Abstract

SummaryBackground This first-in-human phase 1 study assessed the safety of TAS-114, a novel deoxyuridine triphosphatase inhibitor, combined with S-1 to determine its maximum tolerated dose (MTD) and recommended dose (RD). Methods In this dose-escalation study with a 3 + 3 design, TAS-114 and S-1 were concurrently administered orally under fasting conditions at 5–240 mg/m2 and 30–36 mg/m2, respectively, in patients with advanced solid tumors. Safety, efficacy, and pharmacokinetics (PK) were evaluated. Results Seventy-six patients were enrolled. The MTD and RD were TAS-114 200 mg/m2 plus S-1 36 mg/m2 and TAS-114 240 mg/m2 plus S-1 30 mg/m2, respectively. Common treatment-related adverse events were anemia, lymphocytopenia, leukopenia, neutropenia, decreased appetite, rash, nausea, and pigmentation disorder. Partial response (PR) was observed in 10 patients (non-small cell lung cancer [NSCLC], n = 5; pancreatic neuroendocrine tumor, n = 2; gastric cancer, n = 2; gallbladder cancer, n = 1). Of these, four patients achieved PR despite prior treatment history with S-1. Patients administered TAS-114 exhibited linear PK and CYP3A4 induction, with no effect on the PK of S-1. Conclusion TAS-114 plus S-1 showed tolerable, safe, and potentially effective results. To confirm safety and efficacy, two phase 2 studies are ongoing in NSCLC and gastric cancer patients. Clinical trial registration ClinicalTrials.gov (NCT01610479) .

Highlights

  • Given the complexity and variability of the molecular processes that lead to cancer development [1, 2], current cancer therapy is multifaceted

  • This study was conducted in accordance with Declaration of Helsinki and Good Clinical Practice guidelines, and it was approved by the institutional review boards of the participating centers

  • A total of 76 patients were enrolled at four sites in Japan between May 2012 and April 2016, of whom all patients (Part 1, n = 48; Part 2, n = 28) were eligible and received at least 1 dose of TAS-114 and S-1; 64 and 12 patients were assigned to the S-1 30 mg/m2 and S-1 36 mg/m2 cohorts, respectively

Read more

Summary

Introduction

Given the complexity and variability of the molecular processes that lead to cancer development [1, 2], current cancer therapy is multifaceted. It includes molecular targeted therapy [1], immune therapy [2, 3], and other specific agents targeting cellular regulatory factors, oncogenic signalling pathways, tumor-associated angiogenesis, and host immune responses [1–3]. Despite these advances, cytotoxic agents still play an important role in systemic chemotherapy for several tumor types [4–6]. Fluoropyrimidines are the cornerstone therapy for different types of cancer, and various efforts to improve their efficacy have been attempted in the past three decades. There is a need for the development of a new, highly

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call

Disclaimer: All third-party content on this website/platform is and will remain the property of their respective owners and is provided on "as is" basis without any warranties, express or implied. Use of third-party content does not indicate any affiliation, sponsorship with or endorsement by them. Any references to third-party content is to identify the corresponding services and shall be considered fair use under The CopyrightLaw.