Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
J Am Med Inform Assoc ; 31(6): 1303-1312, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38713006

RESUMO

OBJECTIVES: Racial disparities in kidney transplant access and posttransplant outcomes exist between non-Hispanic Black (NHB) and non-Hispanic White (NHW) patients in the United States, with the site of care being a key contributor. Using multi-site data to examine the effect of site of care on racial disparities, the key challenge is the dilemma in sharing patient-level data due to regulations for protecting patients' privacy. MATERIALS AND METHODS: We developed a federated learning framework, named dGEM-disparity (decentralized algorithm for Generalized linear mixed Effect Model for disparity quantification). Consisting of 2 modules, dGEM-disparity first provides accurately estimated common effects and calibrated hospital-specific effects by requiring only aggregated data from each center and then adopts a counterfactual modeling approach to assess whether the graft failure rates differ if NHB patients had been admitted at transplant centers in the same distribution as NHW patients were admitted. RESULTS: Utilizing United States Renal Data System data from 39 043 adult patients across 73 transplant centers over 10 years, we found that if NHB patients had followed the distribution of NHW patients in admissions, there would be 38 fewer deaths or graft failures per 10 000 NHB patients (95% CI, 35-40) within 1 year of receiving a kidney transplant on average. DISCUSSION: The proposed framework facilitates efficient collaborations in clinical research networks. Additionally, the framework, by using counterfactual modeling to calculate the event rate, allows us to investigate contributions to racial disparities that may occur at the level of site of care. CONCLUSIONS: Our framework is broadly applicable to other decentralized datasets and disparities research related to differential access to care. Ultimately, our proposed framework will advance equity in human health by identifying and addressing hospital-level racial disparities.


Assuntos
Algoritmos , Negro ou Afro-Americano , Disparidades em Assistência à Saúde , Transplante de Rim , População Branca , Humanos , Estados Unidos , Disparidades em Assistência à Saúde/etnologia , Adulto , Masculino , Feminino , Rejeição de Enxerto/etnologia , Pessoa de Meia-Idade
2.
Gynecol Oncol ; 178: 119-129, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37862791

RESUMO

OBJECTIVE: This prespecified exploratory analysis evaluated the association of gene expression signatures, tumor mutational burden (TMB), and multiplex immunohistochemistry (mIHC) tumor microenvironment-associated cell phenotypes with clinical outcomes of pembrolizumab in advanced recurrent ovarian cancer (ROC) from the phase II KEYNOTE-100 study. METHODS: Pembrolizumab-treated patients with evaluable RNA-sequencing (n = 317), whole exome sequencing (n = 293), or select mIHC (n = 125) data were evaluated. The association between outcomes (objective response rate [ORR], progression-free survival [PFS], and overall survival [OS]) and gene expression signatures (T-cell-inflamed gene expression profile [TcellinfGEP] and 10 non-TcellinfGEP signatures), TMB, and prespecified mIHC cell phenotype densities as continuous variables was evaluated using logistic (ORR) and Cox proportional hazards regression (PFS; OS). One-sided p-values were calculated at prespecified α = 0.05 for TcellinfGEP, TMB, and mIHC cell phenotypes and at α = 0.10 for non-TcellinfGEP signatures; all but TcellinfGEP and TMB were adjusted for multiplicity. RESULTS: No evidence of associations between ORR and key axes of gene expression was observed. Negative associations were observed between outcomes and TcellinfGEP-adjusted glycolysis (PFS, adjusted-p = 0.019; OS, adjusted-p = 0.085) and hypoxia (PFS, adjusted-p = 0.064) signatures. TMB as a continuous variable was not associated with outcomes (p > 0.05). Positive associations were observed between densities of myeloid cell phenotypes CD11c+ and CD11c+/MHCII-/CD163-/CD68- in the tumor compartment and ORR (adjusted-p = 0.025 and 0.013, respectively). CONCLUSIONS: This exploratory analysis in advanced ROC did not find evidence for associations between gene expression signatures and outcomes of pembrolizumab. mIHC analysis suggests CD11c+ and CD11c+/MHCII-/CD163-/CD68- phenotypes representing myeloid cell populations may be associated with improved outcomes with pembrolizumab in advanced ROC. CLINICAL TRIAL REGISTRATION: ClinicalTrials.gov, NCT02674061.


Assuntos
Antineoplásicos Imunológicos , Neoplasias Ovarianas , Humanos , Feminino , Antineoplásicos Imunológicos/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Intervalo Livre de Progressão , Carcinoma Epitelial do Ovário/tratamento farmacológico , Biomarcadores Tumorais/genética , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/induzido quimicamente , Microambiente Tumoral
3.
NPJ Digit Med ; 5(1): 76, 2022 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-35701668

RESUMO

Integrating real-world data (RWD) from several clinical sites offers great opportunities to improve estimation with a more general population compared to analyses based on a single clinical site. However, sharing patient-level data across sites is practically challenging due to concerns about maintaining patient privacy. We develop a distributed algorithm to integrate heterogeneous RWD from multiple clinical sites without sharing patient-level data. The proposed distributed conditional logistic regression (dCLR) algorithm can effectively account for between-site heterogeneity and requires only one round of communication. Our simulation study and data application with the data of 14,215 COVID-19 patients from 230 clinical sites in the UnitedHealth Group Clinical Research Database demonstrate that the proposed distributed algorithm provides an estimator that is robust to heterogeneity in event rates when efficiently integrating data from multiple clinical sites. Our algorithm is therefore a practical alternative to both meta-analysis and existing distributed algorithms for modeling heterogeneous multi-site binary outcomes.

4.
J Biomed Inform ; 131: 104097, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35643272

RESUMO

BACKGROUND: Observational studies incorporating real-world data from multiple institutions facilitate study of rare outcomes or exposures and improve generalizability of results. Due to privacy concerns surrounding patient-level data sharing across institutions, methods for performing regression analyses distributively are desirable. Meta-analysis of institution-specific estimates is commonly used, but has been shown to produce biased estimates in certain settings. While distributed regression methods are increasingly available, methods for analyzing count outcomes are currently limited. Count data in practice are commonly subject to overdispersion, exhibiting greater variability than expected under a given statistical model. OBJECTIVE: We propose a novel computational method, a one-shot distributed algorithm for quasi-Poisson regression (ODAP), to distributively model count outcomes while accounting for overdispersion. METHODS: ODAP incorporates a surrogate likelihood approach to perform distributed quasi-Poisson regression without requiring patient-level data sharing, only requiring sharing of aggregate data from each participating institution. ODAP requires at most three rounds of non-iterative communication among institutions to generate coefficient estimates and corresponding standard errors. In simulations, we evaluate ODAP under several data scenarios possible in multi-site analyses, comparing ODAP and meta-analysis estimates in terms of error relative to pooled regression estimates, considered the gold standard. In a proof-of-concept real-world data analysis, we similarly compare ODAP and meta-analysis in terms of relative error to pooled estimatation using data from the OneFlorida Clinical Research Consortium, modeling length of stay in COVID-19 patients as a function of various patient characteristics. In a second proof-of-concept analysis, using the same outcome and covariates, we incorporate data from the UnitedHealth Group Clinical Discovery Database together with the OneFlorida data in a distributed analysis to compare estimates produced by ODAP and meta-analysis. RESULTS: In simulations, ODAP exhibited negligible error relative to pooled regression estimates across all settings explored. Meta-analysis estimates, while largely unbiased, were increasingly variable as heterogeneity in the outcome increased across institutions. When baseline expected count was 0.2, relative error for meta-analysis was above 5% in 25% of iterations (250/1000), while the largest relative error for ODAP in any iteration was 3.59%. In our proof-of-concept analysis using only OneFlorida data, ODAP estimates were closer to pooled regression estimates than those produced by meta-analysis for all 15 covariates. In our distributed analysis incorporating data from both OneFlorida and the UnitedHealth Group Clinical Discovery Database, ODAP and meta-analysis estimates were largely similar, while some differences in estimates (as large as 13.8%) could be indicative of bias in meta-analytic estimates. CONCLUSIONS: ODAP performs privacy-preserving, communication-efficient distributed quasi-Poisson regression to analyze count outcomes using data stored within multiple institutions. Our method produces estimates nearly matching pooled regression estimates and sometimes more accurate than meta-analysis estimates, most notably in settings with relatively low counts and high outcome heterogeneity across institutions.


Assuntos
COVID-19 , Algoritmos , COVID-19/epidemiologia , Humanos , Funções Verossimilhança , Modelos Estatísticos , Análise de Regressão
5.
Nat Commun ; 13(1): 1678, 2022 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-35354802

RESUMO

Linear mixed models are commonly used in healthcare-based association analyses for analyzing multi-site data with heterogeneous site-specific random effects. Due to regulations for protecting patients' privacy, sensitive individual patient data (IPD) typically cannot be shared across sites. We propose an algorithm for fitting distributed linear mixed models (DLMMs) without sharing IPD across sites. This algorithm achieves results identical to those achieved using pooled IPD from multiple sites (i.e., the same effect size and standard error estimates), hence demonstrating the lossless property. The algorithm requires each site to contribute minimal aggregated data in only one round of communication. We demonstrate the lossless property of the proposed DLMM algorithm by investigating the associations between demographic and clinical characteristics and length of hospital stay in COVID-19 patients using administrative claims from the UnitedHealth Group Clinical Discovery Database. We extend this association study by incorporating 120,609 COVID-19 patients from 11 collaborative data sources worldwide.


Assuntos
COVID-19 , Algoritmos , COVID-19/epidemiologia , Confidencialidade , Bases de Dados Factuais , Humanos , Modelos Lineares
6.
Sci Rep ; 11(1): 19647, 2021 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-34608222

RESUMO

Clinical research networks (CRNs), made up of multiple healthcare systems each with patient data from several care sites, are beneficial for studying rare outcomes and increasing generalizability of results. While CRNs encourage sharing aggregate data across healthcare systems, individual systems within CRNs often cannot share patient-level data due to privacy regulations, prohibiting multi-site regression which requires an analyst to access all individual patient data pooled together. Meta-analysis is commonly used to model data stored at multiple institutions within a CRN but can result in biased estimation, most notably in rare-event contexts. We present a communication-efficient, privacy-preserving algorithm for modeling multi-site zero-inflated count outcomes within a CRN. Our method, a one-shot distributed algorithm for performing hurdle regression (ODAH), models zero-inflated count data stored in multiple sites without sharing patient-level data across sites, resulting in estimates closely approximating those that would be obtained in a pooled patient-level data analysis. We evaluate our method through extensive simulations and two real-world data applications using electronic health records: examining risk factors associated with pediatric avoidable hospitalization and modeling serious adverse event frequency associated with a colorectal cancer therapy. In simulations, ODAH produced bias less than 0.1% across all settings explored while meta-analysis estimates exhibited bias up to 12.7%, with meta-analysis performing worst in settings with high zero-inflation or low event rates. Across both applied analyses, ODAH estimates had less than 10% bias for 18 of 20 coefficients estimated, while meta-analysis estimates exhibited substantially higher bias. Relative to existing methods for distributed data analysis, ODAH offers a highly accurate, computationally efficient method for modeling multi-site zero-inflated count data.


Assuntos
Algoritmos , Atenção à Saúde/estatística & dados numéricos , Modelos Estatísticos , Big Data , Mineração de Dados/métodos , Registros Eletrônicos de Saúde/estatística & dados numéricos , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA