Abstract

Several studies have estimated the financial inputs for successful drug development. Such analyses do not capture the large investment that patient study participants commit to drug development. To estimate the volume of patients required to achieve a first US Food and Drug Administration (FDA) approval for a new anticancer drug or biologic therapy. This cohort study included a random sample of prelicense oncology drugs and biologics with a trial site in the United States that were launched into clinical efficacy testing between January 1, 2006, and December 31, 2010. Drugs and biologics were identified using ClinicalTrials.gov registration records. Total patient enrollment was captured over an 8-year span, and each intervention was classified based on whether it received FDA approval and was deemed as having intermediate or substantial value according to the American Society of Clinical Oncology Value Framework (ASCO-VF) score. Secondarily, the association between patient numbers and intervention characteristics was tested. Data were analyzed in February 2020. The prespecified primary outcome was the number of patients enrolled in prelicense trials per FDA approval. A total of 120 drugs and biologics were included in our study, with 84 (70.0%) targeted agents, 20 (16.7%) immunotherapies, and 71 (59.2%) novel agents. A total of 13 drugs and biologics (10.8%; 95% CI, 5.3%-16.8%) in our sample gained FDA approval within 8 years, of which 1 (7.7%) was deemed of intermediate value and 3 (23.1%) were deemed of substantial value using ASCO-VF scoring. Overall, 158 810 patients were enrolled in 1335 trials testing these drugs and biologics, 47 913 (30.2%) in trials that led to FDA approval and 110 897 (69.8%) in trials that did not. An estimated 12 217 (95% CI, 7970-22 215) patient study participants contributed to prelicense trials per FDA approval. The estimated number of patients needed to produce a single FDA-approved drug or biologic of intermediate or substantial ASCO-VF clinical value was 39 703 (95% CI, 19 391-177 991). The results of this cohort study make visible the substantial patient investment required for prelicense oncology drug development. Such analyses can be used to devise policies that maximize the clinical impact of research on a per-patient basis.

Highlights

  • Recent studies estimate the median cost of bringing a new drug to market at $985.3 million for all therapeutic agents and $793.4 million for oncology drugs.[1,2] For every 100 drugs entered into phase 1 clinical testing in oncology, fewer than 10 will receive regulatory approval.[3,4,5] A minority of approved cancer drugs provide substantial clinical benefit.[6]cost estimates and molecule success rates do not reflect the full range of inputs societies commit to pharmaceutical research

  • A total of 13 drugs and biologics (10.8%; 95% CI, 5.3%-16.8%) in our sample gained Food and Drug Administration (FDA) approval within 8 years, of which 1 (7.7%) was deemed of intermediate value and 3 (23.1%) were deemed of substantial value using American Society of Clinical Oncology Value Framework (ASCO-VF) scoring

  • 158 810 patients were enrolled in 1335 trials testing these drugs and biologics, 47 913 (30.2%) in trials that led to FDA approval and 110 897 (69.8%) in trials that did not

Read more

Summary

Introduction

Recent studies estimate the median cost of bringing a new drug to market at $985.3 million for all therapeutic agents and $793.4 million for oncology drugs.[1,2] For every 100 drugs entered into phase 1 clinical testing in oncology, fewer than 10 will receive regulatory approval.[3,4,5] A minority of approved cancer drugs provide substantial clinical benefit.[6]cost estimates and molecule success rates do not reflect the full range of inputs societies commit to pharmaceutical research. Recent studies estimate the median cost of bringing a new drug to market at $985.3 million for all therapeutic agents and $793.4 million for oncology drugs.[1,2] For every 100 drugs entered into phase 1 clinical testing in oncology, fewer than 10 will receive regulatory approval.[3,4,5] A minority of approved cancer drugs provide substantial clinical benefit.[6]. Estimates that include all patient study participants can make more visible the degree to which private drug development efforts build on public volunteerism. They can help to identify research activities that more efficiently use this public endowment. The aim of this study was to estimate the number of patient study participants needed to obtain a first FDA approval for a cancer therapeutic

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

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