infiltrating lymphocyte (TIL) density, whole-exome, and transcriptome sequencing on tumor biopsies were also conducted. Forty patients were enrolled with an overall ORR of 50.0% and disease-control rate (DCR) of 87.5%. The median progression free survival (PFS) and overall survival were 7.0 and 23.5 months, respectively. The most common treatment-related adverse effects were leukopenia, neutropenia, anemia, ALT/AST elevation, and nausea. Biomarker analysis showed that none of PD-L1 expression, TMB level, and CD8 + TIL density could serve as a predictive biomarker. Integrated analysis of whole-exome and transcriptome sequencing data revealed that patients with DSPP mutation had a decreased M2 macrophage infiltration and associated with longer PFS than those of wild type. Toripalimab plus chemotherapy showed a promising anti-tumor activity with acceptable safety profiles as the second-line setting in patients with EGFR-mutant NSCLC. DSPP mutation might serve as a potential biomarker for this combination. A phase-III trial to compare toripalimab versus placebo in combination with chemotherapy in this setting is ongoing (NCT03924050).

Conflict of interest statement

Honoraria to Dr. Caicun Zhou: Lilly, Roche, Boehringer Ingelheim, MSD, Hengrui Therapeutics, and QiLu Pharmaceutical; Consulting or advisory role: AmoyDx, Hengrui Therapeutics, Inovent, and QiLu Pharmaceutical. Other authors declared no potential conflicts of interest.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
CONSORT diagram for this phase-II study
Fig. 2
Fig. 2
Summary of treatment response, progression-free and overall survival. a Maximal change of tumor size from baseline assessed by the investigator per RECIST v1.1 (n = 38). The length of the bar represents maximal decrease or minimal increase in target lesion(s). # Unconfirmed partial response classified as stable disease. * Patient with target lesion(s) reduction over 30% but with new lesion(s) or progression of nontarget lesion(s). b Change of individual tumor burden over time from baseline assessed by the investigator per RECIST v1.1 (n = 38). c Progression-free survival by RECIST v1.1 of all 40 enrolled patients. d Overall survival by RECIST v1.1 of all 40 enrolled patients. No. number
Fig. 3
Fig. 3
Whole-exome sequencing. a Highlight mutated genes color-coded by the type of mutations in experimental samples. b According to the P value of PFS between groups, the top three combinations with different gene numbers were selected. The red line in the left panel shows the threshold of the filter. The white number on the barplot in the right panel represents the proportion of the corresponding sample in the total population. c Progression-free survival in patients with DSPP + TP53 comutation versus wild type (log-rank test). d Immune infiltration of DSPP + TP53 comutation versus wild type from EGFR-mutant samples obtained from TCGA (Wilcoxon signed-rank test). e Progression-free survival in patients with DSPP mutation versus wild type (log-rank test). f Immune infiltration of DSPP mutation versus wild type from EGFR mutant samples obtained from TCGA (Wilcoxon signed-rank test). PFS progression free survival; HR hazard ratio; *P < .05, **P < .01, ***P < .001, ****P < .0001
Fig. 4
Fig. 4
Integrated analysis of whole exome and transcriptome sequencing. a Sample correlation heat map based on expression matrix; different colors represent different correlation coefficients. b Volcano plot of eight upregulated and five downregulated differentially expressed genes between PR and non-PR group, each of them labeled the names of the first five genes. c Immune analysis of PR versus non-PR group from our cohort (Wilcoxon signed-rank test). d M1/M2 macrophage ratio of PR versus non-PR from our cohort (Wilcoxon signed-rank test). e M1/M2 macrophage ratio of DSPP-mutant versus wild-type tumors from EGFR-mutant samples obtained from TCGA (Wilcoxon signed-rank test). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001

References

    1. Passaro A, Jänne PA, Mok T, Peters S. Overcoming therapy resistance in EGFR-mutant lung cancer. Nat. Cancer. 2021;2:377–391. doi: 10.1038/s43018-021-00195-8.
    1. Yuan M, Huang LL, Chen JH, Wu J, Xu Q. The emerging treatment landscape of targeted therapy in non-small-cell lung cancer. Signal Transduct. Target Ther. 2019;4:61. doi: 10.1038/s41392-019-0099-9.
    1. Zhou C, et al. Erlotinib versus chemotherapy as first-line treatment for patients with advanced EGFR mutation-positive non-small-cell lung cancer (OPTIMAL, CTONG-0802): a multicentre, open-label, randomised, phase 3 study. Lancet Oncol. 2011;12:735–742. doi: 10.1016/S1470-2045(11)70184-X.
    1. Mitsudomi T, et al. Gefitinib versus cisplatin plus docetaxel in patients with non-small-cell lung cancer harbouring mutations of the epidermal growth factor receptor (WJTOG3405): an open label, randomised phase 3 trial. Lancet Oncol. 2010;11:121–128. doi: 10.1016/S1470-2045(09)70364-X.
    1. Rosell 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;13:239–246. doi: 10.1016/S1470-2045(11)70393-X.
    1. Mok TS, et al. Gefitinib or carboplatin-paclitaxel in pulmonary adenocarcinoma. N. Engl. J. Med. 2009;361:947–957. doi: 10.1056/NEJMoa0810699.
    1. Yang JC, et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol. 2015;16:141–151. doi: 10.1016/S1470-2045(14)71173-8.
    1. Wu YL, et al. Dacomitinib versus gefitinib as first-line treatment for patients with EGFR-mutation-positive non-small-cell lung cancer (ARCHER 1050): a randomised, open-label, phase 3 trial. Lancet Oncol. 2017;18:1454–1466. doi: 10.1016/S1470-2045(17)30608-3.
    1. Soria JC, et al. Osimertinib in untreated EGFR-mutated advanced non-small-cell lung cancer. N. Engl. J. Med. 2018;378:113–125. doi: 10.1056/NEJMoa1713137.
    1. Mok TS, et al. Osimertinib or platinum-pemetrexed in EGFR T790M-positive lung cancer. N. Engl. J. Med. 2017;376:629–640. doi: 10.1056/NEJMoa1612674.
    1. Soria JC, et al. Gefitinib plus chemotherapy versus placebo plus chemotherapy in EGFR-mutation-positive non-small-cell lung cancer after progression on first-line gefitinib (IMPRESS): a phase 3 randomised trial. Lancet Oncol. 2015;16:990–998. doi: 10.1016/S1470-2045(15)00121-7.
    1. Nishimura T, et al. Second-line therapy with first- or second-generation tyrosine kinase inhibitors in EGFR-mutated non-small cell lung cancer patients with T790M-negative or unidentified mutation. Thorac. Cancer. 2021;12:1067–1073. doi: 10.1111/1759-7714.13870.
    1. Oxnard GR, et al. Assessment of resistance mechanisms and clinical implications in patients with EGFR T790M-positive lung cancer and acquired resistance to osimertinib. JAMA Oncol. 2018;4:1527–1534. doi: 10.1001/jamaoncol.2018.2969.
    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. Fehrenbacher L, et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): a multicentre, open-label, phase 2 randomised controlled trial. Lancet. 2016;387:1837–1846. doi: 10.1016/S0140-6736(16)00587-0.
    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. 2016;387:1540–1550. doi: 10.1016/S0140-6736(15)01281-7.
    1. Reck M, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N. Engl. J. Med. 2016;375:1823–1833. doi: 10.1056/NEJMoa1606774.
    1. Herbst RS, et al. Atezolizumab for first-line treatment of PD-L1-selected patients with NSCLC. N. Engl. J. Med. 2020;383:1328–1339. doi: 10.1056/NEJMoa1917346.
    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. Mazieres J, et al. Immune checkpoint inhibitors for patients with advanced lung cancer and oncogenic driver alterations: results from the IMMUNOTARGET registry. Ann. Oncol. 2019;30:1321–1328. doi: 10.1093/annonc/mdz167.
    1. Qiao M, et al. Immune checkpoint inhibitors in EGFR-mutated non-small cell lung cancer: dusk or dawn? J. Thorac. Oncol. 2021;16:1267–1288. doi: 10.1016/j.jtho.2021.04.003.
    1. Oxnard GR, et al. TATTON: a multi-arm, phase Ib trial of osimertinib combined with selumetinib, savolitinib, or durvalumab in EGFR-mutant lung cancer. Ann. Oncol. 2020;31:507–516. doi: 10.1016/j.annonc.2020.01.013.
    1. Oshima Y, Tanimoto T, Yuji K, Tojo A. EGFR-TKI-associated interstitial pneumonitis in nivolumab-treated patients with non-small cell lung cancer. JAMA Oncol. 2018;4:1112–1115. doi: 10.1001/jamaoncol.2017.4526.
    1. Dong ZY, et al. EGFR mutation correlates with uninflamed phenotype and weak immunogenicity, causing impaired response to PD-1 blockade in non-small cell lung cancer. Oncoimmunology. 2017;6:e1356145. doi: 10.1080/2162402X.2017.1356145.
    1. Rottenberg S, Disler C, Perego P. The rediscovery of platinum-based cancer therapy. Nat. Rev. Cancer. 2021;21:37–50. doi: 10.1038/s41568-020-00308-y.
    1. Petroni G, Buque A, Zitvogel L, Kroemer G, Galluzzi L. Immunomodulation by targeted anticancer agents. Cancer Cell. 2021;39:310–345. doi: 10.1016/j.ccell.2020.11.009.
    1. Reck M, et al. Atezolizumab plus bevacizumab and chemotherapy in non-small-cell lung cancer (IMpower150): key subgroup analyses of patients with EGFR mutations or baseline liver metastases in a randomised, open-label phase 3 trial. Lancet. Respir. Med. 2019;7:387–401.
    1. Wang FH, et al. Efficacy, safety, and correlative biomarkers of toripalimab in previously treated recurrent or metastatic nasopharyngeal carcinoma: a phase II clinical trial (POLARIS-02) J. Clin. Oncol. 2021;39:704–712. doi: 10.1200/JCO.20.02712.
    1. Wang Z, et al. Safety, antitumor activity, and pharmacokinetics of toripalimab, a programmed cell death 1 inhibitor, in patients with advanced non-small cell lung cancer: a phase 1 trial. JAMA Netw. Open. 2020;3:e2013770. doi: 10.1001/jamanetworkopen.2020.13770.
    1. Yang J, et al. Safety and clinical efficacy of toripalimab, a PD-1 mAb, in patients with advanced or recurrent malignancies in a phase I study. Eur. J. Cancer. 2020;130:182–192. doi: 10.1016/j.ejca.2020.01.028.
    1. Mayakonda A, Lin DC, Assenov Y, Plass C, Koeffler HP. Maftools: efficient and comprehensive analysis of somatic variants in cancer. Genome Res. 2018;28:1747–1756. doi: 10.1101/gr.239244.118.
    1. Duan Z, Luo Y. Targeting macrophages in cancer immunotherapy. Signal Transduct. Target Ther. 2021;6:127. doi: 10.1038/s41392-021-00506-6.
    1. Salas-Benito, D. et al. Paradigms on immunotherapy combinations with chemotherapy. Cancer Discov., 10.1158/-20-1312 (2021).
    1. Huang MY, Jiang XM, Wang BL, Sun Y, Lu JJ. Combination therapy with PD-1/PD-L1 blockade in non-small cell lung cancer: strategies and mechanisms. Pharm. Ther. 2021;219:107694. doi: 10.1016/j.pharmthera.2020.107694.
    1. Lisberg A, et al. A phase II study of pembrolizumab in EGFR-mutant, PD-L1+, tyrosine kinase inhibitor naive patients with advanced NSCLC. J. Thorac. Oncol. 2018;13:1138–1145. doi: 10.1016/j.jtho.2018.03.035.
    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. 2016;22:4585–4593. doi: 10.1158/1078-0432.CCR-15-3101.
    1. Fuentes-Antras J, Provencio M, Diaz-Rubio E. Hyperprogression as a distinct outcome after immunotherapy. Cancer Treat. Rev. 2018;70:16–21. doi: 10.1016/j.ctrv.2018.07.006.
    1. Mok TSK, et al. Gefitinib plus chemotherapy versus chemotherapy in epidermal growth factor receptor mutation-positive non-small-cell lung cancer resistant to first-line gefitinib (IMPRESS): overall survival and biomarker analyses. J. Clin. Oncol. 2017;35:4027–4034. doi: 10.1200/JCO.2017.73.9250.
    1. Zhou J, et al. An advanced non-small cell lung cancer patient with epidermal growth factor receptor sensitizing mutation responded to toripalimab in combination with chemotherapy after resistance to osimertinib: a case report. Transl. Lung Cancer Res. 2020;9:354–359. doi: 10.21037/tlcr.2020.02.09.
    1. van der Leun AM, Thommen DS, Schumacher TN. CD8(+) T cell states in human cancer: insights from single-cell analysis. Nat. Rev. Cancer. 2020;20:218–232. doi: 10.1038/s41568-019-0235-4.
    1. D’Souza WN, Chang CF, Fischer AM, Li M, Hedrick SM. The Erk2 MAPK regulates CD8 T cell proliferation and survival. J. Immunol. 2008;181:7617–7629. doi: 10.4049/jimmunol.181.11.7617.
    1. Ebert PJR, et al. MAP kinase inhibition promotes T cell and anti-tumor activity in combination with PD-L1 checkpoint blockade. Immunity. 2016;44:609–621. doi: 10.1016/j.immuni.2016.01.024.
    1. Tokito T, et al. Predictive relevance of PD-L1 expression combined with CD8+ TIL density in stage III non-small cell lung cancer patients receiving concurrent chemoradiotherapy. Eur. J. Cancer. 2016;55:7–14. doi: 10.1016/j.ejca.2015.11.020.
    1. Yang H, et al. Prognostic value of PD-L1 expression in combination with CD8(+) TILs density in patients with surgically resected non-small cell lung cancer. Cancer Med. 2018;7:32–45. doi: 10.1002/cam4.1243.
    1. Jiang T, et al. Genomic landscape and its correlations with tumor mutational burden, PD-L1 expression, and immune cells infiltration in Chinese lung squamous cell carcinoma. J. Hematol. Oncol. 2019;12:75. doi: 10.1186/s13045-019-0762-1.

Source: PubMed

3
S'abonner