Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
Sci Rep ; 11(1): 17170, 2021 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-34446747

RESUMO

The present study aimed to construct and evaluate a novel experiment-based hypoxia signature to help evaluations of GBM patient status. First, the 426 proteins, which were previously found to be differentially expressed between normal and hypoxia groups in glioblastoma cells with statistical significance, were converted into the corresponding genes, among which 212 genes were found annotated in TCGA. Second, after evaluated by single-variable Cox analysis, 19 different expressed genes (DEGs) with prognostic value were identified. Based on λ value by LASSO, a gene-based survival risk score model, named RiskScore, was built by 7 genes with LASSO coefficient, which were FKBP2, GLO1, IGFBP5, NSUN5, RBMX, TAGLN2 and UBE2V2. Kaplan-Meier (K-M) survival curve analysis and the area under the curve (AUC) were plotted to further estimate the efficacy of this risk score model. Furthermore, the survival curve analysis was also plotted based on the subtypes of age, IDH, radiotherapy and chemotherapy. Meanwhile, immune infiltration, GSVA, GSEA and chemo drug sensitivity of this risk score model were evaluated. Third, the 7 genes expression were evaluated by AUC, overall survival (OS) and IDH subtype in datasets, importantly, also experimentally verified in GBM cell lines exposed to hypoxic or normal oxygen condition, which showed significant higher expression in hypoxia than in normal group. Last, combing the hypoxia RiskScore with clinical and molecular features, a prognostic composite nomogram was generated, showing the good sensitivity and specificity by AUC and OS. Meanwhile, univariate analysis and multivariate analysis were used for performed to identify variables in nomogram that were significant in independently predicting duration of survival. It is a first time that we successfully established and validated an independent prognostic risk model based on hypoxia microenvironment from glioblastoma cells and public database. The 7 key genes may provide potential directions for future biochemical and pharmaco-therapeutic research.


Assuntos
Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Proteoma/metabolismo , Proteômica/métodos , Microambiente Tumoral/genética , Idoso , Linhagem Celular Tumoral , Bases de Dados Factuais/estatística & dados numéricos , Feminino , Perfilação da Expressão Gênica/estatística & dados numéricos , Glioblastoma/diagnóstico , Glioblastoma/metabolismo , Humanos , Hipóxia , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Nomogramas , Farmacogenética/métodos , Farmacogenética/estatística & dados numéricos , Prognóstico , Proteoma/genética , Proteômica/estatística & dados numéricos
2.
Nat Commun ; 12(1): 1033, 2021 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-33589615

RESUMO

Clinical trials of novel therapeutics for Alzheimer's Disease (AD) have consumed a large amount of time and resources with largely negative results. Repurposing drugs already approved by the Food and Drug Administration (FDA) for another indication is a more rapid and less expensive option. We present DRIAD (Drug Repurposing In AD), a machine learning framework that quantifies potential associations between the pathology of AD severity (the Braak stage) and molecular mechanisms as encoded in lists of gene names. DRIAD is applied to lists of genes arising from perturbations in differentiated human neural cell cultures by 80 FDA-approved and clinically tested drugs, producing a ranked list of possible repurposing candidates. Top-scoring drugs are inspected for common trends among their targets. We propose that the DRIAD method can be used to nominate drugs that, after additional validation and identification of relevant pharmacodynamic biomarker(s), could be readily evaluated in a clinical trial.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Drogas em Investigação/farmacologia , Aprendizado de Máquina , Proteínas do Tecido Nervoso/genética , Fármacos Neuroprotetores/farmacologia , Nootrópicos/farmacologia , Medicamentos sob Prescrição/farmacologia , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Reposicionamento de Medicamentos , Drogas em Investigação/química , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Ensaios de Triagem em Larga Escala , Humanos , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/química , Nootrópicos/química , Farmacogenética/métodos , Farmacogenética/estatística & dados numéricos , Polifarmacologia , Medicamentos sob Prescrição/química , Cultura Primária de Células , Índice de Gravidade de Doença
3.
Pharmacogenomics ; 21(17): 1247-1264, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33124490

RESUMO

Pharmacogenomics (PGx) implementation in clinical practice is steadily increasing. PGx uses genetic information to personalize medication use, which increases medication efficacy and decreases side effects. The availability of clinical PGx guidelines is essential for its implementation in clinical settings. Currently, there are few organizations/associations responsible for releasing those guidelines, including the Clinical Pharmacogenetics Implementation Consortium, Dutch Pharmacogenetics Working Group, the Canadian Pharmacogenomics Network for Drug Safety and the French National Network of Pharmacogenetics. According to the US FDA, oncology medications are highly correlated to PGx biomarkers. Therefore, summarizing the PGx guidelines for oncology drugs will positively impact the clinical decisions for cancer patients. This review aims to scrutinize side-by-side available clinical PGx guidelines in oncology.


Assuntos
Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Guias como Assunto , Oncologia/legislação & jurisprudência , Oncologia/normas , Farmacogenética/legislação & jurisprudência , Farmacogenética/estatística & dados numéricos , Biomarcadores Tumorais , Humanos , Testes Farmacogenômicos , Medicina de Precisão
4.
Surgery ; 166(4): 476-482, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31320226

RESUMO

BACKGROUND: Despite the current strategies aimed at avoiding opioid overprescription by implementing institutional guidelines, the use of opioids after surgical procedures remains highly variable. It is well known that opioids are activated by the cytochrome p450 CYP2D6 enzyme to exert pharmacologic effect. Individual variation in CYP2D6 activity affects drug metabolism, and genotyping can be performed to predict an individual's ability to metabolize CYP2D6 substrates. We postulate that the pharmacogenomic identification of patients with different opioid metabolism capacity may allow for the individualization of postsurgical opioid prescription. METHODS: This study was generated by the unison of data from 2 prior initiatives taking place at our Institution. In the first study, patients undergoing 1 of 25 elective surgical procedures were prospectively identified as part of a quality initiative and surveyed by phone 21 to 35 days after hospital discharge to complete a 29-question survey regarding opioid utilization and pain experience. Additional chart abstraction was conducted to obtain prescribing data and pain scores during the hospitalization. The second study was the Mayo Clinic Right Drug, Right Dose, Right Time study protocol, in which 5 pharmacogenes, including CYP2D6, were genotyped for 1,000 Mayo Clinic Biobank participants. The goal of this study was to implement preemptive pharmacogenomics in an academic health care setting and to generate data for further pharmacogenomic research. Patients were classified by their predicted CYP2D6 activity based on their CYP2D6 genotype. RESULTS: Of the 2,486 patients with prospective opioid utilization data, 21 had pharmacogenetic data available and were included in the analysis. These patients were classified according to their activity as opioid metabolizers, with 10 patients (48%) classified as intermediate, 4 patients (19%) as intermediate to normal, and 7 patients (33%) as normal or extensive. Compared with the intermediate to normal and intermediate phenotypes, normal or extensive patients had the highest percentages of preoperative opioid naivety and recorded pain scores throughout the surgical experience. The percentage of unused opioids for intermediate, intermediate to normal, and normal or extensive categories was 79%, 63%, and 46%, respectively. Moreover, of the 14 patients declaring the highest level of satisfaction for their pain control after discharge, 60% belonged to intermediate, 100% to intermediate to normal, and 57% to the normal or extensive group. CONCLUSION: This study outlines a possible correlation between genetically controlled metabolism and opioid requirements after surgery. In this setting, an increased CYP2D6 enzymatic activity was associated to a greater opioid consumption, lesser amount of unused opioids, and a lower satisfaction level from opioid prescription.


Assuntos
Analgésicos Opioides/uso terapêutico , Citocromo P-450 CYP2D6/genética , Uso de Medicamentos/estatística & dados numéricos , Dor Pós-Operatória/tratamento farmacológico , Medicina de Precisão/métodos , Centros Médicos Acadêmicos , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Bases de Dados Factuais , Feminino , Seguimentos , Genótipo , Humanos , Tempo de Internação , Masculino , Pessoa de Meia-Idade , Minnesota , Manejo da Dor/métodos , Dor Pós-Operatória/diagnóstico , Farmacogenética/estatística & dados numéricos , Padrões de Prática Médica/estatística & dados numéricos , Estudos Prospectivos , Medição de Risco , Estatísticas não Paramétricas
5.
Pac Symp Biocomput ; 24: 248-259, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30864327

RESUMO

The inconsistency of open pharmacogenomics datasets produced by different studies limits the usage of such datasets in many tasks, such as biomarker discovery. Investigation of multiple pharmacogenomics datasets confirmed that the pairwise sensitivity data correlation between drugs, or rows, across different studies (drug-wise) is relatively low, while the pairwise sensitivity data correlation between cell-lines, or columns, across different studies (cell-wise) is considerably strong. This common interesting observation across multiple pharmacogenomics datasets suggests the existence of subtle consistency among the different studies (i.e., strong cell-wise correlation). However, significant noises are also shown (i.e., weak drug-wise correlation) and have prevented researchers from comfortably using the data directly. Motivated by this observation, we propose a novel framework for addressing the inconsistency between large-scale pharmacogenomics data sets. Our method can significantly boost the drug-wise correlation and can be easily applied to re-summarized and normalized datasets proposed by others. We also investigate our algorithm based on many different criteria to demonstrate that the corrected datasets are not only consistent, but also biologically meaningful. Eventually, we propose to extend our main algorithm into a framework, so that in the future when more datasets become publicly available, our framework can hopefully offer a "ground-truth" guidance for references.


Assuntos
Algoritmos , Bases de Dados Genéticas/estatística & dados numéricos , Farmacogenética/estatística & dados numéricos , Linhagem Celular Tumoral , Biologia Computacional/métodos , Bases de Dados de Produtos Farmacêuticos/estatística & dados numéricos , Resistencia a Medicamentos Antineoplásicos/genética , Marcadores Genéticos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Variantes Farmacogenômicos , Medicina de Precisão
6.
Sci Rep ; 8(1): 15742, 2018 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-30356105

RESUMO

Diversity in drug response is attributed to both genetic and non-genetic factors. However, there is paucity of pharmacogenetics information across ethnically and genetically diverse populations of India. Here, we have analyzed 21 SNPs from 12 pharmacogenomics genes in Punjabi Sikhs of Indian origin (N = 1,616), as part of the Sikh Diabetes Study (SDS). We compared the allele frequency of poor metabolism (PM) phenotype among Sikhs across other major global populations from the Exome Aggregation Consortium and 1000 Genomes. The PM phenotype of CYP1A2*1 F for slow metabolism of caffeine and carcinogens was significantly higher in Indians (SDS 42%, GIH [Gujarati] 51%, SAS [Pakistani] 45%) compared to Europeans 29% (pgenotype = 5.3E-05). Similarly, South Asians had a significantly higher frequency of CYP2C9*3 (12% SDS, 13% GIH, 11% SAS) vs. 7% in Europeans (pgenotype = <1.0E-05) and 'T' allele of CYP4F2 (36%) SDS, (43%) GIH, 40% (SAS) vs. (29%) in Europeans (pgenotype = <1.0E-05); both associated with a higher risk of bleeding with warfarin. All South Asians -the Sikhs (0.36), GIH (0.34), and SAS (0.36) had a higher frequency of the NAT2*6 allele (linked with slow acetylation of isoniazid) compared to Europeans (0.29). Additionally, the prevalence of the low activity 'C' allele of MTHFR (rs1801131) was highest in Sikhs compared to all other ethnic groups [SDS (44%), GIH (39%), SAS (42%) and European (32%) (pgenotype = <1.0E-05)]. SNPs in MTHFR affect metabolism of statins, 5-fluorouracil and methotrexate-based cancer drugs. These findings underscore the need for evaluation of other endogamous ethnic groups of India and beyond for establishing a global benchmark for pre-emptive genotyping in drug metabolizing genes before beginning therapeutic intervention.


Assuntos
Etnicidade/genética , Frequência do Gene , Farmacogenética/estatística & dados numéricos , Arilamina N-Acetiltransferase , Citocromo P-450 CYP1A2 , Citocromo P-450 CYP2C9 , Humanos , Índia , Erros Inatos do Metabolismo/genética , Metilenotetra-Hidrofolato Redutase (NADPH2)/genética , Preparações Farmacêuticas/metabolismo , Fenótipo , Polimorfismo de Nucleotídeo Único
7.
Cancer ; 124(14): 3052-3065, 2018 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-29742281

RESUMO

BACKGROUND: Germline and tumor pharmacogenomics impact drug responses, but germline markers less commonly guide oncology prescribing. The authors hypothesized that a critical number of clinically actionable germline pharmacogenomic associations exist, representing clinical implementation opportunities. METHODS: In total, 125 oncology drugs were analyzed for positive germline pharmacogenomic associations in journals with impact factors ≥5. Studies were assessed for design and genotyping quality, clinically relevant outcomes, statistical rigor, and evidence of drug-gene effects. Associations from studies of high methodologic quality were deemed potentially clinically actionable, and translational summaries were written as point-of-care clinical decision support (CDS) tools and formally evaluated using the Appraisal of Guidelines for Research and Evaluation (AGREE) II instrument. RESULTS: The authors identified germline pharmacogenomic results for 56 of 125 oncology drugs (45%) across 173 publications. Actionable associations were detected for 12 drugs, including 6 that had germline pharmacogenomic information within US Food and Drug Administration labels or published guidelines (capecitabine/fluorouracil/dihydropyrimidine dehydrogenase [DPYD], irinotecan/uridine diphosphate glucuronosyltransferase family 1 member A1 [UGT1A1], mercaptopurine/thioguanine/thiopurine S-methyltransferase [TPMT], tamoxifen/cytochrome P450 [CYP] family 2 subfamily D member 6 [CYP2D6]), and 6 others were novel (asparaginase/nuclear factor of activated T-cells 2 [NFATC2]/human leukocyte antigen D-related ß1 [HLA-DRB1], cisplatin/acylphosphatase 2 [ACYP2], doxorubicin/adenosine triphosphate-binding cassette subfamily C member 2/Rac family small guanosine triphosphatase 2/neutrophil cytosolic factor 4 [ABCC2/RAC2/NCF4], lapatinib/human leukocyte antigen DQ α1 [HLA-DQA1], sunitinib/cytochrome P450 family 3 subfamily A member 5 [CYP3A5], vincristine/centrosomal protein 72 [CEP72]). By using AGREE II, the developed CDS summaries had high mean ± standard deviation scores (maximum score, 100) for scope and purpose (92.7 ± 5.1) and rigour of development (87.6 ± 7.4) and moderate yet robust scores for clarity of presentation (58.6 ± 25.1) and applicability (55.9 ± 24.6). The overall mean guideline quality score was 5.2 ± 1.0 (maximum score, 7). Germline pharmacogenomic CDS summaries for these 12 drugs were recommended for implementation. CONCLUSIONS: Several oncology drugs have actionable germline pharmacogenomic information, justifying their delivery through institutional pharmacogenomic implementations to determine clinical utility. Cancer 2018;124:3052-65. © 2018 American Cancer Society.


Assuntos
Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/genética , Neoplasias/tratamento farmacológico , Farmacogenética/estatística & dados numéricos , Medicina de Precisão/normas , Antineoplásicos/farmacologia , Tomada de Decisão Clínica/métodos , Prescrições de Medicamentos/estatística & dados numéricos , Testes Genéticos/normas , Testes Genéticos/estatística & dados numéricos , Técnicas de Genotipagem/normas , Técnicas de Genotipagem/estatística & dados numéricos , Mutação em Linhagem Germinativa/genética , Humanos , Proteína 2 Associada à Farmacorresistência Múltipla , Neoplasias/genética , Seleção de Pacientes , Guias de Prática Clínica como Assunto , Medicina de Precisão/estatística & dados numéricos , Estudos Prospectivos , Resultado do Tratamento
8.
Pac Symp Biocomput ; 23: 44-55, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29218868

RESUMO

A variety of large-scale pharmacogenomic data, such as perturbation experiments and sensitivity profiles, enable the systematical identification of drug mechanism of actions (MoAs), which is a crucial task in the era of precision medicine. However, integrating these complementary pharmacogenomic datasets is inherently challenging due to the wild heterogeneity, high-dimensionality and noisy nature of these datasets. In this work, we develop Mania, a novel method for the scalable integration of large-scale pharmacogenomic data. Mania first constructs a drug-drug similarity network through integrating multiple heterogeneous data sources, including drug sensitivity, drug chemical structure, and perturbation assays. It then learns a compact vector representation for each drug to simultaneously encode its structural and pharmacogenomic properties. Extensive experiments demonstrate that Mania achieves substantially improved performance in both MoAs and targets prediction, compared to predictions based on individual data sources as well as a state-of-the-art integrative method. Moreover, Mania identifies drugs that target frequently mutated cancer genes, which provides novel insights into drug repurposing.


Assuntos
Farmacogenética/estatística & dados numéricos , Algoritmos , Biologia Computacional/métodos , Bases de Dados de Produtos Farmacêuticos/estatística & dados numéricos , Reposicionamento de Medicamentos/estatística & dados numéricos , Ensaios de Seleção de Medicamentos Antitumorais/estatística & dados numéricos , Humanos , Estrutura Molecular , Medicina de Precisão , Integração de Sistemas
10.
Clin Pharmacol Ther ; 99(4): 401-4, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26756170

RESUMO

The field of pharmacogenomics originally emerged in the 1950s from observations that a few rare individuals had unexpected, severe reactions to drugs. As recently as just 6 years ago, prominent views on the subject had largely remained unchanged, with authors from the US Food and Drug Administration (FDA) citing the purpose of pharmacogenetics as "tailoring treatment for the outliers." It should not be surprising if this is the prevailing view--the best-studied pharmacogenomic drug examples are indeed just that, genetic explanations of extreme responses or susceptibilities among usually a very small fraction of the human population. Thiopurine methyltransferase (TPMT) deficiency as a cause of severe myelosuppression upon treatment with azathioprine or mercaptopurine is found as a heterozygous trait in only ∼ 10% of patients, and homozygous (deficiency) carriers are even more rare--occurring in fewer than 1 in 300 patients. Malignant hyperthermia resulting from inhaled anesthetics and succinylcholine is believed to have a genetic incidence of only about 1 in 2000 people.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/genética , Farmacogenética , Interpretação Estatística de Dados , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/diagnóstico , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Predisposição Genética para Doença , Humanos , Modelos Estatísticos , Farmacogenética/estatística & dados numéricos , Fenótipo , Medição de Risco , Fatores de Risco
11.
Biomed Res Int ; 2015: 670691, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26446492

RESUMO

The process for using statistical inference to establish personalized treatment strategies requires specific techniques for data-analysis that optimize the combination of competing therapies with candidate genetic features and characteristics of the patient and disease. A wide variety of methods have been developed. However, heretofore the usefulness of these recent advances has not been fully recognized by the oncology community, and the scope of their applications has not been summarized. In this paper, we provide an overview of statistical methods for establishing optimal treatment rules for personalized medicine and discuss specific examples in various medical contexts with oncology as an emphasis. We also point the reader to statistical software for implementation of the methods when available.


Assuntos
Oncologia/estatística & dados numéricos , Farmacogenética/estatística & dados numéricos , Medicina de Precisão/estatística & dados numéricos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/epidemiologia , Neoplasias/genética
12.
BMC Med Genet ; 16: 32, 2015 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-25956914

RESUMO

BACKGROUND: Pharmacogenetics is a rapidly growing field that aims to identify the genes that influence drug response. This science can be used as a powerful tool to tailor drug treatment to the genetic makeup of individuals. The present study explores the coverage of the topic of pharmacogenetics and its potential benefit in personalised medicine by the UK newsprint media. METHODS: The LexisNexis database was used to identify and retrieve full text articles from the 10 highest circulation national daily newspapers and their Sunday equivalents in the UK. Content analysis of newspaper articles which referenced pharmacogenetic testing was carried out. A second researcher coded a random sample (21%) of newspaper articles to establish the inter-rater reliability of coding. RESULTS: Of the 256 articles captured by the search terms, 96 articles (with pharmacogenetics as a major component) met the study inclusion criteria. The majority of articles over-stated the benefits of pharmacogenetic testing while paying less attention to the associated risks. Overall beneficial effects were mentioned 5.3 times more frequently than risks (p < 0.001). The most common illnesses for which pharmacogenetically based personalised medicine was discussed were cancer, cardiovascular disease and CNS diseases. Only 13% of newspaper articles that cited a specific scientific study mentioned this link in the article. There was a positive correlation between the size of the article and both the number of benefits and risks stated (P < 0.01). CONCLUSION: More comprehensive coverage of the area of personalised medicine within the print media is needed to inform public debate on the inclusion of pharmacogentic testing in routine practice.


Assuntos
Jornais como Assunto , Farmacogenética/educação , Farmacogenética/estatística & dados numéricos , Opinião Pública , Farmacogenética/normas , Medicina de Precisão/normas , Medicina de Precisão/estatística & dados numéricos , Risco
13.
Biometrics ; 71(2): 529-37, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25604216

RESUMO

Pharmacogenetics investigates the relationship between heritable genetic variation and the variation in how individuals respond to drug therapies. Often, gene-drug interactions play a primary role in this response, and identifying these effects can aid in the development of individualized treatment regimes. Haplotypes can hold key information in understanding the association between genetic variation and drug response. However, the standard approach for haplotype-based association analysis does not directly address the research questions dictated by individualized medicine. A complementary post-hoc analysis is required, and this post-hoc analysis is usually under powered after adjusting for multiple comparisons and may lead to seemingly contradictory conclusions. In this work, we propose a penalized likelihood approach that is able to overcome the drawbacks of the standard approach and yield the desired personalized output. We demonstrate the utility of our method by applying it to the Scottish Randomized Trial in Ovarian Cancer. We also conducted simulation studies and showed that the proposed penalized method has comparable or more power than the standard approach and maintains low Type I error rates for both binary and quantitative drug responses. The largest performance gains are seen when the haplotype frequency is low, the difference in effect sizes are small, or the true relationship among the drugs is more complex.


Assuntos
Funções Verossimilhança , Farmacogenética/estatística & dados numéricos , Antineoplásicos/efeitos adversos , Biometria , Simulação por Computador , Feminino , Genes bcl-2 , Haplótipos , Humanos , Modelos Estatísticos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Análise de Regressão
14.
Pac Symp Biocomput ; : 32-43, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25592566

RESUMO

Complex mechanisms involving genomic aberrations in numerous proteins and pathways are believed to be a key cause of many diseases such as cancer. With recent advances in genomics, elucidating the molecular basis of cancer at a patient level is now feasible, and has led to personalized treatment strategies whereby a patient is treated according to his or her genomic profile. However, there is growing recognition that existing treatment modalities are overly simplistic, and do not fully account for the deep genomic complexity associated with sensitivity or resistance to cancer therapies. To overcome these limitations, large-scale pharmacogenomic screens of cancer cell lines--in conjunction with modern statistical learning approaches--have been used to explore the genetic underpinnings of drug response. While these analyses have demonstrated the ability to infer genetic predictors of compound sensitivity, to date most modeling approaches have been data-driven, i.e. they do not explicitly incorporate domain-specific knowledge (priors) in the process of learning a model. While a purely data-driven approach offers an unbiased perspective of the data--and may yield unexpected or novel insights--this strategy introduces challenges for both model interpretability and accuracy. In this study, we propose a novel prior-incorporated sparse regression model in which the choice of informative predictor sets is carried out by knowledge-driven priors (gene sets) in a stepwise fashion. Under regularization in a linear regression model, our algorithm is able to incorporate prior biological knowledge across the predictive variables thereby improving the interpretability of the final model with no loss--and often an improvement--in predictive performance. We evaluate the performance of our algorithm compared to well-known regularization methods such as LASSO, Ridge and Elastic net regression in the Cancer Cell Line Encyclopedia (CCLE) and Genomics of Drug Sensitivity in Cancer (Sanger) pharmacogenomics datasets, demonstrating that incorporation of the biological priors selected by our model confers improved predictability and interpretability, despite much fewer predictors, over existing state-of-the-art methods.


Assuntos
Modelos Lineares , Farmacogenética/estatística & dados numéricos , Algoritmos , Linhagem Celular Tumoral , Biologia Computacional , Bases de Dados Genéticas , Ensaios de Seleção de Medicamentos Antitumorais/estatística & dados numéricos , Humanos , Modelos Genéticos , Neoplasias/tratamento farmacológico , Neoplasias/genética
15.
Eur J Cancer ; 50(15): 2532-43, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25103456

RESUMO

There are an increasing number of studies devoted to the identification of associations between anticancer drug efficacy and toxicity and common polymorphisms present in the patients' genome. However, many articles presenting the results of such studies do not bring the simple and necessary background information allowing the evaluation of the relevance of the study, its significance and its potential importance for patients' treatment. This position paper first addresses clinical oncologists with the aim of giving them the basic knowledge on pharmacogenetics and on the potential use of gene polymorphisms as predictive biomarkers in routine and clinical research. A secondary objective is to give molecular biologists some recommendations on how to conceive protocols and how to publish their results when they develop pharmacogenetic studies appended to clinical trials or with autonomous goals.


Assuntos
Ensaios Clínicos como Assunto/métodos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Polimorfismo Genético , Ensaios Clínicos como Assunto/estatística & dados numéricos , Frequência do Gene , Predisposição Genética para Doença/genética , Genótipo , Humanos , Farmacogenética/métodos , Farmacogenética/estatística & dados numéricos , Resultado do Tratamento
16.
Pac Symp Biocomput ; : 27-38, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24297531

RESUMO

Computational efficiency is important for learning algorithms operating in the "large p, small n" setting. In computational biology, the analysis of data sets containing tens of thousands of features ("large p"), but only a few hundred samples ("small n"), is nowadays routine, and regularized regression approaches such as ridge-regression, lasso, and elastic-net are popular choices. In this paper we propose a novel and highly efficient Bayesian inference method for fitting ridge-regression. Our method is fully analytical, and bypasses the need for expensive tuning parameter optimization, via cross-validation, by employing Bayesian model averaging over the grid of tuning parameters. Additional computational efficiency is achieved by adopting the singular value decomposition reparametrization of the ridge-regression model, replacing computationally expensive inversions of large p × p matrices by efficient inversions of small and diagonal n × n matrices. We show in simulation studies and in the analysis of two large cancer cell line data panels that our algorithm achieves slightly better predictive performance than cross-validated ridge-regression while requiring only a fraction of the computation time. Furthermore, in comparisons based on the cell line data sets, our algorithm systematically out-performs the lasso in both predictive performance and computation time, and shows equivalent predictive performance, but considerably smaller computation time, than the elastic-net.


Assuntos
Algoritmos , Farmacogenética/estatística & dados numéricos , Antineoplásicos/farmacologia , Inteligência Artificial , Teorema de Bayes , Linhagem Celular Tumoral , Biologia Computacional , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Análise de Regressão
17.
Pac Symp Biocomput ; : 63-74, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24297534

RESUMO

Large-scale pharmacogenomic screens of cancer cell lines have emerged as an attractive pre-clinical system for identifying tumor genetic subtypes with selective sensitivity to targeted therapeutic strategies. Application of modern machine learning approaches to pharmacogenomic datasets have demonstrated the ability to infer genomic predictors of compound sensitivity. Such modeling approaches entail many analytical design choices; however, a systematic study evaluating the relative performance attributable to each design choice is not yet available. In this work, we evaluated over 110,000 different models, based on a multifactorial experimental design testing systematic combinations of modeling factors within several categories of modeling choices, including: type of algorithm, type of molecular feature data, compound being predicted, method of summarizing compound sensitivity values, and whether predictions are based on discretized or continuous response values. Our results suggest that model input data (type of molecular features and choice of compound) are the primary factors explaining model performance, followed by choice of algorithm. Our results also provide a statistically principled set of recommended modeling guidelines, including: using elastic net or ridge regression with input features from all genomic profiling platforms, most importantly, gene expression features, to predict continuous-valued sensitivity scores summarized using the area under the dose response curve, with pathway targeted compounds most likely to yield the most accurate predictors. In addition, our study provides a publicly available resource of all modeling results, an open source code base, and experimental design for researchers throughout the community to build on our results and assess novel methodologies or applications in related predictive modeling problems.


Assuntos
Neoplasias/tratamento farmacológico , Neoplasias/genética , Farmacogenética/estatística & dados numéricos , Algoritmos , Inteligência Artificial , Linhagem Celular Tumoral , Biologia Computacional , Bases de Dados Genéticas/estatística & dados numéricos , Resistencia a Medicamentos Antineoplásicos/genética , Perfilação da Expressão Gênica/estatística & dados numéricos , Humanos , Modelos Genéticos , Análise de Regressão
18.
Pac Symp Biocomput ; : 172-82, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24297544

RESUMO

Computational drug repositioning leverages computational technology and high volume of biomedical data to identify new indications for existing drugs. Since it does not require costly experiments that have a high risk of failure, it has attracted increasing interest from diverse fields such as biomedical, pharmaceutical, and informatics areas. In this study, we used pharmacogenomics data generated from pharmacogenomics studies, applied informatics and Semantic Web technologies to address the drug repositioning problem. Specifically, we explored PharmGKB to identify pharmacogenomics related associations as pharmacogenomics profiles for US Food and Drug Administration (FDA) approved breast cancer drugs. We then converted and represented these profiles in Semantic Web notations, which support automated semantic inference. We successfully evaluated the performance and efficacy of the breast cancer drug pharmacogenomics profiles by case studies. Our results demonstrate that combination of pharmacogenomics data and Semantic Web technology/Cheminformatics approaches yields better performance of new indication and possible adverse effects prediction for breast cancer drugs.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Reposicionamento de Medicamentos/estatística & dados numéricos , Bases de Conhecimento , Farmacogenética/estatística & dados numéricos , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Antineoplásicos/farmacologia , Neoplasias da Mama/genética , Biologia Computacional , Feminino , Humanos , Internet , Linguagens de Programação
19.
BMC Med Genomics ; 6 Suppl 3: S4, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24565337

RESUMO

BACKGROUND: During the last few years, the knowledge of drug, disease phenotype and protein has been rapidly accumulated and more and more scientists have been drawn the attention to inferring drug-disease associations by computational method. Development of an integrated approach for systematic discovering drug-disease associations by those informational data is an important issue. METHODS: We combine three different networks of drug, genomic and disease phenotype and assign the weights to the edges from available experimental data and knowledge. Given a specific disease, we use our network propagation approach to infer the drug-disease associations. RESULTS: We apply prostate cancer and colorectal cancer as our test data. We use the manually curated drug-disease associations from comparative toxicogenomics database to be our benchmark. The ranked results show that our proposed method obtains higher specificity and sensitivity and clearly outperforms previous methods. Our result also show that our method with off-targets information gets higher performance than that with only primary drug targets in both test data. CONCLUSIONS: We clearly demonstrate the feasibility and benefits of using network-based analyses of chemical, genomic and phenotype data to reveal drug-disease associations. The potential associations inferred by our method provide new perspectives for toxicogenomics and drug reposition evaluation.


Assuntos
Biologia Computacional/métodos , Descoberta de Drogas/métodos , Predisposição Genética para Doença/genética , Farmacogenética/métodos , Antineoplásicos/uso terapêutico , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Descoberta de Drogas/estatística & dados numéricos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/genética , Redes Reguladoras de Genes/efeitos dos fármacos , Redes Reguladoras de Genes/genética , Estudos de Associação Genética/métodos , Estudos de Associação Genética/estatística & dados numéricos , Humanos , Masculino , Terapia de Alvo Molecular/métodos , Farmacogenética/estatística & dados numéricos , Fenótipo , Neoplasias da Próstata/diagnóstico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Curva ROC , Reprodutibilidade dos Testes , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética
20.
Pac Symp Biocomput ; : 376-87, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22174293

RESUMO

In this paper, we report on adapting the JREX relation extraction engine, originally developed For the elicitation of protein-protein interaction relations, to the domains of pharmacogenetics and pharmacogenomics. We propose an intrinsic and an extrinsic evaluation scenario which is based on knowledge contained in the PharmGKB knowledge base. Porting JREX yields favorable results in the range of 80% F-score for Gene-Disease, Gene-Drug, and Drug-Disease relations.


Assuntos
Bases de Conhecimento , Farmacogenética/estatística & dados numéricos , Hidrocarboneto de Aril Hidroxilases/genética , Hidrocarboneto de Aril Hidroxilases/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Citalopram/farmacocinética , Biologia Computacional , Citocromo P-450 CYP2C19 , Bases de Dados Factuais , Bases de Dados Genéticas , Docetaxel , Feminino , Genes BRCA2 , Predisposição Genética para Doença , Humanos , Obesidade/genética , Farmacogenética/normas , Farmacocinética , Mapeamento de Interação de Proteínas/estatística & dados numéricos , Taxoides/uso terapêutico , Urocortinas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA