Abstract

Evidence of clinical utility is a key issue in translating pharmacogenomics into clinical practice. Appropriately designed randomized controlled trials generally provide the most robust evidence of the clinical utility, but often only data from a pharmacogenomic association study are available. This paper details a method for reframing the results of pharmacogenomic association studies in terms of the comparative treatment effect for a pharmacogenomic subgroup to provide greater insight into the likely clinical utility of a pharmacogenomic marker, its’ likely cost effectiveness, and the value of undertaking the further (often expensive) research required for translation into clinical practice. The method is based on the law of total probability, which relates marginal and conditional probability. It takes as inputs: the prevalence of the pharmacogenomic marker in the patient group of interest, prognostic effect of the pharmacogenomic marker based on observational association studies, and the unstratified comparative treatment effect based on one or more conventional randomized controlled trials. The critical assumption is that of exchangeability across the included studies. The method is demonstrated using a case study of cytochrome P450 (CYP) 2C19 genotype and the anti-platelet agent clopidogrel. Indirect subgroup analysis provided insight into relationship between the clinical utility of genotyping CYP2C19 and the risk ratio of cardiovascular outcomes between CYP2C19 genotypes for individuals using clopidogrel. In this case study the indirect and direct estimates of the treatment effect for the cytochrome P450 2C19 subgroups were similar. In general, however, indirect estimates are likely to have substantially greater risk of bias than an equivalent direct estimate.

Highlights

  • An important element of pharmacogenomics is the use of genomic information to enable stratified or personalised medicine

  • Evidence of clinical utility for a given marker is a key issue in translating pharmacogenomics into clinical practice [1] and the extent to which comparative treatment effect differs between subgroups defined by the marker is an important component of assessing clinical utility

  • We exclude from the concept of clinical utility the dimension of cost effectiveness of the pharmacogenomic marker in producing the health gain, we discuss the application of the method to pharmacoeconomic modelling

Read more

Summary

Introduction

An important element of pharmacogenomics is the use of genomic information (genetic variation and gene expression) to enable stratified or personalised medicine. The results of a pharmacogenomic association study will highlight that individuals with one value for the marker are at higher risk of an event when using a specific drug, compared to individuals who have a different value for the marker This is generally insufficient to inform whether the pharmacogenomic marker identifies subgroups with clinically important and statistically significant differences in comparative treatment effects. A case study for the use of this method is presented, based on the cytochrome P450 (CYP2C19) genotype subgroup analysis of the RCT comparing ticagrelor and clopidogrel for the prevention of cardiovascular (CV) events for individuals with acute coronary syndrome (ACS) Evidence generated using this approach is not a substitute for direct evidence from an RCT; combined with a sensitivity analysis, this indirect method can provide insight into whether the pharmacogenomic marker is likely to have clinical utility and/or be cost-effective, and the value of undertaking further research

Methods
Discussion
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