Clinical and biomarker analyses of sintilimab versus chemotherapy as second-line therapy for advanced or metastatic esophageal squamous cell carcinoma: a randomized, open-label phase 2 study (ORIENT-2)

Jianming Xu, Yi Li, Qingxia Fan, Yongqian Shu, Lei Yang, Tongjian Cui, Kangsheng Gu, Min Tao, Xiuwen Wang, Chengxu Cui, Nong Xu, Juxiang Xiao, Quanli Gao, Yunpeng Liu, Tao Zhang, Yuxian Bai, Wei Li, Yiping Zhang, Guanghai Dai, Dong Ma, Jingdong Zhang, Chunmei Bai, Yunchao Huang, Wangjun Liao, Lin Wu, Xi Chen, Yan Yang, Junye Wang, Shoujian Ji, Hui Zhou, Yan Wang, Zhuo Ma, Yanqi Wang, Bo Peng, Jiya Sun, Christoph Mancao, Jianming Xu, Yi Li, Qingxia Fan, Yongqian Shu, Lei Yang, Tongjian Cui, Kangsheng Gu, Min Tao, Xiuwen Wang, Chengxu Cui, Nong Xu, Juxiang Xiao, Quanli Gao, Yunpeng Liu, Tao Zhang, Yuxian Bai, Wei Li, Yiping Zhang, Guanghai Dai, Dong Ma, Jingdong Zhang, Chunmei Bai, Yunchao Huang, Wangjun Liao, Lin Wu, Xi Chen, Yan Yang, Junye Wang, Shoujian Ji, Hui Zhou, Yan Wang, Zhuo Ma, Yanqi Wang, Bo Peng, Jiya Sun, Christoph Mancao

Abstract

This randomized, open-label, multi-center phase 2 study (NCT03116152) assessed sintilimab, a PD-1 inhibitor, versus chemotherapy in patients with esophageal squamous cell carcinoma after first-line chemotherapy. The primary endpoint was overall survival (OS), while exploratory endpoint was the association of biomarkers with efficacy. The median OS in the sintilimab group was significantly improved compared with the chemotherapy group (median OS 7.2 vs.6.2 months; P = 0.032; HR = 0.70; 95% CI, 0.50-0.97). Incidence of treatment-related adverse events of grade 3-5 was lower with sintilimab than with chemotherapy (20.2 vs. 39.1%). Patients with high T-cell receptor (TCR) clonality and low molecular tumor burden index (mTBI) showed the longest median OS (15.0 months). Patients with NLR < 3 at 6 weeks post-treatment had a significantly prolonged median OS (16.6 months) compared with NLR ≥ 3. The results demonstrate a significant improvement in OS of sintilimab compared to chemotherapy as second-line treatment for advanced or metastatic ESCC.

Conflict of interest statement

H.Z., Y.W., Z.M., Y.Q.W., B.P., J.Y.S., and C.M. are the staff of Innovent Biologics, Inc. Other authors declare no completing interests.

© 2022. The Author(s).

Figures

Fig. 1. CONSORT diagram of patient flow.
Fig. 1. CONSORT diagram of patient flow.
This figure shows reasons for exclusion from the study and the numbers of patients included in the analyses. †, unqualified pathological type; *, patients refused to receive treatment, but received follow-up. Treatment discontinuation occurred due to prespecified conditions such as serious protocol deviation, using prohibited drug in the study, loss of follow-up, life-threatening adverse events and other unacceptable toxicities.
Fig. 2. Kaplan–Meier plots of survival.
Fig. 2. Kaplan–Meier plots of survival.
a Overall survival in the ITT population; b Progression free survival in the ITT population. c Overall survival in the continuous and non-continuous subgroups of the sintilimab group. Log-rank test stratified by Eastern Cooperative Oncology Group performance-status (ECOG PS) score was used (2-sided).
Fig. 3. Forest plot for subgroup analyses…
Fig. 3. Forest plot for subgroup analyses of overall survival.
Dots represent the cohort-specific hazard ratios with error bars corresponding to 95% CI bounds, which were calculated by using the univariate Cox regression model. ECOG PS Eastern Cooperative Oncology Group performance status, HR hazard ratio, CI confidence interval.
Fig. 4. Kaplan–Meier plots of survival in…
Fig. 4. Kaplan–Meier plots of survival in high and low neutrophil-to-lymphocyte ratio (NLR) subgroups of the sintilimab group.
a Overall survival with NLR at baseline. b Progression-free survival with NLR at baseline. c Overall survival with NLR at 6 weeks post treatment. d Progression-free survival with NLR at 6 weeks post treatment. HR hazard ratio. P-values were based on a two-sided log-rank test.
Fig. 5. Heatmap and forest plots of…
Fig. 5. Heatmap and forest plots of the association of tumor microenvironment immune-cell signatures and progression-free survival.
Heat map of immune score of 28 immune-cell populations is shown and forest plots (right panel) displaythe correlation of each immune subtype with progression-free survival. Node position reflects hazard ratio (p-value (the larger the node size, the more significant). P-values were based on a two-sided Wald test. Source data are provided as a Source Data file.
Fig. 6. Kaplan–Meier plots of survival in…
Fig. 6. Kaplan–Meier plots of survival in different TCR clonality and mTBI subgroups of the sintilimab group.
a Overall survival. b Progression-free survival. The high- or low- level groups of TCR clonality or mTBI are split by the respective median value. Group A: high TCR clonality and high mTBI; Group B: high TCR clonality and low mTBI; Group C: low TCR clonality and high mTBI; Group D: low TCR clonality and low mTBI. P-values were based on a two-sided Wald test. Source data are provided as a Source Data file.

References

    1. Sung H, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2021;71:209–249.
    1. Arnold M, Ferlay J, van Berge Henegouwen MI, Soerjomataram I. Global burden of oesophageal and gastric cancer by histology and subsite in 2018. Gut. 2020;69:1564–1571.
    1. The global, regional, and national burden of oesophageal cancer and its attributable risk factors in 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol. Hepatol.5, 582–597 (2020).
    1. Jiao R, et al. Immune checkpoint inhibitors in esophageal squamous cell carcinoma: progress and opportunities. Onco. Targets Ther. 2019;12:6023–6032.
    1. Lordick F, et al. Oesophageal cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2016;27:v50–v57.
    1. Muro K, et al. Pan-Asian adapted ESMO clinical practice guidelines for the management of patients with metastatic oesophageal cancer: a JSMO-ESMO initiative endorsed by CSCO, KSMO, MOS, SSO and TOS. Ann. Oncol. 2019;30:34–43.
    1. National Comprehensive Cancer Network (NCCN). NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines). Esophageal and esophagogastric junction cancers. Version 2 (2021).
    1. Kitagawa Y, et al. Esophageal Cancer Practice Guidelines 2017 edited by the Japan Esophageal Society: part 2. Esophagus. 2019;16:25–43.
    1. Jimenez P, et al. The role of taxanes in the management of gastroesphageal cancer. J. Gastrointest. Oncol. 2011;2:240–249.
    1. Kato K, et al. A phase II study of paclitaxel by weekly 1-h infusion for advanced or recurrent esophageal cancer in patients who had previously received platinum-based chemotherapy. Cancer Chemother. Pharmacol. 2011;67:1265–1272.
    1. Muro K, et al. A phase II study of single-agent docetaxel in patients with metastatic esophageal cancer. Ann. Oncol. 2004;15:955–959.
    1. Kojima T, et al. Randomized phase III KEYNOTE-181 study of pembrolizumab versus chemotherapy in advanced esophageal cancer. J. Clin. Oncol. 2020;38:4138–4148.
    1. Kato K, et al. Nivolumab versus chemotherapy in patients with advanced oesophageal squamous cell carcinoma refractory or intolerant to previous chemotherapy (ATTRACTION-3): a multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2019;20:1506–1517.
    1. Huang J, et al. Camrelizumab versus investigator’s choice of chemotherapy as second-line therapy for advanced or metastatic oesophageal squamous cell carcinoma (Escort): a multicentre, randomised, open-label, phase 3 study. Lancet Oncol. 2020;21:832–842.
    1. Shi Y, et al. Safety and activity of sintilimab in patients with relapsed or refractory classical Hodgkin lymphoma (ORIENT-1): a multicentre, single-arm, phase 2 trial. Lancet Haematol. 2019;6:e12–e19.
    1. Yang Y, et al. Efficacy and Safety of Sintilimab Plus pemetrexed and platinum as First-Line Treatment for Locally Advanced or Metastatic nonsquamous NSCLC: a Randomized, Double-Blind, Phase 3 Study. J. Thorac. Oncol. 2020;15:1636–1646.
    1. Zhou C, et al. Sintilimab plus platinum and gemcitabine as first-line treatment for advanced or metastatic squamous nsclc: results from a randomized, double-blind, phase 3 trial (ORIENT-12) J. Thorac. Oncol. 2021;16:1501–1511.
    1. Ren Z, et al. ORIENT-32 study group. Sintilimab plus a bevacizumab biosimilar (IBI305) versus sorafenib in unresectable hepatocellular carcinoma (ORIENT-32): a randomised, open-label, phase 2-3 study. Lancet Oncol. 2021;22:977–990.
    1. Shen L, et al. RATIONALE 302: randomized, phase 3 study of tislelizumab versus chemotherapy as second-line treatment for advanced unresectable/metastatic esophageal squamous cell carcinoma. J. Clin. Oncol. 2021;39:suppl 4012.
    1. Chen MF, et al. The predictive value of pretreatment neutrophil-to-lymphocyte ratio in esophageal squamous cell carcinoma. Ann. Surg. Oncol. 2019;26:190–199.
    1. Guo JC, et al. Neutrophil-to-lymphocyte ratio and use of antibiotics associated With prognosis in esophageal squamous cell carcinoma patients receiving immune checkpoint inhibitors. Anticancer Res. 2019;39:5675–5682.
    1. Takenaka Y, et al. Prognostic role of neutrophil-to-lymphocyte ratio in head and neck cancer: a meta-analysis. Head. Neck. 2018;40:647–655.
    1. Ota Y, et al. Changes in the neutrophil-to-lymphocyte ratio during nivolumab monotherapy are associated with gastric cancer survival. Cancer Chemother. Pharmacol. 2020;85:265–272.
    1. Pirozzolo G, et al. Neutrophil-to-lymphocyte ratio as prognostic marker in esophageal cancer: a systematic review and meta-analysis. J. Thorac. Dis. 2019;11:3136–3145.
    1. Gregory AD, Houghton AM. Tumor-associated neutrophils: new targets for cancer therapy. Cancer Res. 2011;71:2411–2416.
    1. Shau HY, Kim A. Suppression of lymphokine-activated killer induction by neutrophils. J. Immunol. 1988;141:4395–4402.
    1. An X, et al. Elevated neutrophil to lymphocyte ratio predicts survival in advanced pancreatic cancer. Biomarkers. 2010;15:516–522.
    1. Cabrita R, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577:561–565.
    1. Helmink BA, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature. 2020;577:549–555.
    1. Petitprez F, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020;577:556–560.
    1. Sautès-Fridman, et al. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat. Rev. Cancer. 2019;19:307–325.
    1. Barros LRC, et al. High infiltration of B cells in tertiary lymphoid structures, TCR oligoclonality, and neoantigens are part of esophageal squamous cell carcinoma microenvironment. J. Leukoc. Biol. 2020;108:1307–1318.
    1. Gattinoni L, et al. Wnt signaling arrests effector T cell differentiation and generates CD8+ memory stem cells. Nat. Med. 2009;15:808–813.
    1. Gattinoni L, et al. Wnt/beta-catenin signaling in T-cell immunity and cancer immunotherapy. Clin. Cancer Res. 2010;16:4695–4701.
    1. Chen YQ, et al. Wnt pathway activator TWS119 enhances the proliferation and cytolytic activity of human γδT cells against colon cancer. Exp. Cell Res. 2018;362:63–71.
    1. Martínez-Rey D, et al. Extracellular superoxide dismutase, the endothelial basement membrane, and the wnt pathway: new players in vascular normalization and tumor infiltration by T-cells. Front Immunol. 2020;11:579552.
    1. Albuquerque CPebre, Pereira L. Wnt signalling-targeted therapy in the CMS2 tumor subtype: a new paradigm in CRC treatment? Adv. Exp. Med. Biol. 2018;1110:75–100.
    1. Wang Y, et al. Circulating tumor DNA analyses predict progressive disease and indicate trastuzumab-resistant mechanism in advanced gastric cancer. EBiomedicine. 2019;43:261–269.
    1. Cha E, et al. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci. Transl. Med. 2014;6:238ra70.
    1. Hopkins AC, et al. T cell receptor repertoire features associated with survival in immunotherapy-treated pancreatic ductal adenocarcinoma. JCI Insight. 2018;3:e122092.
    1. Han J, et al. TCR repertoire diversity of peripheral PD-1(+)CD8(+) T cells predicts clinical outcomes after immunotherapy in patients with non-small cell lung cancer. Cancer Immunol. Res. 2020;8:146–154.
    1. Lee CM, Tannock IF. The distribution of the therapeutic monoclonal antibodies cetuximab and rastuzumab within solid tumors. BMC Cancer. 2010;10:255.
    1. Huang AC, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature. 2017;545:60–65.
    1. Yu JW, et al. Tumor-immune profiling of murine syngeneic tumor models as a framework to guide mechanistic studies and predict therapy response in distinct tumor microenvironments. PLoS ONE. 2018;13:e0206223.
    1. Najjar YG, et al. Myeloid-derived suppressor cell subset accumulation in renal cell carcinoma parenchyma is associated with intratumoral expression of IL1β, IL8, CXCL5, and Mip-1α. Clin. Cancer Res. 2017;23:2346–2355.
    1. Shabaneh TB, et al. Oncogenic BRAFV600E governs regulatory T-cell recruitment during melanoma tumorigenesis. Cancer Res. 2018;78:5038–5049.
    1. Guisier F, et al. A rationale for surgical debulking to improve anti-PD1 therapy outcome in non small cell lung cancer. Sci. Rep. 2019;9:16902.
    1. Colak S, Ten Dijke P. Targeting TGF-β signaling in cancer. Trends Cancer. 2017;3:56–71.
    1. Lee S, et al. Prognostic significance of neutrophil lymphocyte ratio and platelet lymphocyte ratio in advanced gastric cancer patients treated with FOLFOX chemotherapy. BMC Cancer. 2013;13:350.
    1. Babraham Bioinformatics. (2021).
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30:2114–2120.
    1. Dobin A, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29:15–21.
    1. Anders S, Pyl PT, Huber W. HTSeq–a Python framework to work with high-throughput sequencing data. Bioinformatics. 2015;31:166–169.
    1. CRAN. (2021).
    1. Hänzelmann S, et al. GSVA: gene set variation analysis for microarray and RNA-Seq data. BMC Bioinforma. 2013;14:7.
    1. Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25:1754–1760.
    1. Van der Auwera GA, et al. From FastQ data to high confidence variant calls: the Genome Analysis Toolkit best practices pipeline. Curr. Protoc. Bioinforma. 2013;43:11.10.1–11.10.33.
    1. Roth A, et al. PyClone: statistical inference of clonal population structure in cancer. Nat. Methods. 2014;11:396–398.
    1. Zhang J, et al. PEAR: a fast and accurate Illumina paired-end read merger. Bioinformatics. 2014;30:614–620.
    1. Bolotin DA, et al. MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods. 2015;12:380–381.
    1. CRAN. (2021).

Source: PubMed

3
Sottoscrivi