Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Ann Rheum Dis ; 83(8): 1018-1027, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38569851

RESUMEN

INTRODUCTION: Anifrolumab is a type I interferon (IFN) receptor 1 (IFNAR1) blocking antibody approved for treating patients with systemic lupus erythematosus (SLE). Here, we investigated the immunomodulatory mechanisms of anifrolumab using longitudinal transcriptomic and proteomic analyses of the 52-week, randomised, phase 3 TULIP-1 and TULIP-2 trials. METHODS: Patients with moderate to severe SLE were enrolled in TULIP-1 and TULIP-2 and received intravenous anifrolumab or placebo alongside standard therapy. Whole-blood expression of 18 017 genes using genome-wide RNA sequencing (RNA-seq) (pooled TULIP; anifrolumab, n=244; placebo, n=258) and 184 plasma proteins using Olink and Simoa panels (TULIP-1; anifrolumab, n=124; placebo, n=132) were analysed. We compared treatment groups via gene set enrichment analysis using MetaBase pathway analysis, blood transcriptome modules, in silico deconvolution of RNA-seq and longitudinal linear mixed effect models for gene counts and protein levels. RESULTS: Compared with placebo, anifrolumab modulated >2000 genes by week 24, with overlapping results at week 52, and 41 proteins by week 52. IFNAR1 blockade with anifrolumab downregulated multiple type I and II IFN-induced gene modules/pathways and type III IFN-λ protein levels, and impacted apoptosis-associated and neutrophil extracellular traps-(NET)osis-associated transcriptional pathways, innate cell activating chemokines and receptors, proinflammatory cytokines and B-cell activating cytokines. In silico deconvolution of RNA-seq data indicated an increase from baseline of mucosal-associated invariant and γδT cells and a decrease of monocytes following anifrolumab treatment. DISCUSSION: Type I IFN blockade with anifrolumab modulated multiple inflammatory pathways downstream of type I IFN signalling, including apoptotic, innate and adaptive mechanisms that play key roles in SLE immunopathogenesis.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Interferón Tipo I , Lupus Eritematoso Sistémico , Proteómica , Humanos , Lupus Eritematoso Sistémico/tratamiento farmacológico , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Femenino , Masculino , Adulto , Persona de Mediana Edad , Receptor de Interferón alfa y beta/genética , Transcriptoma
2.
Genet Epidemiol ; 2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38606643

RESUMEN

Recent advancement in genome-wide association studies (GWAS) comes from not only increasingly larger sample sizes but also the shift in focus towards underrepresented populations. Multipopulation GWAS increase power to detect novel risk variants and improve fine-mapping resolution by leveraging evidence and differences in linkage disequilibrium (LD) from diverse populations. Here, we expand upon our previous approach for single-population fine-mapping through Joint Analysis of Marginal SNP Effects (JAM) to a multipopulation analysis (mJAM). Under the assumption that true causal variants are common across studies, we implement a hierarchical model framework that conditions on multiple SNPs while explicitly incorporating the different LD structures across populations. The mJAM framework can be used to first select index variants using the mJAM likelihood with different feature selection approaches. In addition, we present a novel approach leveraging the ideas of mediation to construct credible sets for these index variants. Construction of such credible sets can be performed given any existing index variants. We illustrate the implementation of the mJAM likelihood through two implementations: mJAM-SuSiE (a Bayesian approach) and mJAM-Forward selection. Through simulation studies based on realistic effect sizes and levels of LD, we demonstrated that mJAM performs well for constructing concise credible sets that include the underlying causal variants. In real data examples taken from the most recent multipopulation prostate cancer GWAS, we showed several practical advantages of mJAM over other existing multipopulation methods.

3.
Genet Epidemiol ; 47(1): 3-25, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36273411

RESUMEN

Mendelian randomization (MR) is the use of genetic variants to assess the existence of a causal relationship between a risk factor and an outcome of interest. Here, we focus on two-sample summary-data MR analyses with many correlated variants from a single gene region, particularly on cis-MR studies which use protein expression as a risk factor. Such studies must rely on a small, curated set of variants from the studied region; using all variants in the region requires inverting an ill-conditioned genetic correlation matrix and results in numerically unstable causal effect estimates. We review methods for variable selection and estimation in cis-MR with summary-level data, ranging from stepwise pruning and conditional analysis to principal components analysis, factor analysis, and Bayesian variable selection. In a simulation study, we show that the various methods have comparable performance in analyses with large sample sizes and strong genetic instruments. However, when weak instrument bias is suspected, factor analysis and Bayesian variable selection produce more reliable inferences than simple pruning approaches, which are often used in practice. We conclude by examining two case studies, assessing the effects of low-density lipoprotein-cholesterol and serum testosterone on coronary heart disease risk using variants in the HMGCR and SHBG gene regions, respectively.


Asunto(s)
Análisis de la Aleatorización Mendeliana , Modelos Genéticos , Humanos , Análisis de la Aleatorización Mendeliana/métodos , Teorema de Bayes , Factores de Riesgo , Causalidad
4.
Biostatistics ; 24(1): 85-107, 2022 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-34363680

RESUMEN

Risk prediction models are a crucial tool in healthcare. Risk prediction models with a binary outcome (i.e., binary classification models) are often constructed using methodology which assumes the costs of different classification errors are equal. In many healthcare applications, this assumption is not valid, and the differences between misclassification costs can be quite large. For instance, in a diagnostic setting, the cost of misdiagnosing a person with a life-threatening disease as healthy may be larger than the cost of misdiagnosing a healthy person as a patient. In this article, we present Tailored Bayes (TB), a novel Bayesian inference framework which "tailors" model fitting to optimize predictive performance with respect to unbalanced misclassification costs. We use simulation studies to showcase when TB is expected to outperform standard Bayesian methods in the context of logistic regression. We then apply TB to three real-world applications, a cardiac surgery, a breast cancer prognostication task, and a breast cancer tumor classification task and demonstrate the improvement in predictive performance over standard methods.


Asunto(s)
Neoplasias de la Mama , Modelos Estadísticos , Humanos , Femenino , Teorema de Bayes , Modelos Logísticos , Simulación por Computador , Neoplasias de la Mama/diagnóstico
5.
Biometrics ; 78(1): 141-150, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33448327

RESUMEN

High-dimensional biomarkers such as genomics are increasingly being measured in randomized clinical trials. Consequently, there is a growing interest in developing methods that improve the power to detect biomarker-treatment interactions. We adapt recently proposed two-stage interaction detecting procedures in the setting of randomized clinical trials. We also propose a new stage 1 multivariate screening strategy using ridge regression to account for correlations among biomarkers. For this multivariate screening, we prove the asymptotic between-stage independence, required for familywise error rate control, under biomarker-treatment independence. Simulation results show that in various scenarios, the ridge regression screening procedure can provide substantially greater power than the traditional one-biomarker-at-a-time screening procedure in highly correlated data. We also exemplify our approach in two real clinical trial data applications.


Asunto(s)
Genómica , Biomarcadores , Simulación por Computador , Ensayos Clínicos Controlados Aleatorios como Asunto
6.
Stat Med ; 40(23): 5025-5045, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34155684

RESUMEN

Mendelian randomization is the use of genetic variants as instruments to assess the existence of a causal relationship between a risk factor and an outcome. A Mendelian randomization analysis requires a set of genetic variants that are strongly associated with the risk factor and only associated with the outcome through their effect on the risk factor. We describe a novel variable selection algorithm for Mendelian randomization that can identify sets of genetic variants which are suitable in both these respects. Our algorithm is applicable in the context of two-sample summary-data Mendelian randomization and employs a recently proposed theoretical extension of the traditional Bayesian statistics framework, including a loss function to penalize genetic variants that exhibit pleiotropic effects. The algorithm offers robust inference through the use of model averaging, as we illustrate by running it on a range of simulation scenarios and comparing it against established pleiotropy-robust Mendelian randomization methods. In a real-data application, we study the effect of systolic and diastolic blood pressure on the risk of suffering from coronary heart disease (CHD). Based on a recent large-scale GWAS for blood pressure, we use 395 genetic variants for systolic and 391 variants for diastolic blood pressure. Both traits are shown to have significant risk-increasing effects on CHD risk.


Asunto(s)
Pleiotropía Genética , Análisis de la Aleatorización Mendeliana , Teorema de Bayes , Causalidad , Variación Genética , Estudio de Asociación del Genoma Completo , Humanos , Factores de Riesgo
7.
Am J Epidemiol ; 190(6): 1148-1158, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33404048

RESUMEN

Previous research has demonstrated the usefulness of hierarchical modeling for incorporating a flexible array of prior information in genetic association studies. When this prior information consists of estimates from association analyses of single-nucleotide polymorphisms (SNP)-intermediate or SNP-gene expression, a hierarchical model is equivalent to a 2-stage instrumental or transcriptome-wide association study (TWAS) analysis, respectively. We propose to extend our previous approach for the joint analysis of marginal summary statistics to incorporate prior information via a hierarchical model (hJAM). In this framework, the use of appropriate estimates as prior information yields an analysis similar to Mendelian randomization (MR) and TWAS approaches. hJAM is applicable to multiple correlated SNPs and intermediates to yield conditional estimates for the intermediates on the outcome, thus providing advantages over alternative approaches. We investigated the performance of hJAM in comparison with existing MR and TWAS approaches and demonstrated that hJAM yields an unbiased estimate, maintains correct type-I error, and has increased power across extensive simulations. We applied hJAM to 2 examples: estimating the causal effects of body mass index (GIANT Consortium) and type 2 diabetes (DIAGRAM data set, GERA Cohort, and UK Biobank) on myocardial infarction (UK Biobank) and estimating the causal effects of the expressions of the genes for nuclear casein kinase and cyclin dependent kinase substrate 1 and peptidase M20 domain containing 1 on the risk of prostate cancer (PRACTICAL and GTEx).


Asunto(s)
Interpretación Estadística de Datos , Perfilación de la Expresión Génica/métodos , Análisis de la Aleatorización Mendeliana/métodos , Modelos Genéticos , Amidohidrolasas/análisis , Sesgo , Índice de Masa Corporal , Diabetes Mellitus Tipo 2/genética , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Infarto del Miocardio/genética , Proteínas Nucleares/análisis , Fosfoproteínas/análisis , Polimorfismo de Nucleótido Simple , Neoplasias de la Próstata/genética
8.
Genet Epidemiol ; 43(7): 730-741, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31328830

RESUMEN

The heritability of most complex traits is driven by variants throughout the genome. Consequently, polygenic risk scores, which combine information on multiple variants genome-wide, have demonstrated improved accuracy in genetic risk prediction. We present a new two-step approach to constructing genome-wide polygenic risk scores from meta-GWAS summary statistics. Local linkage disequilibrium (LD) is adjusted for in Step 1, followed by, uniquely, long-range LD in Step 2. Our algorithm is highly parallelizable since block-wise analyses in Step 1 can be distributed across a high-performance computing cluster, and flexible, since sparsity and heritability are estimated within each block. Inference is obtained through a formal Bayesian variable selection framework, meaning final risk predictions are averaged over competing models. We compared our method to two alternative approaches: LDPred and lassosum using all seven traits in the Welcome Trust Case Control Consortium as well as meta-GWAS summaries for type 1 diabetes (T1D), coronary artery disease, and schizophrenia. Performance was generally similar across methods, although our framework provided more accurate predictions for T1D, for which there are multiple heterogeneous signals in regions of both short- and long-range LD. With sufficient compute resources, our method also allows the fastest runtimes.


Asunto(s)
Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Herencia Multifactorial/genética , Área Bajo la Curva , Estudios de Casos y Controles , Enfermedad de la Arteria Coronaria/genética , Diabetes Mellitus Tipo 1/genética , Humanos , Modelos Genéticos , Polimorfismo de Nucleótido Simple/genética , Curva ROC , Factores de Riesgo , Esquizofrenia/genética
9.
BMC Genomics ; 20(1): 77, 2019 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-30674271

RESUMEN

BACKGROUND: Hi-C and capture Hi-C (CHi-C) are used to map physical contacts between chromatin regions in cell nuclei using high-throughput sequencing. Analysis typically proceeds considering the evidence for contacts between each possible pair of fragments independent from other pairs. This can produce long runs of fragments which appear to all make contact with the same baited fragment of interest. RESULTS: We hypothesised that these long runs could result from a smaller subset of direct contacts and propose a new method, based on a Bayesian sparse variable selection approach, which attempts to fine map these direct contacts. Our model is conceptually novel, exploiting the spatial pattern of counts in CHi-C data. Although we use only the CHi-C count data in fitting the model, we show that the fragments prioritised display biological properties that would be expected of true contacts: for bait fragments corresponding to gene promoters, we identify contact fragments with active chromatin and contacts that correspond to edges found in previously defined enhancer-target networks; conversely, for intergenic bait fragments, we identify contact fragments corresponding to promoters for genes expressed in that cell type. We show that long runs of apparently co-contacting fragments can typically be explained using a subset of direct contacts consisting of <10% of the number in the full run, suggesting that greater resolution can be extracted from existing datasets. CONCLUSIONS: Our results appear largely complementary to those from a per-fragment analytical approach, suggesting that they provide an additional level of interpretation that may be used to increase resolution for mapping direct contacts in CHi-C experiments.


Asunto(s)
Cromatina/química , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Análisis de Secuencia de ADN/métodos , Linfocitos T CD4-Positivos , Macrófagos , Modelos Estadísticos , Regiones Promotoras Genéticas
10.
Nat Commun ; 9(1): 2256, 2018 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-29892050

RESUMEN

Prostate cancer is a polygenic disease with a large heritable component. A number of common, low-penetrance prostate cancer risk loci have been identified through GWAS. Here we apply the Bayesian multivariate variable selection algorithm JAM to fine-map 84 prostate cancer susceptibility loci, using summary data from a large European ancestry meta-analysis. We observe evidence for multiple independent signals at 12 regions and 99 risk signals overall. Only 15 original GWAS tag SNPs remain among the catalogue of candidate variants identified; the remainder are replaced by more likely candidates. Biological annotation of our credible set of variants indicates significant enrichment within promoter and enhancer elements, and transcription factor-binding sites, including AR, ERG and FOXA1. In 40 regions at least one variant is colocalised with an eQTL in prostate cancer tissue. The refined set of candidate variants substantially increase the proportion of familial relative risk explained by these known susceptibility regions, which highlights the importance of fine-mapping studies and has implications for clinical risk profiling.


Asunto(s)
Neoplasias de la Próstata/genética , Algoritmos , Teorema de Bayes , Población Negra/genética , Mapeo Cromosómico , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Anotación de Secuencia Molecular , Análisis Multivariante , Polimorfismo de Nucleótido Simple , Sitios de Carácter Cuantitativo , Riesgo , Población Blanca/genética
11.
Clin Cancer Res ; 24(9): 2110-2115, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29444929

RESUMEN

Purpose: To compare PREDICT and CancerMath, two widely used prognostic models for invasive breast cancer, taking into account their clinical utility. Furthermore, it is unclear whether these models could be improved.Experimental Design: A dataset of 5,729 women was used for model development. A Bayesian variable selection algorithm was implemented to stochastically search for important interaction terms among the predictors. The derived models were then compared in three independent datasets (n = 5,534). We examined calibration, discrimination, and performed decision curve analysis.Results: CancerMath demonstrated worse calibration performance compared with PREDICT in estrogen receptor (ER)-positive and ER-negative tumors. The decline in discrimination performance was -4.27% (-6.39 to -2.03) and -3.21% (-5.9 to -0.48) for ER-positive and ER-negative tumors, respectively. Our new models matched the performance of PREDICT in terms of calibration and discrimination, but offered no improvement. Decision curve analysis showed predictions for all models were clinically useful for treatment decisions made at risk thresholds between 5% and 55% for ER-positive tumors and at thresholds of 15% to 60% for ER-negative tumors. Within these threshold ranges, CancerMath provided the lowest clinical utility among all the models.Conclusions: Survival probabilities from PREDICT offer both improved accuracy and discrimination over CancerMath. Using PREDICT to make treatment decisions offers greater clinical utility than CancerMath over a range of risk thresholds. Our new models performed as well as PREDICT, but no better, suggesting that, in this setting, including further interaction terms offers no predictive benefit. Clin Cancer Res; 24(9); 2110-5. ©2018 AACR.


Asunto(s)
Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Adulto , Anciano , Algoritmos , Teorema de Bayes , Neoplasias de la Mama/epidemiología , Femenino , Humanos , Persona de Mediana Edad , Modelos Estadísticos , Clasificación del Tumor , Metástasis de la Neoplasia , Estadificación de Neoplasias , Pronóstico , Vigilancia en Salud Pública , Reproducibilidad de los Resultados , Tasa de Supervivencia
12.
Sci Rep ; 7(1): 9214, 2017 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-28835676

RESUMEN

SLC10A1 codes for the sodium-taurocholate cotransporting polypeptide (NTCP), which is a hepatocellular transporter for bile acids (BAs) and the receptor for hepatitis B and D viruses. NTCP is also a target of multiple drugs. We aimed to evaluate the medical consequences of the loss of function mutation p.Ser267Phe in SLC10A1. We identified eight individuals with homozygous p.Ser267Phe mutation in SLC10A1 and followed up for 8-90 months. We compared their total serum BAs and 6 species of BAs with 170 wild-type and 107 heterozygous healthy individuals. We performed in-depth medical examinations and exome sequencing in the homozygous individuals. All homozygous individuals had persistent hypercholanemia (P = 5.8 × 10-29). Exome sequencing excluded the involvement of other BA metabolism-associated genes in the hypercholanemia. Although asymptomatic, all individuals had low vitamin D levels. Of six adults that were subjected to bone mineral density analysis, three presented with osteoporosis/osteopenia. Sex hormones and blood lipids were deviated in all subjects. Homozygosity of p.Ser267Phe in SLC10A1 is associated with asymptomatic hypercholanemia. Individuals with homozygous p.Ser267Phe in SLC10A1 are prone to vitamin D deficiency, deviated sex hormones and blood lipids. Surveillance of these parameters may also be needed in patients treated with drugs targeting NTCP.


Asunto(s)
Alelos , Sustitución de Aminoácidos , Homocigoto , Hipercolesterolemia/sangre , Hipercolesterolemia/genética , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Medicina de Precisión , Simportadores/genética , Adolescente , Adulto , Ácidos y Sales Biliares/sangre , Ácidos y Sales Biliares/metabolismo , Densidad Ósea , Niño , Femenino , Estudios de Seguimiento , Humanos , Metabolismo de los Lípidos , Masculino , Persona de Mediana Edad , Secuenciación del Exoma , Adulto Joven
13.
Genet Epidemiol ; 40(3): 188-201, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27027514

RESUMEN

Recently, large scale genome-wide association study (GWAS) meta-analyses have boosted the number of known signals for some traits into the tens and hundreds. Typically, however, variants are only analysed one-at-a-time. This complicates the ability of fine-mapping to identify a small set of SNPs for further functional follow-up. We describe a new and scalable algorithm, joint analysis of marginal summary statistics (JAM), for the re-analysis of published marginal summary statistics under joint multi-SNP models. The correlation is accounted for according to estimates from a reference dataset, and models and SNPs that best explain the complete joint pattern of marginal effects are highlighted via an integrated Bayesian penalized regression framework. We provide both enumerated and Reversible Jump MCMC implementations of JAM and present some comparisons of performance. In a series of realistic simulation studies, JAM demonstrated identical performance to various alternatives designed for single region settings. In multi-region settings, where the only multivariate alternative involves stepwise selection, JAM offered greater power and specificity. We also present an application to real published results from MAGIC (meta-analysis of glucose and insulin related traits consortium) - a GWAS meta-analysis of more than 15,000 people. We re-analysed several genomic regions that produced multiple significant signals with glucose levels 2 hr after oral stimulation. Through joint multivariate modelling, JAM was able to formally rule out many SNPs, and for one gene, ADCY5, suggests that an additional SNP, which transpired to be more biologically plausible, should be followed up with equal priority to the reported index.


Asunto(s)
Teorema de Bayes , Estudio de Asociación del Genoma Completo/métodos , Polimorfismo de Nucleótido Simple/genética , Adenilil Ciclasas/genética , Algoritmos , Simulación por Computador , Ayuno/metabolismo , Genómica , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Modelos Genéticos , Fenotipo
14.
PLoS Genet ; 12(3): e1005908, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27015630

RESUMEN

Genome-wide association studies (GWAS) have transformed our understanding of the genetics of complex traits such as autoimmune diseases, but how risk variants contribute to pathogenesis remains largely unknown. Identifying genetic variants that affect gene expression (expression quantitative trait loci, or eQTLs) is crucial to addressing this. eQTLs vary between tissues and following in vitro cellular activation, but have not been examined in the context of human inflammatory diseases. We performed eQTL mapping in five primary immune cell types from patients with active inflammatory bowel disease (n = 91), anti-neutrophil cytoplasmic antibody-associated vasculitis (n = 46) and healthy controls (n = 43), revealing eQTLs present only in the context of active inflammatory disease. Moreover, we show that following treatment a proportion of these eQTLs disappear. Through joint analysis of expression data from multiple cell types, we reveal that previous estimates of eQTL immune cell-type specificity are likely to have been exaggerated. Finally, by analysing gene expression data from multiple cell types, we find eQTLs not previously identified by database mining at 34 inflammatory bowel disease-associated loci. In summary, this parallel eQTL analysis in multiple leucocyte subsets from patients with active disease provides new insights into the genetic basis of immune-mediated diseases.


Asunto(s)
Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/genética , Estudios de Asociación Genética , Enfermedades Inflamatorias del Intestino/genética , Sitios de Carácter Cuantitativo/genética , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/inmunología , Vasculitis Asociada a Anticuerpos Citoplasmáticos Antineutrófilos/patología , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Masculino , Monocitos/inmunología , Monocitos/metabolismo , Neutrófilos/inmunología , Neutrófilos/metabolismo , Fenotipo , Linfocitos T/inmunología , Linfocitos T/metabolismo
15.
Hum Hered ; 80(4): 178-86, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-27576758

RESUMEN

OBJECTIVE: Gene scores are often used to model the combined effects of genetic variants. When variants are in linkage disequilibrium, it is common to prune all variants except the most strongly associated. This avoids duplicating information but discards information when variants have independent effects. However, joint modelling of correlated variants increases the sampling error in the gene score. In recent applications, joint modelling has offered only small improvements in accuracy over pruning. We aimed to quantify the relationship between pruning and joint modelling in relation to sample size. METHODS: We derived the coefficient of determination R2 for a gene score constructed from pruned markers, and for one constructed from correlated markers with jointly estimated effects. RESULTS: Pruned scores tend to have slightly lower R2 than jointly modelled scores, but the differences are small at sample sizes up to 100,000. If the proportion of correlated variants is high, joint modelling can obtain modest improvements asymptotically. CONCLUSIONS: The small gains observed to date from joint modelling can be explained by sample size. As studies become larger, joint modelling will be useful for traits affected by many correlated variants, but the improvements may remain small. Pruning remains a useful heuristic for current studies.


Asunto(s)
Marcadores Genéticos/genética , Desequilibrio de Ligamiento/genética , Modelos Genéticos , Variación Genética , Humanos , Carácter Cuantitativo Heredable , Tamaño de la Muestra
16.
Retina ; 34(2): 288-97, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23842101

RESUMEN

PURPOSE: To investigate the association between genetic risk variants for age-related macular degeneration (AMD) and response to intravitreal ranibizumab in Korean patients with neovascular AMD. METHODS: This prospective study included 273 treatment-naive patients (273 eyes) who underwent 5 monthly injections (Months 0, 1, 2, 3, and 4) of intravitreal ranibizumab for neovascular AMD. Patients were genotyped for 23 single-nucleotide polymorphisms within 12 AMD-relevant genes. For each polymorphism, genotypic association with good response at Month 5, predetermined as visual improvement of ≥ 8 Early Treatment Diabetic Retinopathy Study letters from baseline, was investigated with logistic regression analysis adjusted for age, gender, smoking, baseline Early Treatment Diabetic Retinopathy Study letter, central retinal thickness, lesion area, and type of choroidal neovascularization. RESULTS: At Month 5, visual acuity improved by 9.1 ± 17.6 letters from baseline, and 136 patients (49.8%) were classified as good responders. In logistic regression, no tested polymorphism showed statistically significant association with favorable visual outcome at Month 5. When unadjusted for multiple tests, AA genotype for VEGF rs699947 had an increased chance of good response compared with other genotypes (odds ratio, 3.61; 95% confidence interval, 1.42-9.18; P = 0.0071). CONCLUSION: In this Korean neovascular AMD cohort, there was no statistically significant effect of genotype on early visual outcome after ranibizumab treatment.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos Monoclonales Humanizados/uso terapéutico , Proteínas del Ojo/genética , Polimorfismo de Nucleótido Simple , Degeneración Macular Húmeda/tratamiento farmacológico , Degeneración Macular Húmeda/genética , Anciano , Anciano de 80 o más Años , Pueblo Asiatico , Colorantes , Femenino , Angiografía con Fluoresceína , Marcadores Genéticos , Genotipo , Humanos , Verde de Indocianina , Inyecciones Intravítreas , Masculino , Persona de Mediana Edad , Farmacogenética , Reacción en Cadena de la Polimerasa , Estudios Prospectivos , Ranibizumab , República de Corea , Factores de Riesgo , Tomografía de Coherencia Óptica , Resultado del Tratamiento , Agudeza Visual/fisiología , Degeneración Macular Húmeda/fisiopatología
17.
Genet Epidemiol ; 36(1): 71-83, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22890972

RESUMEN

We present the most comprehensive comparison to date of the predictive benefit of genetics in addition to currently used clinical variables, using genotype data for 33 single-nucleotide polymorphisms (SNPs) in 1,547 Caucasian men from the placebo arm of the REduction by DUtasteride of prostate Cancer Events (REDUCE®) trial. Moreover, we conducted a detailed comparison of three techniques for incorporating genetics into clinical risk prediction. The first method was a standard logistic regression model, which included separate terms for the clinical covariates and for each of the genetic markers. This approach ignores a substantial amount of external information concerning effect sizes for these Genome Wide Association Study (GWAS)-replicated SNPs. The second and third methods investigated two possible approaches to incorporating meta-analysed external SNP effect estimates - one via a weighted PCa 'risk' score based solely on the meta analysis estimates, and the other incorporating both the current and prior data via informative priors in a Bayesian logistic regression model. All methods demonstrated a slight improvement in predictive performance upon incorporation of genetics. The two methods that incorporated external information showed the greatest receiver-operating-characteristic AUCs increase from 0.61 to 0.64. The value of our methods comparison is likely to lie in observations of performance similarities, rather than difference, between three approaches of very different resource requirements. The two methods that included external information performed best, but only marginally despite substantial differences in complexity.


Asunto(s)
Teorema de Bayes , Predisposición Genética a la Enfermedad , Modelos Logísticos , Neoplasias de la Próstata/genética , Anciano , Algoritmos , Área Bajo la Curva , Calibración , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Persona de Mediana Edad , Modelos Genéticos , Modelos Estadísticos , Polimorfismo de Nucleótido Simple , Curva ROC , Ensayos Clínicos Controlados Aleatorios como Asunto , Población Blanca/genética
18.
Am J Ophthalmol ; 154(3): 568-578.e12, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22704140

RESUMEN

PURPOSE: To develop comprehensive predictive models for choroidal neovascularization (CNV) and geographic atrophy (GA) incidence within 3 years that can be applied realistically to clinical practice. DESIGN: Retrospective evaluation of data from a longitudinal study to develop and validate predictive models of CNV and GA. METHODS: The predictive performance of clinical, environmental, demographic, and genetic risk factors was explored in regression models, using data from both eyes of 2011 subjects from the Age-Related Eye Disease Study (AREDS). The performance of predictive models was compared using 10-fold cross-validated receiver operating characteristic curves in the training data, followed by comparisons in an independent validation dataset (1410 AREDS subjects). Bayesian trial simulations were used to compare the usefulness of predictive models to screen patients for inclusion in prevention clinical trials. RESULTS: Logistic regression models that included clinical, demographic, and environmental factors had better predictive performance for 3-year CNV and GA incidence (area under the receiver operating characteristic curve of 0.87 and 0.89, respectively), compared with simple clinical criteria (AREDS simplified severity scale). Although genetic markers were associated significantly with 3-year CNV (CFH: Y402H; ARMS2: A69S) and GA incidence (CFH: Y402H), the inclusion of genetic factors in the models provided only marginal improvements in predictive performance. CONCLUSIONS: The logistic regression models combine good predictive performance with greater flexibility to optimize clinical trial design compared with simple clinical models (AREDS simplified severity scale). The benefit of including genetic factors to screen patients for recruitment to CNV prevention studies is marginal and is dependent on individual clinical trial economics.


Asunto(s)
Neovascularización Coroidal/diagnóstico , Ensayos Clínicos como Asunto , Atrofia Geográfica/diagnóstico , Modelos Estadísticos , Proyectos de Investigación , Anciano , Área Bajo la Curva , Neovascularización Coroidal/genética , Reacciones Falso Positivas , Femenino , Marcadores Genéticos , Genotipo , Atrofia Geográfica/genética , Humanos , Incidencia , Masculino , Polimorfismo Genético , Valor Predictivo de las Pruebas , Curva ROC , Estudios Retrospectivos , Factores de Riesgo
19.
Eur Urol ; 62(6): 953-61, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22652152

RESUMEN

BACKGROUND: Several germline single nucleotide polymorphisms (SNPs) have been consistently associated with prostate cancer (PCa) risk. OBJECTIVE: To determine whether there is an improvement in PCa risk prediction by adding these SNPs to existing predictors of PCa. DESIGN, SETTING, AND PARTICIPANTS: Subjects included men in the placebo arm of the randomized Reduction by Dutasteride of Prostate Cancer Events (REDUCE) trial in whom germline DNA was available. All men had an initial negative prostate biopsy and underwent study-mandated biopsies at 2 yr and 4 yr. Predictive performance of baseline clinical parameters and/or a genetic score based on 33 established PCa risk-associated SNPs was evaluated. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Area under the receiver operating characteristic curves (AUC) were used to compare different models with different predictors. Net reclassification improvement (NRI) and decision curve analysis (DCA) were used to assess changes in risk prediction by adding genetic markers. RESULTS AND LIMITATIONS: Among 1654 men, genetic score was a significant predictor of positive biopsy, even after adjusting for known clinical variables and family history (p = 3.41 × 10(-8)). The AUC for the genetic score exceeded that of any other PCa predictor at 0.59. Adding the genetic score to the best clinical model improved the AUC from 0.62 to 0.66 (p<0.001), reclassified PCa risk in 33% of men (NRI: 0.10; p=0.002), resulted in higher net benefit from DCA, and decreased the number of biopsies needed to detect the same number of PCa instances. The benefit of adding the genetic score was greatest among men at intermediate risk (25th percentile to 75th percentile). Similar results were found for high-grade (Gleason score ≥ 7) PCa. A major limitation of this study was its focus on white patients only. CONCLUSIONS: Adding genetic markers to current clinical parameters may improve PCa risk prediction. The improvement is modest but may be helpful for better determining the need for repeat prostate biopsy. The clinical impact of these results requires further study.


Asunto(s)
Próstata/patología , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Biopsia , Reacciones Falso Negativas , Marcadores Genéticos , Humanos , Masculino , Valor Predictivo de las Pruebas , Pronóstico , Ensayos Clínicos Controlados Aleatorios como Asunto , Medición de Riesgo/métodos
20.
Genet Epidemiol ; 35(5): 333-40, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21400586

RESUMEN

We present a Bayesian semiparametric model for the meta-analysis of candidate gene studies with a binary outcome. Such studies often report results from association tests for different, possibly study-specific and non-overlapping genetic markers in the same genetic region. Meta-analyses of the results at each marker in isolation are seldom appropriate as they ignore the correlation that may exist between markers due to linkage disequilibrium (LD) and cannot assess the relative importance of variants at each marker. Also such marker-wise meta-analyses are restricted to only those studies that have typed the marker in question, with a potential loss of power. A better strategy is one which incorporates information about the LD between markers so that any combined estimate of the effect of each variant is corrected for the effect of other variants, as in multiple regression. Here we develop a Bayesian semiparametric model which models the observed genotype group frequencies conditional to the case/control status and uses pairwise LD measurements between markers as prior information to make posterior inference on adjusted effects. The approach allows borrowing of strength across studies and across markers. The analysis is based on a mixture of Dirichlet processes model as the underlying semiparametric model. Full posterior inference is performed through Markov chain Monte Carlo algorithms. The approach is demonstrated on simulated and real data.


Asunto(s)
Estudio de Asociación del Genoma Completo/estadística & datos numéricos , Algoritmos , Teorema de Bayes , Simulación por Computador , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 4 , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Humanos , Funciones de Verosimilitud , Desequilibrio de Ligamiento , Cadenas de Markov , Metaanálisis como Asunto , Modelos Genéticos , Modelos Estadísticos , Método de Montecarlo , Análisis Multivariante , Accidente Cerebrovascular/enzimología , Accidente Cerebrovascular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA