Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 55(1): 56-64.e4, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-34986342

RESUMO

We evaluated the impact of class I and class II human leukocyte antigen (HLA) genotypes, heterozygosity, and diversity on the efficacy of pembrolizumab. Seventeen pembrolizumab clinical trials across eight tumor types and one basket trial in patients with advanced solid tumors were included (n > 3,500 analyzed). Germline DNA was genotyped using a custom genotyping array. HLA diversity (measured by heterozygosity and evolutionary divergence) across class I loci was not associated with improved response to pembrolizumab, either within each tumor type evaluated or across all patients. Similarly, HLA heterozygosity at each class I and class II gene was not associated with response to pembrolizumab after accounting for the number of tests conducted. No conclusive association between HLA genotype and response to pembrolizumab was identified in this dataset. Germline HLA genotype or diversity alone is not an important independent determinant of response to pembrolizumab and should not be used for clinical decision-making in patients treated with pembrolizumab.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Genótipo , Mutação em Linhagem Germinativa/genética , Antígenos HLA/genética , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias/tratamento farmacológico , Fatores Etários , Feminino , Estudos de Associação Genética , Heterozigoto , Humanos , Masculino , Neoplasias/diagnóstico , Neoplasias/mortalidade , Polimorfismo Genético , Prognóstico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Fatores Sexuais , Análise de Sobrevida , Resultado do Tratamento
2.
Am J Hum Genet ; 109(3): 433-445, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35196515

RESUMO

Biobanks linked to massive, longitudinal electronic health record (EHR) data make numerous new genetic research questions feasible. One among these is the study of biomarker trajectories. For example, high blood pressure measurements over visits strongly predict stroke onset, and consistently high fasting glucose and Hb1Ac levels define diabetes. Recent research reveals that not only the mean level of biomarker trajectories but also their fluctuations, or within-subject (WS) variability, are risk factors for many diseases. Glycemic variation, for instance, is recently considered an important clinical metric in diabetes management. It is crucial to identify the genetic factors that shift the mean or alter the WS variability of a biomarker trajectory. Compared to traditional cross-sectional studies, trajectory analysis utilizes more data points and captures a complete picture of the impact of time-varying factors, including medication history and lifestyle. Currently, there are no efficient tools for genome-wide association studies (GWASs) of biomarker trajectories at the biobank scale, even for just mean effects. We propose TrajGWAS, a linear mixed effect model-based method for testing genetic effects that shift the mean or alter the WS variability of a biomarker trajectory. It is scalable to biobank data with 100,000 to 1,000,000 individuals and many longitudinal measurements and robust to distributional assumptions. Simulation studies corroborate that TrajGWAS controls the type I error rate and is powerful. Analysis of eleven biomarkers measured longitudinally and extracted from UK Biobank primary care data for more than 150,000 participants with 1,800,000 observations reveals loci that significantly alter the mean or WS variability.


Assuntos
Bancos de Espécimes Biológicos , Estudo de Associação Genômica Ampla , Biomarcadores , Estudos Transversais , Registros Eletrônicos de Saúde , Humanos , Estudos Longitudinais
3.
Biostatistics ; 25(2): 504-520, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-36897773

RESUMO

Identifying genotype-by-environment interaction (GEI) is challenging because the GEI analysis generally has low power. Large-scale consortium-based studies are ultimately needed to achieve adequate power for identifying GEI. We introduce Multi-Trait Analysis of Gene-Environment Interactions (MTAGEI), a powerful, robust, and computationally efficient framework to test gene-environment interactions on multiple traits in large data sets, such as the UK Biobank (UKB). To facilitate the meta-analysis of GEI studies in a consortium, MTAGEI efficiently generates summary statistics of genetic associations for multiple traits under different environmental conditions and integrates the summary statistics for GEI analysis. MTAGEI enhances the power of GEI analysis by aggregating GEI signals across multiple traits and variants that would otherwise be difficult to detect individually. MTAGEI achieves robustness by combining complementary tests under a wide spectrum of genetic architectures. We demonstrate the advantages of MTAGEI over existing single-trait-based GEI tests through extensive simulation studies and the analysis of the whole exome sequencing data from the UKB.


Assuntos
Interação Gene-Ambiente , Estudo de Associação Genômica Ampla , Humanos , Fenótipo , Simulação por Computador
4.
Brief Bioinform ; 25(1)2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-38152980

RESUMO

Polygenic risk scores (PRSs) have emerged as promising tools for the prediction of human diseases and complex traits in disease genome-wide association studies (GWAS). Applying PRSs to pharmacogenomics (PGx) studies has begun to show great potential for improving patient stratification and drug response prediction. However, there are unique challenges that arise when applying PRSs to PGx GWAS beyond those typically encountered in disease GWAS (e.g. Eurocentric or trans-ethnic bias). These challenges include: (i) the lack of knowledge about whether PGx or disease GWAS/variants should be used in the base cohort (BC); (ii) the small sample sizes in PGx GWAS with corresponding low power and (iii) the more complex PRS statistical modeling required for handling both prognostic and predictive effects simultaneously. To gain insights in this landscape about the general trends, challenges and possible solutions, we first conduct a systematic review of both PRS applications and PRS method development in PGx GWAS. To further address the challenges, we propose (i) a novel PRS application strategy by leveraging both PGx and disease GWAS summary statistics in the BC for PRS construction and (ii) a new Bayesian method (PRS-PGx-Bayesx) to reduce Eurocentric or cross-population PRS prediction bias. Extensive simulations are conducted to demonstrate their advantages over existing PRS methods applied in PGx GWAS. Our systematic review and methodology research work not only highlights current gaps and key considerations while applying PRS methods to PGx GWAS, but also provides possible solutions for better PGx PRS applications and future research.


Assuntos
Estratificação de Risco Genético , Estudo de Associação Genômica Ampla , Humanos , Teorema de Bayes , Predisposição Genética para Doença , Herança Multifatorial , Farmacogenética , Revisões Sistemáticas como Assunto
5.
Brief Bioinform ; 24(1)2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36545787

RESUMO

Genotype-by-environment interaction (GEI or GxE) plays an important role in understanding complex human traits. However, it is usually challenging to detect GEI signals efficiently and accurately while adjusting for population stratification and sample relatedness in large-scale genome-wide association studies (GWAS). Here we propose a fast and powerful linear mixed model-based approach, fastGWA-GE, to test for GEI effect and G + GxE joint effect. Our extensive simulations show that fastGWA-GE outperforms other existing GEI test methods by controlling genomic inflation better, providing larger power and running hundreds to thousands of times faster. We performed a fastGWA-GE analysis of ~7.27 million variants on 452 249 individuals of European ancestry for 13 quantitative traits and five environment variables in the UK Biobank GWAS data and identified 96 significant signals (72 variants across 57 loci) with GEI test P-values < 1 × 10-9, including 27 novel GEI associations, which highlights the effectiveness of fastGWA-GE in GEI signal discovery in large-scale GWAS.


Assuntos
Interação Gene-Ambiente , Estudo de Associação Genômica Ampla , Humanos , Fenótipo , Genótipo , Modelos Lineares , Polimorfismo de Nucleotídeo Único
6.
Brief Bioinform ; 24(4)2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37200155

RESUMO

Polygenic risk score (PRS) has been recently developed for predicting complex traits and drug responses. It remains unknown whether multi-trait PRS (mtPRS) methods, by integrating information from multiple genetically correlated traits, can improve prediction accuracy and power for PRS analysis compared with single-trait PRS (stPRS) methods. In this paper, we first review commonly used mtPRS methods and find that they do not directly model the underlying genetic correlations among traits, which has been shown to be useful in guiding multi-trait association analysis in the literature. To overcome this limitation, we propose a mtPRS-PCA method to combine PRSs from multiple traits with weights obtained from performing principal component analysis (PCA) on the genetic correlation matrix. To accommodate various genetic architectures covering different effect directions, signal sparseness and across-trait correlation structures, we further propose an omnibus mtPRS method (mtPRS-O) by combining P values from mtPRS-PCA, mtPRS-ML (mtPRS based on machine learning) and stPRSs using Cauchy Combination Test. Our extensive simulation studies show that mtPRS-PCA outperforms other mtPRS methods in both disease and pharmacogenomics (PGx) genome-wide association studies (GWAS) contexts when traits are similarly correlated, with dense signal effects and in similar effect directions, and mtPRS-O is consistently superior to most other methods due to its robustness under various genetic architectures. We further apply mtPRS-PCA, mtPRS-O and other methods to PGx GWAS data from a randomized clinical trial in the cardiovascular domain and demonstrate performance improvement of mtPRS-PCA in both prediction accuracy and patient stratification as well as the robustness of mtPRS-O in PRS association test.


Assuntos
Estudo de Associação Genômica Ampla , Herança Multifatorial , Humanos , Estudo de Associação Genômica Ampla/métodos , Farmacogenética , Polimorfismo de Nucleotídeo Único , Fenótipo , Predisposição Genética para Doença
7.
Bioinformatics ; 39(1)2023 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-36661328

RESUMO

MOTIVATION: Pharmacogenomics (PGx) research holds the promise for detecting association between genetic variants and drug responses in randomized clinical trials, but it is limited by small populations and thus has low power to detect signals. It is critical to increase the power of PGx genome-wide association studies (GWAS) with small sample sizes so that variant-drug-response association discoveries are not limited to common variants with extremely large effect. RESULTS: In this article, we first discuss the challenges of PGx GWAS studies and then propose the adaptively weighted joint test (AWOT) and Cauchy Weighted jOint Test (CWOT), which are two flexible and robust joint tests of the single nucleotide polymorphism main effect and genotype-by-treatment interaction effect for continuous and binary endpoints. Two analytic procedures are proposed to accurately calculate the joint test P-value. We evaluate AWOT and CWOT through extensive simulations under various scenarios. The results show that the proposed AWOT and CWOT control type I error well and outperform existing methods in detecting the most interesting signal patterns in PGx settings (i.e. with strong genotype-by-treatment interaction effects, but weak genotype main effects). We demonstrate the value of AWOT and CWOT by applying them to the PGx GWAS from the Bezlotoxumab Clostridium difficile MODIFY I/II Phase 3 trials. AVAILABILITY AND IMPLEMENTATION: The R package COWT is publicly available on CRAN https://cran.r-project.org/web/packages/cwot/index.html. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Estudo de Associação Genômica Ampla , Farmacogenética , Estudo de Associação Genômica Ampla/métodos , Fenótipo , Genótipo , Polimorfismo de Nucleotídeo Único
8.
Clin Trials ; : 17407745231222448, 2024 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-38305269

RESUMO

In randomized clinical trials, analyses of time-to-event data without risk stratification, or with stratification based on pre-selected factors revealed at the end of the trial to be at most weakly associated with risk, are quite common. We caution that such analyses are likely delivering hazard ratio estimates that unwittingly dilute the evidence of benefit for the test relative to the control treatment. To make our case, first, we use a hypothetical scenario to contrast risk-unstratified and risk-stratified hazard ratios. Thereafter, we draw attention to the previously published 5-step stratified testing and amalgamation routine (5-STAR) approach in which a pre-specified treatment-blinded algorithm is applied to survival times from the trial to partition patients into well-separated risk strata using baseline covariates determined to be jointly strongly prognostic for event risk. After treatment unblinding, a treatment comparison is done within each risk stratum and stratum-level results are averaged for overall inference. For illustration, we use 5-STAR to reanalyze data for the primary and key secondary time-to-event endpoints from three published cardiovascular outcomes trials. The results show that the 5-STAR estimate is typically smaller (i.e. more in favor of the test treatment) than the originally reported (traditional) estimate. This is not surprising because 5-STAR mitigates the presumed dilution bias in the traditional hazard ratio estimate caused by no or inadequate risk stratification, as evidenced by two detailed examples. Pre-selection of stratification factors at the trial design stage to achieve adequate risk stratification for the analysis will often be challenging. In such settings, an objective risk stratification approach such as 5-STAR, which is partly aligned with guidance from the US Food and Drug Administration on covariate-adjustment in clinical trials, is worthy of consideration.

9.
Pharm Stat ; 22(6): 1076-1088, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37550963

RESUMO

Sample sizes of Phase 2 dose-finding studies, usually determined based on a power requirement to detect a significant dose-response relationship, will generally not provide adequate precision for Phase 3 target dose selection. We propose to calculate the sample size of a dose-finding study based on the probability of successfully identifying the target dose within an acceptable range (e.g., 80%-120% of the target) using the multiple comparison and modeling procedure (MCP-Mod). With the proposed approach, different design options for the Phase 2 dose-finding study can also be compared. Due to inherent uncertainty around an assumed true dose-response relationship, sensitivity analyses to assess the robustness of the sample size calculations to deviations from modeling assumptions are recommended. Planning for a hypothetical Phase 2 dose-finding study is used to illustrate the main points. Codes for the proposed approach is available at https://github.com/happysundae/posMCPMod.


Assuntos
Projetos de Pesquisa , Humanos , Tamanho da Amostra , Relação Dose-Resposta a Droga , Probabilidade , Incerteza
10.
Stat Med ; 41(21): 4227-4244, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-35799329

RESUMO

Personalized medicine, a paradigm of medicine tailored to a patient's characteristics, is an increasingly attractive field in health care. An important goal of personalized medicine is to identify a subgroup of patients, based on baseline covariates, that benefits more from the targeted treatment than other comparative treatments. Most of the current subgroup identification methods only focus on obtaining a subgroup with an enhanced treatment effect without paying attention to subgroup size. Yet, a clinically meaningful subgroup learning approach should identify the maximum number of patients who can benefit from the better treatment. In this article, we present an optimal subgroup selection rule (SSR) that maximizes the number of selected patients, and in the meantime, achieves the pre-specified clinically meaningful mean outcome, such as the average treatment effect. We derive two equivalent theoretical forms of the optimal SSR based on the contrast function that describes the treatment-covariates interaction in the outcome. We further propose a constrained policy tree search algorithm (CAPITAL) to find the optimal SSR within the interpretable decision tree class. The proposed method is flexible to handle multiple constraints that penalize the inclusion of patients with negative treatment effects, and to address time to event data using the restricted mean survival time as the clinically interesting mean outcome. Extensive simulations, comparison studies, and real data applications are conducted to demonstrate the validity and utility of our method.


Assuntos
Algoritmos , Medicina de Precisão , Humanos , Políticas , Medicina de Precisão/métodos , Projetos de Pesquisa
11.
Ann Intern Med ; 174(2): 221-228, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33090877

RESUMO

Several vaccine candidates to protect against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or coronavirus disease 2019 (COVID-19) have entered or will soon enter large-scale, phase 3, placebo-controlled randomized clinical trials. To facilitate harmonized evaluation and comparison of the efficacy of these vaccines, a general set of clinical endpoints is proposed, along with considerations to guide the selection of the primary endpoints on the basis of clinical and statistical reasoning. The plausibility that vaccine protection against symptomatic COVID-19 could be accompanied by a shift toward more SARS-CoV-2 infections that are asymptomatic is highlighted, as well as the potential implications of such a shift.


Assuntos
Vacinas contra COVID-19/uso terapêutico , COVID-19/prevenção & controle , Ensaios Clínicos Controlados Aleatórios como Assunto/métodos , Infecções Assintomáticas , COVID-19/diagnóstico , Teste para COVID-19 , Vacinas contra COVID-19/efeitos adversos , Ensaios Clínicos Fase III como Assunto/métodos , Humanos , SARS-CoV-2 , Índice de Gravidade de Doença
12.
Ann Intern Med ; 174(8): 1118-1125, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33844575

RESUMO

Multiple candidate vaccines to prevent COVID-19 have entered large-scale phase 3 placebo-controlled randomized clinical trials, and several have demonstrated substantial short-term efficacy. At some point after demonstration of substantial efficacy, placebo recipients should be offered the efficacious vaccine from their trial, which will occur before longer-term efficacy and safety are known. The absence of a placebo group could compromise assessment of longer-term vaccine effects. However, by continuing follow-up after vaccination of the placebo group, this study shows that placebo-controlled vaccine efficacy can be mathematically derived by assuming that the benefit of vaccination over time has the same profile for the original vaccine recipients and the original placebo recipients after their vaccination. Although this derivation provides less precise estimates than would be obtained by a standard trial where the placebo group remains unvaccinated, this proposed approach allows estimation of longer-term effect, including durability of vaccine efficacy and whether the vaccine eventually becomes harmful for some. Deferred vaccination, if done open-label, may lead to riskier behavior in the unblinded original vaccine group, confounding estimates of long-term vaccine efficacy. Hence, deferred vaccination via blinded crossover, where the vaccine group receives placebo and vice versa, would be the preferred way to assess vaccine durability and potential delayed harm. Deferred vaccination allows placebo recipients timely access to the vaccine when it would no longer be proper to maintain them on placebo, yet still allows important insights about immunologic and clinical effectiveness over time.


Assuntos
Vacinas contra COVID-19/administração & dosagem , COVID-19/prevenção & controle , Ensaios Clínicos Fase III como Assunto/normas , Ensaios Clínicos Controlados Aleatórios como Assunto/normas , Ensaios Clínicos Fase III como Assunto/métodos , Estudos Cross-Over , Método Duplo-Cego , Esquema de Medicação , Seguimentos , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto/métodos , Projetos de Pesquisa/normas , SARS-CoV-2 , Resultado do Tratamento
13.
Clin Infect Dis ; 73(8): 1540-1544, 2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-33340397

RESUMO

A large number of studies are being conducted to evaluate the efficacy and safety of candidate vaccines against coronavirus disease 2019 (COVID-19). Most phase 3 trials have adopted virologically confirmed symptomatic COVID-19 as the primary efficacy end point, although laboratory-confirmed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is also of interest. In addition, it is important to evaluate the effect of vaccination on disease severity. To provide a full picture of vaccine efficacy and make efficient use of available data, we propose using SARS-CoV-2 infection, symptomatic COVID-19, and severe COVID-19 as dual or triple primary end points. We demonstrate the advantages of this strategy through realistic simulation studies. Finally, we show how this approach can provide rigorous interim monitoring of the trials and efficient assessment of the durability of vaccine efficacy.


Assuntos
COVID-19 , Vacinas , Vacinas contra COVID-19 , Humanos , SARS-CoV-2 , Resultado do Tratamento
14.
Bioinformatics ; 36(10): 3162-3168, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32101275

RESUMO

MOTIVATION: It is of substantial interest to discover novel genetic markers that influence drug response in order to develop personalized treatment strategies that maximize therapeutic efficacy and safety. To help enable such discoveries, we focus on testing the association between the cumulative effect of multiple single nucleotide polymorphisms (SNPs) in a particular genomic region and a drug response of interest. However, the currently existing methods are either computational inefficient or not able to control type I error and provide decent power for whole exome or genome analysis in Pharmacogenetics (PGx) studies with small sample sizes. RESULTS: In this article, we propose the Composite Kernel Association Test (CKAT), a flexible and robust kernel machine-based approach to jointly test the genetic main effect and SNP-treatment interaction effect for SNP-sets in Pharmacogenetics (PGx) assessments embedded within randomized clinical trials. An analytic procedure is developed to accurately calculate the P-value so that computationally extensive procedures (e.g. permutation or perturbation) can be avoided. We evaluate CKAT through extensive simulation studies and application to the gene-level association test of the reduction in Clostridium difficile infection recurrence in patients treated with bezlotoxumab. The results demonstrate that the proposed CKAT controls type I error well for PGx studies, is efficient for whole exome/genome association analysis and provides better power performance than existing methods across multiple scenarios. AVAILABILITY AND IMPLEMENTATION: The R package CKAT is publicly available on CRAN https://cran.r-project.org/web/packages/CKAT/. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Farmacogenética , Polimorfismo de Nucleotídeo Único , Estudo de Associação Genômica Ampla , Genótipo , Humanos , Fenótipo , Ensaios Clínicos Controlados Aleatórios como Assunto
15.
Pharm Stat ; 20(4): 737-751, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33624407

RESUMO

A randomized trial allows estimation of the causal effect of an intervention compared to a control in the overall population and in subpopulations defined by baseline characteristics. Often, however, clinical questions also arise regarding the treatment effect in subpopulations of patients, which would experience clinical or disease related events post-randomization. Events that occur after treatment initiation and potentially affect the interpretation or the existence of the measurements are called intercurrent events in the ICH E9(R1) guideline. If the intercurrent event is a consequence of treatment, randomization alone is no longer sufficient to meaningfully estimate the treatment effect. Analyses comparing the subgroups of patients without the intercurrent events for intervention and control will not estimate a causal effect. This is well known, but post-hoc analyses of this kind are commonly performed in drug development. An alternative approach is the principal stratum strategy, which classifies subjects according to their potential occurrence of an intercurrent event on both study arms. We illustrate with examples that questions formulated through principal strata occur naturally in drug development and argue that approaching these questions with the ICH E9(R1) estimand framework has the potential to lead to more transparent assumptions as well as more adequate analyses and conclusions. In addition, we provide an overview of assumptions required for estimation of effects in principal strata. Most of these assumptions are unverifiable and should hence be based on solid scientific understanding. Sensitivity analyses are needed to assess robustness of conclusions.


Assuntos
Desenvolvimento de Medicamentos , Projetos de Pesquisa , Causalidade , Interpretação Estatística de Dados , Humanos
16.
Stat Med ; 39(30): 4724-4744, 2020 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-32954531

RESUMO

Randomized clinical trials are often designed to assess whether a test treatment prolongs survival relative to a control treatment. Increased patient heterogeneity, while desirable for generalizability of results, can weaken the ability of common statistical approaches to detect treatment differences, potentially hampering the regulatory approval of safe and efficacious therapies. A novel solution to this problem is proposed. A list of baseline covariates that have the potential to be prognostic for survival under either treatment is pre-specified in the analysis plan. At the analysis stage, using all observed survival times but blinded to patient-level treatment assignment, "noise" covariates are removed with elastic net Cox regression. The shortened covariate list is used by a conditional inference tree algorithm to segment the heterogeneous trial population into subpopulations of prognostically homogeneous patients (risk strata). After patient-level treatment unblinding, a treatment comparison is done within each formed risk stratum and stratum-level results are combined for overall statistical inference. The impressive power-boosting performance of our proposed 5-step stratified testing and amalgamation routine (5-STAR), relative to that of the logrank test and other common approaches that do not leverage inherently structured patient heterogeneity, is illustrated using a hypothetical and two real datasets along with simulation results. Furthermore, the importance of reporting stratum-level comparative treatment effects (time ratios from accelerated failure time model fits in conjunction with model averaging and, as needed, hazard ratios from Cox proportional hazard model fits) is highlighted as a potential enabler of personalized medicine. An R package is available at https://github.com/rmarceauwest/fiveSTAR.


Assuntos
Ensaios Clínicos Controlados Aleatórios como Assunto , Simulação por Computador , Humanos , Prognóstico , Modelos de Riscos Proporcionais , Análise de Sobrevida
17.
Clin Trials ; 16(4): 339-344, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30977390

RESUMO

In the second half of 2014, the Steering Committee of the International Council for Harmonisation endorsed the formation of an expert working group to develop an addendum to the International Council for Harmonisation E9 guideline (Statistical Principles for Clinical Trials). The addendum was to focus on two clinical trial topics: estimands and sensitivity analysis. A draft of the addendum, referred to as E9/R1, was developed by the expert working group and made available for public comments across the International Council for Harmonisation regions in the second half of 2017. A structured framework for clinical trial design and analysis proposed in the draft addendum are briefly described, including four key inputs for developing objective-driven estimands and strategies for tackling one of the inputs ('intercurrent events'). The proposed framework aligns each clinical trial objective with the corresponding statistical target of estimation (estimand), trial design and data to be collected, main method of estimation/inference, and sensitivity analysis to pressure test key analytic assumption(s) in the main analysis. A case study from the diabetes therapeutic area illustrates how the framework can be implemented in practice. International Council for Harmonisation E9/R1 is expected to enable better planning, conduct, analysis, and interpretation of randomised clinical trials. This will facilitate improvements in new drug applications and strengthen understanding of decision making by regulatory authorities and advisory committees.


Assuntos
Comitês de Monitoramento de Dados de Ensaios Clínicos/normas , Ensaios Clínicos como Assunto/estatística & dados numéricos , Interpretação Estatística de Dados , Guias como Assunto , Indústria Farmacêutica , Humanos , Modelos Estatísticos , Ensaios Clínicos Controlados Aleatórios como Assunto/estatística & dados numéricos , Projetos de Pesquisa , Estatística como Assunto
18.
Pharm Stat ; 18(3): 366-376, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30706642

RESUMO

The stratified Cox model is commonly used for stratified clinical trials with time-to-event endpoints. The estimated log hazard ratio is approximately a weighted average of corresponding stratum-specific Cox model estimates using inverse-variance weights; the latter are optimal only under the (often implausible) assumption of a constant hazard ratio across strata. Focusing on trials with limited sample sizes (50-200 subjects per treatment), we propose an alternative approach in which stratum-specific estimates are obtained using a refined generalized logrank (RGLR) approach and then combined using either sample size or minimum risk weights for overall inference. Our proposal extends the work of Mehrotra et al, to incorporate the RGLR statistic, which outperforms the Cox model in the setting of proportional hazards and small samples. This work also entails development of a remarkably accurate plug-in formula for the variance of RGLR-based estimated log hazard ratios. We demonstrate using simulations that our proposed two-step RGLR analysis delivers notably better results through smaller estimation bias and mean squared error and larger power than the stratified Cox model analysis when there is a treatment-by-stratum interaction, with similar performance when there is no interaction. Additionally, our method controls the type I error rate while the stratified Cox model does not in small samples. We illustrate our method using data from a clinical trial comparing two treatments for colon cancer.


Assuntos
Simulação por Computador/estatística & dados numéricos , Determinação de Ponto Final/estatística & dados numéricos , Ensaios Clínicos Controlados Aleatórios como Assunto/estatística & dados numéricos , Neoplasias do Colo/epidemiologia , Neoplasias do Colo/terapia , Determinação de Ponto Final/métodos , Humanos , Modelos de Riscos Proporcionais , Ensaios Clínicos Controlados Aleatórios como Assunto/métodos , Tamanho da Amostra
19.
Stat Med ; 37(25): 3547-3556, 2018 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-29900572

RESUMO

The medical literature contains numerous examples of randomized clinical trials with time-to-event endpoints in which large numbers of events accrued over relatively short follow-up periods, resulting in many tied event times. A generally common feature across such examples was that the logrank test was used for hypothesis testing and the Cox proportional hazards model was used for hazard ratio estimation. We caution that this common practice is particularly risky in the setting of many tied event times for two reasons. First, the estimator of the hazard ratio can be severely biased if the Breslow tie-handling approximation for the Cox model (the default in SAS and Stata software) is used. Second, the 95% confidence interval for the hazard ratio can include one even when the corresponding logrank test p-value is less than 0.05. To help establish a better practice, with applicability for both superiority and noninferiority trials, we use theory and simulations to contrast Wald and score tests based on well-known tie-handling approximations for the Cox model. Our recommendation is to report the Wald test p-value and corresponding confidence interval based on the Efron approximation. The recommended test is essentially as powerful as the logrank test, the accompanying point and interval estimates of the hazard ratio have excellent statistical properties even in settings with many tied event times, inferential alignment between the p-value and confidence interval is guaranteed, and implementation is straightforward using commonly used software.


Assuntos
Ensaios Clínicos como Assunto/métodos , Modelos de Riscos Proporcionais , Intervalos de Confiança , Interpretação Estatística de Dados , Humanos , Fatores de Tempo
20.
Stat Med ; 37(23): 3280-3292, 2018 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-29888552

RESUMO

Two-period two-treatment (2×2) crossover designs are commonly used in clinical trials. For continuous endpoints, it has been shown that baseline (pretreatment) measurements collected before the start of each treatment period can be useful in improving the power of the analysis. Methods to achieve a corresponding gain for censored time-to-event endpoints have not been adequately studied. We propose a method in which censored values are treated as missing data and multiply imputed using prespecified parametric event time models. The event times in each imputed data set are then log-transformed and analyzed using a linear model suitable for a 2×2 crossover design with continuous endpoints, with the difference in period-specific baselines included as a covariate. Results obtained from the imputed data sets are synthesized for point and confidence interval estimation of the treatment ratio of geometric mean event times using model averaging in conjunction with Rubin's combination rule. We use simulations to illustrate the favorable operating characteristics of our method relative to two other methods for crossover trials with censored time-to-event data, ie, a hierarchical rank test that ignores the baselines and a stratified Cox model that uses each study subject as a stratum and includes period-specific baselines as a covariate. Application to a real data example is provided.


Assuntos
Estudos Cross-Over , Ensaios Clínicos Controlados Aleatórios como Assunto/estatística & dados numéricos , Análise de Variância , Bioestatística , Simulação por Computador , Interpretação Estatística de Dados , Determinação de Ponto Final/estatística & dados numéricos , Humanos , Modelos Lineares , Modelos Estatísticos , Modelos de Riscos Proporcionais , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa