Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Clin Trials ; : 17407745231222448, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38305269

ABSTRACT

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.

2.
Biostatistics ; 25(2): 504-520, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-36897773

ABSTRACT

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.


Subject(s)
Gene-Environment Interaction , Genome-Wide Association Study , Humans , Phenotype , Computer Simulation
3.
Brief Bioinform ; 25(1)2023 11 22.
Article in English | MEDLINE | ID: mdl-38152980

ABSTRACT

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.


Subject(s)
Genetic Risk Score , Genome-Wide Association Study , Humans , Bayes Theorem , Genetic Predisposition to Disease , Multifactorial Inheritance , Pharmacogenetics , Systematic Reviews as Topic
4.
Pharm Stat ; 22(6): 1076-1088, 2023.
Article in English | MEDLINE | ID: mdl-37550963

ABSTRACT

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.


Subject(s)
Research Design , Humans , Sample Size , Dose-Response Relationship, Drug , Probability , Uncertainty
5.
Stat Methods Med Res ; 32(10): 1961-1972, 2023 10.
Article in English | MEDLINE | ID: mdl-37519295

ABSTRACT

In the era of precision medicine, many biomarkers have been discovered to be associated with drug efficacy and safety responses, which can be used for patient stratification and drug response prediction. Due to the small sample size and limited power of randomized clinical studies, meta-analysis is usually conducted to aggregate all available studies to maximize the power for identifying prognostic and predictive biomarkers. However, it is often challenging to find an independent study to replicate the discoveries from the meta-analysis (e.g. meta-analysis of pharmacogenomics genome-wide association studies (PGx GWAS)), which seriously limits the potential impacts of the discovered biomarkers. To overcome this challenge, we develop a novel statistical framework, MAJAR (meta-analysis of joint effect associations for biomarker replicability assessment), to jointly test prognostic and predictive effects and assess the replicability of identified biomarkers by implementing an enhanced expectation-maximization algorithm and calculating their posterior-probability-of-replicabilities and Bayesian false discovery rates (Fdr). Extensive simulation studies were conducted to compare the performance of MAJAR and existing methods in terms of Fdr, power, and computational efficiency. The simulation results showed improved statistical power with well-controlled Fdr of MAJAR over existing methods and robustness to outliers under different data generation processes. We further demonstrated the advantages of MAJAR over existing methods by applying MAJAR to the PGx GWAS summary statistics data from a large cardiovascular randomized clinical trial. Compared to testing main effects only, MAJAR identified 12 novel variants associated with the treatment-related low-density lipoprotein cholesterol reduction from baseline.


Subject(s)
Genome-Wide Association Study , Polymorphism, Single Nucleotide , Humans , Phenotype , Bayes Theorem , Biomarkers , Randomized Controlled Trials as Topic
6.
Brief Bioinform ; 24(4)2023 07 20.
Article in English | MEDLINE | ID: mdl-37200155

ABSTRACT

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.


Subject(s)
Genome-Wide Association Study , Multifactorial Inheritance , Humans , Genome-Wide Association Study/methods , Pharmacogenetics , Polymorphism, Single Nucleotide , Phenotype , Genetic Predisposition to Disease
7.
Bioinformatics ; 39(1)2023 01 01.
Article in English | MEDLINE | ID: mdl-36661328

ABSTRACT

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.


Subject(s)
Genome-Wide Association Study , Pharmacogenetics , Genome-Wide Association Study/methods , Phenotype , Genotype , Polymorphism, Single Nucleotide
8.
Brief Bioinform ; 24(1)2023 01 19.
Article in English | MEDLINE | ID: mdl-36545787

ABSTRACT

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.


Subject(s)
Gene-Environment Interaction , Genome-Wide Association Study , Humans , Phenotype , Genotype , Linear Models , Polymorphism, Single Nucleotide
9.
Nat Commun ; 13(1): 5278, 2022 09 08.
Article in English | MEDLINE | ID: mdl-36075892

ABSTRACT

Polygenic risk scores (PRS) have been successfully developed for the prediction of human diseases and complex traits in the past years. For drug response prediction in randomized clinical trials, a common practice is to apply PRS built from a disease genome-wide association study (GWAS) directly to a corresponding pharmacogenomics (PGx) setting. Here, we show that such an approach relies on stringent assumptions about the prognostic and predictive effects of the selected genetic variants. We propose a shift from disease PRS to PGx PRS approaches by simultaneously modeling both the prognostic and predictive effects and further make this shift possible by developing a series of PRS-PGx methods, including a novel Bayesian regression approach (PRS-PGx-Bayes). Simulation studies show that PRS-PGx methods generally outperform the disease PRS methods and PRS-PGx-Bayes is superior to all other PRS-PGx methods. We further apply the PRS-PGx methods to PGx GWAS data from a large cardiovascular randomized clinical trial (IMPROVE-IT) to predict treatment related LDL cholesterol reduction. The results demonstrate substantial improvement of PRS-PGx-Bayes in both prediction accuracy and the capability of capturing the treatment-specific predictive effects while compared with the disease PRS approaches.


Subject(s)
Genome-Wide Association Study , Pharmacogenetics , Bayes Theorem , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Humans , Multifactorial Inheritance/genetics , Polymorphism, Single Nucleotide , Risk Factors
10.
Stat Med ; 41(21): 4227-4244, 2022 09 20.
Article in English | MEDLINE | ID: mdl-35799329

ABSTRACT

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.


Subject(s)
Algorithms , Precision Medicine , Humans , Policy , Precision Medicine/methods , Research Design
11.
NPJ Genom Med ; 7(1): 33, 2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35680959

ABSTRACT

In pharmacogenetic (PGx) studies, drug response phenotypes are often measured in the form of change in a quantitative trait before and after treatment. There is some debate in recent literature regarding baseline adjustment, or inclusion of pre-treatment or baseline value as a covariate, in PGx genome-wide association studies (GWAS) analysis. Here, we provide a clear statistical perspective on this baseline adjustment issue by running extensive simulations based on nine statistical models to evaluate the influence of baseline adjustment on type I error and power. We then apply these nine models to analyzing the change in low-density lipoprotein cholesterol (LDL-C) levels with ezetimibe + simvastatin combination therapy compared with simvastatin monotherapy therapy in the 5661 participants of the IMPROVE-IT (IMProved Reduction of Outcomes: Vytroin Efficacy International Trial) PGx GWAS, supporting the conclusions drawn from our simulations. Both simulations and GWAS analyses consistently show that baseline-unadjusted models inflate type I error for the variants associated with baseline value if the baseline value is also associated with change from baseline (e.g., when baseline value is a mediator between a variant and change from baseline), while baseline-adjusted models can control type I error in various scenarios. We thus recommend performing baseline-adjusted analyses in PGx GWASs of quantitative change.

12.
Am J Hum Genet ; 109(3): 433-445, 2022 03 03.
Article in English | MEDLINE | ID: mdl-35196515

ABSTRACT

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.


Subject(s)
Biological Specimen Banks , Genome-Wide Association Study , Biomarkers , Cross-Sectional Studies , Electronic Health Records , Humans , Longitudinal Studies
13.
Immunity ; 55(1): 56-64.e4, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34986342

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Genotype , Germ-Line Mutation/genetics , HLA Antigens/genetics , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Age Factors , Female , Genetic Association Studies , Heterozygote , Humans , Male , Neoplasms/diagnosis , Neoplasms/mortality , Polymorphism, Genetic , Prognosis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Sex Factors , Survival Analysis , Treatment Outcome
14.
Stat Methods Med Res ; 30(11): 2447-2458, 2021 11.
Article in English | MEDLINE | ID: mdl-34520293

ABSTRACT

Non-proportional hazards data are routinely encountered in randomized clinical trials. In such cases, classic Cox proportional hazards model can suffer from severe power loss, with difficulty in interpretation of the estimated hazard ratio since the treatment effect varies over time. We propose CauchyCP, an omnibus test of change-point Cox regression models, to overcome both challenges while detecting signals of non-proportional hazards patterns. Extensive simulation studies demonstrate that, compared to existing treatment comparison tests under non-proportional hazards, the proposed CauchyCP test (a) controls the type I error better at small α levels (<0.01); (b) increases the power of detecting time-varying effects; and (c) is more computationally efficient than popular methods like MaxCombo for large-scale data analysis. The superior performance of CauchyCP is further illustrated using retrospective analyses of two randomized clinical trial datasets and a pharmacogenetic biomarker study dataset. The R package CauchyCP is publicly available on CRAN.


Subject(s)
Retrospective Studies , Computer Simulation , Proportional Hazards Models
16.
Ann Intern Med ; 174(8): 1118-1125, 2021 08.
Article in English | MEDLINE | ID: mdl-33844575

ABSTRACT

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.


Subject(s)
COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , Clinical Trials, Phase III as Topic/standards , Randomized Controlled Trials as Topic/standards , Clinical Trials, Phase III as Topic/methods , Cross-Over Studies , Double-Blind Method , Drug Administration Schedule , Follow-Up Studies , Humans , Randomized Controlled Trials as Topic/methods , Research Design/standards , SARS-CoV-2 , Treatment Outcome
17.
Pharm Stat ; 20(4): 737-751, 2021 07.
Article in English | MEDLINE | ID: mdl-33624407

ABSTRACT

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.


Subject(s)
Drug Development , Research Design , Causality , Data Interpretation, Statistical , Humans
18.
Clin Infect Dis ; 73(8): 1540-1544, 2021 10 20.
Article in English | MEDLINE | ID: mdl-33340397

ABSTRACT

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.


Subject(s)
COVID-19 , Vaccines , COVID-19 Vaccines , Humans , SARS-CoV-2 , Treatment Outcome
19.
Ann Appl Stat ; 15(4): 1652-1672, 2021 Dec.
Article in English | MEDLINE | ID: mdl-35198092

ABSTRACT

Single nucleotide polymorphism (SNP) set analysis aggregates both common and rare variants and tests for association between phenotype(s) of interest and a set. However, multiple SNP-sets, such as genes, pathways, or sliding windows are usually investigated across the whole genome in which all groups are tested separately, followed by multiple testing adjustments. We propose a novel method to prioritize SNP-sets in a joint multivariate variance component model. Each SNP-set corresponds to a variance component (or kernel), and model selection is achieved by incorporating either convex or nonconvex penalties. The uniqueness of this variance component selection framework, which we call VCSEL, is that it naturally encompasses multivariate traits (VCSEL-M) and SNP-set-treatment or -environment interactions (VCSEL-I). We devise an optimization algorithm scalable to many variance components, based on the majorization-minimization (MM) principle. Simulation studies demonstrate the superiority of our methods in model selection performance, as measured by the area under the precision-recall (PR) curve, compared to the commonly used marginal testing and group penalization methods. Finally, we apply our methods to a real pharmacogenomics study and a real whole exome sequencing study. Some top ranked genes by VCSEL are detected as insignificant by the marginal test methods which emphasizes formal inference of individual genes with a strict significance threshold. This provides alternative insights for biologists to prioritize follow-up studies and develop polygenic risk score models.

20.
Ann Intern Med ; 174(2): 221-228, 2021 02.
Article in English | MEDLINE | ID: mdl-33090877

ABSTRACT

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.


Subject(s)
COVID-19 Vaccines/therapeutic use , COVID-19/prevention & control , Randomized Controlled Trials as Topic/methods , Asymptomatic Infections , COVID-19/diagnosis , COVID-19 Testing , COVID-19 Vaccines/adverse effects , Clinical Trials, Phase III as Topic/methods , Humans , SARS-CoV-2 , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...