Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 213
Filtrar
1.
Nature ; 629(8012): 624-629, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38632401

RESUMEN

The cost of drug discovery and development is driven primarily by failure1, with only about 10% of clinical programmes eventually receiving approval2-4. We previously estimated that human genetic evidence doubles the success rate from clinical development to approval5. In this study we leverage the growth in genetic evidence over the past decade to better understand the characteristics that distinguish clinical success and failure. We estimate the probability of success for drug mechanisms with genetic support is 2.6 times greater than those without. This relative success varies among therapy areas and development phases, and improves with increasing confidence in the causal gene, but is largely unaffected by genetic effect size, minor allele frequency or year of discovery. These results indicate we are far from reaching peak genetic insights to aid the discovery of targets for more effective drugs.


Asunto(s)
Ensayos Clínicos como Asunto , Aprobación de Drogas , Descubrimiento de Drogas , Resultado del Tratamiento , Humanos , Alelos , Ensayos Clínicos como Asunto/economía , Ensayos Clínicos como Asunto/estadística & datos numéricos , Aprobación de Drogas/economía , Descubrimiento de Drogas/economía , Descubrimiento de Drogas/métodos , Descubrimiento de Drogas/estadística & datos numéricos , Descubrimiento de Drogas/tendencias , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Terapia Molecular Dirigida , Probabilidad , Factores de Tiempo , Insuficiencia del Tratamiento
2.
Comput Math Methods Med ; 2021: 9949328, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34938362

RESUMEN

Developing new treatments for emerging infectious diseases in infectious and noninfectious diseases has attracted a particular attention. The emergence of viral diseases is expected to accelerate; these data indicate the need for a proactive approach to develop widely active family specific and cross family therapies for future disease outbreaks. Viral disease such as pneumonia, severe acute respiratory syndrome type 2, HIV infection, and Hepatitis-C virus can cause directly and indirectly cardiovascular disease (CVD). Emphasis should be placed not only on the development of broad-spectrum molecules and antibodies but also on host factor therapy, including the reutilization of previously approved or developing drugs. Another new class of therapeutics with great antiviral therapeutic potential is molecular communication networks using deep learning autoencoder (DL-AEs). The use of DL-AEs for diagnosis and prognosis prediction of infectious and noninfectious diseases has attracted a particular attention. MCN is map to molecular signaling and communication that are found inside and outside the human body where the goal is to develop a new black box mechanism that can serve the future robust healthcare industry (HCI). MCN has the ability to characterize the signaling process between cells and infectious disease locations at various levels of the human body called point-to-point MCN through DL-AE and provide targeted drug delivery (TDD) environment. Through MCN, and DL-AE healthcare provider can remotely measure biological signals and control certain processes in the required organism for the maintenance of the patient's health state. We use biomicrodevices to promote the real-time monitoring of human health and storage of the gathered data in the cloud. In this paper, we use the DL-based AE approach to design and implement a new drug source and target for the MCN under white Gaussian noise. Simulation results show that transceiver executions for a given medium model that reduces the bit error rate which can be learned. Then, next development of molecular diagnosis such as heart sounds is classified. Furthermore, biohealth interface for the inside and outside human body mechanism is presented, comparative perspective with up-to-date current situation about MCN.


Asunto(s)
Enfermedades Transmisibles Emergentes/tratamiento farmacológico , Aprendizaje Profundo , Virosis/tratamiento farmacológico , Antivirales/uso terapéutico , Enfermedades Transmisibles Emergentes/epidemiología , Biología Computacional , Simulación por Computador , Sistemas de Liberación de Medicamentos , Descubrimiento de Drogas/métodos , Descubrimiento de Drogas/estadística & datos numéricos , Epidemias , Humanos , Microtecnología , Redes Neurales de la Computación , Biología Sintética , Virosis/epidemiología
3.
Biomed Pharmacother ; 141: 111638, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34153846

RESUMEN

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.


Asunto(s)
Simulación por Computador , Descubrimiento de Drogas/métodos , Reposicionamiento de Medicamentos/métodos , Aprendizaje Automático , Preparaciones Farmacéuticas/administración & dosificación , Animales , Macrodatos , Simulación por Computador/estadística & datos numéricos , Sistemas de Liberación de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/estadística & datos numéricos , Descubrimiento de Drogas/estadística & datos numéricos , Reposicionamiento de Medicamentos/estadística & datos numéricos , Humanos , Aprendizaje Automático/estadística & datos numéricos
4.
Brief Bioinform ; 22(5)2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-33418563

RESUMEN

Matched molecular pairs analysis (MMPA) has become a powerful tool for automatically and systematically identifying medicinal chemistry transformations from compound/property datasets. However, accurate determination of matched molecular pair (MMP) transformations largely depend on the size and quality of existing experimental data. Lack of high-quality experimental data heavily hampers the extraction of more effective medicinal chemistry knowledge. Here, we developed a new strategy called quantitative structure-activity relationship (QSAR)-assisted-MMPA to expand the number of chemical transformations and took the logD7.4 property endpoint as an example to demonstrate the reliability of the new method. A reliable logD7.4 consensus prediction model was firstly established, and its applicability domain was strictly assessed. By applying the reliable logD7.4 prediction model to screen two chemical databases, we obtained more high-quality logD7.4 data by defining a strict applicability domain threshold. Then, MMPA was performed on the predicted data and experimental data to derive more chemical rules. To validate the reliability of the chemical rules, we compared the magnitude and directionality of the property changes of the predicted rules with those of the measured rules. Then, we compared the novel chemical rules generated by our proposed approach with the published chemical rules, and found that the magnitude and directionality of the property changes were consistent, indicating that the proposed QSAR-assisted-MMPA approach has the potential to enrich the collection of rule types or even identify completely novel rules. Finally, we found that the number of the MMP rules derived from the experimental data could be amplified by the predicted data, which is helpful for us to analyze the medicinal chemical rules in local chemical environment. In summary, the proposed QSAR-assisted-MMPA approach could be regarded as a very promising strategy to expand the chemical transformation space for lead optimization, especially when no enough experimental data can support MMPA.


Asunto(s)
Técnicas de Química Sintética/métodos , Química Farmacéutica/métodos , Descubrimiento de Drogas/métodos , Drogas en Investigación/síntesis química , Modelos Estadísticos , Biotransformación , Bases de Datos de Compuestos Químicos , Conjuntos de Datos como Asunto , Descubrimiento de Drogas/estadística & datos numéricos , Drogas en Investigación/metabolismo , Humanos , Estructura Molecular , Relación Estructura-Actividad Cuantitativa , Reproducibilidad de los Resultados
6.
Nucleic Acids Res ; 49(D1): D1160-D1169, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33151287

RESUMEN

DrugCentral is a public resource (http://drugcentral.org) that serves the scientific community by providing up-to-date drug information, as described in previous papers. The current release includes 109 newly approved (October 2018 through March 2020) active pharmaceutical ingredients in the US, Europe, Japan and other countries; and two molecular entities (e.g. mefuparib) of interest for COVID19. New additions include a set of pharmacokinetic properties for ∼1000 drugs, and a sex-based separation of side effects, processed from FAERS (FDA Adverse Event Reporting System); as well as a drug repositioning prioritization scheme based on the market availability and intellectual property rights forFDA approved drugs. In the context of the COVID19 pandemic, we also incorporated REDIAL-2020, a machine learning platform that estimates anti-SARS-CoV-2 activities, as well as the 'drugs in news' feature offers a brief enumeration of the most interesting drugs at the present moment. The full database dump and data files are available for download from the DrugCentral web portal.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Bases de Datos Farmacéuticas/estadística & datos numéricos , Aprobación de Drogas/estadística & datos numéricos , Descubrimiento de Drogas/estadística & datos numéricos , Reposicionamiento de Medicamentos/estadística & datos numéricos , SARS-CoV-2/efectos de los fármacos , Antivirales/efectos adversos , Antivirales/farmacocinética , COVID-19/epidemiología , COVID-19/virología , Aprobación de Drogas/métodos , Descubrimiento de Drogas/métodos , Reposicionamiento de Medicamentos/métodos , Epidemias , Europa (Continente) , Humanos , Almacenamiento y Recuperación de la Información/métodos , Internet , Japón , SARS-CoV-2/fisiología , Estados Unidos
7.
Nucleic Acids Res ; 49(D1): D1388-D1395, 2021 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-33151290

RESUMEN

PubChem (https://pubchem.ncbi.nlm.nih.gov) is a popular chemical information resource that serves the scientific community as well as the general public, with millions of unique users per month. In the past two years, PubChem made substantial improvements. Data from more than 100 new data sources were added to PubChem, including chemical-literature links from Thieme Chemistry, chemical and physical property links from SpringerMaterials, and patent links from the World Intellectual Properties Organization (WIPO). PubChem's homepage and individual record pages were updated to help users find desired information faster. This update involved a data model change for the data objects used by these pages as well as by programmatic users. Several new services were introduced, including the PubChem Periodic Table and Element pages, Pathway pages, and Knowledge panels. Additionally, in response to the coronavirus disease 2019 (COVID-19) outbreak, PubChem created a special data collection that contains PubChem data related to COVID-19 and the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).


Asunto(s)
COVID-19/prevención & control , Bases de Datos de Compuestos Químicos , Almacenamiento y Recuperación de la Información/estadística & datos numéricos , SARS-CoV-2/aislamiento & purificación , Interfaz Usuario-Computador , COVID-19/epidemiología , COVID-19/virología , Descubrimiento de Drogas/estadística & datos numéricos , Epidemias , Humanos , Almacenamiento y Recuperación de la Información/métodos , Internet , Salud Pública/estadística & datos numéricos , SARS-CoV-2/fisiología , Programas Informáticos
8.
Malar J ; 19(1): 421, 2020 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-33228666

RESUMEN

To maintain momentum towards improved malaria control and elimination, a vaccine would be a key addition to the intervention toolkit. Two approaches are recommended: (1) promote the development and short to medium term deployment of first generation vaccine candidates and (2) support innovation and discovery to identify and develop highly effective, long-lasting and affordable next generation malaria vaccines.


Asunto(s)
Investigación Biomédica , Descubrimiento de Drogas/estadística & datos numéricos , Vacunas contra la Malaria , Vacunas contra la Malaria/análisis , Vacunas contra la Malaria/química , Vacunas contra la Malaria/aislamiento & purificación , Vacunas contra la Malaria/farmacología
9.
Comput Math Methods Med ; 2020: 1862168, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32952598

RESUMEN

The Traditional Chinese Medicine (TCM) formula is the main treatment method of TCM. A formula often contains multiple herbs where core herbs play a critical therapeutic effect for treating diseases. It is of great significance to find out the core herbs in formulae for providing evidences and references for the clinical application of Chinese herbs and formulae. In this paper, we propose a core herb discovery model CHDSC based on semantic analysis and community detection to discover the core herbs for treating a certain disease from large-scale literature, which includes three stages: corpus construction, herb network establishment, and core herb discovery. In CHDSC, two artificial intelligence modules are used, where the Chinese word embedding algorithm ESSP2VEC is designed to analyse the semantics of herbs in Chinese literature based on the stroke, structure, and pinyin features of Chinese characters, and the label propagation-based algorithm LILPA is adopted to detect herb communities and core herbs in the herbal semantic network constructed from large-scale literature. To validate the proposed model, we choose chronic glomerulonephritis (CGN) as an example, search 1126 articles about how to treat CGN in TCM from the China National Knowledge Infrastructure (CNKI), and apply CHDSC to analyse the collected literature. Experimental results reveal that CHDSC discovers three major herb communities and eighteen core herbs for treating different CGN syndromes with high accuracy. The community size, degree, and closeness centrality distributions of the herb network are analysed to mine the laws of core herbs. As a result, we can observe that core herbs mainly exist in the communities with more than 25 herbs. The degree and closeness centrality of core herb nodes concentrate on the range of [15, 40] and [0.25, 0.45], respectively. Thus, semantic analysis and community detection are helpful for mining effective core herbs for treating a certain disease from large-scale literature.


Asunto(s)
Descubrimiento de Drogas/métodos , Medicamentos Herbarios Chinos/clasificación , Medicamentos Herbarios Chinos/uso terapéutico , Glomerulonefritis/tratamiento farmacológico , Fitoterapia , Algoritmos , Inteligencia Artificial , China , Enfermedad Crónica , Biología Computacional , Minería de Datos , Bases de Datos Farmacéuticas , Descubrimiento de Drogas/estadística & datos numéricos , Humanos , Conceptos Matemáticos , Medicina Tradicional China/métodos , Medicina Tradicional China/estadística & datos numéricos , Semántica
10.
Methods ; 179: 55-64, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32446957

RESUMEN

At the early stages of the drug discovery, molecule toxicity prediction is crucial to excluding drug candidates that are likely to fail in clinical trials. In this paper, we presented a novel molecular representation method and developed a corresponding deep learning-based framework called TOP (the abbreviation of TOxicity Prediction). TOP integrates specifically designed data preprocessing methods, an RNN based on bidirectional gated recurrent unit (BiGRU), and fully connected neural networks for end-to-end molecular representation learning and chemical toxicity prediction. TOP can automatically learn a mixed molecular representation from not only SMILES contextual information that describes the molecule structure, but also physiochemical properties. Therefore, TOP can overcome the drawbacks of existing methods that use either of them, thus greatly promotes toxicity prediction accuracy. We conducted extensive experiments over 14 classic toxicity prediction tasks on three different benchmark datasets, including balanced and imbalanced ones. The results show that, with the help of the novel molecular representation method, TOP significantly outperforms not only three baseline machine learning methods, but also five state-of-the-art methods.


Asunto(s)
Quimioinformática/métodos , Aprendizaje Profundo , Descubrimiento de Drogas/métodos , Farmacología Clínica/métodos , Pruebas de Toxicidad/métodos , Conjuntos de Datos como Asunto , Descubrimiento de Drogas/estadística & datos numéricos , Predicción/métodos , Humanos , Farmacología Clínica/estadística & datos numéricos , Pruebas de Toxicidad/estadística & datos numéricos
11.
SLAS Discov ; 25(9): 1009-1017, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32468893

RESUMEN

During drug discovery, compounds/biologics are screened against biological targets of interest to find drug candidates with the most desirable activity profile. The compounds are tested at multiple concentrations to understand the dose-response relationship, often summarized as AC50 values and used directly in ranking compounds. Differences between compound repeats are inevitable because of experimental noise and/or systematic error; however, it is often desired to detect the latter when it occurs. To address this, the ß-expectation tolerance interval is proposed in this article. Besides the classical acceptance criteria on assay performance, based on control compounds (e.g., quality control samples), this metric permits us to compare new estimates against historical estimates of the same study compound. It provides a measure that detects whether observed differences are likely due to systematic error. The challenge here is that limited information is available to build such compound-specific acceptance limits. To this end, we propose the use of Bayesian ß-expectation tolerance intervals to validate agreement between replicate potency estimates for individual study compounds. This approach allows the variability of the compound-testing process to be estimated from reference compounds within the assay and used as prior knowledge in the computation of compound-specific intervals as from the first repeat of the compound and then continuously updated as more information is acquired with subsequent repeats. A repeat is then flagged when it is not within limits. Unlike a fixed threshold such as 0.5log, which is often used in practice, this approach identifies unexpected deviations on each compound repeat given the observed variability of the assay.


Asunto(s)
Teorema de Bayes , Biofarmacia , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas/estadística & datos numéricos , Sesgo , Humanos , Estándares de Referencia
12.
PLoS One ; 15(5): e0232989, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32407402

RESUMEN

Multi drug treatments are increasingly used in the clinic to combat complex and co-occurring diseases. However, most drug combination discovery efforts today are mainly focused on anticancer therapy and rarely examine the potential of using more than two drugs simultaneously. Moreover, there is currently no reported methodology for performing second- and higher-order drug combination analysis of secretomic patterns, meaning protein concentration profiles released by the cells. Here, we introduce COMBSecretomics (https://github.com/EffieChantzi/COMBSecretomics.git), the first pragmatic methodological framework designed to search exhaustively for second- and higher-order mixtures of candidate treatments that can modify, or even reverse malfunctioning secretomic patterns of human cells. This framework comes with two novel model-free combination analysis methods; a tailor-made generalization of the highest single agent principle and a data mining approach based on top-down hierarchical clustering. Quality control procedures to eliminate outliers and non-parametric statistics to quantify uncertainty in the results obtained are also included. COMBSecretomics is based on a standardized reproducible format and could be employed with any experimental platform that provides the required protein release data. Its practical use and functionality are demonstrated by means of a proof-of-principle pharmacological study related to cartilage degradation. COMBSecretomics is the first methodological framework reported to enable secretome-related second- and higher-order drug combination analysis. It could be used in drug discovery and development projects, clinical practice, as well as basic biological understanding of the largely unexplored changes in cell-cell communication that occurs due to disease and/or associated pharmacological treatment conditions.


Asunto(s)
Combinación de Medicamentos , Descubrimiento de Drogas/métodos , Metabolómica/métodos , Cartílago/efectos de los fármacos , Cartílago/metabolismo , Simulación por Computador , Descubrimiento de Drogas/estadística & datos numéricos , Evaluación Preclínica de Medicamentos/métodos , Evaluación Preclínica de Medicamentos/estadística & datos numéricos , Humanos , Técnicas In Vitro , Metabolómica/estadística & datos numéricos , Modelos Biológicos , Osteoartritis/tratamiento farmacológico , Osteoartritis/metabolismo , Proteómica/métodos , Proteómica/estadística & datos numéricos , Programas Informáticos
13.
J Comput Aided Mol Des ; 34(7): 731-746, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32297073

RESUMEN

In drug development, late stage toxicity issues of a compound are the main cause of failure in clinical trials. In silico methods are therefore of high importance to guide the early design process to reduce time, costs and animal testing. Technical advances and the ever growing amount of available toxicity data enabled machine learning, especially neural networks, to impact the field of predictive toxicology. In this study, cytotoxicity prediction, one of the earliest handles in drug discovery, is investigated using a deep learning approach trained on a highly consistent in-house data set of over 34,000 compounds with a share of less than 5% of cytotoxic molecules. The model reached a balanced accuracy of over 70%, similar to previously reported studies using Random Forest. Albeit yielding good results, neural networks are often described as a black box lacking deeper mechanistic understanding of the underlying model. To overcome this absence of interpretability, a Deep Taylor Decomposition method is investigated to identify substructures that may be responsible for the cytotoxic effects, the so-called toxicophores. Furthermore, this study introduces cytotoxicity maps which provide a visual structural interpretation of the relevance of these substructures. Using this approach could be helpful in drug development to predict the potential toxicity of a compound as well as to generate new insights into the toxic mechanism. Moreover, it could also help to de-risk and optimize compounds.


Asunto(s)
Citotoxinas/química , Citotoxinas/toxicidad , Aprendizaje Profundo , Descubrimiento de Drogas/métodos , Supervivencia Celular/efectos de los fármacos , Diseño Asistido por Computadora , Diseño de Fármacos , Descubrimiento de Drogas/estadística & datos numéricos , Células HEK293 , Células Hep G2 , Humanos , Modelos Biológicos , Redes Neurales de la Computación , Bibliotecas de Moléculas Pequeñas , Programas Informáticos , Toxicología/estadística & datos numéricos
14.
Parasitology ; 147(6): 611-633, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32046803

RESUMEN

During three decades, only about 20 new drugs have been developed for malaria, tuberculosis and all neglected tropical diseases (NTDs). This critical situation was reached because NTDs represent only 10% of health research investments; however, they comprise about 90% of the global disease burden. Computational simulations applied in virtual screening (VS) strategies are very efficient tools to identify pharmacologically active compounds or new indications for drugs already administered for other diseases. One of the advantages of this approach is the low time-consuming and low-budget first stage, which filters for testing experimentally a group of candidate compounds with high chances of binding to the target and present trypanocidal activity. In this work, we review the most common VS strategies that have been used for the identification of new drugs with special emphasis on those applied to trypanosomiasis and leishmaniasis. Computational simulations based on the selected protein targets or their ligands are explained, including the method selection criteria, examples of successful VS campaigns applied to NTDs, a list of validated molecular targets for drug development and repositioned drugs for trypanosomatid-caused diseases. Thereby, here we present the state-of-the-art of VS and drug repurposing to conclude pointing out the future perspectives in the field.


Asunto(s)
Biología Computacional/estadística & datos numéricos , Descubrimiento de Drogas/estadística & datos numéricos , Leishmaniasis/tratamiento farmacológico , Tripanocidas/farmacología , Tripanosomiasis/tratamiento farmacológico , Animales , Simulación por Computador , Humanos , Ratones
15.
Molecules ; 25(3)2020 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-32050446

RESUMEN

During 2019, the US Food and Drug Administration (FDA) approved 48 new drugs (38 New Chemical Entities and 10 Biologics). Although this figure is slightly lower than that registered in 2018 (59 divided between 42 New Chemical Entities and 17 Biologics), a year that broke a record with respect to new drugs approved by this agency, it builds on the trend initiated in 2017, when 46 drugs were approved. Of note, three antibody drug conjugates, three peptides, and two oligonucleotides were approved in 2019. This report analyzes the 48 new drugs of the class of 2019 from a strictly chemical perspective. The classification, which was carried out on the basis of chemical structure, includes the following: Biologics (antibody drug conjugates, antibodies, and proteins); TIDES (peptide and oligonucleotides); drug combinations; natural products; and small molecules.


Asunto(s)
Aprobación de Drogas/estadística & datos numéricos , Descubrimiento de Drogas/estadística & datos numéricos , Industria Farmacéutica/tendencias , United States Food and Drug Administration/estadística & datos numéricos , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/uso terapéutico , Productos Biológicos/química , Productos Biológicos/uso terapéutico , Aprobación de Drogas/historia , Aprobación de Drogas/legislación & jurisprudencia , Combinación de Medicamentos , Descubrimiento de Drogas/historia , Industria Farmacéutica/historia , Drogas en Investigación/química , Drogas en Investigación/uso terapéutico , Historia del Siglo XXI , Humanos , Inmunoconjugados/química , Inmunoconjugados/uso terapéutico , Estructura Molecular , Oligonucleótidos/química , Oligonucleótidos/uso terapéutico , Péptidos/química , Péptidos/uso terapéutico , Relación Estructura-Actividad , Estados Unidos , United States Food and Drug Administration/historia , United States Food and Drug Administration/legislación & jurisprudencia
16.
CPT Pharmacometrics Syst Pharmacol ; 9(3): 143-152, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31920008

RESUMEN

Differences in the effect of gefitinib and chemotherapy on tumor burden in non-small cell lung cancer remain to be fully understood. Using a Bayesian hierarchical model of tumor size dynamics, we estimated the rates of tumor growth and treatment resistance for patients in the Iressa Pan-Asia Study study (NCT00322452). The following relationships characterize greater efficacy of gefitinib in epidermal growth factor receptor (EGFR) positive tumors: Maximum drug effect is, in decreasing order, gefitinib in EGFR-positive, chemotherapy in EGFR-positive, chemotherapy in EGFR-negative, and gefitinib in EGFR-negative tumors; the rate of resistance emergence is, in increasing order: gefitinib in EGFR positive, chemotherapy in EGFR positive, while each is plausibly similar to the rate in EGFR negative tumors, which are estimated with less certainty. The rate of growth is smaller in EGFR-positive than in EGFR-negative fully resistant tumors, regardless of treatment. The model can be used to compare treatment effects and resistance dynamics among different drugs.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Receptores ErbB/efectos de los fármacos , Gefitinib/farmacología , Neoplasias Pulmonares/patología , Algoritmos , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Asia/epidemiología , Teorema de Bayes , Carboplatino/farmacología , Carboplatino/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Supervivencia sin Enfermedad , Descubrimiento de Drogas/estadística & datos numéricos , Resistencia a Medicamentos/fisiología , Receptores ErbB/metabolismo , Gefitinib/uso terapéutico , Humanos , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Carga Tumoral/efectos de los fármacos
17.
J Comput Aided Mol Des ; 34(7): 717-730, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31960253

RESUMEN

Machine learning methods may have the potential to significantly accelerate drug discovery. However, the increasing rate of new methodological approaches being published in the literature raises the fundamental question of how models should be benchmarked and validated. We reanalyze the data generated by a recently published large-scale comparison of machine learning models for bioactivity prediction and arrive at a somewhat different conclusion. We show that the performance of support vector machines is competitive with that of deep learning methods. Additionally, using a series of numerical experiments, we question the relevance of area under the receiver operating characteristic curve as a metric in virtual screening. We further suggest that area under the precision-recall curve should be used in conjunction with the receiver operating characteristic curve. Our numerical experiments also highlight challenges in estimating the uncertainty in model performance via scaffold-split nested cross validation.


Asunto(s)
Aprendizaje Profundo , Descubrimiento de Drogas/métodos , Aprendizaje Automático , Algoritmos , Área Bajo la Curva , Benchmarking , Simulación por Computador , Descubrimiento de Drogas/normas , Descubrimiento de Drogas/estadística & datos numéricos , Evaluación Preclínica de Medicamentos , Humanos , Curva ROC , Máquina de Vectores de Soporte , Interfaz Usuario-Computador
18.
J Biopharm Stat ; 30(1): 104-120, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31462134

RESUMEN

Identification of genomic biomarkers is an important area of research in the context of drug discovery experiments. These experiments typically consist of several high dimensional datasets that contain information about a set of drugs (compounds) under development. This type of data structure introduces the challenge of multi-source data integration. High-Performance Computing (HPC) has become an important tool for everyday research tasks. In the context of drug discovery, high dimensional multi-source data needs to be analyzed to identify the biological pathways related to the new set of drugs under development. In order to process all information contained in the datasets, HPC techniques are required. Even though R packages for parallel computing are available, they are not optimized for a specific setting and data structure. In this article, we propose a new framework, for data analysis, to use R in a computer cluster. The proposed data analysis workflow is applied to a multi-source high dimensional drug discovery dataset and compared with a few existing R packages for parallel computing.


Asunto(s)
Descubrimiento de Drogas/estadística & datos numéricos , Marcadores Genéticos , Genómica/estadística & datos numéricos , Proyectos de Investigación/estadística & datos numéricos , Macrodatos , Interpretación Estadística de Datos , Bases de Datos Genéticas , Humanos , Flujo de Trabajo
19.
J Comput Aided Mol Des ; 34(7): 805-815, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31407224

RESUMEN

Generative topographic mapping was used to investigate the possibility to diversify the in-house compounds collection of Boehringer Ingelheim (BI). For this purpose, a 2D map covering the relevant chemical space was trained, and the BI compound library was compared to the Aldrich-Market Select (AMS) database of more than 8M purchasable compounds. In order to discover new (sub)structures, the "AutoZoom" tool was developed and applied in order to analyze chemotypes of molecules residing in heavily populated zones of a map and to extract the corresponding maximum common substructures. A set of 401K new structures from the AMS database was retrieved and checked for drug-likeness and biological activity.


Asunto(s)
Descubrimiento de Drogas/métodos , Bibliotecas de Moléculas Pequeñas , Algoritmos , Diseño Asistido por Computadora/estadística & datos numéricos , Bases de Datos de Compuestos Químicos/estadística & datos numéricos , Bases de Datos Farmacéuticas/estadística & datos numéricos , Diseño de Fármacos , Desarrollo de Medicamentos/estadística & datos numéricos , Descubrimiento de Drogas/estadística & datos numéricos , Humanos , Estructura Molecular , Programas Informáticos , Interfaz Usuario-Computador
20.
J Comput Aided Mol Des ; 34(7): 769-782, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31677002

RESUMEN

We present a Focused Library Generator that is able to create from scratch new molecules with desired properties. After training the Generator on the ChEMBL database, transfer learning was used to switch the generator to producing new Mdmx inhibitors that are a promising class of anticancer drugs. Lilly medicinal chemistry filters, molecular docking, and a QSAR IC50 model were used to refine the output of the Generator. Pharmacophore screening and molecular dynamics (MD) simulations were then used to further select putative ligands. Finally, we identified five promising hits with equivalent or even better predicted binding free energies and IC50 values than known Mdmx inhibitors. The source code of the project is available on https://github.com/bigchem/online-chem.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Diseño de Fármacos , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas , Antineoplásicos/química , Antineoplásicos/farmacología , Sitios de Unión , Proteínas de Ciclo Celular/química , Diseño Asistido por Computadora/estadística & datos numéricos , Bases de Datos de Compuestos Químicos/estadística & datos numéricos , Bases de Datos Farmacéuticas , Descubrimiento de Drogas/métodos , Descubrimiento de Drogas/estadística & datos numéricos , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Redes Neurales de la Computación , Unión Proteica , Proteínas Proto-Oncogénicas/química , Relación Estructura-Actividad Cuantitativa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...