S-1/temozolomide versus S-1/temozolomide plus thalidomide in advanced pancreatic and non-pancreatic neuroendocrine tumours (STEM): A randomised, open-label, multicentre phase 2 trial

Yihebali Chi, Lijie Song, Weili Liu, Yuhong Zhou, Yadong Miao, Weijia Fang, Huangying Tan, Susheng Shi, Hai Jiang, Jianming Xu, Ru Jia, Bo Zheng, Liming Jiang, Jiuda Zhao, Rui Zhang, Huijing Tan, Yuehua Wang, Qichen Chen, Minjie Yang, Xi Guo, Zhou Tong, Zhirong Qi, Fuxing Zhao, Xiaofei Yan, Hong Zhao, Yihebali Chi, Lijie Song, Weili Liu, Yuhong Zhou, Yadong Miao, Weijia Fang, Huangying Tan, Susheng Shi, Hai Jiang, Jianming Xu, Ru Jia, Bo Zheng, Liming Jiang, Jiuda Zhao, Rui Zhang, Huijing Tan, Yuehua Wang, Qichen Chen, Minjie Yang, Xi Guo, Zhou Tong, Zhirong Qi, Fuxing Zhao, Xiaofei Yan, Hong Zhao

Abstract

Background: There are currently limited systemic treatment options for patients with advanced neuroendocrine tumours (NETS) and the efficacy of existing treatments is sub-optimal. We evaluated the efficacy and safety of Tegafur/gimeracil/oteracil/potassium capsules (S-1)/Temozolomide with or without thalidomide for the treatment of NETS (STEM trial).

Methods: A randomised, controlled, open-label, phase 2 trial conducted at eight hospitals in China. Adults (≥18 years) with unresectable/metastatic, pancreatic or non-pancreatic NETS, with an Eastern Cooperative Oncology Group (ECOG) PS of 0-1, and progression on ≤2 previous therapies were randomised (1:1, using hierarchical block randomization with block length 4, stratified by pancreatic/non-pancreatic disease to receive S-1 40-60 mg orally twice daily on days 1-14 plus temozolomide 200 mg orally daily on days 10-14 in a 21-day cycle OR S-1 and temozolomide plus thalidomide orally nightly (100 mg on days 1-7, 200 mg on days 8-14, and 300 mg from day 15), until disease progression, death, intolerable toxicity, withdrawal of informed consent or at the investigator's discretion. The primary endpoint was objective response rate (ORR) by RECIST 1.1 in an intention-to-treat population. Safety was assessed in all patients who received treatment. The study was registered at ClinicalTrials.gov: NCT03204019 (pancreatic group) and NCT03204032 (non-pancreatic group).

Findings: Between March 23, 2017 and November 16, 2020, 187 patients were screened and 140 were randomly assigned to S-1/temozolomide plus thalidomide (n = 69) or S-1/temozolomide (n =71). After a median follow-up of 12·1 months (IQR: 8·4-16·6), the ORR was comparable in the S-1/temozolomide plus thalidomide and S-1/temozolomide groups 26·1% [95% CI 17·2-37·5] versus 25·4% [95% CI 16·7-36·6]; odds ratio: 1·03 [95% CI 0·48-2·22]; P = 0·9381). In the S-1/temozolomide plus thalidomide group, the most common grade 3-4 treatment-related adverse event was fatigue (2/68, 3%), and in the control group were thrombocytopenia and diarrhea (both 1/71, 2%). There were no treatment-related deaths in either group.

Interpretation: S-1/temozolomide with or without thalidomide leads to a comparable treatment response in patients with advanced/metastatic NETS.

Funding: This work was supported by CAMS Innovation Fund for Medical Sciences (CIFMS,2021-I2M-1-066, 2017-I2M-4-002, 2021-I2M-1-019, 2017-I2M-1-001), the National Natural Science Foundation of China (81972311, 82141127, 31970794,), the State Key Project on Infection Diseases of China (2017ZX10201021-007-003), the Non-profit Central Research Institute Fund of Chinese Academy of Medical Sciences (2019PT310026), Sanming Project of Medicine in Shenzhen (SZSM202011010), and the State Key Laboratory Special fund from the Ministry of Science (2060204).

Keywords: Neuroendocrine tumours; S-1; Temozolomide; Thalidomide.

Conflict of interest statement

The authors have no conflicts of interest to declare.

© 2022 The Authors.

Figures

Figure 1
Figure 1
Trial profile. S1+TMZ, S-1/temozolomide; S1+TMZ+Thal, S-1/temozolomide plus thalidomide.
Figure 2
Figure 2
Kaplan-Meier estimates of progression-free survival. A) patients in the intention-to-treat population assigned to S1+TMZ and S1+TMZ+Thal; B) patients with non-pancreatic disease assigned to S1+TMZ and S1+TMZ+Thal; C) patients with pancreatic disease assigned to S1+TMZ and S1+TMZ+Thal and D) patients with MGMT positive or negative status. S1+TMZ, S-1/temozolomide; S1+TMZ+Thal, S-1/temozolomide plus thalidomide; MGMT, O6-methylguanine-DNA methyltransferase.

References

    1. Basu B, Sirohi B, Corrie P. Systemic therapy for neuroendocrine tumours of gastroenteropancreatic origin. Endocr Relat Cancer. 2010;17:R75–R90. doi: 10.1677/ERC-09-0108.
    1. Oberg KE, Reubi J-C, Kwekkeboom DJ, Krenning EP. Role of somatostatins in gastroenteropancreatic neuroendocrine tumour development and therapy. Gastroenterology. 2010;139:742–753. doi: 10.1053/j.gastro.2010.07.002. , 753.e1.
    1. Dasari A, Shen C, Halperin D, et al. Trends in the Incidence, Prevalence, and Survival Outcomes in Patients With Neuroendocrine tumours in the United States. JAMA Oncol. 2017;3:1335–1342. doi: 10.1001/jamaoncol.2017.0589.
    1. Halperin DM, Kulke MH, Yao JC. A tale of two tumours: treating pancreatic and extrapancreatic neuroendocrine tumours. Annu Rev Med. 2015;66:1–16. doi: 10.1146/annurev-med-061813-012908.
    1. Rinke A, Müller H-H, Schade-Brittinger C, et al. Placebo-controlled, double-blind, prospective, randomised study on the effect of octreotide LAR in the control of tumour growth in patients with metastatic neuroendocrine midgut tumours: a report from the PROMID Study Group. J Clin Oncol. 2009;27:4656–4663. doi: 10.1200/JCO.2009.22.8510.
    1. Caplin ME, Pavel M, Ruszniewski P. Lanreotide in metastatic enteropancreatic neuroendocrine tumours. N Engl J Med. 2014;371:1556–1557. doi: 10.1056/NEJMc1409757.
    1. Yao JC, Fazio N, Singh S, et al. Everolimus for the treatment of advanced, non-functional neuroendocrine tumours of the lung or gastrointestinal tract (RADIANT-4): a randomised, placebo-controlled, phase 3 study. Lancet. 2016;387:968–977. doi: 10.1016/S0140-6736(15)00817-X.
    1. Lu Y, Zhao Z, Wang J, et al. Safety and efficacy of combining capecitabine and temozolomide (CAPTEM) to treat advanced neuroendocrine neoplasms: a meta-analysis. Medicine. 2018;97:e12784. doi: 10.1097/MD.0000000000012784.
    1. Cives M, Strosberg JR. Gastroenteropancreatic Neuroendocrine tumours. CA. 2018;68:471–487. doi: 10.3322/caac.21493.
    1. Chatzellis E, Angelousi A, Daskalakis K, et al. Activity and safety of standard and prolonged capecitabine/temozolomide administration in patients with advanced neuroendocrine neoplasms. Neuroendocrinology. 2019;109:333–345. doi: 10.1159/000500135.
    1. Hirata K, Horikoshi N, Aiba K, et al. Pharmacokinetic study of S-1, a novel oral fluorouracil antitumour drug. Clin Cancer Res. 1999;5:2000–2005.
    1. Miura K, Kinouchi M, Ishida K, et al. 5-FU metabolism in cancer and orally-administrable 5-FU drugs. Cancers. 2010;2:1717–1730. doi: 10.3390/cancers2031717.
    1. Shirasaka T, Nakano K, Takechi T, et al. Antitumour activity of 1 M tegafur-0.4 M 5-chloro-2,4-dihydroxypyridine-1 M potassium oxonate (S-1) against human colon carcinoma orthotopically implanted into nude rats. Cancer Res. 1996;56:2602–2606.
    1. Vaflard P, Ederhy S, Torregrosa C, André T, Cohen R, Lopez-Trabada D. La toxicité cardiaque des fluoropyrimidines: 5-fluorouracile, capécitabine, le composé S-1 et le trifluridine/tipiracil. Bulletin du Cancer. 2018;105:707–719. doi: 10.1016/j.bulcan.2018.05.005.
    1. ter Veer E, Ngai LL, Valkenhoef G van, et al. Capecitabine, 5-fluorouracil and S-1 based regimens for previously untreated advanced oesophagogastric cancer: a network meta-analysis. Sci Rep. 2017;7:7142. doi: 10.1038/s41598-017-07750-3.
    1. Zhao J, Zhao H, Chi Y. Safety and efficacy of the S-1/temozolomide regimen in patients with metastatic neuroendocrine tumours. Neuroendocrinology. 2018;106:318–323. doi: 10.1159/000480402.
    1. Sherbet GV. Therapeutic potential of thalidomide and its analogues in the treatment of cancer. Anticancer Res. 2015;35:5767–5772.
    1. Kulke MH, Stuart K, Enzinger PC, et al. Phase II study of temozolomide and thalidomide in patients with metastatic neuroendocrine tumours. JCO. 2006;24:401–406. doi: 10.1200/JCO.2005.03.6046.
    1. Pfeifer G. Defining driver DNA methylation changes in human cancer. IJMS. 2018;19:1166. doi: 10.3390/ijms19041166.
    1. Paz MF, Yaya-Tur R, Rojas-Marcos I, et al. CpG island hypermethylation of the DNA repair enzyme methyltransferase predicts response to temozolomide in primary gliomas. Clin Cancer Res. 2004;10:4933–4938. doi: 10.1158/1078-0432.CCR-04-0392.
    1. Campana D, Walter T, Pusceddu S, et al. Correlation between MGMT promoter methylation and response to temozolomide-based therapy in neuroendocrine neoplasms: an observational retrospective multicentre study. Endocrine. 2018;60:490–498. doi: 10.1007/s12020-017-1474-3.
    1. Lloyd RV, Osamura RY, Klöppel G, Rosai J, editors. WHO Classification of Tumours of Endocrine Organs. 4th Ed. International Agency for Research on Cancer; Lyon: 2017.
    1. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1) Eur J Cancer. 2009;45:228–247. doi: 10.1016/j.ejca.2008.10.026.
    1. Xu J, Shen L, Zhou Z, et al. Surufatinib in advanced extrapancreatic neuroendocrine tumours (SANET-ep): a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2020;21:1500–1512. doi: 10.1016/S1470-2045(20)30496-4.
    1. Okusaka T, Ueno H, Morizane C, et al. Cytotoxic chemotherapy for pancreatic neuroendocrine tumours. J Hepatobiliary Pancreat Sci. 2015;22:628–633. doi: 10.1002/jhbp.257.
    1. Yao JC, Shah MH, Ito T, et al. Everolimus for advanced pancreatic neuroendocrine tumours. N Engl J Med. 2011;364:514–523. doi: 10.1056/NEJMoa1009290.
    1. Pietrantonio F, Lobefaro R, Antista M, et al. Capecitabine and temozolomide versus FOLFIRI in RAS-mutated, MGMT-methylated metastatic colorectal cancer. Clin Cancer Res. 2020;26:1017–1024. doi: 10.1158/1078-0432.CCR-19-3024.
    1. Walter T, van Brakel B, Vercherat C, et al. O6-Methylguanine-DNA methyltransferase status in neuroendocrine tumours: prognostic relevance and association with response to alkylating agents. Br J Cancer. 2015;112:523–531. doi: 10.1038/bjc.2014.660.
    1. Cives M, Ghayouri M, Morse B, et al. Analysis of potential response predictors to capecitabine/temozolomide in metastatic pancreatic neuroendocrine tumours. Endocrine-Related Cancer. 2016;23:759–767. doi: 10.1530/ERC-16-0147.
    1. Kunz PL, Graham N, Catalano PJ, et al. A randomised study of temozolomide or temozolomide and capecitabine in patients with advanced pancreatic neuroendocrine tumours: final analysis of efficacy and evaluation of MGMT (ECOG-ACRIN E2211) JCO. 2022;40 doi: 10.1200/JCO.2022.40.16_suppl.4004. 4004–4004.
    1. Kawakami H, Takeno A, Endo S, et al. Randomised, open-label phase II study comparing capecitabine-cisplatin every 3 weeks with s-1-cisplatin every 5 weeks in chemotherapy-naïve patients with her2-negative advanced gastric cancer: OGSG1105, HERBIS-4A trial. Oncologist. 2018;23:1411–e147. doi: 10.1634/theoncologist.2018-0175.
    1. Lee J-L, Kang Y-K, Kang HJ, et al. A randomised multicentre phase II trial of capecitabine vs S-1 as first-line treatment in elderly patients with metastatic or recurrent unresectable gastric cancer. Br J Cancer. 2008;99:584–590. doi: 10.1038/sj.bjc.6604536.
    1. Fine RL, Fogelman DR, Schreibman SM. Effective treatment of neuroendocrine tumortumours With temozolomide and capecitabine. JCO. 2005;23:4216. doi: 10.1200/jco.2005.23.16_suppl.4216. –4216.
    1. Al-Toubah T, Pelle E, Valone T, Haider M, Strosberg JR. Efficacy and toxicity analysis of capecitabine and temozolomide in neuroendocrine neoplasms. J Nat Comprehens Cancer Network. 2022;20:29–36. doi: 10.6004/jnccn.2021.7017.

Source: PubMed

3
Suscribir