Preoperative pembrolizumab combined with chemoradiotherapy for esophageal squamous cell carcinoma: Trial design

Yuyan Zheng, Chengqiang Li, Bentong Yu, Shengguang Zhao, Jian Li, Xiaoyan Chen, Hecheng Li, Yuyan Zheng, Chengqiang Li, Bentong Yu, Shengguang Zhao, Jian Li, Xiaoyan Chen, Hecheng Li

Abstract

Objective: The safety and feasibility of preoperative pembrolizumab combined with chemoradiotherapy (PPCT) for resectable esophageal squamous cell carcinoma have been confirmed by the prior Preoperative Anti-PD-1 Antibody combined with Chemoradiotherapy for Locally Advanced Squmous Cell Carcinoma of Esophageus (PALACE)-1 trial. Potential therapeutic benefit was also observed with a pathologic complete response rate of 55.6% after PPCT. We will conduct the multicenter single-arm PALACE-2 study to investigate the efficacy and to further confirm the safety of PPCT (ClinicalTrials.gov ID: NCT04435197).

Methods: A total of 143 patients with previously untreated, locally advanced, and surgically resectable esophageal squamous cell carcinoma (T2 through T4a, N0 through N+, M0) will be enrolled in PALACE-2. Main exclusion criteria are autoimmune disease, interstitial lung disease, ongoing immunosuppressive therapy, and having received chemotherapy, radiotherapy, target therapy, or immune therapy for this or any other malignancies. Positive programmed cell death ligand 1 expression is not mandatory for enrollment. Patients will receive PPCT, which includes concurrent pembrolizumab (200 mg on day 1 and day 22), carboplatin (area under the curve = 2, once a week for 5 weeks), nab-paclitaxel (50 mg/m2, once a week for 5 weeks), and radiotherapy (23 fractions of 1.8 Gy, 5 fractions a week). Esophagectomy will be performed within 4 to 6 weeks after the completion of PPCT.

Results: The primary end point is the rate of pathologic complete response. Secondary outcome measures are 3-year disease-free survival rate, 3-year overall survival rate, R0 resection rate, and adverse events during neoadjuvant and perioperative periods.

Conclusions: PPCT was preliminarily demonstrated to be safe, feasible, and to provide potential therapeutic benefits by the PALACE-1 trial. The subsequent multicenter PALACE-2 study will investigate the efficacy and further confirm the safety of PPCT for locally advanced, resectable esophageal squamous cell carcinoma.

Keywords: AEs, adverse events; CROSS, Chemoradiotherapy for Oesophageal Cancer Followed by Surgery Study; CT, computed tomography; ESCC, esophageal squamous cell carcinoma; ICI, immune checkpoint inhibitor; OS, overall survival; PALACE, Preoperative Anti-PD-1 Antibody combined with Chemoradiotherapy for Locally Advanced Squmous Cell Carcinoma of Esophageus; PD-1, programmed cell death protein 1; PD-L1, programmed cell death ligand 1; PET-CT, positron emission tomography-computed tomography; PPCT, preoperative pembrolizumab combined with chemoradiotherapy; chemoradiotherapy; esophageal squamous cell carcinoma; immunotherapy; nCRT, neoadjuvant chemoradiotherapy; neoadjuvant therapy; pCR, pathologic complete response.

© 2021 The Author(s).

Figures

Graphical abstract
Graphical abstract
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/9390428/bin/fx2.jpg
Flowchart of the PALACE-2 trial.
Figure 1
Figure 1
Flowchart of the Preoperative Anti-PD-1 Antibody combined with Chemoradiotherapy for Locally Advanced Squmous Cell Carcinoma of Esophageus-2 trial. ESCC, Esophageal squamous cell carcinoma; AUC, area under the curve; PET-CT, positron emission tomography-computed tomography.
Figure 2
Figure 2
Trial design and flow chart of the Preoperative Anti-PD-1 Antibody combined with Chemoradiotherapy for Locally Advanced Squmous Cell Carcinoma of Esophageus-2 trial. ESCC, Esophageal squamous cell carcinoma; AUC, area under the curve; pCR, pathologic complete response; DFS, disease-free survival; OS, overall survival; AEs, adverse events.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/9390428/bin/fx3.jpg

References

    1. Bray F., Ferlay J., Soerjomataram I., Siegel R.L., Torre L.A., Jemal A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2018;68:394–424.
    1. Abnet C.C., Arnold M., Wei W.Q. Epidemiology of esophageal squamous cell carcinoma. Gastroenterology. 2018;154:360–373.
    1. van Hagen P., Hulshof M.C., van Lanschot J.J., Steyerberg E.W., van Berge Henegouwen M.I., Wijnhoven B.P., et al. Preoperative chemoradiotherapy for esophageal or junctional cancer. N Engl J Med. 2012;366:2074–2084.
    1. Shapiro J., van Lanschot J.J.B., Hulshof M., van Hagen P., van Berge Henegouwen M.I., Wijnhoven B.P.L., et al. Neoadjuvant chemoradiotherapy plus surgery versus surgery alone for oesophageal or junctional cancer (CROSS): long-term results of a randomised controlled trial. Lancet Oncol. 2015;16:1090–1098.
    1. Lin D.C., Hao J.J., Nagata Y., Xu L., Shang L., Meng X., et al. Genomic and molecular characterization of esophageal squamous cell carcinoma. Nat Genet. 2014;46:467–473.
    1. Song Y., Li L., Ou Y., Gao Z., Li E., Li X., et al. Identification of genomic alterations in oesophageal squamous cell cancer. Nature. 2014;509:91–95.
    1. Zhang L., Zhou Y., Cheng C., Cui H., Cheng L., Kong P., et al. Genomic analyses reveal mutational signatures and frequently altered genes in esophageal squamous cell carcinoma. Am J Hum Genet. 2015;96:597–611.
    1. Cancer Genome Atlas Research Network, Analysis Working Group: Asan University, BC Cancer Agency, Brigham and Women's Hospital, Broad Institute, Brown University, et al. Integrated genomic characterization of oesophageal carcinoma. Nature. 2017;541:169–175.
    1. Ohigashi Y., Sho M., Yamada Y., Tsurui Y., Hamada K., Ikeda N., et al. Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand-2 expression in human esophageal cancer. Clin Cancer Res. 2005;11:2947–2953.
    1. Sholl L.M., Hirsch F.R., Hwang D., Botling J., Lopez-Rios F., Bubendorf L., et al. The promises and challenges of tumor mutation burden as an immunotherapy biomarker: a perspective from the International Association for the Study of Lung Cancer Pathology Committee. J Thorac Oncol. 2020;15:1409–1424.
    1. Yarchoan M., Johnson B.A., III, Lutz E.R., Laheru D.A., Jaffee E.M. Targeting neoantigens to augment antitumour immunity. Nat Rev Cancer. 2017;17:209–222.
    1. Jardim D.L., Goodman A., de Melo Gagliato D., Kurzrock R. The challenges of tumor mutational burden as an immunotherapy biomarker. Cancer Cell. 2021;39:154–173.
    1. Shah M.A., Kojima T., Hochhauser D., Enzinger P., Raimbourg J., Hollebecque A., et al. Efficacy and safety of pembrolizumab for heavily pretreated patients with advanced, metastatic adenocarcinoma or squamous cell carcinoma of the esophagus: the phase 2 KEYNOTE-180 Study. JAMA Oncol. 2019;5:546–550.
    1. Kojima T., Shah M.A., Muro K., Francois E., Adenis A., Hsu C.H., et al. Randomized phase III KEYNOTE-181 study of pembrolizumab versus chemotherapy in advanced esophageal cancer. J Clin Oncol. 2020;38:4138–4148.
    1. Sharma P., Allison J.P. The future of immune checkpoint therapy. Science. 2015;348:56–61.
    1. Sharabi A.B., Lim M., DeWeese T.L., Drake C.G. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 2015;16:e498–e509.
    1. Li C., Zhao S., Zheng Y., Han Y., Chen X., Cheng Z., et al. Preoperative Pembrolizumab Combined with Chemoradiotherapy for Oesophageal Squamous Cell Carcinoma (PALACE-1) Eur J Cancer. 2021;144:232–241.
    1. Rice T.W., Ishwaran H., Ferguson M.K., Blackstone E.H., Goldstraw P. Cancer of the esophagus and esophagogastric junction: an eighth edition staging primer. J Thorac Oncol. 2017;12:36–42.
    1. Chirieac L.R., Swisher S.G., Ajani J.A., Komaki R.R., Correa A.M., Morris J.S., et al. Posttherapy pathologic stage predicts survival in patients with esophageal carcinoma receiving preoperative chemoradiation. Cancer. 2005;103:1347–1355.
    1. Li C.Y., Huang P.M., Chu P.Y., Chen P.M., Lin M.W., Kuo S.W., et al. Predictors of survival in esophageal squamous cell carcinoma with pathologic major response after neoadjuvant chemoradiation therapy and surgery: the impact of chemotherapy protocols. Biomed Res Int. 2016;2016:6423297.
    1. Travis W.D., Dacic S., Wistuba I., Sholl L., Adusumilli P., Bubendorf L., et al. IASLC multidisciplinary recommendations for pathologic assessment of lung cancer resection specimens after neoadjuvant therapy. J Thorac Oncol. 2020;15:709–740.
    1. Nishimaki T., Tanaka O., Ando N., Ide H., Watanabe H., Shinoda M., et al. Evaluation of the accuracy of preoperative staging in thoracic esophageal cancer. Ann Thorac Surg. 1999;68:2059–2064.
    1. Elliott J.A., O'Farrell N.J., King S., Halpenny D., Malik V., Muldoon C., et al. Value of CT-PET after neoadjuvant chemoradiation in the prediction of histological tumour regression, nodal status and survival in oesophageal adenocarcinoma. Br J Surg. 2014;101:1702–1711.
    1. Ajani J.A., D'Amico T.A., Bentrem D.J., Chao J., Corvera C., Das P., et al. Esophageal and esophagogastric junction cancers, version 2.2019, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2019;17:855–883.
    1. Yang H., Liu H., Chen Y., Zhu C., Fang W., Yu Z., et al. Neoadjuvant chemoradiotherapy followed by surgery versus surgery alone for locally advanced squamous cell carcinoma of the esophagus (NEOCRTEC5010): a phase iii multicenter, randomized, open-label clinical trial. J Clin Oncol. 2018;36:2796–2803.
    1. Davern M., Lysaght J. Cooperation between chemotherapy and immunotherapy in gastroesophageal cancers. Cancer Lett. 2020;495:89–99.
    1. Shurin G.V., Tourkova I.L., Kaneno R., Shurin M.R. Chemotherapeutic agents in noncytotoxic concentrations increase antigen presentation by dendritic cells via an IL-12-dependent mechanism. J Immunol. 2009;183:137–144.
    1. Ma Y., Adjemian S., Mattarollo S.R., Yamazaki T., Aymeric L., Yang H., et al. Anticancer chemotherapy-induced intratumoral recruitment and differentiation of antigen-presenting cells. Immunity. 2013;38:729–741.
    1. Wang Y.J., Fletcher R., Yu J., Zhang L. Immunogenic effects of chemotherapy-induced tumor cell death. Genes Dis. 2018;5:194–203.
    1. Reits E.A., Hodge J.W., Herberts C.A., Groothuis T.A., Chakraborty M., Wansley E.K., et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J Exp Med. 2006;203:1259–1271.
    1. Matsumura S., Wang B., Kawashima N., Braunstein S., Badura M., Cameron T.O., et al. Radiation-induced CXCL16 release by breast cancer cells attracts effector T cells. J Immunol. 2008;181:3099–3107.
    1. Hallahan D., Kuchibhotla J., Wyble C. Cell adhesion molecules mediate radiation-induced leukocyte adhesion to the vascular endothelium. Cancer Res. 1996;56:5150–5155.
    1. Dovedi S.J., Adlard A.L., Lipowska-Bhalla G., McKenna C., Jones S., Cheadle E.J., et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 2014;74:5458–5468.
    1. Tran L., Allen C.T., Xiao R., Moore E., Davis R., Park S.J., et al. Cisplatin alters antitumor immunity and synergizes with PD-1/PD-L1 inhibition in head and neck squamous cell carcinoma. Cancer Immunol Res. 2017;5:1141–1151.
    1. Ng H.Y., Li J., Tao L., Lam A.K., Chan K.W., Ko J.M.Y., et al. Chemotherapeutic treatments increase PD-L1 expression in esophageal squamous cell carcinoma through EGFR/ERK activation. Transl Oncol. 2018;11:1323–1333.

Source: PubMed

3
Prenumerera