Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Comput Math Methods Med ; 2022: 6783659, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35140805

RESUMO

Rheumatoid arthritis (RA) is an autoimmune and inflammatory disease for which there is a lack of therapeutic options. Genome-wide association studies (GWASs) have identified over 100 genetic loci associated with RA susceptibility; however, the most causal risk genes (RGs) associated with, and molecular mechanism underlying, RA remain unknown. In this study, we collected 95 RA-associated loci from multiple GWASs and detected 87 candidate high-confidence risk genes (HRGs) from these loci via integrated multiomics data (the genome-scale chromosome conformation capture data, enhancer-promoter linkage data, and gene expression data) using the Bayesian integrative risk gene selector (iRIGS). Analysis of these HRGs indicates that these genes were indeed, markedly associated with different aspects of RA. Among these, 36 and 46 HRGs have been reported to be related to RA and autoimmunity, respectively. Meanwhile, most novel HRGs were also involved in the significantly enriched RA-related biological functions and pathways. Furthermore, drug repositioning prediction of the HRGs revealed three potential targets (ERBB2, IL6ST, and MAPK1) and nine possible drugs for RA treatment, of which two IL-6 receptor antagonists (tocilizumab and sarilumab) have been approved for RA treatment and four drugs (trastuzumab, lapatinib, masoprocol, and arsenic trioxide) have been reported to have a high potential to ameliorate RA. In summary, we believe that this study provides new clues for understanding the pathogenesis of RA and is important for research regarding the mechanisms underlying RA and the development of therapeutics for this condition.


Assuntos
Artrite Reumatoide/genética , Antirreumáticos/farmacologia , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/imunologia , Autoimunidade/genética , Teorema de Bayes , Biologia Computacional , Desenvolvimento de Medicamentos/estatística & dados numéricos , Reposicionamento de Medicamentos/estatística & dados numéricos , Redes Reguladoras de Genes , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla/estatística & dados numéricos , Humanos , Fatores de Risco
2.
Biomed Pharmacother ; 141: 111638, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34153846

RESUMO

Repositioning or "repurposing" of existing therapies for indications of alternative disease is an attractive approach that can generate lower costs and require a shorter approval time than developing a de novo drug. The development of experimental drugs is time-consuming, expensive, and limited to a fairly small number of targets. The incorporation of separate and complementary data should be used, as each type of data set exposes a specific feature of organism knowledge Drug repurposing opportunities are often focused on sporadic findings or on time-consuming pre-clinical drug tests which are often not guided by hypothesis. In comparison, repurposing in-silico drugs is a new, hypothesis-driven method that takes advantage of big-data use. Nonetheless, the widespread use of omics technology, enhanced data storage, data sense, machine learning algorithms, and computational modeling all give unparalleled knowledge of the methods of action of biological processes and drugs, providing wide availability, for both disease-related data and drug-related data. This review has taken an in-depth look at the current state, possibilities, and limitations of further progress in the field of drug repositioning.


Assuntos
Simulação por Computador , Descoberta de Drogas/métodos , Reposicionamento de Medicamentos/métodos , Aprendizado de Máquina , Preparações Farmacêuticas/administração & dosagem , Animais , Big Data , Simulação por Computador/estatística & dados numéricos , Sistemas de Liberação de Medicamentos/métodos , Sistemas de Liberação de Medicamentos/estatística & dados numéricos , Descoberta de Drogas/estatística & dados numéricos , Reposicionamento de Medicamentos/estatística & dados numéricos , Humanos , Aprendizado de Máquina/estatística & dados numéricos
3.
J Immunother Cancer ; 9(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34117116

RESUMO

SARS-CoV-2 is the virus responsible for the COVID-19 pandemic. COVID-19 has highly variable disease severity and a bimodal course characterized by acute respiratory viral infection followed by hyperinflammation in a subset of patients with severe disease. This immune dysregulation is characterized by lymphocytopenia, elevated levels of plasma cytokines and proliferative and exhausted T cells, among other dysfunctional cell types. Immunocompromised persons often fare worse in the context of acute respiratory infections, but preliminary data suggest this may not hold true for COVID-19. In this review, we explore the effect of SARS-CoV-2 infection on mortality in four populations with distinct forms of immunocompromise: (1) persons with hematological malignancies (HM) and hematopoietic stem cell transplant (HCT) recipients; (2) solid organ transplant recipients (SOTRs); (3) persons with rheumatological diseases; and (4) persons living with HIV (PLWH). For each population, key immunological defects are described and how these relate to the immune dysregulation in COVID-19. Next, outcomes including mortality after SARS-CoV-2 infection are described for each population, giving comparisons to the general population of age-matched and comorbidity-matched controls. In these four populations, iatrogenic or disease-related immunosuppression is not clearly associated with poor prognosis in HM, HCT, SOTR, rheumatological diseases, or HIV. However, certain individual immunosuppressants or disease states may be associated with harmful or beneficial effects, including harm from severe CD4 lymphocytopenia in PLWH and possible benefit to the calcineurin inhibitor ciclosporin in SOTRs, or tumor necrosis factor-α inhibitors in persons with rheumatic diseases. Lastly, insights gained from clinical and translational studies are explored as to the relevance for repurposing of immunosuppressive host-directed therapies for the treatment of hyperinflammation in COVID-19 in the general population.


Assuntos
COVID-19 , Reposicionamento de Medicamentos , Hospedeiro Imunocomprometido , Imunossupressores/uso terapêutico , Imunoterapia , COVID-19/epidemiologia , COVID-19/imunologia , COVID-19/terapia , Comorbidade , Reposicionamento de Medicamentos/métodos , Reposicionamento de Medicamentos/estatística & dados numéricos , Infecções por HIV/epidemiologia , Infecções por HIV/imunologia , Neoplasias Hematológicas/epidemiologia , Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas/estatística & dados numéricos , Humanos , Hospedeiro Imunocomprometido/fisiologia , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Imunoterapia/estatística & dados numéricos , Mortalidade , Pandemias , Prognóstico , Doenças Reumáticas/epidemiologia , SARS-CoV-2/fisiologia , Transplantados/estatística & dados numéricos
4.
Biochem Pharmacol ; 178: 114057, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32470547

RESUMO

COVID-19 is an ongoing viral pandemic disease that is caused by SARS-CoV2, inducing severe pneumonia in humans. However, several classes of repurposed drugs have been recommended, no specific vaccines or effective therapeutic interventions for COVID-19 are developed till now. Viral dependence on ACE-2, as entry receptors, drove the researchers into RAS impact on COVID-19 pathogenesis. Several evidences have pointed at Neprilysin (NEP) as one of pulmonary RAS components. Considering the protective effect of NEP against pulmonary inflammatory reactions and fibrosis, it is suggested to direct the future efforts towards its potential role in COVID-19 pathophysiology. Thus, the review aimed to shed light on the potential beneficial effects of NEP pathways as a novel target for COVID-19 therapy by summarizing its possible molecular mechanisms. Additional experimental and clinical studies explaining more the relationships between NEP and COVID-19 will greatly benefit in designing the future treatment approaches.


Assuntos
Antivirais/uso terapêutico , Betacoronavirus/efeitos dos fármacos , Infecções por Coronavirus/prevenção & controle , Reposicionamento de Medicamentos/métodos , Neprilisina/antagonistas & inibidores , Pandemias/prevenção & controle , Pneumonia Viral/prevenção & controle , Transdução de Sinais/efeitos dos fármacos , Angiotensina I/farmacologia , Angiotensina I/uso terapêutico , Antagonistas de Receptores de Angiotensina/farmacologia , Antagonistas de Receptores de Angiotensina/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Antivirais/farmacologia , Betacoronavirus/fisiologia , COVID-19 , Infecções por Coronavirus/fisiopatologia , Infecções por Coronavirus/virologia , Reposicionamento de Medicamentos/estatística & dados numéricos , Reposicionamento de Medicamentos/tendências , Humanos , Neprilisina/metabolismo , Fragmentos de Peptídeos/farmacologia , Fragmentos de Peptídeos/uso terapêutico , Pneumonia Viral/fisiopatologia , Pneumonia Viral/virologia , SARS-CoV-2
5.
Ann Rheum Dis ; 78(8): 1127-1134, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31092410

RESUMO

OBJECTIVES: There is a need to identify effective treatments for rheumatic diseases, and while genetic studies have been successful it is unclear which genes contribute to the disease. Using our existing Capture Hi-C data on three rheumatic diseases, we can identify potential causal genes which are targets for existing drugs and could be repositioned for use in rheumatic diseases. METHODS: High confidence candidate causal genes were identified using Capture Hi-C data from B cells and T cells. These genes were used to interrogate drug target information from DrugBank to identify existing treatments, which could be repositioned to treat these diseases. The approach was refined using Ingenuity Pathway Analysis to identify enriched pathways and therefore further treatments relevant to the disease. RESULTS: Overall, 454 high confidence genes were identified. Of these, 48 were drug targets (108 drugs) and 11 were existing therapies used in the treatment of rheumatic diseases. After pathway analysis refinement, 50 genes remained, 13 of which were drug targets (33 drugs). However considering targets across all enriched pathways, a further 367 drugs were identified for potential repositioning. CONCLUSION: Capture Hi-C has the potential to identify therapies which could be repositioned to treat rheumatic diseases. This was particularly successful for rheumatoid arthritis, where six effective, biologic treatments were identified. This approach may therefore yield new ways to treat patients, enhancing their quality of life and reducing the economic impact on healthcare providers. As additional cell types and other epigenomic data sets are generated, this prospect will improve further.


Assuntos
Antirreumáticos/uso terapêutico , Cromatina/genética , Reposicionamento de Medicamentos/estatística & dados numéricos , Terapia de Alvo Molecular/métodos , Receptores de Estrogênio/efeitos dos fármacos , Doenças Reumáticas/genética , Cromatina/efeitos dos fármacos , Estudos de Coortes , Reposicionamento de Medicamentos/métodos , Feminino , Estudos de Associação Genética , Estudo de Associação Genômica Ampla , Humanos , Masculino , Receptores de Estrogênio/genética , Doenças Reumáticas/tratamento farmacológico , Sensibilidade e Especificidade
6.
Acta Psychiatr Scand ; 139(1): 68-77, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30182363

RESUMO

OBJECTIVE: To investigate whether continued use of non-aspirin NSAID, low-dose aspirin, high-dose aspirin, statins, allopurinol and angiotensin agents decreases the rate of incident depression using Danish nationwide population-based registers. METHODS: All persons in Denmark who purchased the exposure medications of interest between 1995 and 2015 and a random sample of 30% of the Danish population was included in the study. Two different outcome measures were included, (i) a diagnosis of depressive disorder at a psychiatric hospital as in-patient or out-patient and (ii) a combined measure of a diagnosis of depression or use of antidepressants. RESULTS: A total of 1 576 253 subjects were exposed to one of the six drugs of interest during the exposure period from 2005 to 2015. Continued use of low-dose aspirin, statins, allopurinol and angiotensin agents was associated with a decreased rate of incident depression according to both outcome measures. Continued uses of non-aspirin NSAIDs as well as high-dose aspirin were associated with an increased rate of incident depression. CONCLUSION: The findings support the potential of agents acting on inflammation and the stress response system in depression as well as the potential of population-based registers to systematically identify drugs with repurposing potential.


Assuntos
Depressão/tratamento farmacológico , Transtorno Depressivo/tratamento farmacológico , Reposicionamento de Medicamentos/métodos , Estresse Fisiológico/efeitos dos fármacos , Adulto , Idoso , Alopurinol/efeitos adversos , Alopurinol/uso terapêutico , Angiotensinas/efeitos adversos , Angiotensinas/uso terapêutico , Anti-Inflamatórios não Esteroides/efeitos adversos , Anti-Inflamatórios não Esteroides/uso terapêutico , Antidepressivos/uso terapêutico , Aspirina/efeitos adversos , Aspirina/uso terapêutico , Dinamarca/epidemiologia , Depressão/diagnóstico , Depressão/epidemiologia , Transtorno Depressivo/diagnóstico , Transtorno Depressivo/epidemiologia , Reposicionamento de Medicamentos/estatística & dados numéricos , Feminino , Supressores da Gota/efeitos adversos , Supressores da Gota/uso terapêutico , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Incidência , Inflamação/tratamento farmacológico , Masculino , Pessoa de Meia-Idade , Avaliação de Resultados em Cuidados de Saúde , Sistema de Registros
7.
Int J Parasitol Drugs Drug Resist ; 8(3): 440-450, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30396011

RESUMO

The metacestode stage of the fox tapeworm Echinococcus multilocularis causes the lethal disease alveolar echinococcosis. Current chemotherapeutic treatment options are based on benzimidazoles (albendazole and mebendazole), which are insufficient and hence alternative drugs are needed. In this study, we screened the 400 compounds of the Medicines for Malaria Venture (MMV) Pathogen Box against E. multilocularis metacestodes. For the screen, we employed the phosphoglucose isomerase (PGI) assay which assesses drug-induced damage on metacestodes, and identified ten new compounds with activity against the parasite. The anti-theilerial drug MMV689480 (buparvaquone) and MMV671636 (ELQ-400) were the most promising compounds, with an IC50 of 2.87 µM and 0.02 µM respectively against in vitro cultured E. multilocularis metacestodes. Both drugs suggested a therapeutic window based on their cytotoxicity against mammalian cells. Transmission electron microscopy revealed that treatment with buparvaquone impaired parasite mitochondria early on and additional tests showed that buparvaquone had a reduced activity under anaerobic conditions. Furthermore, we established a system to assess mitochondrial respiration in isolated E. multilocularis cells in real time using the Seahorse XFp Analyzer and demonstrated inhibition of the cytochrome bc1 complex by buparvaquone. Mice with secondary alveolar echinococcosis were treated with buparvaquone (100 mg/kg per dose, three doses per week, four weeks of treatment), but the drug failed to reduce the parasite burden in vivo. Future studies will reveal whether improved formulations of buparvaquone could increase its effectivity.


Assuntos
Antiprotozoários/farmacologia , Reposicionamento de Medicamentos/métodos , Echinococcus multilocularis/efeitos dos fármacos , Naftoquinonas/farmacologia , Naftoquinonas/uso terapêutico , Albendazol/farmacologia , Albendazol/uso terapêutico , Animais , Anti-Helmínticos/farmacologia , Antiprotozoários/química , Antiprotozoários/isolamento & purificação , Antiprotozoários/uso terapêutico , Avaliação Pré-Clínica de Medicamentos/métodos , Reposicionamento de Medicamentos/estatística & dados numéricos , Equinococose/tratamento farmacológico , Echinococcus multilocularis/patogenicidade , Complexo III da Cadeia de Transporte de Elétrons/efeitos dos fármacos , Complexo III da Cadeia de Transporte de Elétrons/farmacologia , Glucose-6-Fosfato Isomerase/metabolismo , Concentração Inibidora 50 , Estágios do Ciclo de Vida/efeitos dos fármacos , Camundongos , Microscopia Eletrônica de Transmissão , Naftoquinonas/química , Carga Parasitária , Éteres Fenílicos/farmacologia , Quinolonas/farmacologia
8.
J Biomed Inform ; 85: 114-125, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30092360

RESUMO

Molecular Property Diagnostic Suite - Diabetes Mellitus (MPDSDM) is a Galaxy-based, open source disease-specific web portal for diabetes. It consists of three modules namely (i) data library (ii) data processing and (iii) data analysis tools. The data library (target library and literature) module provide extensive and curated information about the genes involved in type 1 and type 2 diabetes onset and progression stage (available at http://www.mpds-diabetes.in). The database also contains information on drug targets, biomarkers, therapeutics and associated genes specific to type 1, and type 2 diabetes. A unique MPDS identification number has been assigned for each gene involved in diabetes mellitus and the corresponding card contains chromosomal data, gene information, protein UniProt ID, functional domains, druggability and related pathway information. One of the objectives of the web portal is to have an open source data repository that contains all information on diabetes and use this information for developing therapeutics to cure diabetes. We also make an attempt for computational drug repurposing for the validated diabetes targets. We performed virtual screening of 1455 FDA approved drugs on selected 20 type 1 and type 2 diabetes proteins using docking protocol and their biological activity was predicted using "PASS Online" server (http://www.way2drug.com/passonline) towards anti-diabetic activity, resulted in the identification of 41 drug molecules. Five drug molecules (which are earlier known for anti-malarial/microbial, anti-viral, anti-cancer, anti-pulmonary activities) were proposed to have a better repurposing potential for type 2 anti-diabetic activity and good binding affinity towards type 2 diabetes target proteins.


Assuntos
Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/genética , Descoberta de Drogas , Reposicionamento de Medicamentos , Biologia Computacional , Diabetes Mellitus/diagnóstico , Descoberta de Drogas/estatística & dados numéricos , Avaliação Pré-Clínica de Medicamentos , Reposicionamento de Medicamentos/estatística & dados numéricos , Humanos , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Internet , Técnicas de Diagnóstico Molecular/estatística & dados numéricos , Simulação de Acoplamento Molecular , Interface Usuário-Computador
9.
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.
J Mol Biol ; 430(15): 2266-2273, 2018 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-29237557

RESUMO

About 7000 rare, or orphan, diseases affect more than 350 million people worldwide. Although these conditions collectively pose significant health care problems, drug companies seldom develop drugs for orphan diseases due to extremely limited individual markets. Consequently, developing new treatments for often life-threatening orphan diseases is primarily contingent on financial incentives from governments, special research grants, and private philanthropy. Computer-aided drug repositioning is a cheaper and faster alternative to traditional drug discovery offering a promising venue for orphan drug research. Here, we present eRepo-ORP, a comprehensive resource constructed by a large-scale repositioning of existing drugs to orphan diseases with a collection of structural bioinformatics tools, including eThread, eFindSite, and eMatchSite. Specifically, a systematic exploration of 320,856 possible links between known drugs in DrugBank and orphan proteins obtained from Orphanet reveals as many as 18,145 candidates for repurposing. In order to illustrate how potential therapeutics for rare diseases can be identified with eRepo-ORP, we discuss the repositioning of a kinase inhibitor for Ras-associated autoimmune leukoproliferative disease. The eRepo-ORP data set is available through the Open Science Framework at https://osf.io/qdjup/.


Assuntos
Biologia Computacional/métodos , Descoberta de Drogas/métodos , Reposicionamento de Medicamentos/métodos , Doenças Raras/tratamento farmacológico , Síndrome Linfoproliferativa Autoimune/tratamento farmacológico , Síndrome Linfoproliferativa Autoimune/metabolismo , Descoberta de Drogas/economia , Descoberta de Drogas/estatística & dados numéricos , Reposicionamento de Medicamentos/economia , Reposicionamento de Medicamentos/estatística & dados numéricos , Humanos , Internet , Inibidores de Proteínas Quinases/uso terapêutico , Reprodutibilidade dos Testes , Proteínas ras/antagonistas & inibidores , Proteínas ras/metabolismo
11.
Braz. J. Pharm. Sci. (Online) ; 53(2): e16087, 2017. tab, graf
Artigo em Inglês | LILACS | ID: biblio-839493

RESUMO

ABSTRACT The discovery of arteannuin (qinghaosu) in the 20th Century was a major advance for medicine. Besides functioning as a malaria therapy, arteannuin is a pharmacological agent in a range of other diseases, but its mechanism of action remains obscure. In this study, the reverse docking server PharmMapper was used to identify potential targets of arteannuin. The results were checked using the chemical-protein interactome servers DRAR-CPI and DDI-CPI, and verified by AutoDock Vina. The results showed that neprilysin (also known as CD10), a common acute lymphoblastic leukaemia antigen, was the top disease-related target of arteannuin. The chemical-protein interactome and docking results agreed with those of PharmMapper, further implicating neprilysin as a potential target. Although experimental verification is required, this study provides guidance for future pharmacological investigations into novel clinical applications for arteannuin.


Assuntos
Simulação por Computador/classificação , Neprilisina/farmacologia , Artemisininas/análise , Reposicionamento de Medicamentos/estatística & dados numéricos
12.
PLoS One ; 11(1): e0144797, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26735301

RESUMO

BACKGROUNDS: Based on in vitro data and results of a recent drug repositioning study, some medications approved by the FDA for the treatment of various non-malignant disorders were demonstrated to have anti-SCLC activity in preclinical models. The aim of our study is to confirm whether use of these medications is associated with survival benefit. METHODS: Consecutive patients with pathologically confirmed, stage 4 SCLC were analyzed in this retrospective study. Patients that were prescribed statins, aspirin, clomipramine (tricyclic antidepressant; TCA), selective serotonin reuptake inhibitors (SSRIs), doxazosin or prazosin (α1-adrenergic receptor antagonists; ADRA1) were identified. RESULTS: There were a total of 876 patients. Aspirin, statins, SSRIs, ADRA1, and TCA were administered in 138, 72, 20, 28, and 5 cases, respectively. A statistically significant increase in median OS was observed only in statin-treated patients when compared to those not receiving any of the aforementioned medications (OS, 8.4 vs. 6.1 months, respectively; p = 0.002). The administration of SSRIs, aspirin, and ADRA1 did not result in a statistically significant OS benefit (median OS, 8.5, 6.8, and 6.0 months, respectively). The multivariate Cox model showed that, besides age and ECOG PS, radiotherapy was an independent survival predictor (Hazard Ratio, 2.151; 95% confidence interval, 1.828-2.525; p <0.001). CONCLUSIONS: Results of drug repositioning studies using only preclinical data or small numbers of patients should be treated with caution before application in the clinic. Our data demonstrated that radiotherapy appears to be an independent survival predictor in stage 4 SCLC, therefore confirming the results of other prospective and retrospective studies.


Assuntos
Reposicionamento de Medicamentos/estatística & dados numéricos , Neoplasias Pulmonares/terapia , Carcinoma de Pequenas Células do Pulmão/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Feminino , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/química , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Estimativa de Kaplan-Meier , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Modelos de Riscos Proporcionais , Estudos Retrospectivos , Carcinoma de Pequenas Células do Pulmão/mortalidade , Carcinoma de Pequenas Células do Pulmão/patologia
13.
Brief Bioinform ; 17(1): 2-12, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25832646

RESUMO

Computational drug repositioning or repurposing is a promising and efficient tool for discovering new uses from existing drugs and holds the great potential for precision medicine in the age of big data. The explosive growth of large-scale genomic and phenotypic data, as well as data of small molecular compounds with granted regulatory approval, is enabling new developments for computational repositioning. To achieve the shortest path toward new drug indications, advanced data processing and analysis strategies are critical for making sense of these heterogeneous molecular measurements. In this review, we show recent advancements in the critical areas of computational drug repositioning from multiple aspects. First, we summarize available data sources and the corresponding computational repositioning strategies. Second, we characterize the commonly used computational techniques. Third, we discuss validation strategies for repositioning studies, including both computational and experimental methods. Finally, we highlight potential opportunities and use-cases, including a few target areas such as cancers. We conclude with a brief discussion of the remaining challenges in computational drug repositioning.


Assuntos
Reposicionamento de Medicamentos/tendências , Biologia Computacional/tendências , Mineração de Dados , Combinação de Medicamentos , Reposicionamento de Medicamentos/estatística & dados numéricos , Genômica , Humanos , Aprendizado de Máquina , Estrutura Molecular , Fenótipo , Inquéritos e Questionários
14.
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
15.
Clin Ther ; 35(6): 808-18, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23726388

RESUMO

BACKGROUND: Much of the literature on trends and factors affecting biopharmaceutical innovation has focused overwhelmingly on the development and approval of never-before approved drugs and biologics. Little attention has been paid to new uses for already-approved compounds, which can be an important form of innovation. OBJECTIVE: This paper aimed to determine and analyze recent trends in the number and type of new or modified US indication approvals for drugs and biologics. We also examine regulatory approval-phase times for new-use efficacy supplements and compare them to approval-phase times for original-use approvals over the same period. METHODS: We developed a data set of efficacy supplements approved by the US Food and Drug Administration (FDA) from 1998 to 2011 that includes information on the type, approval-phase time (time from submission to the FDA of an application for marketing approval to approval of the application), and FDA therapeutic-significance rating for the approved application, which we obtained from an FDA Web site. This data set was merged with a Tufts Center for the Study of Drug Development (CSDD) data set of US new drug and biologics approvals. We developed descriptive statistics on trends in the number and type of new-use efficacy supplements, on US regulatory approval-phase times for the supplements, and on original new drug and biologics approvals over the study period and for the time from original- to new-use approval. RESULTS: The total number of new-use efficacy-supplement approvals did not exhibit a marked trend, but the number of new pediatric-indication approvals increased substantially. Approval-phase times for new-use supplements varied by therapeutic class and FDA therapeutic-significance rating. Mean approval-phase times were highest for central nervous system compounds (13.8 months) and lowest for antineoplastics (8.9 months). The mean time from original to supplement approval was substantially longer for new pediatric indications than for other new uses. Mean approval-phase time during the study period for applications that received a standard review rating from the FDA was substantially shorter for supplements compared to original uses, but the differences for applications that received a priority review rating from the FDA were negligible. CONCLUSIONS: Development of and regulatory approval for new uses of already-approved drugs and biologics is an important source of innovation by biopharmaceutical firms. Despite rising development costs, the output of new-use approvals has remained stable in recent years, driven largely by the pursuit of new pediatric indications. FDA approval-phase times have generally declined substantially for all types of applications since the mid-1990s following legislation that provided a new source of income for the agency. However, while the resources needed to review supplemental applications are likely lower in general than for original-use approvals, the approval-phase times for important new uses are no lower than for important original-use applications.


Assuntos
Bases de Dados de Produtos Farmacêuticos/estatística & dados numéricos , Aprovação de Drogas/estatística & dados numéricos , Reposicionamento de Medicamentos/tendências , United States Food and Drug Administration , Fatores Biológicos/uso terapêutico , Bases de Dados de Produtos Farmacêuticos/economia , Bases de Dados de Produtos Farmacêuticos/tendências , Aprovação de Drogas/economia , Reposicionamento de Medicamentos/economia , Reposicionamento de Medicamentos/estatística & dados numéricos , Humanos , Marketing , Fatores de Tempo , Estados Unidos
16.
J Chem Inf Model ; 53(4): 753-62, 2013 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-23527559

RESUMO

Prediction of polypharmacological profiles of drugs enables us to investigate drug side effects and further find their new indications, i.e. drug repositioning, which could reduce the costs while increase the productivity of drug discovery. Here we describe a new computational framework to predict polypharmacological profiles of drugs by the integration of chemical, side effect, and therapeutic space. On the basis of our previous developed drug side effects database, named MetaADEDB, a drug side effect similarity inference (DSESI) method was developed for drug-target interaction (DTI) prediction on a known DTI network connecting 621 approved drugs and 893 target proteins. The area under the receiver operating characteristic curve was 0.882 ± 0.011 averaged from 100 simulated tests of 10-fold cross-validation for the DSESI method, which is comparative with drug structural similarity inference and drug therapeutic similarity inference methods. Seven new predicted candidate target proteins for seven approved drugs were confirmed by published experiments, with the successful hit rate more than 15.9%. Moreover, network visualization of drug-target interactions and off-target side effect associations provide new mechanism-of-action of three approved antipsychotic drugs in a case study. The results indicated that the proposed methods could be helpful for prediction of polypharmacological profiles of drugs.


Assuntos
Antipsicóticos/química , Reposicionamento de Medicamentos/estatística & dados numéricos , Modelos Estatísticos , Medicamentos sob Prescrição/química , Algoritmos , Antipsicóticos/efeitos adversos , Área Sob a Curva , Simulação por Computador , Mineração de Dados , Bases de Dados de Produtos Farmacêuticos , Humanos , Ligantes , Valor Preditivo dos Testes , Medicamentos sob Prescrição/efeitos adversos , Transtornos Psicóticos/tratamento farmacológico , Relação Quantitativa Estrutura-Atividade , Curva ROC
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA