0
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Evaluation of the Cost-effectiveness of Doublet Therapy in Metastatic BRAF Variant Colorectal Cancer

      research-article
      , BS 1 , , MD 1 , , MD 1 , , MD 2 , , MD, MPH, MSc 1 , 3 ,
      JAMA Network Open
      American Medical Association

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          This economic evaluation uses Markov modeling to identify and compare the cost and utility of doublet and standard treatments for patients with BRAF V600E variant colorectal cancer.

          Key Points

          Question

          Is the use of doublet therapy (encorafenib and cetuximab) compared with standard chemotherapy (with cetuximab and irinotecan-based regimens) a cost-effective treatment strategy in patients with metastatic BRAF variant colorectal cancer?

          Findings

          In this economic evaluation of the cost-effectiveness of 2 treatment strategies, doublet therapy was associated with an incremental cost of $78 233, an incremental effectiveness of 0.15 quality-adjusted life years, and an incremental cost-effectiveness ratio of $523 374 per quality-adjusted life year gained.

          Meaning

          Results of this study suggest that doublet therapy for metastatic BRAF variant colorectal cancer is unlikely to be a cost-effective treatment under current pricing.

          Abstract

          Importance

          The BEACON trial showed that combination therapy with encorafenib (BRAF inhibitor) and cetuximab (EGFR inhibitor) was associated with prolonged overall survival compared with standard chemotherapy in patients with metastatic BRAF variant colorectal cancer. However, the cost-effectiveness of using these agents in this clinical setting is unknown.

          Objective

          To create a cost-effectiveness model to compare doublet therapy (encorafenib plus cetuximab) with standard chemotherapy (cetuximab plus irinotecan or cetuximab plus folinic acid, fluorouracil, and irinotecan) in treating patients with metastatic BRAF variant colorectal cancer.

          Design, Setting, and Participants

          This economic evaluation constructed a Markov model to compare the lifetime cost and utility of doublet therapy and standard chemotherapy. Parametric survival modeling was used to extrapolate the effectiveness of each line of therapy from large clinical trials. One-way and probabilistic sensitivity analyses assessed the uncertainty in the model. Patients mirrored the cohorts in the BEACON trial: they had metastatic BRAF variant colorectal cancer and were followed up as they progressed through multiple lines of therapy, best supportive care, and death. Data collection and data analysis were performed from November 15, 2019, to July 14, 2020.

          Main Outcomes and Measures

          The main outcome was the incremental cost-effectiveness ratio, which was calculated using the cumulative cost and effectiveness in quality-adjusted life years (QALYs), of doublet therapy compared with standard chemotherapy.

          Results

          The model patient cohort had a mean age of 61 years, and 53% of the patients were women, 66% had 1 previous line of therapy, and 8% had high microsatellite instability. Doublet therapy was associated with an improvement of 0.15 QALYs compared with standard chemotherapy. However, the incremental cost of doublet therapy was $78 233, leading to an incremental cost-effectiveness ratio of $523 374 per QALY gained. Concomitant decreases in the price of encorafenib and cetuximab are needed to achieve cost-effectiveness at a willingness-to-pay threshold of $150 000 per QALY gained.

          Conclusions and Relevance

          This study found that doublet therapy for metastatic BRAF variant colorectal cancer was unlikely to be cost-effective under current pricing. Cost-effectiveness needs to be considered in clinical trial design, particularly when combining new therapies with non-cost-effective treatments that are coadministered without a fixed duration.

          Related collections

          Most cited references28

          • Record: found
          • Abstract: found
          • Article: not found

          Recommendations for Conduct, Methodological Practices, and Reporting of Cost-effectiveness Analyses: Second Panel on Cost-Effectiveness in Health and Medicine.

          Since publication of the report by the Panel on Cost-Effectiveness in Health and Medicine in 1996, researchers have advanced the methods of cost-effectiveness analysis, and policy makers have experimented with its application. The need to deliver health care efficiently and the importance of using analytic techniques to understand the clinical and economic consequences of strategies to improve health have increased in recent years.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Durable Clinical Benefit With Nivolumab Plus Ipilimumab in DNA Mismatch Repair–Deficient/Microsatellite Instability–High Metastatic Colorectal Cancer

            Purpose Nivolumab provides clinical benefit (objective response rate [ORR], 31%; 95% CI, 20.8 to 42.9; disease control rate, 69%; 12-month overall survival [OS], 73%) in previously treated patients with DNA mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H) metastatic colorectal cancer (mCRC); nivolumab plus ipilimumab may improve these outcomes. Efficacy and safety results for the nivolumab plus ipilimumab cohort of CheckMate-142, the largest single-study report of an immunotherapy combination in dMMR/MSI-H mCRC, are reported. Patients and Methods Patients received nivolumab 3 mg/kg plus ipilimumab 1 mg/kg once every 3 weeks (four doses) followed by nivolumab 3 mg/kg once every 2 weeks. Primary end point was investigator-assessed ORR. Results Of 119 patients, 76% had received ≥ two prior systemic therapies. At median follow-up of 13.4 months, investigator-assessed ORR was 55% (95% CI, 45.2 to 63.8), and disease control rate for ≥ 12 weeks was 80%. Median duration of response was not reached; most responses (94%) were ongoing at data cutoff. Progression-free survival rates were 76% (9 months) and 71% (12 months); respective OS rates were 87% and 85%. Statistically significant and clinically meaningful improvements were observed in patient-reported outcomes, including functioning, symptoms, and quality of life. Grade 3 to 4 treatment-related adverse events (AEs) occurred in 32% of patients and were manageable. Patients (13%) who discontinued treatment because of study drug-related AEs had an ORR (63%) consistent with that of the overall population. Conclusion Nivolumab plus ipilimumab demonstrated high response rates, encouraging progression-free survival and OS at 12 months, manageable safety, and meaningful improvements in key patient-reported outcomes. Indirect comparisons suggest combination therapy provides improved efficacy relative to anti-programmed death-1 monotherapy and has a favorable benefit-risk profile. Nivolumab plus ipilimumab provides a promising new treatment option for patients with dMMR/MSI-H mCRC.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Enhanced secondary analysis of survival data: reconstructing the data from published Kaplan-Meier survival curves

              Background The results of Randomized Controlled Trials (RCTs) on time-to-event outcomes that are usually reported are median time to events and Cox Hazard Ratio. These do not constitute the sufficient statistics required for meta-analysis or cost-effectiveness analysis, and their use in secondary analyses requires strong assumptions that may not have been adequately tested. In order to enhance the quality of secondary data analyses, we propose a method which derives from the published Kaplan Meier survival curves a close approximation to the original individual patient time-to-event data from which they were generated. Methods We develop an algorithm that maps from digitised curves back to KM data by finding numerical solutions to the inverted KM equations, using where available information on number of events and numbers at risk. The reproducibility and accuracy of survival probabilities, median survival times and hazard ratios based on reconstructed KM data was assessed by comparing published statistics (survival probabilities, medians and hazard ratios) with statistics based on repeated reconstructions by multiple observers. Results The validation exercise established there was no material systematic error and that there was a high degree of reproducibility for all statistics. Accuracy was excellent for survival probabilities and medians, for hazard ratios reasonable accuracy can only be obtained if at least numbers at risk or total number of events are reported. Conclusion The algorithm is a reliable tool for meta-analysis and cost-effectiveness analyses of RCTs reporting time-to-event data. It is recommended that all RCTs should report information on numbers at risk and total number of events alongside KM curves.
                Bookmark

                Author and article information

                Journal
                JAMA Netw Open
                JAMA Netw Open
                JAMA Netw Open
                JAMA Network Open
                American Medical Association
                2574-3805
                12 January 2021
                January 2021
                12 January 2021
                : 4
                : 1
                : e2033441
                Affiliations
                [1 ]Department of Hematology/Oncology, Yale University School of Medicine, New Haven, Connecticut
                [2 ]Department of Hematology/Oncology, University of Pennsylvania Perelman School of Medicine, Philadelphia
                [3 ]Yale Cancer Outcomes, Public Policy and Effectiveness Research Center, New Haven, Connecticut
                Author notes
                Article Information
                Accepted for Publication: November 20, 2020.
                Published: January 12, 2021. doi:10.1001/jamanetworkopen.2020.33441
                Open Access: This is an open access article distributed under the terms of the CC-BY License. © 2021 Patel KK et al. JAMA Network Open.
                Corresponding Author: Scott F. Huntington, MD, MPH, MSc, Department of Hematology/Oncology, Yale University School of Medicine, 333 Cedar St, PO Box 208028, New Haven, CT 06520 ( scott.huntington@ 123456yale.edu ).
                Author Contributions: Mr Patel and Dr Huntington had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.
                Concept and design: Patel, Huntington.
                Acquisition, analysis, or interpretation of data: All authors.
                Drafting of the manuscript: Patel, Huntington.
                Critical revision of the manuscript for important intellectual content: All authors.
                Statistical analysis: Patel.
                Obtained funding: Patel.
                Supervision: Huntington.
                Conflict of Interest Disclosures: Dr Stein reported receiving personal fees from Genentech, QED, Bayer, and Esiai outside the submitted work. Dr Lacy reported receiving personal fees from AstraZeneca Advisory Board, Celgene Advisory Board, Merck Advisory Board, Deciphera Advisory Board, and Ipsen Advisory Board outside the submitted work. Dr O’Hara reported receiving grants from Bristol Myers Squibb; nonfinancial support from AstraZeneca; grants from Celldex; and personal fees from Exelixis, Karyopharm, and Geneos outside the submitted work. Dr Huntington reported receiving grants from the American Society of Hematology during the conduct of the study as well as personal fees from Pharmacyclics, AbbVie, Flatiron Health, Genentech, AstraZeneca, and Bayer outside the submitted work.
                Funding/Support: This study was funded in part by a Physician-Scientist Career Development Award from the American Society of Hematology (Mr Patel).
                Role of the Funder/Sponsor: The funder had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.
                Article
                zoi201020
                10.1001/jamanetworkopen.2020.33441
                7804917
                33433598
                d4b67c16-746e-4fab-9318-16ad407f8901
                Copyright 2021 Patel KK et al. JAMA Network Open.

                This is an open access article distributed under the terms of the CC-BY License.

                History
                : 16 July 2020
                : 20 November 2020
                Categories
                Research
                Original Investigation
                Online Only
                Oncology

                Comments

                Comment on this article