Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Divers ; 26(5): 2523-2534, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34802116

RESUMEN

Hypertension is a medical condition that affects millions of people worldwide. Despite the high efficacy of the current antihypertensive drugs, they are associated with serious side effects. Peptides constitute attractive options for chemical therapy against hypertension, and computational models can accelerate the design of antihypertensive peptides. Yet, to the best of our knowledge, all the in silico models predict only the antihypertensive activity of peptides while neglecting their inherent toxic potential to red blood cells. In this work, we report the first sequence-based model that combines perturbation theory and machine learning through multilayer perceptron networks (SB-PTML-MLP) to enable the simultaneous screening of antihypertensive activity and hemotoxicity of peptides. We have interpreted the molecular descriptors present in the model from a physicochemical and structural point of view. By strictly following such interpretations as guidelines, we performed two tasks. First, we selected amino acids with favorable contributions to both the increase of the antihypertensive activity and the diminution of hemotoxicity. Then, we assembled those suitable amino acids, virtually designing peptides that were predicted by the SB-PTML-MLP model as antihypertensive agents exhibiting low hemotoxicity. The potentiality of the SB-PTML-MLP model as a tool for designing potent and safe antihypertensive peptides was confirmed by predictions performed by online computational tools reported in the scientific literature. The methodology presented here can be extended to other pharmacological applications of peptides.


Asunto(s)
Antihipertensivos , Hipertensión , Aminoácidos , Antihipertensivos/química , Antihipertensivos/farmacología , Humanos , Hipertensión/tratamiento farmacológico , Aprendizaje Automático , Péptidos/química , Péptidos/farmacología
2.
J Chem Inf Model ; 56(3): 588-98, 2016 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-26960000

RESUMEN

Antimicrobial peptides (AMPs) have emerged as promising therapeutic alternatives to fight against the diverse infections caused by different pathogenic microorganisms. In this context, theoretical approaches in bioinformatics have paved the way toward the creation of several in silico models capable of predicting antimicrobial activities of peptides. All current models have several significant handicaps, which prevent the efficient search for highly active AMPs. Here, we introduce the first multitarget (mt) chemo-bioinformatic model devoted to performing alignment-free prediction of antibacterial activity of peptides against multiple Gram-positive bacterial strains. The model was constructed from a data set containing 2488 cases of AMPs sequences assayed against at least 1 out of 50 Gram-positive bacterial strains. This mt-chemo-bioinformatic model displayed percentages of correct classification higher than 90.00% in both training and prediction (test) sets. For the first time, two computational approaches derived from basic concepts in genetics and molecular biology were applied, allowing the calculations of the relative contributions of any amino acid (in a defined position) to the antibacterial activity of an AMP and depending on the bacterial strain used in the biological assay. The present mt-chemo-bioinformatic model constitutes a powerful tool to enable the discovery of potent and versatile AMPs.


Asunto(s)
Antibacterianos/farmacología , Biología Computacional , Bacterias Grampositivas/efectos de los fármacos , Péptidos/farmacología , Pruebas de Sensibilidad Microbiana
3.
Environ Sci Technol ; 48(24): 14686-94, 2014 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-25384130

RESUMEN

Nanomaterials have revolutionized modern science and technology due to their multiple applications in engineering, physics, chemistry, and biomedicine. Nevertheless, the use and manipulation of nanoparticles (NPs) can bring serious damages to living organisms and their ecosystems. For this reason, ecotoxicity and cytotoxicity assays are of special interest in order to determine the potential harmful effects of NPs. Processes based on ecotoxicity and cytotoxicity tests can significantly consume time and financial resources. In this sense, alternative approaches such as quantitative structure-activity/toxicity relationships (QSAR/QSTR) modeling have provided important insights for the better understanding of the biological behavior of NPs that may be responsible for causing toxicity. Until now, QSAR/QSTR models have predicted ecotoxicity or cytotoxicity separately against only one organism (bioindicator species or cell line) and have not reported information regarding the quantitative influence of characteristics other than composition or size. In this work, we developed a unified QSTR-perturbation model to simultaneously probe ecotoxicity and cytotoxicity of NPs under different experimental conditions, including diverse measures of toxicities, multiple biological targets, compositions, sizes and conditions to measure those sizes, shapes, times during which the biological targets were exposed to NPs, and coating agents. The model was created from 36488 cases (NP-NP pairs) and exhibited accuracies higher than 98% in both training and prediction sets. The model was used to predict toxicities of several NPs that were not included in the original data set. The results of the predictions suggest that the present QSTR-perturbation model can be employed as a highly promising tool for the fast and efficient assessment of ecotoxicity and cytotoxicity of NPs.


Asunto(s)
Nanoestructuras/química , Nanoestructuras/toxicidad , Relación Estructura-Actividad Cuantitativa , Medición de Riesgo/métodos , Animales , Ecotoxicología/métodos , Nanopartículas/química , Nanopartículas/toxicidad
4.
Curr Top Med Chem ; 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39171594

RESUMEN

BACKGROUND: Cancers are complex multi-genetic diseases that should be tackled in multi-target drug discovery scenarios. Computational methods are of great importance to accelerate the discovery of multi-target anticancer agents. Here, we employed a ligand-based approach by combining a perturbation-theory machine learning model derived from an ensemble of multilayer perceptron networks (PTML-EL-MLP) with the Fragment-Based Topological Design (FBTD) approach to rationally design and predict triple-target inhibitors against the cancerrelated proteins named Tropomyosin Receptor Kinase A (TRKA), poly[ADP-ribose] polymerase 1 (PARP-1), and Insulin-like Growth Factor 1 Receptor (IGF1R). METHODS: We extracted the chemical and biological data from ChEMBL. We applied the Box- Jenkins approach to generate multi-label topological indices and subsequently created the PTML-EL-MLP model. RESULTS: Our PTML-EL-MLP model exhibited an accuracy of around 80%. The application FBTD permitted the physicochemical and structural interpretation of the PTML-EL-MLP model, thus enabling a) the chemistry-driven analysis of different molecular fragments with a positive influence on the multi-target activity and b) the use of those favorable fragments as building blocks to virtually design four new drug-like molecules. The designed molecules were predicted as triple-target inhibitors against the aforementioned cancer-related proteins. CONCLUSION: Our study envisages the capabilities of combining PTML modeling with FBTD for the generation of new chemical diversity for multi-target drug discovery in oncology research and beyond.

5.
Bioorg Med Chem ; 21(10): 2727-32, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23582445

RESUMEN

Streptococci are a group of Gram-positive bacteria which are responsible for causing many diverse diseases in humans and other animals worldwide. The high prevalence of resistance of these bacteria to current antibacterial drugs is an alarming problem for the scientific community. The battle against streptococci by using antimicrobial chemotherapies will depend on the design of new chemicals with high inhibitory activity, having also as low toxicity as possible. Multi-target approaches based on quantitative-structure activity relationships (mt-QSAR) have played a very important role, providing a better knowledge about the molecular patterns related with the appearance of different pharmacological profiles including antimicrobial activity. Until now, almost all mt-QSAR models have considered the study of biological activity or toxicity separately. In the present study, we develop by the first time, a unified multitasking (mtk) QSAR model for the simultaneous prediction of anti-streptococci activity and toxic effects against biological models like Mus musculus and Rattus norvegicus. The mtk-QSAR model was created by using artificial neural networks (ANN) analysis for the classification of compounds as positive (high biological activity and/or low toxicity) or negative (otherwise) under diverse sets of experimental conditions. Our mtk-QSAR model, correctly classified more than 97% of the cases in the whole database (more than 11,500 cases), serving as a promising tool for the virtual screening of potent and safe anti-streptococci drugs.


Asunto(s)
Antibacterianos/química , Antibacterianos/farmacología , Animales , Animales de Laboratorio , Diseño de Fármacos , Descubrimiento de Drogas/métodos , Humanos , Informática/métodos , Modelos Biológicos , Relación Estructura-Actividad Cuantitativa , Streptococcus/química , Streptococcus/efectos de los fármacos , Relación Estructura-Actividad
6.
Expert Opin Drug Discov ; 18(11): 1231-1243, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37639708

RESUMEN

INTRODUCTION: Drug discovery has provided modern societies with the means to fight against many diseases. In this sense, computational methods have been at the forefront, playing an important role in rationalizing the search for novel drugs. Yet, tackling phenomena such as the multi-genic nature of diseases and drug resistance are limitations of the current computational methods. Multi-tasking models for quantitative structure-biological effect relationships (mtk-QSBER) have emerged to overcome such limitations. AREAS COVERED: The present review describes an update on the fundamentals and applications of the mtk-QSBER models as tools to accelerate multiple stages/substages of the drug discovery process. EXPERT OPINION: Computational approaches are extremely important for the rationalization of the search for novel and efficacious therapeutic agents. However, they need to focus more on the multi-target drug discovery paradigm. In this sense, mtk-QSBER models are particularly suited for multi-target drug discovery, offering encouraging opportunities across multiple therapeutic areas and scientific disciplines associated with drug discovery.


Asunto(s)
Descubrimiento de Drogas , Relación Estructura-Actividad Cuantitativa , Humanos , Descubrimiento de Drogas/métodos , Sistemas de Liberación de Medicamentos , Diseño de Fármacos
7.
Bioorg Med Chem ; 20(15): 4848-55, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22750007

RESUMEN

The discovery of new and more potent anti-cancer agents constitutes one of the most active fields of research in chemotherapy. Colorectal cancer (CRC) is one of the most studied cancers because of its high prevalence and number of deaths. In the current pharmaceutical design of more efficient anti-CRC drugs, the use of methodologies based on Chemoinformatics has played a decisive role, including Quantitative-Structure-Activity Relationship (QSAR) techniques. However, until now, there is no methodology able to predict anti-CRC activity of compounds against more than one CRC cell line, which should constitute the principal goal. In an attempt to overcome this problem we develop here the first multi-target (mt) approach for the virtual screening and rational in silico discovery of anti-CRC agents against ten cell lines. Here, two mt-QSAR classification models were constructed using a large and heterogeneous database of compounds. The first model was based on linear discriminant analysis (mt-QSAR-LDA) employing fragment-based descriptors while the second model was obtained using artificial neural networks (mt-QSAR-ANN) with global 2D descriptors. Both models correctly classified more than 90% of active and inactive compounds in training and prediction sets. Some fragments were extracted from the molecules and their contributions to anti-CRC activity were calculated using mt-QSAR-LDA model. Several fragments were identified as potential substructural features responsible for the anti-CRC activity and new molecules designed from those fragments with positive contributions were suggested and correctly predicted by the two models as possible potent and versatile anti-CRC agents.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Diseño de Fármacos , Relación Estructura-Actividad Cuantitativa , Protocolos de Quimioterapia Combinada Antineoplásica/síntesis química , Protocolos de Quimioterapia Combinada Antineoplásica/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Relación Estructura-Actividad
8.
Mol Divers ; 16(1): 183-91, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22020812

RESUMEN

Rational design of entry inhibitors is an active area for the discovery of new and effective anti-HIV agents. C-C Chemokine receptors represent key targets for the HIV entry process. Several of these proteins with features to be HIV co-receptors have not been sufficiently studied or used for the design of novel entry inhibitors. With the purpose to overcome this problem, we develop here a fragment-based approach for the design of multi-target inhibitors against four C-C chemokine receptors. This approach was focused on the construction of a multi-target QSAR discriminant model using a large and heterogeneous database of compounds and substructural descriptors for the classification and prediction of inhibitors for C-C chemokine receptors. The model correctly classified more than 89% of active and inactive compounds in both: training and prediction series. As principal advantage, this model permitted the automatic and fast extraction of fragments responsible for the inhibitory activity against the different C-C chemokine receptors under study and new molecular entities were suggested as possible versatile inhibitors for these proteins.


Asunto(s)
Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacología , Quimiocinas CC/antagonistas & inhibidores , Biología Computacional/métodos , Diseño de Fármacos , Quimiocinas CC/metabolismo , Humanos , Modelos Moleculares , Relación Estructura-Actividad Cuantitativa , Curva ROC
9.
Ecotoxicol Environ Saf ; 80: 308-13, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22521812

RESUMEN

Agriculture is needed to deal with crop losses caused by biotic stresses like pests. The use of pesticides has played a vital role, contributing to improve crop production and harvest productivity, providing a better crop quality and supply, and consequently contributing with the improvement of the human health. An important group of these pesticides is fungicides. However, the use of these agrochemical fungicides is an important source of contamination, damaging the ecosystems. Several studies have been realized for the assessment of the toxicity in agrochemical fungicides, but the principal limitation is the use of structurally related compounds against usually one indicator species. In order to overcome this problem, we explore the quantitative structure-toxicity relationships (QSTR) in agrochemical fungicides. Here, we developed the first multi-species (ms) chemoinformatic approach for the prediction multiple ecotoxicological profiles of fungicides against 20 indicators species and their classifications in toxic or nontoxic. The ms-QSTR discriminant model was based on substructural descriptors and a heterogeneous database of compounds. The percentages of correct classification were higher than 90% for both, training and prediction series. Also, substructural alerts responsible for the toxicity/no toxicity in fungicides respect all ecotoxicological profiles, were extracted and analyzed.


Asunto(s)
Organismos Acuáticos/efectos de los fármacos , Ecotoxicología/métodos , Fungicidas Industriales/toxicidad , Animales , Ecosistema , Fungicidas Industriales/química , Fungicidas Industriales/clasificación , Relación Estructura-Actividad Cuantitativa , Pruebas de Toxicidad , Contaminantes Químicos del Agua/química , Contaminantes Químicos del Agua/clasificación , Contaminantes Químicos del Agua/toxicidad
10.
Biomedicines ; 10(2)2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35203699

RESUMEN

Pancreatic cancer (PANC) is a dangerous type of cancer that is a major cause of mortality worldwide and exhibits a remarkably poor prognosis. To date, discovering anti-PANC agents remains a very complex and expensive process. Computational approaches can accelerate the search for anti-PANC agents. We report for the first time two models that combined perturbation theory with machine learning via a multilayer perceptron network (PTML-MLP) to perform the virtual design and prediction of molecules that can simultaneously inhibit multiple PANC cell lines and PANC-related proteins, such as caspase-1, tumor necrosis factor-alpha (TNF-alpha), and the insulin-like growth factor 1 receptor (IGF1R). Both PTML-MLP models exhibited accuracies higher than 78%. Using the interpretation from one of the PTML-MLP models as a guideline, we extracted different molecular fragments desirable for the inhibition of the PANC cell lines and the aforementioned PANC-related proteins and then assembled some of those fragments to form three new molecules. The two PTML-MLP models predicted the designed molecules as potentially versatile anti-PANC agents through inhibition of the three PANC-related proteins and multiple PANC cell lines. Conclusions: This work opens new horizons for the application of the PTML modeling methodology to anticancer research.

11.
ACS Omega ; 7(36): 32119-32130, 2022 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-36120024

RESUMEN

Respiratory viruses are infectious agents, which can cause pandemics. Although nowadays the danger associated with respiratory viruses continues to be evidenced by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) as the virus responsible for the current COVID-19 pandemic, other viruses such as SARS-CoV-1, the influenza A and B viruses (IAV and IBV, respectively), and the respiratory syncytial virus (RSV) can lead to globally spread viral diseases. Also, from a biological point of view, most of these viruses can cause an organ-damaging hyperinflammatory response known as the cytokine storm (CS). Computational approaches constitute an essential component of modern drug development campaigns, and therefore, they have the potential to accelerate the discovery of chemicals able to simultaneously inhibit multiple molecular and nonmolecular targets. We report here the first multicondition model based on quantitative structure-activity relationships and an artificial neural network (mtc-QSAR-ANN) for the virtual design and prediction of molecules with dual pan-antiviral and anti-CS profiles. Our mtc-QSAR-ANN model exhibited an accuracy higher than 80%. By interpreting the different descriptors present in the mtc-QSAR-ANN model, we could retrieve several molecular fragments whose assembly led to new molecules with drug-like properties and predicted pan-antiviral and anti-CS activities.

12.
Bioorg Med Chem ; 19(21): 6239-44, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21967806

RESUMEN

Prostate cancer (PCa) is the second-leading cause of cancer deaths among men in the around the world. Understanding the biology of PCa is essential to the development of novel therapeutic strategies, in order to prevent this disease. However, after PCa make metastases, chemotherapy plays an extremely important role. With the pass of the time, PCa cell lines become resistant to the current anti-PCa drugs. For this reason, there is a necessity to develop new anti-PCa agents with the ability to be active against several PCa cell lines. The present work is an effort to overcome this problem. We introduce here the first multi-target approach for the design and prediction of anti-PCa agents against several cell lines. Here, a fragment-based QSAR model was developed. The model had a sensitivity of 88.36% and specificity 89.81% in training series. Also, the model showed 94.06% and 92.92% for sensitivity and specificity, respectively. Some fragments were extracted from the molecules and their contributions to anti-PCa activity were calculated. Several fragments were identified as potential substructural features responsible of anti-PCa activity and new molecular entities designed from fragments with positive contributions were suggested as possible anti-PCa agents.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Descubrimiento de Drogas/métodos , Terapia Molecular Dirigida/métodos , Neoplasias de la Próstata/tratamiento farmacológico , Línea Celular Tumoral , Análisis Discriminante , Humanos , Concentración 50 Inhibidora , Masculino , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Relación Estructura-Actividad Cuantitativa , Curva ROC , Sensibilidad y Especificidad
13.
Mol Divers ; 15(4): 901-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21633788

RESUMEN

The increasing resistance of several phytopathogenic fungal species to the existing agrochemical fungicides has alarmed to the worldwide scientific community. There is no available methodology to predict in an efficient way if a new fungicide will have resistance risk due to fungal species which cause considerable crop losses. In an attempt to overcome this problem, a multi-resistance risk QSAR model, based on substructural descriptors was developed from a heterogeneous database of compounds. The purpose of this model is the classification, design, and prediction of agrochemical fungicides according to resistance risk categories. The QSAR model classified correctly 85.11% of the fungicides and the 85.07% of the inactive compounds in the training series, for an accuracy of 85.08%. In the prediction series, the percentages of correct classification were 85.71 and 86.55% for fungicides and inactive compounds, respectively, with an accuracy of 86.39%. Some fragments were extracted and their quantitative contributions to the fungicidal activity were calculated taking into consideration the different resistance risk categories for agrochemical fungicides. In the same way, some fragments present in molecules with fungicidal activity and with negative contributions were analyzed like structural alerts responsible of resistance risk.


Asunto(s)
Diseño de Fármacos , Farmacorresistencia Fúngica/efectos de los fármacos , Fungicidas Industriales/química , Fungicidas Industriales/farmacología , Relación Estructura-Actividad Cuantitativa , Análisis Discriminante , Plantas/microbiología , Curva ROC , Riesgo
14.
Antibiotics (Basel) ; 10(8)2021 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-34439055

RESUMEN

Tuberculosis remains the most afflicting infectious disease known by humankind, with one quarter of the population estimated to have it in the latent state. Discovering antituberculosis drugs is a challenging, complex, expensive, and time-consuming task. To overcome the substantial costs and accelerate drug discovery and development, drug repurposing has emerged as an attractive alternative to find new applications for "old" drugs and where computational approaches play an essential role by filtering the chemical space. This work reports the first multi-condition model based on quantitative structure-activity relationships and an ensemble of neural networks (mtc-QSAR-EL) for the virtual screening of potential antituberculosis agents able to act as multi-strain inhibitors. The mtc-QSAR-EL model exhibited an accuracy higher than 85%. A physicochemical and fragment-based structural interpretation of this model was provided, and a large dataset of agency-regulated chemicals was virtually screened, with the mtc-QSAR-EL model identifying already proven antituberculosis drugs while proposing chemicals with great potential to be experimentally repurposed as antituberculosis (multi-strain inhibitors) agents. Some of the most promising molecules identified by the mtc-QSAR-EL model as antituberculosis agents were also confirmed by another computational approach, supporting the capabilities of the mtc-QSAR-EL model as an efficient tool for computational drug repurposing.

15.
Biomolecules ; 11(12)2021 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-34944476

RESUMEN

Inflammation involves a complex biological response of the body tissues to damaging stimuli. When dysregulated, inflammation led by biomolecular mediators such as caspase-1 and tumor necrosis factor-alpha (TNF-alpha) can play a detrimental role in the progression of different medical conditions such as cancer, neurological disorders, autoimmune diseases, and cytokine storms caused by viral infections such as COVID-19. Computational approaches can accelerate the search for dual-target drugs able to simultaneously inhibit the aforementioned proteins, enabling the discovery of wide-spectrum anti-inflammatory agents. This work reports the first multicondition model based on quantitative structure-activity relationships and a multilayer perceptron neural network (mtc-QSAR-MLP) for the virtual screening of agency-regulated chemicals as versatile anti-inflammatory therapeutics. The mtc-QSAR-MLP model displayed accuracy higher than 88%, and was interpreted from a physicochemical and structural point of view. When using the mtc-QSAR-MLP model as a virtual screening tool, we could identify several agency-regulated chemicals as dual inhibitors of caspase-1 and TNF-alpha, and the experimental information later retrieved from the scientific literature converged with our computational results. This study supports the capabilities of our mtc-QSAR-MLP model in anti-inflammatory therapy with direct applications to current health issues such as the COVID-19 pandemic.


Asunto(s)
Antiinflamatorios/farmacología , Inhibidores de Caspasas/farmacología , Reposicionamiento de Medicamentos/métodos , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Antiinflamatorios/química , Caspasa 1/metabolismo , Inhibidores de Caspasas/química , Humanos , Inflamación/tratamiento farmacológico , Simulación del Acoplamiento Molecular , Relación Estructura-Actividad Cuantitativa , Factor de Necrosis Tumoral alfa/metabolismo , Tratamiento Farmacológico de COVID-19
16.
Curr Top Med Chem ; 21(30): 2687-2693, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34636311

RESUMEN

Respiratory viruses continue to afflict mankind. Among them, pathogens such as coronaviruses [including the current pandemic agent known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)] and the one causing influenza A (IAV) are highly contagious and deadly. These can evade the immune system defenses while causing a hyperinflammatory response that can damage different tissues/organs. Simultaneously targeting several immunomodulatory proteins is a plausible antiviral strategy since it could lead to the discovery of indirect-acting pan-antiviral (IAPA) agents for the treatment of diseases caused by respiratory viruses. In this context, computational approaches, which are an essential part of the modern drug discovery campaigns, could accelerate the identification of multi-target immunomodulators. This perspective discusses the usefulness of computational multi-target drug discovery for the virtual screening (drug repurposing) of IAPA agents capable of boosting the immune system through the activation of the toll-like receptor 7 (TLR7) and/or the stimulator of interferon genes (STING) while inhibiting key inflammation-related proteins such as caspase-1 and tumor necrosis factor-alpha (TNF-α).


Asunto(s)
Antivirales , Descubrimiento de Drogas , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Antivirales/farmacología , COVID-19 , Biología Computacional , Evaluación Preclínica de Medicamentos , Humanos , Pandemias , Infecciones del Sistema Respiratorio/virología , SARS-CoV-2/efectos de los fármacos
17.
Curr Top Med Chem ; 21(7): 661-675, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33463472

RESUMEN

BACKGROUND: Cyclin-dependent kinase 4 (CDK4) and the human epidermal growth factor receptor 2 (HER2) are two of the most promising targets in oncology research. Thus, a series of computational approaches have been applied to the search for more potent inhibitors of these cancerrelated proteins. However, current approaches have focused on chemical analogs while predicting the inhibitory activity against only one of these targets, but never against both. AIMS: We report the first perturbation model combined with machine learning (PTML) to enable the design and prediction of dual inhibitors of CDK4 and HER2. METHODS: Inhibition data for CDK4 and HER2 were extracted from ChEMBL. The PTML model relied on artificial neural networks to allow the classification/prediction of molecules as active or inactive against CDK4 and/or HER2. RESULTS: The PTML model displayed sensitivity and specificity higher than 80% in the training set. The same statistical metrics had values above 75% in the test set. We extracted several molecular fragments and estimated their quantitative contributions to the inhibitory activity against CDK4 and HER2. Guided by the physicochemical and structural interpretations of the molecular descriptors in the PTML model, we designed six molecules by assembling several fragments with positive contributions. Three of these molecules were predicted as potent dual inhibitors of CDK4 and HER2, while the other three were predicted as inhibitors of at least one of these proteins. All the molecules complied with Lipinski's rule of five and its variants. CONCLUSION: The present work represents an encouraging alternative for future anticancer chemotherapies.


Asunto(s)
Descubrimiento de Drogas/métodos , Inhibidores Enzimáticos/química , Lenguajes de Programación , Antineoplásicos/química , Antineoplásicos/farmacología , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Humanos , Estructura Molecular , Redes Neurales de la Computación , Receptor ErbB-2/antagonistas & inhibidores
18.
Front Chem ; 9: 634663, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33777898

RESUMEN

Parasitic diseases remain as unresolved health issues worldwide. While for some parasites the treatments involve drug combinations with serious side effects, for others, chemical therapies are inefficient due to the emergence of drug resistance. This urges the search for novel antiparasitic agents able to act through multiple mechanisms of action. Here, we report the first multi-target model based on quantitative structure-activity relationships and a multilayer perceptron neural network (mt-QSAR-MLP) to virtually design and predict versatile inhibitors of proteins involved in the survival and/or infectivity of different pathogenic parasites. The mt-QSAR-MLP model exhibited high accuracy (>80%) in both training and test sets for the classification/prediction of protein inhibitors. Several fragments were directly extracted from the physicochemical and structural interpretations of the molecular descriptors in the mt-QSAR-MLP model. Such interpretations enabled the generation of four molecules that were predicted as multi-target inhibitors against at least three of the five parasitic proteins reported here with two of the molecules being predicted to inhibit all the proteins. Docking calculations converged with the mt-QSAR-MLP model regarding the multi-target profile of the designed molecules. The designed molecules exhibited drug-like properties, complying with Lipinski's rule of five, as well as Ghose's filter and Veber's guidelines.

19.
Mini Rev Med Chem ; 20(14): 1357-1374, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32013845

RESUMEN

Fragment-Based Drug Design (FBDD) has established itself as a promising approach in modern drug discovery, accelerating and improving lead optimization, while playing a crucial role in diminishing the high attrition rates at all stages in the drug development process. On the other hand, FBDD has benefited from the application of computational methodologies, where the models derived from the Quantitative Structure-Activity Relationships (QSAR) have become consolidated tools. This mini-review focuses on the evolution and main applications of the QSAR paradigm in the context of FBDD in the last five years. This report places particular emphasis on the QSAR models derived from fragment-based topological approaches to extract physicochemical and/or structural information, allowing to design potentially novel mono- or multi-target inhibitors from relatively large and heterogeneous databases. Here, we also discuss the need to apply multi-scale modeling, to exemplify how different datasets based on target inhibition can be simultaneously integrated and predicted together with other relevant endpoints such as the biological activity against non-biomolecular targets, as well as in vitro and in vivo toxicity and pharmacokinetic properties. In this context, seminal papers are briefly analyzed. As huge amounts of data continue to accumulate in the domains of the chemical, biological and biomedical sciences, it has become clear that drug discovery must be viewed as a multi-scale optimization process. An ideal multi-scale approach should integrate diverse chemical and biological data and also serve as a knowledge generator, enabling the design of potentially optimal chemicals that may become therapeutic agents.


Asunto(s)
Diseño de Fármacos , Modelos Moleculares , Relación Estructura-Actividad Cuantitativa , Antibacterianos/química , Antibacterianos/farmacología , Antivirales/química , Antivirales/farmacología , Antivirales/uso terapéutico , Bacterias Gramnegativas/efectos de los fármacos , Infecciones por VIH/tratamiento farmacológico , Hepacivirus/efectos de los fármacos , Humanos , Redes Neurales de la Computación
20.
Curr Top Med Chem ; 20(19): 1661-1676, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32515311

RESUMEN

BACKGROUND: Alzheimer's disease is characterized by a progressive pattern of cognitive and functional impairment, which ultimately leads to death. Computational approaches have played an important role in the context of drug discovery for anti-Alzheimer's therapies. However, most of the computational models reported to date have been focused on only one protein associated with Alzheimer's, while relying on small datasets of structurally related molecules. OBJECTIVE: We introduce the first model combining perturbation theory and machine learning based on artificial neural networks (PTML-ANN) for simultaneous prediction and design of inhibitors of three Alzheimer's disease-related proteins, namely glycogen synthase kinase 3 beta (GSK3B), histone deacetylase 1 (HDAC1), and histone deacetylase 6 (HDAC6). METHODS: The PTML-ANN model was obtained from a dataset retrieved from ChEMBL, and it relied on a classification approach to predict chemicals as active or inactive. RESULTS: The PTML-ANN model displayed sensitivity and specificity higher than 85% in both training and test sets. The physicochemical and structural interpretation of the molecular descriptors in the model permitted the direct extraction of fragments suggested to favorably contribute to enhancing the multitarget inhibitory activity. Based on this information, we assembled ten molecules from several fragments with positive contributions. Seven of these molecules were predicted as triple target inhibitors while the remaining three were predicted as dual-target inhibitors. The estimated physicochemical properties of the designed molecules complied with Lipinski's rule of five and its variants. CONCLUSION: This work opens new horizons toward the design of multi-target inhibitors for anti- Alzheimer's therapies.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 6/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Aprendizaje Automático , Redes Neurales de la Computación , Inhibidores de Proteínas Quinasas/farmacología , Enfermedad de Alzheimer/metabolismo , Diseño de Fármacos , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 6/metabolismo , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/química , Humanos , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA