Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Yearb Med Inform ; 31(1): 184-198, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36463877

RESUMO

OBJECTIVES: To review current studies about designing and implementing clinician-facing clinical decision support (CDS) integrated or interoperable with an electronic health record (EHR) to improve health care for populations facing disparities. METHODS: We searched PubMed to identify studies published between January 1, 2011 and October 22, 2021 about clinician-facing CDS integrated or interoperable with an EHR. We screened abstracts and titles and extracted study data from articles using a protocol developed by team consensus. Extracted data included patient population characteristics, clinical specialty, setting, EHR, clinical problem, CDS type, reported user-centered design, implementation strategies, and outcomes. RESULTS: There were 28 studies (36 articles) included. Most studies were performed at safety net institutions (14 studies) or Indian Health Service sites (6 studies). CDS tools were implemented in primary care outpatient settings in 24 studies (86%) for screening or treatment. CDS included point-of-care alerts (93%), order facilitators (46%), workflow support (39%), relevant information display (36%), expert systems (11%), and medication dosing support (7%). Successful outcomes were reported in 19 of 26 studies that reported outcomes (73%). User-centered design was reported during CDS planning (39%), development (32%), and implementation phase (25%). Most frequent implementation strategies were education (89%) and consensus facilitation (50%). CONCLUSIONS: CDS tools may improve health equity and outcomes for patients who face disparities. The present review underscores the need for high-quality analyses of CDS-associated health outcomes, reporting of user-centered design and implementation strategies used in low-resource settings, and methods to disseminate CDS created to improve health equity.


Assuntos
Sistemas de Apoio a Decisões Clínicas , Equidade em Saúde , Estados Unidos , Humanos , Registros Eletrônicos de Saúde , Disparidades em Assistência à Saúde , Sistemas Inteligentes
2.
Bioinformatics ; 37(Suppl_1): i67-i75, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34252934

RESUMO

MOTIVATION: Identifying altered transcripts between very small human cohorts is particularly challenging and is compounded by the low accrual rate of human subjects in rare diseases or sub-stratified common disorders. Yet, single-subject studies (S3) can compare paired transcriptome samples drawn from the same patient under two conditions (e.g. treated versus pre-treatment) and suggest patient-specific responsive biomechanisms based on the overrepresentation of functionally defined gene sets. These improve statistical power by: (i) reducing the total features tested and (ii) relaxing the requirement of within-cohort uniformity at the transcript level. We propose Inter-N-of-1, a novel method, to identify meaningful differences between very small cohorts by using the effect size of 'single-subject-study'-derived responsive biological mechanisms. RESULTS: In each subject, Inter-N-of-1 requires applying previously published S3-type N-of-1-pathways MixEnrich to two paired samples (e.g. diseased versus unaffected tissues) for determining patient-specific enriched genes sets: Odds Ratios (S3-OR) and S3-variance using Gene Ontology Biological Processes. To evaluate small cohorts, we calculated the precision and recall of Inter-N-of-1 and that of a control method (GLM+EGS) when comparing two cohorts of decreasing sizes (from 20 versus 20 to 2 versus 2) in a comprehensive six-parameter simulation and in a proof-of-concept clinical dataset. In simulations, the Inter-N-of-1 median precision and recall are > 90% and >75% in cohorts of 3 versus 3 distinct subjects (regardless of the parameter values), whereas conventional methods outperform Inter-N-of-1 at sample sizes 9 versus 9 and larger. Similar results were obtained in the clinical proof-of-concept dataset. AVAILABILITY AND IMPLEMENTATION: R software is available at Lussierlab.net/BSSD.


Assuntos
Perfilação da Expressão Gênica , Doenças Raras , Ontologia Genética , Humanos , Doenças Raras/genética , Transcriptoma
3.
BMJ Health Care Inform ; 28(1)2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33980502

RESUMO

OBJECTIVES: Prior research has reported an increased risk of fatality for patients with cancer, but most studies investigated the risk by comparing cancer to non-cancer patients among COVID-19 infections, where cancer might have contributed to the increased risk. This study is to understand COVID-19's imposed HR of fatality while controlling for covariates, such as age, sex, metastasis status and cancer type. METHODS: We conducted survival analyses of 4606 cancer patients with COVID-19 test results from 16 March to 11 October 2020 in UK Biobank and estimated the overall HR of fatality with and without COVID-19 infection. We also examined the HRs of 13 specific cancer types with at least 100 patients using a stratified analysis. RESULTS: COVID-19 resulted in an overall HR of 7.76 (95% CI 5.78 to 10.40, p<10-10) by following 4606 patients with cancer for 21 days after the tests. The HR varied among cancer type, with over a 10-fold increase in fatality rate (false discovery rate ≤0.02) for melanoma, haematological malignancies, uterine cancer and kidney cancer. Although COVID-19 imposed a higher risk for localised versus distant metastasis cancers, those of distant metastases yielded higher overall fatality rates due to their multiplicative effects. DISCUSSION: The results confirmed prior reports for the increased risk of fatality for patients with COVID-19 plus hematological malignancies and demonstrated similar findings of COVID-19 on melanoma, uterine, and kidney cancers. CONCLUSION: The results highlight the heightened risk that COVID-19 imposes on localised and haematological cancer patients and the necessity to vaccinate uninfected patients with cancer promptly, particularly for the cancer types most influenced by COVID-19. Results also suggest the importance of timely care for patients with localised cancer, whether they are infected by COVID-19 or not.


Assuntos
COVID-19/mortalidade , Nível de Saúde , Neoplasias/mortalidade , Vigilância em Saúde Pública , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Masculino , Neoplasias/patologia , Medição de Risco , Fatores de Risco , Análise de Sobrevida , Adulto Jovem
4.
BMC Bioinformatics ; 21(1): 495, 2020 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-33138767

RESUMO

An amendment to this paper has been published and can be accessed via the original article.

5.
BMC Bioinformatics ; 21(1): 374, 2020 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-32859146

RESUMO

BACKGROUND: In this era of data science-driven bioinformatics, machine learning research has focused on feature selection as users want more interpretation and post-hoc analyses for biomarker detection. However, when there are more features (i.e., transcripts) than samples (i.e., mice or human samples) in a study, it poses major statistical challenges in biomarker detection tasks as traditional statistical techniques are underpowered in high dimension. Second and third order interactions of these features pose a substantial combinatoric dimensional challenge. In computational biology, random forest (RF) classifiers are widely used due to their flexibility, powerful performance, their ability to rank features, and their robustness to the "P > > N" high-dimensional limitation that many matrix regression algorithms face. We propose binomialRF, a feature selection technique in RFs that provides an alternative interpretation for features using a correlated binomial distribution and scales efficiently to analyze multiway interactions. RESULTS: In both simulations and validation studies using datasets from the TCGA and UCI repositories, binomialRF showed computational gains (up to 5 to 300 times faster) while maintaining competitive variable precision and recall in identifying biomarkers' main effects and interactions. In two clinical studies, the binomialRF algorithm prioritizes previously-published relevant pathological molecular mechanisms (features) with high classification precision and recall using features alone, as well as with their statistical interactions alone. CONCLUSION: binomialRF extends upon previous methods for identifying interpretable features in RFs and brings them together under a correlated binomial distribution to create an efficient hypothesis testing algorithm that identifies biomarkers' main effects and interactions. Preliminary results in simulations demonstrate computational gains while retaining competitive model selection and classification accuracies. Future work will extend this framework to incorporate ontologies that provide pathway-level feature selection from gene expression input data.


Assuntos
Algoritmos , Biomarcadores/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/diagnóstico , Biologia Computacional/métodos , Feminino , Humanos , Neoplasias Renais/diagnóstico
6.
J Pers Med ; 11(1)2020 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-33396440

RESUMO

Background: Developing patient-centric baseline standards that enable the detection of clinically significant outlier gene products on a genome-scale remains an unaddressed challenge required for advancing personalized medicine beyond the small pools of subjects implied by "precision medicine". This manuscript proposes a novel approach for reference standard development to evaluate the accuracy of single-subject analyses of transcriptomes and offers extensions into proteomes and metabolomes. In evaluation frameworks for which the distributional assumptions of statistical testing imperfectly model genome dynamics of gene products, artefacts and biases are confounded with authentic signals. Model confirmation biases escalate when studies use the same analytical methods in the discovery sets and reference standards. In such studies, replicated biases are confounded with measures of accuracy. We hypothesized that developing method-agnostic reference standards would reduce such replication biases. We propose to evaluate discovery methods with a reference standard derived from a consensus of analytical methods distinct from the discovery one to minimize statistical artefact biases. Our methods involve thresholding effect-size and expression-level filtering of results to improve consensus between analytical methods. We developed and released an R package "referenceNof1" to facilitate the construction of robust reference standards. Results: Since RNA-Seq data analysis methods often rely on binomial and negative binomial assumptions to non-parametric analyses, the differences create statistical noise and make the reference standards method dependent. In our experimental design, the accuracy of 30 distinct combinations of fold changes (FC) and expression counts (hereinafter "expression") were determined for five types of RNA analyses in two different datasets. This design was applied to two distinct datasets: Breast cancer cell lines and a yeast study with isogenic biological replicates in two experimental conditions. Furthermore, the reference standard (RS) comprised all RNA analytical methods with the exception of the method testing accuracy. To mitigate biases towards a specific analytical method, the pairwise Jaccard Concordance Index between observed results of distinct analytical methods were calculated for optimization. Optimization through thresholding effect-size and expression-level reduced the greatest discordances between distinct methods' analytical results and resulted in a 65% increase in concordance. Conclusions: We have demonstrated that comparing accuracies of different single-subject analysis methods for clinical optimization in transcriptomics requires a new evaluation framework. Reliable and robust reference standards, independent of the evaluated method, can be obtained under a limited number of parameter combinations: Fold change (FC) ranges thresholds, expression level cutoffs, and exclusion of the tested method from the RS development process. When applying anticonservative reference standard frameworks (e.g., using the same method for RS development and prediction), most of the concordant signal between prediction and Gold Standard (GS) cannot be confirmed by other methods, which we conclude as biased results. Statistical tests to determine DEGs from a single-subject study generate many biased results requiring subsequent filtering to increase reliability. Conventional single-subject studies pertain to one or a few patient's measures over time and require a substantial conceptual framework extension to address the numerous measures in genome-wide analyses of gene products. The proposed referenceNof1 framework addresses some of the inherent challenges for improving transcriptome scale single-subject analyses by providing a robust approach to constructing reference standards.

7.
BMC Med Genomics ; 12(Suppl 5): 96, 2019 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-31296218

RESUMO

BACKGROUND: Gene expression profiling has benefited medicine by providing clinically relevant insights at the molecular candidate and systems levels. However, to adopt a more 'precision' approach that integrates individual variability including 'omics data into risk assessments, diagnoses, and therapeutic decision making, whole transcriptome expression needs to be interpreted meaningfully for single subjects. We propose an "all-against-one" framework that uses biological replicates in isogenic conditions for testing differentially expressed genes (DEGs) in a single subject (ss) in the absence of an appropriate external reference standard or replicates. To evaluate our proposed "all-against-one" framework, we construct reference standards (RSs) with five conventional replicate-anchored analyses (NOISeq, DEGseq, edgeR, DESeq, DESeq2) and the remainder were treated separately as single-subject sample pairs for ss analyses (without replicates). RESULTS: Eight ss methods (NOISeq, DEGseq, edgeR, mixture model, DESeq, DESeq2, iDEG, and ensemble) for identifying genes with differential expression were compared in Yeast (parental line versus snf2 deletion mutant; n = 42/condition) and a MCF7 breast-cancer cell line (baseline versus stimulated with estradiol; n = 7/condition). Receiver-operator characteristic (ROC) and precision-recall plots were determined for eight ss methods against each of the five RSs in both datasets. Consistent with prior analyses of these data, ~ 50% and ~ 15% DEGs were obtained in Yeast and MCF7 datasets respectively, regardless of the RSs method. NOISeq, edgeR, and DESeq were the most concordant for creating a RS. Single-subject versions of NOISeq, DEGseq, and an ensemble learner achieved the best median ROC-area-under-the-curve to compare two transcriptomes without replicates regardless of the RS method and dataset (> 90% in Yeast, > 0.75 in MCF7). Further, distinct specific single-subject methods perform better according to different proportions of DEGs. CONCLUSIONS: The "all-against-one" framework provides a honest evaluation framework for single-subject DEG studies since these methods are evaluated, by design, against reference standards produced by unrelated DEG methods. The ss-ensemble method was the only one to reliably produce higher accuracies in all conditions tested in this conservative evaluation framework. However, single-subject methods for identifying DEGs from paired samples need improvement, as no method performed with precision> 90% and obtained moderate levels of recall. http://www.lussiergroup.org/publications/EnsembleBiomarker.


Assuntos
Perfilação da Expressão Gênica/métodos , Medicina de Precisão , Perfilação da Expressão Gênica/normas , Humanos , Padrões de Referência
8.
Front Genet ; 10: 414, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31143202

RESUMO

RNA-Sequencing data offers an opportunity to enable precision medicine, but most methods rely on gene expression alone. To date, no methodology exists to identify and interpret alternative splicing patterns within pathways for an individual patient. This study develops methodology and conducts computational experiments to test the hypothesis that pathway aggregation of subject-specific alternatively spliced genes (ASGs) can inform upon disease mechanisms and predict survival. We propose the N-of-1-pathways Alternatively Spliced (N1PAS) method that takes an individual patient's paired-sample RNA-Seq isoform expression data (e.g., tumor vs. non-tumor, before-treatment vs. during-therapy) and pathway annotations as inputs. N1PAS quantifies the degree of alternative splicing via Hellinger distances followed by two-stage clustering to determine pathway enrichment. We provide a clinically relevant "odds ratio" along with statistical significance to quantify pathway enrichment. We validate our method in clinical samples and find that our method selects relevant pathways (p < 0.05 in 4/6 data sets). Extensive Monte Carlo studies show N1PAS powerfully detects pathway enrichment of ASGs while adequately controlling false discovery rates. Importantly, our studies also unveil highly heterogeneous single-subject alternative splicing patterns that cohort-based approaches overlook. Finally, we apply our patient-specific results to predict cancer survival (FDR < 20%) while providing diagnostics in pursuit of translating transcriptome data into clinically actionable information. Software available at https://github.com/grizant/n1pas/tree/master.

9.
Brief Bioinform ; 20(3): 789-805, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-29272327

RESUMO

The development of computational methods capable of analyzing -omics data at the individual level is critical for the success of precision medicine. Although unprecedented opportunities now exist to gather data on an individual's -omics profile ('personalome'), interpreting and extracting meaningful information from single-subject -omics remain underdeveloped, particularly for quantitative non-sequence measurements, including complete transcriptome or proteome expression and metabolite abundance. Conventional bioinformatics approaches have largely been designed for making population-level inferences about 'average' disease processes; thus, they may not adequately capture and describe individual variability. Novel approaches intended to exploit a variety of -omics data are required for identifying individualized signals for meaningful interpretation. In this review-intended for biomedical researchers, computational biologists and bioinformaticians-we survey emerging computational and translational informatics methods capable of constructing a single subject's 'personalome' for predicting clinical outcomes or therapeutic responses, with an emphasis on methods that provide interpretable readouts. Key points: (i) the single-subject analytics of the transcriptome shows the greatest development to date and, (ii) the methods were all validated in simulations, cross-validations or independent retrospective data sets. This survey uncovers a growing field that offers numerous opportunities for the development of novel validation methods and opens the door for future studies focusing on the interpretation of comprehensive 'personalomes' through the integration of multiple -omics, providing valuable insights into individual patient outcomes and treatments.


Assuntos
Medicina de Precisão , Transcriptoma , Humanos
10.
AMIA Annu Symp Proc ; 2019: 582-591, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32308852

RESUMO

Calculating Differentially Expressed Genes (DEGs) from RNA-sequencing requires replicates to estimate gene-wise variability, a requirement that is at times financially or physiologically infeasible in clinics. By imposing restrictive transcriptome-wide assumptions limiting inferential opportunities of conventional methods (edgeR, NOISeq-sim, DESeq, DEGseq), comparing two conditions without replicates (TCWR) has been proposed, but not evaluated. Under TCWR conditions (e.g., unaffected tissue vs. tumor), differences of transformed expression of the proposed individualized DEG (iDEG) method follow a distribution calculated across a local partition of related transcripts at baseline expression; thereafter the probability of each DEG is estimated by empirical Bayes with local false discovery rate control using a two-group mixture model. In extensive simulation studies of TCWR methods, iDEG and NOISeq are more accurate at 5%90%, recall>75%, false_positive_rate<1%) and 30%

Assuntos
Algoritmos , Perfilação da Expressão Gênica , Análise de Sequência de RNA/métodos , Transcriptoma , Teorema de Bayes , Genômica , Humanos , Conceitos Matemáticos , Modelos Teóricos , Medicina de Precisão
11.
Artigo em Inglês | MEDLINE | ID: mdl-29888037

RESUMO

The transition of procedure coding from ICD-9-CM-Vol-3 to ICD-10-PCS has generated problems for the medical community at large resulting from the lack of clarity required to integrate two non-congruent coding systems. We hypothesized that quantifying these issues with network topology analyses offers a better understanding of the issues, and therefore we developed solutions (online tools) to empower hospital administrators and researchers to address these challenges. Five topologies were identified: "identity"(I), "class-to-subclass"(C2S), "subclass-toclass"(S2C), "convoluted(C)", and "no mapping"(NM). The procedure codes in the 2010 Illinois Medicaid dataset (3,290 patients, 116 institutions) were categorized as C=55%, C2S=40%, I=3%, NM=2%, and S2C=1%. Majority of the problematic and ambiguous mappings (convoluted) pertained to operations in ophthalmology cardiology, urology, gyneco-obstetrics, and dermatology. Finally, the algorithms were expanded into a user-friendly tool to identify problematic topologies and specify lists of procedural codes utilized by medical professionals and researchers for mitigating error-prone translations, simplifying research, and improving quality.http://www.lussiergroup.org/transition-to-ICD10PCS.

12.
BMC Med Genomics ; 11(Suppl 6): 112, 2018 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-30598089

RESUMO

BACKGROUND: Forty-two percent of patients experience disease comorbidity, contributing substantially to mortality rates and increased healthcare costs. Yet, the possibility of underlying shared mechanisms for diseases remains not well established, and few studies have confirmed their molecular predictions with clinical datasets. METHODS: In this work, we integrated genome-wide association study (GWAS) associating diseases and single nucleotide polymorphisms (SNPs) with transcript regulatory activity from expression quantitative trait loci (eQTL). This allowed novel mechanistic insights for noncoding and intergenic regions. We then analyzed pairs of SNPs across diseases to identify shared molecular effectors robust to multiple test correction (False Discovery Rate FDReRNA < 0.05). We hypothesized that disease pairs found to be molecularly convergent would also be significantly overrepresented among comorbidities in clinical datasets. To assess our hypothesis, we used clinical claims datasets from the Healthcare Cost and Utilization Project (HCUP) and calculated significant disease comorbidities (FDRcomorbidity < 0.05). We finally verified if disease pairs resulting molecularly convergent were also statistically comorbid more than by chance using the Fisher's Exact Test. RESULTS: Our approach integrates: (i) 6175 SNPs associated with 238 diseases from ~ 1000 GWAS, (ii) eQTL associations from 19 tissues, and (iii) claims data for 35 million patients from HCUP. Logistic regression (controlled for age, gender, and race) identified comorbidities in HCUP, while enrichment analyses identified cis- and trans-eQTL downstream effectors of GWAS-identified variants. Among ~ 16,000 combinations of diseases, 398 disease-pairs were prioritized by both convergent eQTL-genetics (RNA overlap enrichment, FDReRNA < 0.05) and clinical comorbidities (OR > 1.5, FDRcomorbidity < 0.05). Case studies of comorbidities illustrate specific convergent noncoding regulatory elements. An intergenic architecture of disease comorbidity was unveiled due to GWAS and eQTL-derived convergent mechanisms between distinct diseases being overrepresented among observed comorbidities in clinical datasets (OR = 8.6, p-value = 6.4 × 10- 5 FET). CONCLUSIONS: These comorbid diseases with convergent eQTL genetic mechanisms suggest clinical syndromes. While it took over a decade to confirm the genetic underpinning of the metabolic syndrome, this study is likely highlighting hundreds of new ones. Further, this knowledge may improve the clinical management of comorbidities with precision and shed light on novel approaches of drug repositioning or SNP-guided precision molecular therapy inclusive of intergenic risks.


Assuntos
Comorbidade , Doença/genética , Estudo de Associação Genômica Ampla , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Biologia Computacional , Conjuntos de Dados como Assunto , Feminino , Regulação da Expressão Gênica , Humanos , Masculino , Medicina Molecular/métodos , RNA , Síndrome , Unified Medical Language System
13.
J Am Med Inform Assoc ; 24(6): 1116-1126, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-29016970

RESUMO

OBJECTIVE: To introduce a disease prognosis framework enabled by a robust classification scheme derived from patient-specific transcriptomic response to stimulation. MATERIALS AND METHODS: Within an illustrative case study to predict asthma exacerbation, we designed a stimulation assay that reveals individualized transcriptomic response to human rhinovirus. Gene expression from peripheral blood mononuclear cells was quantified from 23 pediatric asthmatic patients and stimulated in vitro with human rhinovirus. Responses were obtained via the single-subject gene set testing methodology "N-of-1-pathways." The classifier was trained on a related independent training dataset (n = 19). Novel visualizations of personal transcriptomic responses are provided. RESULTS: Of the 23 pediatric asthmatic patients, 12 experienced recurrent exacerbations. Our classifier, using individualized responses and trained on an independent dataset, obtained 74% accuracy (area under the receiver operating curve of 71%; 2-sided P = .039). Conventional classifiers using messenger RNA (mRNA) expression within the viral-exposed samples were unsuccessful (all patients predicted to have recurrent exacerbations; accuracy of 52%). DISCUSSION: Prognosis based on single time point, static mRNA expression alone neglects the importance of dynamic genome-by-environment interplay in phenotypic presentation. Individualized transcriptomic response quantified at the pathway (gene sets) level reveals interpretable signals related to clinical outcomes. CONCLUSION: The proposed framework provides an innovative approach to precision medicine. We show that quantifying personal pathway-level transcriptomic response to a disease-relevant environmental challenge predicts disease progression. This genome-by-environment interaction assay offers a noninvasive opportunity to translate omics data to clinical practice by improving the ability to predict disease exacerbation and increasing the potential to produce more effective treatment decisions.


Assuntos
Asma/genética , Interação Gene-Ambiente , Medicina de Precisão , Transcriptoma , Asma/classificação , Teorema de Bayes , Criança , Conjuntos de Dados como Assunto , Árvores de Decisões , Progressão da Doença , Feminino , Humanos , Leucócitos Mononucleares/metabolismo , Masculino , Modelos Estatísticos , Modelagem Computacional Específica para o Paciente , Prognóstico , RNA Mensageiro/metabolismo , Curva ROC , Rhinovirus , Máquina de Vetores de Suporte , Transcriptoma/imunologia , Transcriptoma/fisiologia
14.
BMC Med Genomics ; 10(Suppl 1): 27, 2017 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-28589853

RESUMO

BACKGROUND: Transcriptome analytic tools are commonly used across patient cohorts to develop drugs and predict clinical outcomes. However, as precision medicine pursues more accurate and individualized treatment decisions, these methods are not designed to address single-patient transcriptome analyses. We previously developed and validated the N-of-1-pathways framework using two methods, Wilcoxon and Mahalanobis Distance (MD), for personal transcriptome analysis derived from a pair of samples of a single patient. Although, both methods uncover concordantly dysregulated pathways, they are not designed to detect dysregulated pathways with up- and down-regulated genes (bidirectional dysregulation) that are ubiquitous in biological systems. RESULTS: We developed N-of-1-pathways MixEnrich, a mixture model followed by a gene set enrichment test, to uncover bidirectional and concordantly dysregulated pathways one patient at a time. We assess its accuracy in a comprehensive simulation study and in a RNA-Seq data analysis of head and neck squamous cell carcinomas (HNSCCs). In presence of bidirectionally dysregulated genes in the pathway or in presence of high background noise, MixEnrich substantially outperforms previous single-subject transcriptome analysis methods, both in the simulation study and the HNSCCs data analysis (ROC Curves; higher true positive rates; lower false positive rates). Bidirectional and concordant dysregulated pathways uncovered by MixEnrich in each patient largely overlapped with the quasi-gold standard compared to other single-subject and cohort-based transcriptome analyses. CONCLUSION: The greater performance of MixEnrich presents an advantage over previous methods to meet the promise of providing accurate personal transcriptome analysis to support precision medicine at point of care.


Assuntos
Perfilação da Expressão Gênica/métodos , Neoplasias de Cabeça e Pescoço/genética , Humanos , Neoplasias de Células Escamosas/genética , Medicina de Precisão , Curva ROC
15.
J Biomed Inform ; 66: 32-41, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28007582

RESUMO

MOTIVATION: Understanding dynamic, patient-level transcriptomic response to therapy is an important step forward for precision medicine. However, conventional transcriptome analysis aims to discover cohort-level change, lacking the capacity to unveil patient-specific response to therapy. To address this gap, we previously developed two N-of-1-pathways methods, Wilcoxon and Mahalanobis distance, to detect unidirectionally responsive transcripts within a pathway using a pair of samples from a single subject. Yet, these methods cannot recognize bidirectionally (up and down) responsive pathways. Further, our previous approaches have not been assessed in presence of background noise and are not designed to identify differentially expressed mRNAs between two samples of a patient taken in different contexts (e.g. cancer vs non cancer), which we termed responsive transcripts (RTs). METHODS: We propose a new N-of-1-pathways method, k-Means Enrichment (kMEn), that detects bidirectionally responsive pathways, despite background noise, using a pair of transcriptomes from a single patient. kMEn identifies transcripts responsive to the stimulus through k-means clustering and then tests for an over-representation of the responsive genes within each pathway. The pathways identified by kMEn are mechanistically interpretable pathways significantly responding to a stimulus. RESULTS: In ∼9000 simulations varying six parameters, superior performance of kMEn over previous single-subject methods is evident by: (i) improved precision-recall at various levels of bidirectional response and (ii) lower rates of false positives (1-specificity) when more than 10% of genes in the genome are differentially expressed (background noise). In a clinical proof-of-concept, personal treatment-specific pathways identified by kMEn correlate with therapeutic response (p-value<0.01). CONCLUSION: Through improved single-subject transcriptome dynamics of bidirectionally-regulated signals, kMEn provides a novel approach to identify mechanism-level biomarkers.


Assuntos
Perfilação da Expressão Gênica , Medicina de Precisão , Transcriptoma , Análise por Conglomerados , Interpretação Estatística de Dados , Humanos , RNA Mensageiro
16.
Bioinformatics ; 32(12): i80-i89, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27307648

RESUMO

MOTIVATION: As 'omics' biotechnologies accelerate the capability to contrast a myriad of molecular measurements from a single cell, they also exacerbate current analytical limitations for detecting meaningful single-cell dysregulations. Moreover, mRNA expression alone lacks functional interpretation, limiting opportunities for translation of single-cell transcriptomic insights to precision medicine. Lastly, most single-cell RNA-sequencing analytic approaches are not designed to investigate small populations of cells such as circulating tumor cells shed from solid tumors and isolated from patient blood samples. RESULTS: In response to these characteristics and limitations in current single-cell RNA-sequencing methodology, we introduce an analytic framework that models transcriptome dynamics through the analysis of aggregated cell-cell statistical distances within biomolecular pathways. Cell-cell statistical distances are calculated from pathway mRNA fold changes between two cells. Within an elaborate case study of circulating tumor cells derived from prostate cancer patients, we develop analytic methods of aggregated distances to identify five differentially expressed pathways associated to therapeutic resistance. Our aggregation analyses perform comparably with Gene Set Enrichment Analysis and better than differentially expressed genes followed by gene set enrichment. However, these methods were not designed to inform on differential pathway expression for a single cell. As such, our framework culminates with the novel aggregation method, cell-centric statistics (CCS). CCS quantifies the effect size and significance of differentially expressed pathways for a single cell of interest. Improved rose plots of differentially expressed pathways in each cell highlight the utility of CCS for therapeutic decision-making. AVAILABILITY AND IMPLEMENTATION: http://www.lussierlab.org/publications/CCS/ CONTACT: yves@email.arizona.edu or piegorsch@math.arizona.edu SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Células Neoplásicas Circulantes/efeitos dos fármacos , Análise de Sequência de RNA , Transcriptoma , Perfilação da Expressão Gênica , Humanos , Masculino , Neoplasias da Próstata/tratamento farmacológico , RNA
17.
J Am Med Inform Assoc ; 22(3): 730-7, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25681260

RESUMO

In the United States, International Classification of Disease Clinical Modification (ICD-9-CM, the ninth revision) diagnosis codes are commonly used to identify patient cohorts and to conduct financial analyses related to disease. In October 2015, the healthcare system of the United States will transition to ICD-10-CM (the tenth revision) diagnosis codes. One challenge posed to clinical researchers and other analysts is conducting diagnosis-related queries across datasets containing both coding schemes. Further, healthcare administrators will manage growth, trends, and strategic planning with these dually-coded datasets. The majority of the ICD-9-CM to ICD-10-CM translations are complex and nonreciprocal, creating convoluted representations and meanings. Similarly, mapping back from ICD-10-CM to ICD-9-CM is equally complex, yet different from mapping forward, as relationships are likewise nonreciprocal. Indeed, 10 of the 21 top clinical categories are complex as 78% of their diagnosis codes are labeled as "convoluted" by our analyses. Analysis and research related to external causes of morbidity, injury, and poisoning will face the greatest challenges due to 41 745 (90%) convolutions and a decrease in the number of codes. We created a web portal tool and translation tables to list all ICD-9-CM diagnosis codes related to the specific input of ICD-10-CM diagnosis codes and their level of complexity: "identity" (reciprocal), "class-to-subclass," "subclass-to-class," "convoluted," or "no mapping." These tools provide guidance on ambiguous and complex translations to reveal where reports or analyses may be challenging to impossible.Web portal: http://www.lussierlab.org/transition-to-ICD9CM/Tables annotated with levels of translation complexity: http://www.lussierlab.org/publications/ICD10to9.


Assuntos
Codificação Clínica/métodos , Classificação Internacional de Doenças , Humanos , Classificação Internacional de Doenças/economia , Internet , Estados Unidos
18.
J Am Med Inform Assoc ; 22(1): 19-28, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25186492

RESUMO

Reporting of hospital adverse events relies on Patient Safety Indicators (PSIs) using International Classification of Diseases, Ninth Edition, Clinical Modification (ICD-9-CM) codes. The US transition to ICD-10-CM in 2015 could result in erroneous comparisons of PSIs. Using the General Equivalent Mappings (GEMs), we compared the accuracy of ICD-9-CM coded PSIs against recommended ICD-10-CM codes from the Centers for Medicaid/Medicare Services (CMS). We further predict their impact in a cohort of 38,644 patients (1,446,581 visits and 399 hospitals). We compared the predicted results to the published PSI related ICD-10-CM diagnosis codes. We provide the first report of substantial hospital safety reporting errors with five direct comparisons from the 23 types of PSIs (transfusion and anesthesia related PSIs). One PSI was excluded from the comparison between code sets due to reorganization, while 15 additional PSIs were inaccurate to a lesser degree due to the complexity of the coding translation. The ICD-10-CM translations proposed by CMS pose impending risks for (1) comparing safety incidents, (2) inflating the number of PSIs, and (3) increasing the variability of calculations attributable to the abundance of coding system translations. Ethical organizations addressing 'data-, process-, and system-focused' improvements could be penalized using the new ICD-10-CM Agency for Healthcare Research and Quality PSIs because of apparent increases in PSIs bearing the same PSI identifier and label, yet calculated differently. Here we investigate which PSIs would reliably transition between ICD-9-CM and ICD-10-CM, and those at risk of under-reporting and over-reporting adverse events while the frequency of these adverse events remain unchanged.


Assuntos
Classificação Internacional de Doenças , Segurança do Paciente , Indicadores de Qualidade em Assistência à Saúde , Humanos , Informática Médica
19.
AMIA Annu Symp Proc ; 2014: 855-64, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25954392

RESUMO

Earlier studies on hospitalization risk are largely based on regression models. To our knowledge, network modeling of multiple comorbidities is novel and inherently enables multidimensional scoring and unbiased feature reduction. Network modeling was conducted using an independent validation design starting from 38,695 patients, 1,446,581 visits, and 430 distinct clinical facilities/hospitals. Odds ratios (OR) were calculated for every pair of comorbidity using patient counts and compared their tendency with hospitalization rates and ED visits. Network topology analyses were performed, defining significant comorbidity associations as having OR≥5 & False-Discovery-Rate≤10(-7). Four COPD-associated comorbidity sub-networks emerged, incorporating multiple clinical systems: (i) metabolic syndrome, (ii) substance abuse and mental disorder, (iii) pregnancy-associated conditions, and (iv) fall-related injury. The latter two have not been reported yet. Features prioritized from the network are predictive of hospitalizations in an independent set (p<0.004). Therefore, we suggest that network topology is a scalable and generalizable method predictive of hospitalization.


Assuntos
Comorbidade , Hospitalização , Doença Pulmonar Obstrutiva Crônica , Acidentes por Quedas , Diabetes Mellitus/epidemiologia , Feminino , Humanos , Gravidez , Complicações na Gravidez/epidemiologia , Risco , Transtornos Relacionados ao Uso de Substâncias/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...