59 research outputs found

    Bayesian meta-analysis for evaluating treatment effectiveness in biomarker subgroups using trials of mixed patient populations

    Full text link
    During drug development, evidence can emerge to suggest a treatment is more effective in a specific patient subgroup. Whilst early trials may be conducted in biomarker-mixed populations, later trials are more likely to enrol biomarker-positive patients alone, thus leading to trials of the same treatment investigated in different populations. When conducting a meta-analysis, a conservative approach would be to combine only trials conducted in the biomarker-positive subgroup. However, this discards potentially useful information on treatment effects in the biomarker-positive subgroup concealed within observed treatment effects in biomarker-mixed populations. We extend standard random-effects meta-analysis to combine treatment effects obtained from trials with different populations to estimate pooled treatment effects in a biomarker subgroup of interest. The model assumes a systematic difference in treatment effects between biomarker-positive and biomarker-negative subgroups, which is estimated from trials which report either or both treatment effects. The estimated systematic difference and proportion of biomarker-negative patients in biomarker-mixed studies are used to interpolate treatment effects in the biomarker-positive subgroup from observed treatment effects in the biomarker-mixed population. The developed methods are applied to an illustrative example in metastatic colorectal cancer and evaluated in a simulation study. In the example, the developed method resulted in improved precision of the pooled treatment effect estimate compared to standard random-effects meta-analysis of trials investigating only biomarker-positive patients. The simulation study confirmed that when the systematic difference in treatment effects between biomarker subgroups is not very large, the developed method can improve precision of estimation of pooled treatment effects while maintaining low bias

    A novel approach to bivariate meta-analysis of binary outcomes and its application in the context of surrogate endpoints

    Get PDF
    Bivariate meta-analysis provides a useful framework for combining information across related studies and has been widely utilised to combine evidence from clinical studies in order to evaluate treatment efficacy. Bivariate meta-analysis has also been used to investigate surrogacy patterns between treatment effects on the surrogate and the final outcome. Surrogate endpoints play an important role in drug development when they can be used to measure treatment effect early compared to the final clinical outcome and to predict clinical benefit or harm. The standard bivariate meta-analytic approach models the observed treatment effects on the surrogate and final outcomes jointly, at both the within-study and between-studies levels, using a bivariate normal distribution. For binomial data a normal approximation can be used on log odds ratio scale, however, this method may lead to biased results when the proportions of events are close to one or zero, affecting the validation of surrogate endpoints. In this paper, two Bayesian meta-analytic approaches are introduced which allow for modelling the within-study variability using binomial data directly. The first uses independent binomial likelihoods to model the within-study variability avoiding to approximate the observed treatment effects, however, ignores the within-study association. The second, models the summarised events in each arm jointly using a bivariate copula with binomial marginals. This allows the model to take into account the within-study association through the copula dependence parameter. We applied the methods to an illustrative example in chronic myeloid leukemia to investigate the surrogate relationship between complete cytogenetic response (CCyR) and event-free-survival (EFS).Comment: 20 pages, 6 figure

    Bivariate network meta-analysis for surrogate endpoint evaluation

    Full text link
    Surrogate endpoints are very important in regulatory decision-making in healthcare, in particular if they can be measured early compared to the long-term final clinical outcome and act as good predictors of clinical benefit. Bivariate meta-analysis methods can be used to evaluate surrogate endpoints and to predict the treatment effect on the final outcome from the treatment effect measured on a surrogate endpoint. However, candidate surrogate endpoints are often imperfect, and the level of association between the treatment effects on the surrogate and final outcomes may vary between treatments. This imposes a limitation on the pairwise methods which do not differentiate between the treatments. We develop bivariate network meta-analysis (bvNMA) methods which combine data on treatment effects on the surrogate and final outcomes, from trials investigating heterogeneous treatment contrasts. The bvNMA methods estimate the effects on both outcomes for all treatment contrasts individually in a single analysis. At the same time, they allow us to model the surrogacy patterns across multiple trials (different populations) within a treatment contrast and across treatment contrasts, thus enabling predictions of the treatment effect on the final outcome for a new study in a new population or investigating a new treatment. Modelling assumptions about the between-studies heterogeneity and the network consistency, and their impact on predictions, are investigated using simulated data and an illustrative example in advanced colorectal cancer. When the strength of the surrogate relationships varies across treatment contrasts, bvNMA has the advantage of identifying treatments for which surrogacy holds, thus leading to better predictions

    Combining individual patient data from randomized and non-randomized studies to predict real-world effectiveness of interventions.

    Get PDF
    Meta-analysis of randomized controlled trials is generally considered the most reliable source of estimates of relative treatment effects. However, in the last few years, there has been interest in using non-randomized studies to complement evidence from randomized controlled trials. Several meta-analytical models have been proposed to this end. Such models mainly focussed on estimating the average relative effects of interventions. In real-life clinical practice, when deciding on how to treat a patient, it might be of great interest to have personalized predictions of absolute outcomes under several available treatment options. This paper describes a general framework for developing models that combine individual patient data from randomized controlled trials and non-randomized study when aiming to predict outcomes for a set of competing medical interventions applied in real-world clinical settings. We also discuss methods for measuring the models' performance to identify the optimal model to use in each setting. We focus on the case of continuous outcomes and illustrate our methods using a data set from rheumatoid arthritis, comprising patient-level data from three randomized controlled trials and two registries from Switzerland and Britain

    Network meta-analysis of multiple outcome measures accounting for borrowing of information across outcomes

    Get PDF
    Background: Network meta-analysis (NMA) enables simultaneous comparison of multiple treatments while preserving randomisation. When summarising evidence to inform an economic evaluation, it is important that the analysis accurately reflects the dependency structure within the data, as correlations between outcomes may have implication for estimating the net benefit associated with treatment. A multivariate NMA offers a framework for evaluating multiple treatments across multiple outcome measures while accounting for the correlation structure between outcomes. Methods: The standard NMA model is extended to multiple outcome settings in two stages. In the first stage, information is borrowed across outcomes as well across studies through modelling the within-study and between-study correlation structure. In the second stage, we make use of the additional assumption that intervention effects are exchangeable between outcomes to predict effect estimates for all outcomes, including effect estimates on outcomes where evidence is either sparse or the treatment had not been considered by any one of the studies included in the analysis. We apply the methods to binary outcome data from a systematic review evaluating the effectiveness of nine home safety interventions on uptake of three poisoning prevention practices (safe storage of medicines, safe storage of other household products, and possession of poison centre control telephone number) in households with children. Analyses are conducted in WinBUGS using Markov Chain Monte Carlo (MCMC) simulations. Results: Univariate and the first stage multivariate models produced broadly similar point estimates of intervention effects but the uncertainty around the multivariate estimates varied depending on the prior distribution specified for the between-study covariance structure. The second stage multivariate analyses produced more precise effect estimates while enabling intervention effects to be predicted for all outcomes, including intervention effects on outcomes not directly considered by the studies included in the analysis. Conclusions: Accounting for the dependency between outcomes in a multivariate meta-analysis may or may not improve the precision of effect estimates from a network meta-analysis compared to analysing each outcome separately

    Hierarchical network meta-analysis models for synthesis of evidence from randomised and non-randomised studies

    Get PDF
    BACKGROUND: With the increased interest in the inclusion of non-randomised data in network meta-analyses (NMAs) of randomised controlled trials (RCTs), analysts need to consider the implications of the differences in study designs as such data can be prone to increased bias due to the lack of randomisation and unmeasured confounding. This study aims to explore and extend a number of NMA models that account for the differences in the study designs, assessing their impact on the effect estimates and uncertainty. METHODS: Bayesian random-effects meta-analytic models, including naïve pooling and hierarchical models differentiating between the study designs, were extended to allow for the treatment class effect and accounting for bias, with further extensions allowing for bias terms to vary depending on the treatment class. Models were applied to an illustrative example in type 2 diabetes; using data from a systematic review of RCTs and non-randomised studies of two classes of glucose-lowering medications: sodium-glucose co-transporter 2 inhibitors and glucagon-like peptide-1 receptor agonists. RESULTS: Across all methods, the estimated mean differences in glycated haemoglobin after 24 and 52 weeks remained similar with the inclusion of observational data. The uncertainty around these estimates reduced when conducting naïve pooling, compared to NMA of RCT data alone, and remained similar when applying hierarchical model allowing for class effect. However, the uncertainty around these effect estimates increased when fitting hierarchical models allowing for the differences in study design. The impact on uncertainty varied between treatments when applying the bias adjustment models. Hierarchical models and bias adjustment models all provided a better fit in comparison to the naïve-pooling method. CONCLUSIONS: Hierarchical and bias adjustment NMA models accounting for study design may be more appropriate when conducting a NMA of RCTs and observational studies. The degree of uncertainty around the effectiveness estimates varied depending on the method but use of hierarchical models accounting for the study design resulted in increased uncertainty. Inclusion of non-randomised data may, however, result in inferences that are more generalisable and the models accounting for the differences in the study design allow for more detailed and appropriate modelling of complex data, preventing overly optimistic conclusions

    Bridging disconnected networks of first and second lines of biologic therapies in rheumatoid arthritis with registry data : Bayesian evidence synthesis with target trial emulation

    Get PDF
    We aim to utilise real world data in evidence synthesis to optimise an evidence base for the effectiveness of biologic therapies in rheumatoid arthritis in order to allow for evidence on first-line therapies to inform second-line effectiveness estimates. We use data from the British Society for Rheumatology Biologics Register for Rheumatoid Arthritis (BSRBR-RA) to supplement RCT evidence obtained from the literature, by emulating target trials of treatment sequences to estimate treatment effects in each line of therapy. Treatment effects estimates from the target trials inform a bivariate network meta-analysis (NMA) of first and second-line treatments. Summary data were obtained from 21 trials of biologic therapies including 2 for second-line treatment and results from six emulated target trials of both treatment lines. Bivariate NMA resulted in a decrease in uncertainty around the effectiveness estimates of the second-line therapies, when compared to the results of univariate NMA, and allowed for predictions of treatment effects not evaluated in second-line RCTs. Bivariate NMA provides effectiveness estimates for all treatments in first- and second-line, including predicted effects in second-line where these estimates did not exist in the data. This novel methodology may have further applications, for example for bridging networks of trials in children and adults. [Abstract copyright: Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.

    Methods for the inclusion of real-world evidence in network meta-analysis

    Get PDF
    Background Network Meta-Analysis (NMA) is a key component of submissions to reimbursement agencies world-wide, especially when there is limited direct head-to-head evidence for multiple technologies from randomised controlled trials (RCTs). Many NMAs include only data from RCTs. However, real-world evidence (RWE) is also becoming widely recognised as a valuable source of clinical data. This study aims to investigate methods for the inclusion of RWE in NMA and its impact on the level of uncertainty around the effectiveness estimates, with particular interest in effectiveness of fingolimod. Methods A range of methods for inclusion of RWE in evidence synthesis were investigated by applying them to an illustrative example in relapsing remitting multiple sclerosis (RRMS). A literature search to identify RCTs and RWE evaluating treatments in RRMS was conducted. To assess the impact of inclusion of RWE on the effectiveness estimates, Bayesian hierarchical and adapted power prior models were applied. The effect of the inclusion of RWE was investigated by varying the degree of down weighting of this part of evidence by the use of a power prior. Results Whilst the inclusion of the RWE led to an increase in the level of uncertainty surrounding effect estimates in this example, this depended on the method of inclusion adopted for the RWE. ‘Power prior’ NMA model resulted in stable effect estimates for fingolimod yet increasing the width of the credible intervals with increasing weight given to RWE data. The hierarchical NMA models were effective in allowing for heterogeneity between study designs, however, this also increased the level of uncertainty. Conclusion The ‘power prior’ method for the inclusion of RWE in NMAs indicates that the degree to which RWE is taken into account can have a significant impact on the overall level of uncertainty. The hierarchical modelling approach further allowed for accommodating differences between study types. Consequently, further work investigating both empirical evidence for biases associated with individual RWE studies and methods of elicitation from experts on the extent of such biases is warranted
    • …
    corecore