Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Molecules ; 26(1)2021 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-33401494

RESUMEN

Amorphous solid dispersions (ASDs) have emerged as widespread formulations for drug delivery of poorly soluble active pharmaceutical ingredients (APIs). Predicting the API solubility with various carriers in the API-carrier mixture and the principal API-carrier non-bonding interactions are critical factors for rational drug development and formulation decisions. Experimental determination of these interactions, solubility, and dissolution mechanisms is time-consuming, costly, and reliant on trial and error. To that end, molecular modeling has been applied to simulate ASD properties and mechanisms. Quantum mechanical methods elucidate the strength of API-carrier non-bonding interactions, while molecular dynamics simulations model and predict ASD physical stability, solubility, and dissolution mechanisms. Statistical learning models have been recently applied to the prediction of a variety of drug formulation properties and show immense potential for continued application in the understanding and prediction of ASD solubility. Continued theoretical progress and computational applications will accelerate lead compound development before clinical trials. This article reviews in silico research for the rational formulation design of low-solubility drugs. Pertinent theoretical groundwork is presented, modeling applications and limitations are discussed, and the prospective clinical benefits of accelerated ASD formulation are envisioned.


Asunto(s)
Simulación por Computador , Composición de Medicamentos , Excipientes/química , Modelos Químicos , Polímeros/química , Química Farmacéutica , Solubilidad
2.
Molecules ; 26(7)2021 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-33805419

RESUMEN

The COVID-19 pandemic has reached over 100 million worldwide. Due to the multi-targeted nature of the virus, it is clear that drugs providing anti-COVID-19 effects need to be developed at an accelerated rate, and a combinatorial approach may stand to be more successful than a single drug therapy. Among several targets and pathways that are under investigation, the renin-angiotensin system (RAS) and specifically angiotensin-converting enzyme (ACE), and Ca2+-mediated SARS-CoV-2 cellular entry and replication are noteworthy. A combination of ACE inhibitors and calcium channel blockers (CCBs), a critical line of therapy for pulmonary hypertension, has shown therapeutic relevance in COVID-19 when investigated independently. To that end, we conducted in silico modeling using BIOiSIM, an AI-integrated mechanistic modeling platform by utilizing known preclinical in vitro and in vivo datasets to accurately simulate systemic therapy disposition and site-of-action penetration of the CCBs and ACEi compounds to tissues implicated in COVID-19 pathogenesis.


Asunto(s)
Antivirales/farmacocinética , Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos/métodos , Hipertensión Pulmonar/tratamiento farmacológico , Inhibidores de la Enzima Convertidora de Angiotensina/farmacocinética , Antivirales/sangre , Biosimilares Farmacéuticos , COVID-19/complicaciones , Bloqueadores de los Canales de Calcio/farmacocinética , Simulación por Computador , Bases de Datos Farmacéuticas , Desarrollo de Medicamentos/métodos , Humanos , Hipertensión Pulmonar/virología , Distribución Tisular
3.
Drug Metab Rev ; 52(2): 283-298, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32083960

RESUMEN

Historically, failure rates in drug development are high; increased sophistication and investment throughout the process has shifted the reasons for attrition, but the overall success rates have remained stubbornly and consistently low. Only 8% of new entities entering clinical testing gain regulatory approval, indicating that significant obstacles still exist for efficient therapeutic development. The continued high failure rate can be partially attributed to the inability to link drug exposure with the magnitude of observed safety and efficacy-related pharmacodynamic (PD) responses; frequently, this is a result of nonclinical models exhibiting poor prediction of human outcomes across a wide range of disease conditions, resulting in faulty evaluation of drug toxicology and efficacy. However, the increasing quality and standardization of experimental methods in preclinical stages of testing has created valuable data sets within companies that can be leveraged to further improve the efficiency and accuracy of preclinical prediction for both pharmacokinetics (PK) and PD. Models of Quantitative structure-activity relationships (QSAR), physiologically based pharmacokinetics (PBPK), and PK/PD relationships have also improved efficiency. Founded on a core understanding of biochemistry and physiological interactions of xenobiotics, these in silico methods have the potential to increase the probability of compound success in clinical trials. Integration of traditional computational methods with machine-learning approaches and existing internal pharma databases stands to make a fundamental impact on the speed and accuracy of predictions during the process of drug development and approval.


Asunto(s)
Desarrollo de Medicamentos/métodos , Modelos Biológicos , Modelos Químicos , Animales , Inteligencia Artificial , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos , Humanos , Aprendizaje Automático , Farmacocinética , Relación Estructura-Actividad Cuantitativa
4.
Drug Discov Today ; 26(6): 1459-1465, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33609781

RESUMEN

The development of successful drugs is expensive and time-consuming because of high clinical attrition rates. This is caused partially by the rupture seen in the translatability of the drug from the bench to the clinic in the context of personalized medicine. Artificial intelligence (AI)-driven platforms integrated with mechanistic modeling have become instrumental in accelerating the drug development process by leveraging data ubiquitously across the various phases. AI can counter the deficiencies and ambiguities that arise during the classical drug development process while reducing human intervention and bridging the translational gap in discovering the connections between drugs and diseases.


Asunto(s)
Inteligencia Artificial , Desarrollo de Medicamentos/métodos , Medicina de Precisión/métodos , Animales , Simulación por Computador , Humanos , Investigación Biomédica Traslacional
5.
Pharmaceutics ; 13(2)2021 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-33669957

RESUMEN

The use of opioid analgesics in treating severe pain is frequently associated with putative adverse effects in humans. Topical agents that are shown to have high efficacy with a favorable safety profile in clinical settings are great alternatives for pain management of multimodal analgesia. However, the risk of side effects induced by transdermal absorption and systemic exposure is of great concern as they are challenging to predict. The present study aimed to use "BIOiSIM" an artificial intelligence-integrated biosimulation platform to predict the transdermal disposition of opioid analgesics. The model successfully predicted their exposure following the topical application of central opioid agonist buprenorphine and peripheral agonist oxycodone in healthy human subjects with simulation of intra-skin exposure in subjects with burns and pressure wounds. The predicted plasma levels of analgesics were used to evaluate the safety of the therapeutic pain control in patients with the dermal structural impairments caused by acute (burns) or chronic cutaneous lesions (pressure wounds) with topical opioid analgesics.

6.
Sci Rep ; 11(1): 11143, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34045592

RESUMEN

Prediction of the first-in-human dosing regimens is a critical step in drug development and requires accurate quantitation of drug distribution. Traditional in vivo studies used to characterize clinical candidate's volume of distribution are error-prone, time- and cost-intensive and lack reproducibility in clinical settings. The paper demonstrates how a computational platform integrating machine learning optimization with mechanistic modeling can be used to simulate compound plasma concentration profile and predict tissue-plasma partition coefficients with high accuracy by varying the lipophilicity descriptor logP. The approach applied to chemically diverse small molecules resulted in comparable geometric mean fold-errors of 1.50 and 1.63 in pharmacokinetic outputs for direct tissue:plasma partition and hybrid logP optimization, with the latter enabling prediction of tissue permeation that can be used to guide toxicity and efficacy dosing in human subjects. The optimization simulations required to achieve these results were parallelized on the AWS cloud and generated outputs in under 5 h. Accuracy, speed, and scalability of the framework indicate that it can be used to assess the relevance of other mechanistic relationships implicated in pharmacokinetic-pharmacodynamic phenomena with a lower risk of overfitting datasets and generate large database of physiologically-relevant drug disposition for further integration with machine learning models.

7.
Drug Des Devel Ther ; 14: 2307-2317, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32606600

RESUMEN

INTRODUCTION: Transdermal drug delivery is gaining popularity as an alternative to traditional routes of administration. It can increase patient compliance because of its painless and noninvasive nature, aid compounds in bypassing presystemic metabolic effects, and reduce the likelihood of adverse effects through decreased systemic exposure. In silico physiological modeling is critical to predicting dermal exposure for a therapeutic and assessing the impact of different formulations on transdermal disposition. METHODS: The present study aimed at developing a physiologically based transdermal platform, "BIOiSIM", that could be globally applied to a wide variety of compounds to predict their transdermal disposition. The platform integrates a 16-compartment model of compound pharmacokinetics and was used to simulate and predict drug exposure of three chemically and biologically distinct drug-like compounds. Machine learning optimization was composed of two components: exhaustive search algorithm (coarse-tuning) and descent (fine-tuning) integrated with the platform used to quantitatively determine parameters influencing pharmacokinetics (eg permeability, kperm) of test compounds. RESULTS: The model successfully predicted drug exposure (AUC, Cmax and Tmax) following transdermal application of morphine, buprenorphine and nicotine in human subjects, mostly with less than two-fold absolute average fold error (AAFE). The model was further able to successfully characterize the relationship between observed systemic exposure and intended pharmacological effect. The predicted systemic concentration of morphine and plasma levels of endogenous pain biomarkers were used to estimate the effectiveness of a given therapeutic regimen. CONCLUSION: BIOiSIM marks a novel approach to in silico prediction that will enable leveraging of machine learning technology in the pharmaceutical space. The approach to model development outlined results in scalable, accurate models and enables the generation of large parameter/coefficient datasets from in vivo clinical data that can be used in future work to train quantitative structure activity relationship (QSAR) models for predicting likelihood of compound utility as a transdermally administered therapeutic.


Asunto(s)
Buprenorfina/metabolismo , Modelos Biológicos , Morfina/metabolismo , Nicotina/metabolismo , Administración Cutánea , Buprenorfina/administración & dosificación , Buprenorfina/farmacocinética , Simulación por Computador , Humanos , Morfina/administración & dosificación , Morfina/farmacocinética , Nicotina/administración & dosificación , Nicotina/farmacocinética , Permeabilidad , Relación Estructura-Actividad Cuantitativa
8.
Tissue Eng Part C Methods ; 21(3): 274-83, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25091645

RESUMEN

INTRODUCTION: Endothelial colony-forming cells (ECFCs) are endothelial progenitors that circulate in peripheral blood and are currently the subject of intensive investigation due to their therapeutic potential. However, in adults, ECFCs comprise a very small subset among circulating cells, which makes their isolation a challenge. MATERIALS AND METHODS: Currently, the standard method for ECFC isolation relies on the separation of mononuclear cells and erythrocyte lysis, steps that are time consuming and known to increase cell loss. Alternatively, we previously developed a novel disposable microfluidic platform containing antibody-functionalized degradable hydrogel coatings that is ideally suited for capturing low-abundance circulating cells from unprocessed blood. In this study, we reasoned that this microfluidic approach could effectively isolate rare ECFCs by virtue of their CD34 expression. RESULTS: We conducted preclinical experiments with peripheral blood from four adult volunteers and demonstrated that the actual microfluidic capture of circulating CD34(+) cells from unprocessed blood was compatible with the subsequent differentiation of these cells into ECFCs. Moreover, the ECFC yield obtained with the microfluidic system was comparable to that of the standard method. Importantly, we unequivocally validated the phenotypical and functional properties of the captured ECFCs, including the ability to form microvascular networks following transplantation into immunodeficient mice. DISCUSSION: We showed that the simplicity and versatility of our microfluidic system could be very instrumental for ECFC isolation while preserving their therapeutic potential. We anticipate our results will facilitate additional development of clinically suitable microfluidic devices by the vascular therapeutic and diagnostic industry.


Asunto(s)
Células Sanguíneas/citología , Ensayo de Unidades Formadoras de Colonias/métodos , Células Endoteliales/citología , Microfluídica/métodos , Adulto , Animales , Humanos , Ratones Desnudos , Neovascularización Fisiológica , Fenotipo , Reproducibilidad de los Resultados
9.
Sci Rep ; 5: 13317, 2015 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-26304831

RESUMEN

Many studies have suggested the significance of glycosyltransferase-mediated macromolecule glycosylation in the regulation of pluripotent states in human pluripotent stem cells (hPSCs). Here, we observed that the sialyltransferase ST6GAL1 was preferentially expressed in undifferentiated hPSCs compared to non-pluripotent cells. A lectin which preferentially recognizes α-2,6 sialylated galactosides showed strong binding reactivity with undifferentiated hPSCs and their glycoproteins, and did so to a much lesser extent with differentiated cells. In addition, downregulation of ST6GAL1 in undifferentiated hPSCs led to a decrease in POU5F1 (also known as OCT4) protein and significantly altered the expression of many genes that orchestrate cell morphogenesis during differentiation. The induction of cellular pluripotency in somatic cells was substantially impeded by the shRNA-mediated suppression of ST6GAL1, partially through interference with the expression of endogenous POU5F1 and SOX2. Targeting ST6GAL1 activity with a sialyltransferase inhibitor during cell reprogramming resulted in a dose-dependent reduction in the generation of human induced pluripotent stem cells (hiPSCs). Collectively, our data indicate that ST6GAL1 plays an important role in the regulation of pluripotency and differentiation in hPSCs, and the pluripotent state in human cells can be modulated using pharmacological tools to target sialyltransferase activity.


Asunto(s)
Antígenos CD/metabolismo , Diferenciación Celular/fisiología , Lectinas/metabolismo , Ácido N-Acetilneuramínico/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Sialiltransferasas/metabolismo , Activación Enzimática , Regulación del Desarrollo de la Expresión Génica/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Glicosilación , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA