Cost Effectiveness of Sequencing 34 Cancer-Associated Genes as an Aid for Treatment Selection in Patients with Metastatic Melanoma.

Abstract

ObjectiveTo determine whether a next-generation sequencing (NGS) panel of 34 cancer-associated genes would cost-effectively aid in the treatment selection for patients with metastatic melanoma, compared with a single-site BRAF V600 mutation test.MethodsA decision model was developed to estimate the costs and health outcomes of the two test strategies. The cost effectiveness of these two strategies was analyzed from a payer perspective over a 2-year time horizon with model parameters taken from the literature.ResultsIn the base case, the gene sequencing panel strategy resulted in a cost of US120,022and0.721quality−adjustedlifeyears(QALYs)perpatient,whereasthesingle−sitemutationteststrategyresultedinacostofUS120,022 and 0.721 quality-adjusted life years (QALYs) per patient, whereas the single-site mutation test strategy resulted in a cost of US128,965 and 0.704 QALYs. Thus, the gene sequencing panel strategy cost US8943lessperpatientandincreasedQALYsby0.0174perpatient.Sensitivityanalysesshowedthat,comparedwiththesingle−sitemutationteststrategy,thegenesequencingpanelstrategyhada90.98943 less per patient and increased QALYs by 0.0174 per patient. Sensitivity analyses showed that, compared with the single-site mutation test strategy, the gene sequencing panel strategy had a 90.9% chance of having reduced costs and increased QALYs, with the cost of the gene sequencing panel test having minimal effect on the incremental cost.ConclusionCompared with the single-site mutation test, the use of an NGS panel of 34 cancer-associated genes as an aid in selecting therapy for patients with metastatic melanoma reduced costs and increased QALYs. If the base-case results were applied to the 8900 patients diagnosed with metastatic melanoma in the USA each year, the gene sequencing panel strategy could result in an annual savings of US79.6 million and a gain of 155 QALYs

    Similar works