Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Annu Rev Pharmacol Toxicol ; 53: 451-73, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23140241

RESUMO

To achieve sensitive and specific mechanism-based prediction of drug toxicity, the tools of systems pharmacology will be integrated using structured ontological approaches, analytics, mathematics, and statistics. Success of this effort is based on the assumption that a systems network that consists of drug-induced perturbations of physiological functions can be characterized. This network spans the hierarchy of biological organization, from gene to mRNA to protein to intracellular organelle to cell to organ to organism. It is populated with data from each of these levels of biological organization. These data, from disparate sources, include the published literature, drug development archives of all approved drugs and drug candidates that did not complete development, and various toxicity databases and adverse event reporting systems. The network contains interrelated genomics, transcriptomics, and metabolomics data, as well as organ and physiological functional data that are derived from the universe of information that describes and analyzes drug toxicity. Here we describe advances in bioinformatics, computer sciences, next-generation sequencing, and systems biology that create the opportunity for integrated systems pharmacology-based prediction of drug safety.


Assuntos
Biologia Computacional/métodos , Descoberta de Drogas/métodos , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/etiologia , Farmacologia/métodos , Biologia de Sistemas/métodos , Animais , Humanos
2.
Chem Res Toxicol ; 29(5): 914-23, 2016 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-27063352

RESUMO

Drug toxicity presents a major challenge in drug development and patient care. We set to build upon previous works regarding select drug-induced toxicities to find common patterns in the mode of action of the drugs associated with these toxicities. In particular, we focused on five disparate organ toxicities, peripheral neuropathy (PN), rhabdomyolysis (RM), Stevens-Johnson syndrome/toxic epidermal necrosis (SJS/TEN), lung injury (LI), and heart contraction-related cardiotoxicity (CT), and identified biological commonalities between and among the toxicities in terms of pharmacological targets and nearest neighbors (indirect effects) using the hyper-geometric test and a distance metric of Spearman correlation. There were 20 significant protein targets associated with two toxicities and 0 protein targets associated with three or more toxicities. Per Spearman distance, PN was closest to SJS/TEN compared to other pairs, whereas the pairs involving RM were more different than others excluding RM. The significant targets associated with RM outnumbered those associated with every one of the other four toxicities. Enrichment analysis of drug targets that are expressed in corresponding organ/tissues determined proteins that should be avoided in drug discovery. The identified biological patterns emerging from the mode of action of these drugs are statistically associated with these serious toxicities and could potentially be used as predictors for new drug candidates. The predictive power and usefulness of these biological patterns will increase with the database of these five toxicities. Furthermore, extension of our approach to all severe adverse reactions will produce useful biological commonalities for reference in drug discovery and development.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Bases de Dados Factuais , Humanos
3.
Chem Res Toxicol ; 28(5): 927-34, 2015 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-25811541

RESUMO

Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are serious cutaneous adverse reactions. We mined the approved labels in Drugs@FDA, identified the SJS/TEN list of 259 small molecular drugs and biologics, and conducted systems pharmacological network analyses. Pharmacological network analysis revealed that drugs with treatment-related SJS and/or TEN are pharmacologically diverse and that the largest subnetwork is associated with antiepileptic drugs and their pharmacological targets. Our pharmacological network analysis identified CTNNB1 [catenin (cadherin-associated protein), beta 1, 88 kDa] as a significant intermediator. This protein is involved in maintaining the functional integrity of the epithelium through regulating cell growth and adhesion between cells in various organs, including the skin. Leveraging a publicly accessible genome-wide transcriptional expression database, we found that human leukocyte antigen-related (HLA) genes were significantly perturbed by various SJS/TEN-inducing drugs. Notably, carbamazepine (CBZ) perturbed several HLA genes, among which HLA-DQB1*0201 was reportedly shown to be associated with CBZ-induced SJS/TEN in caucasians. In short, systems analysis by leveraging a publicly accessible knowledge base and databases could produce meaningful results for further mechanistic investigation. Our study sheds light on the utility of systems pharmacology analysis for gaining insight into clinical drug toxicity.


Assuntos
Anticonvulsivantes/efeitos adversos , Bibliotecas de Moléculas Pequenas/efeitos adversos , Síndrome de Stevens-Johnson/etiologia , Carbamazepina/efeitos adversos , Bases de Dados de Produtos Farmacêuticos , Regulação da Expressão Gênica/efeitos dos fármacos , Cadeias beta de HLA-DQ/genética , Humanos , Fatores de Risco , Síndrome de Stevens-Johnson/genética , Síndrome de Stevens-Johnson/metabolismo , beta Catenina/metabolismo
4.
Chem Res Toxicol ; 27(3): 421-32, 2014 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-24422454

RESUMO

The goal of this study was to integrate systems pharmacology and biochemical flux to delineate drug-induced rhabdomyolysis by leveraging prior knowledge and publicly accessible data. A list of 211 rhabdomyolysis-inducing drugs (RIDs) was compiled and curated from multiple sources. Extended pharmacological network analysis revealed that the intermediators directly interacting with the pharmacological targets of RIDs were significantly enriched with functions such as regulation of cell cycle, apoptosis, and ubiquitin-mediated proteolysis. A total of 78 intermediators were shown to be significantly connected to at least five RIDs, including estrogen receptor 1 (ESR1), synuclein gamma (SNCG), and janus kinase 2 (JAK2). Transcriptomic analysis of RIDs profiled in Connectivity Map on the global scale revealed that multiple pathways are perturbed by RIDs, including ErbB signaling and lipid metabolism pathways, and that carnitine palmitoyl transferase 2 (CPT2) was in the top 1 percent of the most differentially perturbed genes. CPT2 was downregulated by nine drugs that perturbed the genes significantly enriched in oxidative phosphorylation and energy-metabolism pathways. With statins as the use case, biochemical pathway analysis on the local scale implicated a role for CPT2 in statin-induced perturbation of energy homeostasis, which is in agreement with reports of statin-CPT2 interaction. Considering the complexity of human biology, an integrative multiple-approach analysis composed of a biochemical flux network, pharmacological on- and off-target networks, and transcriptomic signature is important for understanding drug safety and for providing insight into clinical gene-drug interactions.


Assuntos
Preparações Farmacêuticas/metabolismo , Rabdomiólise/metabolismo , Apoptose/efeitos dos fármacos , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Bases de Dados Factuais , Regulação para Baixo/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Perfilação da Expressão Gênica , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/química , Inibidores de Hidroximetilglutaril-CoA Redutases/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/toxicidade , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Fosforilação Oxidativa/efeitos dos fármacos , Preparações Farmacêuticas/química , Proteólise/efeitos dos fármacos , Rabdomiólise/induzido quimicamente , Rabdomiólise/patologia , gama-Sinucleína/genética , gama-Sinucleína/metabolismo
5.
Biopharm Drug Dispos ; 35(1): 1-14, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24136298

RESUMO

Advances in systems biology in conjunction with the expansion in knowledge of drug effects and diseases present an unprecedented opportunity to extend traditional pharmacokinetic and pharmacodynamic modeling/analysis to conduct systems pharmacology modeling. Many drugs that cause liver injury and myopathies have been studied extensively. Mitochondrion-centric systems pharmacology modeling is important since drug toxicity across a large number of pharmacological classes converges to mitochondrial injury and death. Approaches to systems pharmacology modeling of drug effects need to consider drug exposure, organelle and cellular phenotypes across all key cell types of human organs, organ-specific clinical biomarkers/phenotypes, gene-drug interaction and immune responses. Systems modeling approaches, that leverage the knowledge base constructed from curating a selected list of drugs across a wide range of pharmacological classes, will provide a critically needed blueprint for making informed decisions to reduce the rate of attrition for drugs in development and increase the number of drugs with an acceptable benefit/risk ratio.


Assuntos
Modelos Biológicos , Farmacologia Clínica/métodos , Biologia de Sistemas/métodos , Biomarcadores , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/prevenção & controle , Humanos , Fenótipo
6.
Clin Pharmacol Ther ; 115(2): 201-205, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37984065

RESUMO

One of the goals of the Accelerating Rare Disease Cures (ARC) program in the Center for Drug Evaluation and Research (CDER) at the US Food and Drug Administration (FDA) is the development and use of regulatory and scientific tools, including drug/disease modeling, dose selection, and translational medicine tools. To facilitate achieving this goal, the FDA in collaboration with the University of Maryland Center of Excellence in Regulatory Science and Innovation (M-CERSI) hosted a virtual public workshop on May 11, 2023, entitled "Creating a Roadmap to Quantitative Systems Pharmacology-Informed Rare Disease Drug Development." This workshop engaged scientists from pharmaceutical companies, academic institutes, and the FDA to discuss the potential utility of quantitative systems pharmacology (QSP) in rare disease drug development and identify potential challenges and solutions to facilitate its use. Here, we report the main findings from this workshop, highlight the key takeaways, and propose a roadmap to facilitate the use of QSP in rare disease drug development.


Assuntos
Farmacologia em Rede , Doenças Raras , Humanos , Preparações Farmacêuticas , Doenças Raras/tratamento farmacológico , Desenvolvimento de Medicamentos , Desenho de Fármacos
7.
J Pharm Sci ; 112(4): 904-908, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36279954

RESUMO

Advances in biomedical and computer technologies have presented the modeling community the opportunity for mechanistically modeling and simulating the variability in a disease phenotype or in a drug response. The capability to quantify response variability can inform a drug development program. Quantitative systems pharmacology scientists have published various computational approaches for creating virtual patient populations (VPops) to model and simulate drug response variability. Genomic variations can impact disease characteristics and drug exposure and response. Quantitative proteomics technologies are increasingly used to facilitate drug discovery and development and inform patient care. Incorporating variations in genomics and quantitative proteomics may potentially inform creation of VPops to model and simulate virtual patient trials, and may help account for, in a predictive manner, phenotypic variations observed clinically.


Assuntos
Genômica , Proteômica , Desenvolvimento de Medicamentos , Fenótipo , Variação Biológica da População
8.
Clin Pharmacol Ther ; 113(1): 71-79, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36282186

RESUMO

US Food and Drug Administration (FDA) guidance outlines how biosimilars can be developed based on pharmacokinetic (PK) and pharmacodynamic (PD) similarity study data in lieu of a comparative clinical efficacy study. There is a paucity of PD comparability studies in biosimilar development, leaving open questions about how best to plan these studies. To that end, we conducted a randomized, double-blinded, placebo-controlled, single-dose, parallel-arm clinical study in healthy participants to evaluate approaches to address information gaps, inform analysis best practices, and apply emerging technologies in biomarker characterization. Seventy-two healthy participants (n = 8 per arm) received either placebo or one of four doses of the proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors alirocumab (15-100 mg) or evolocumab (21-140 mg) to evaluate the maximum change from baseline (ΔPDmax ) and the baseline-adjusted area under the effect curve (AUEC) for the biomarkers low-density lipoprotein cholesterol (LDL-C) and apolipoprotein B (apoB) in serum. We investigated approaches to minimize variability in PD measures. Coefficient of variation was lower for LDL-C than apoB at therapeutic doses. Modeling and simulation were used to establish the dose-response relationship and provided support that therapeutic doses for these products are adequately sensitive and are on the steep part of the dose-response curves. Similar dose-response relationships were observed for both biomarkers. ΔPDmax plateaued at lower doses than AUEC. In summary, this study illustrates how pilot study data can be leveraged to inform appropriate dosing and data analyses for a PK and PD similarity study.


Assuntos
Anticolesterolemiantes , Medicamentos Biossimilares , Humanos , Medicamentos Biossimilares/efeitos adversos , Inibidores de PCSK9 , LDL-Colesterol , Pró-Proteína Convertase 9 , Anticorpos Monoclonais/farmacocinética , Projetos Piloto , Apolipoproteínas B , Biomarcadores , Resultado do Tratamento , Anticolesterolemiantes/farmacocinética
9.
Mol Pharm ; 9(12): 3495-505, 2012 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-23051182

RESUMO

Prolonged use of proton pump inhibitors has reportedly caused rare clinically symptomatic hypomagnesemia. A review of the literature suggests PPI drugs may impair intestinal magnesium absorption. With the goal of preventing PPI-induced hypomagnesemia, an oral absorption-centric model was developed by referencing literature data. Our modeling with human data reveals that magnesium absorption is substantial in the distal intestine. We then perform simulations by referring to the reported reduction in mid to distal intestinal pH caused by one week of oral esomeprazole, and to reported reduction of the divalent cation-sensitive current when the carboxyl side chains of glutamic and aspartic residues in the binding channels of TRPM6/TRPM7 were neutralized. Our simulations reveal that short-term PPI therapy may cause a very small reduction (5%) in the serum magnesium level, which is qualitatively consistent with the reported 1% reduction in magnesium absorption following 1 week of omeprazole in humans. Simulations provide insight into the benefit of frequent but small dose of magnesium supplementation in maintaining the serum magnesium level when magnesium deficiency occurs.


Assuntos
Simulação por Computador , Absorção Intestinal/efeitos dos fármacos , Deficiência de Magnésio/tratamento farmacológico , Deficiência de Magnésio/metabolismo , Magnésio/farmacocinética , Omeprazol/farmacologia , Inibidores da Bomba de Prótons/farmacologia , Administração Oral , Homeostase , Humanos , Magnésio/metabolismo , Modelos Biológicos , Proteínas Serina-Treonina Quinases , Canais de Cátion TRPM/metabolismo , Distribuição Tecidual
10.
Methods Mol Biol ; 2486: 87-104, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35437720

RESUMO

Viruses can cause many diseases resulting in disabilities and death. Fortunately, advances in systems medicine enable the development of effective therapies for treating viral diseases, of vaccines to prevent viral infections, as well as of diagnostic tools to mitigate the risk and reduce the death toll. Characterizing the SARS-CoV-2 gene sequence and the role of its spike protein in infection informs development of small molecule drugs, antibodies, and vaccines to combat infection and complication, as well as to end the pandemic. Drug repurposing of small molecule drugs is a viable strategy to combat viral diseases; the key concepts include (1) linking a drug candidate's pharmacological network to its pharmacodynamic response in patients; (2) linking a drug candidate's physicochemical properties to its pharmacokinetic characteristics; and (3) optimizing the safe and effective dosing regimen within its therapeutic window. Computational integration of drug-induced signaling pathways with clinical outcomes is useful to inform selection of potential drug candidates with respect to safety and effectiveness. Key pharmacokinetic and pharmacodynamic principles for computational optimization of drug development include a drug candidate's Cminss/IC95 ratio, pharmacokinetic characteristics, and systemic exposure-response relationship, where Cminss is the trough concentration following multiple dosing. In summary, systems medicine approaches play a vital role in global success in combating viral diseases, including global real-time information sharing, development of test kits, drug repurposing, discovery and development of safe, effective therapies, detection of highly transmissible and deadly variants, and development of vaccines.


Assuntos
Tratamento Farmacológico da COVID-19 , SARS-CoV-2 , Reposicionamento de Medicamentos , Humanos , Pandemias/prevenção & controle , SARS-CoV-2/genética , Análise de Sistemas
11.
CPT Pharmacometrics Syst Pharmacol ; 10(12): 1479-1484, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34734497

RESUMO

Quantitative systems pharmacology (QSP) has been proposed as a scientific domain that can enable efficient and informative drug development. During the past several years, there has been a notable increase in the number of regulatory submissions that contain QSP, including Investigational New Drug Applications (INDs), New Drug Applications (NDAs), and Biologics License Applications (BLAs) to the US Food and Drug Administration. However, there has been no comprehensive characterization of the nature of these regulatory submissions regarding model details and intended applications. To address this gap, a landscape analysis of all the QSP submissions as of December 2020 was conducted. This report summarizes the (1) yearly trend of submissions, (2) proportion of submissions between INDs and NDAs/BLAs, (3) percentage distribution along the stages of drug development, (4) percentage distribution across various therapeutic areas, and (5) nature of QSP applications. In brief, QSP is increasingly applied to model and simulate both drug effectiveness and safety throughout the drug development process across disease areas.


Assuntos
Desenvolvimento de Medicamentos/estatística & dados numéricos , Farmacologia em Rede/estatística & dados numéricos , United States Food and Drug Administration/estatística & dados numéricos , Humanos , Estados Unidos
12.
AAPS J ; 23(3): 60, 2021 04 30.
Artigo em Inglês | MEDLINE | ID: mdl-33931790

RESUMO

The pharmaceutical industry is actively applying quantitative systems pharmacology (QSP) to make internal decisions and guide drug development. To facilitate the eventual development of a common framework for assessing the credibility of QSP models for clinical drug development, scientists from US Food and Drug Administration and the pharmaceutical industry organized a full-day virtual Scientific Exchange on July 1, 2020. An assessment form was used to ensure consistency in the evaluation process. Among the cases presented, QSP was applied to various therapeutic areas. Applications mostly focused on phase 2 dose selection. Model transparency, including details on expert knowledge and data used for model development, was identified as a major factor for robust model assessment. The case studies demonstrated some commonalities in the workflow of QSP model development, calibration, and validation but differ in the size, scope, and complexity of QSP models, in the acceptance criteria for model calibration and validation, and in the algorithms/approaches used for creating virtual patient populations. Though efforts are being made to build the credibility of QSP models and the confidence is increasing in applying QSP for internal decisions at the clinical stages of drug development, there are still many challenges facing QSP application to late stage drug development. The QSP community needs a strategic plan that includes the ability and flexibility to Adapt, to establish Common expectations for model Credibility needed to inform drug Labeling and patient care, and to AIM to achieve the goal (ACCLAIM).


Assuntos
Desenvolvimento de Medicamentos/métodos , Colaboração Intersetorial , Modelos Biológicos , Biologia de Sistemas/métodos , Congressos como Assunto , Indústria Farmacêutica/organização & administração , Humanos , Estados Unidos , United States Food and Drug Administration/organização & administração
13.
Front Physiol ; 12: 637999, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33841175

RESUMO

Mathematical biology and pharmacology models have a long and rich history in the fields of medicine and physiology, impacting our understanding of disease mechanisms and the development of novel therapeutics. With an increased focus on the pharmacology application of system models and the advances in data science spanning mechanistic and empirical approaches, there is a significant opportunity and promise to leverage these advancements to enhance the development and application of the systems pharmacology field. In this paper, we will review milestones in the evolution of mathematical biology and pharmacology models, highlight some of the gaps and challenges in developing and applying systems pharmacology models, and provide a vision for an integrated strategy that leverages advances in adjacent fields to overcome these challenges.

14.
Artigo em Inglês | MEDLINE | ID: mdl-31674729

RESUMO

The substantial progress made in the basic sciences of the brain has yet to be adequately translated to successful clinical therapeutics to treat central nervous system (CNS) diseases. Possible explanations include the lack of quantitative and validated biomarkers, the subjective nature of many clinical endpoints, and complex pharmacokinetic/pharmacodynamic relationships, but also the possibility that highly selective drugs in the CNS do not reflect the complex interactions of different brain circuits. Although computational systems pharmacology modeling designed to capture essential components of complex biological systems has been increasingly accepted in pharmaceutical research and development for oncology, inflammation, and metabolic disorders, the uptake in the CNS field has been very modest. In this article, a cross-disciplinary group with representatives from academia, pharma, regulatory, and funding agencies make the case that the identification and exploitation of CNS therapeutic targets for drug discovery and development can benefit greatly from a system and network approach that can span the gap between molecular pathways and the neuronal circuits that ultimately regulate brain activity and behavior. The National Institute of Neurological Disorders and Stroke (NINDS), in collaboration with the National Institute on Aging (NIA), National Institute of Mental Health (NIMH), National Institute on Drug Abuse (NIDA), and National Center for Advancing Translational Sciences (NCATS), convened a workshop to explore and evaluate the potential of a quantitative systems pharmacology (QSP) approach to CNS drug discovery and development. The objective of the workshop was to identify the challenges and opportunities of QSP as an approach to accelerate drug discovery and development in the field of CNS disorders. In particular, the workshop examined the potential for computational neuroscience to perform QSP-based interrogation of the mechanism of action for CNS diseases, along with a more accurate and comprehensive method for evaluating drug effects and optimizing the design of clinical trials. Following up on an earlier white paper on the use of QSP in general disease mechanism of action and drug discovery, this report focuses on new applications, opportunities, and the accompanying limitations of QSP as an approach to drug development in the CNS therapeutic area based on the discussions in the workshop with various stakeholders.


Assuntos
Fármacos do Sistema Nervoso Central/farmacologia , Doenças do Sistema Nervoso Central/tratamento farmacológico , Desenvolvimento de Medicamentos/métodos , Descoberta de Drogas/métodos , Animais , Humanos , Farmacologia/métodos , Biologia de Sistemas
15.
J Pharm Sci ; 108(2): 798-806, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30244014

RESUMO

There are no approved drugs or biologics to treat Ebola virus disease (EVD). Literature reviews identified a list of 141 drugs with reports of preliminary in vitro potency and in vivo effectiveness in animals or with reports of clinical use/trials in EVD patients. The majority of these drugs have been individually approved by the U.S. Food and Drug Administration for treating various non-EVD diseases. The anti-Ebola potency data of these drugs were curated from literature and publicly accessible databases, along with their individual biopharmaceutical and pharmacokinetic characteristics. To facilitate the development of antiviral drugs including anti-EVD drugs, highlights include optimization of the exposure-response relationship, design of a safe and effective clinical dosing regimen to achieve an adequate high ratio of clinical Cmin to a plasma protein binding-adjusted EC95, and the pharmacokinetic studies needed in animal models (healthy and affected) and in healthy volunteers. The exposure/response relationship for human dose selection is summarized, as described in the U.S. Food and Drug Administration "Animal Rule'' guidance when human efficacy studies are not ethical or feasible.


Assuntos
Antivirais/uso terapêutico , Reposicionamento de Medicamentos/métodos , Doença pelo Vírus Ebola/tratamento farmacológico , Animais , Antivirais/farmacocinética , Aprovação de Drogas/métodos , Humanos , Estados Unidos , United States Food and Drug Administration
16.
AAPS J ; 21(4): 72, 2019 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-31161268

RESUMO

Systems pharmacology approaches have the capability of quantitatively linking the key biological molecules relevant to a drug candidate's mechanism of action (drug-induced signaling pathways) to the clinical biomarkers associated with the proposed target disease, thereby quantitatively facilitating its development and life cycle management. In this review, the model attributes of published quantitative systems pharmacology (QSP) modeling for lowering cholesterol, treating salt-sensitive hypertension, and treating rare diseases as well as describing bone homeostasis and related pharmacological effects are critically reviewed with respect to model quality, calibration, validation, and performance. We further reviewed the common practices in optimizing QSP modeling and prediction. Notably, leveraging genetics and genomic studies for model calibration and validation is common. Statistical and quantitative assessment of QSP prediction and handling of model uncertainty are, however, mostly lacking as are the quantitative and statistical criteria for assessing QSP predictions and the covariance matrix of coefficients between the parameters in a validated virtual population. To accelerate advances and application of QSP with consistent quality, a list of key questions is proposed to be addressed when assessing the quality of a QSP model in hopes of stimulating the scientific community to set common expectations. The common expectations as to what constitutes the best QSP modeling practices, which the scientific community supports, will advance QSP modeling in the realm of informed drug development. In the long run, good practices will extend the life cycles of QSP models beyond the life cycles of individual drugs.


Assuntos
Desenvolvimento de Medicamentos/métodos , Modelos Biológicos , Farmacologia/métodos , Biologia de Sistemas/métodos , Pesquisa Translacional Biomédica/métodos , Desenvolvimento de Medicamentos/normas , Drogas em Investigação/farmacologia , Humanos , Pesquisa Translacional Biomédica/normas
17.
Methods Mol Biol ; 1939: 181-198, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30848462

RESUMO

In the era of big data and informatics, computational integration of data across the hierarchical structures of human biology enables discovery of new druggable targets of disease and new mode of action of a drug. We present herein a computational framework and guide of integrating drug targets, gene expression data, transcription factors, and prior knowledge of protein interactions to computationally construct the signaling network (mode of action) of a drug. In a similar manner, a disease network is constructed using its disease targets. And then, drug candidates are computationally prioritized by computationally ranking the closeness between a disease network and a drug's signaling network. Furthermore, we describe the use of the most perturbed HLA genes to assess the safety risk for immune-mediated adverse reactions such as Stevens-Johnson syndrome/toxic epidermal necrolysis.


Assuntos
Biologia Computacional/métodos , Descoberta de Drogas/métodos , Big Data , Bases de Dados Factuais , Humanos , Medicina de Precisão/métodos , Programação Linear , Mapas de Interação de Proteínas/efeitos dos fármacos , Síndrome de Stevens-Johnson/etiologia , Transcriptoma/efeitos dos fármacos
20.
CPT Pharmacometrics Syst Pharmacol ; 7(3): 166-174, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29341478

RESUMO

Drug-induced cardiomyopathy contributes to drug attrition. We compared two pipelines of predictive modeling: (1) applying elastic net (EN) to differentially expressed genes (DEGs) of drugs; (2) applying integer linear programming (ILP) to construct each drug's signaling pathway starting from its targets to downstream proteins, to transcription factors, and to its DEGs in human cardiomyocytes, and then subjecting the genes/proteins in the drugs' signaling networks to EN regression. We classified 31 drugs with availability of DEGs into 13 toxic and 18 nontoxic drugs based on a clinical cardiomyopathy incidence cutoff of 0.1%. The ILP-augmented modeling increased prediction accuracy from 79% to 88% (sensitivity: 88%; specificity: 89%) under leave-one-out cross validation. The ILP-constructed signaling networks of drugs were better predictors than DEGs. Per literature, the microRNAs that reportedly regulate expression of our six top predictors are of diagnostic value for natural heart failure or doxorubicin-induced cardiomyopathy. This translational predictive modeling might uncover potential biomarkers.


Assuntos
Cardiomiopatias/induzido quimicamente , Doxorrubicina/farmacologia , Redes Reguladoras de Genes/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Cardiomiopatias/metabolismo , Cardiotoxicidade , Biologia Computacional , Doxorrubicina/efeitos adversos , Humanos , Modelos Biológicos , Terapia de Alvo Molecular , Análise de Regressão , Pesquisa Translacional Biomédica
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa