Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Front Pharmacol ; 13: 1040838, 2022.
Article En | MEDLINE | ID: mdl-36339562

Assessing drug permeability across the blood-brain barrier (BBB) is important when evaluating the abuse potential of new pharmaceuticals as well as developing novel therapeutics that target central nervous system disorders. One of the gold-standard in vivo methods for determining BBB permeability is rodent log BB; however, like most in vivo methods, it is time-consuming and expensive. In the present study, two statistical-based quantitative structure-activity relationship (QSAR) models were developed to predict BBB permeability of drugs based on their chemical structure. The in vivo BBB permeability data were harvested for 921 compounds from publicly available literature, non-proprietary drug approval packages, and University of Washington's Drug Interaction Database. The cross-validation performance statistics for the BBB models ranged from 82 to 85% in sensitivity and 80-83% in negative predictivity. Additionally, the performance of newly developed models was assessed using an external validation set comprised of 83 chemicals. Overall, performance of individual models ranged from 70 to 75% in sensitivity, 70-72% in negative predictivity, and 78-86% in coverage. The predictive performance was further improved to 93% in coverage by combining predictions across the two software programs. These new models can be rapidly deployed to predict blood brain barrier permeability of pharmaceutical candidates and reduce the use of experimental animals.

2.
Regul Toxicol Pharmacol ; 125: 105006, 2021 Oct.
Article En | MEDLINE | ID: mdl-34273441

The ICH M7 (R1) guideline recommends the use of complementary (Q)SAR models to assess the mutagenic potential of drug impurities as a state-of-the-art, high-throughput alternative to empirical testing. Additionally, it includes a provision for the application of expert knowledge to increase prediction confidence and resolve conflicting calls. Expert knowledge, which considers structural analogs and mechanisms of activity, has been valuable when models return an indeterminate (equivocal) result or no prediction (out-of-domain). A retrospective analysis of 1002 impurities evaluated in drug regulatory applications between April 2017 and March 2019 assessed the impact of expert review on (Q)SAR predictions. Expert knowledge overturned the default predictions for 26% of the impurities and resolved 91% of equivocal predictions and 75% of out-of-domain calls. Of the 261 overturned default predictions, 15% were upgraded to equivocal or positive and 79% were downgraded to equivocal or negative. Chemical classes with the most overturns were primary aromatic amines (46%), aldehydes (45%), Michael-reactive acceptors (37%), and non-primary alkyl halides (33%). Additionally, low confidence predictions were the most often overturned. Collectively, the results suggest that expert knowledge continues to play an important role in an ICH M7 (Q)SAR prediction workflow and triaging predictions based on chemical class and probability can improve (Q)SAR review efficiency.


Drug Contamination , Mutagens/chemistry , Quantitative Structure-Activity Relationship , Computer Simulation , Mutagenicity Tests , Retrospective Studies , Risk Assessment
3.
Comput Toxicol ; 202021 Nov.
Article En | MEDLINE | ID: mdl-35340402

Hepatotoxicity is one of the most frequently observed adverse effects resulting from exposure to a xenobiotic. For example, in pharmaceutical research and development it is one of the major reasons for drug withdrawals, clinical failures, and discontinuation of drug candidates. The development of faster and cheaper methods to assess hepatotoxicity that are both more sustainable and more informative is critically needed. The biological mechanisms and processes underpinning hepatotoxicity are summarized and experimental approaches to support the prediction of hepatotoxicity are described, including toxicokinetic considerations. The paper describes the increasingly important role of in silico approaches and highlights challenges to the adoption of these methods including the lack of a commonly agreed upon protocol for performing such an assessment and the need for in silico solutions that take dose into consideration. A proposed framework for the integration of in silico and experimental information is provided along with a case study describing how computational methods have been used to successfully respond to a regulatory question concerning non-genotoxic impurities in chemically synthesized pharmaceuticals.

4.
PLoS One ; 15(3): e0229646, 2020.
Article En | MEDLINE | ID: mdl-32126112

Kratom is a botanical substance that is marketed and promoted in the US for pharmaceutical opioid indications despite having no US Food and Drug Administration approved uses. Kratom contains over forty alkaloids including two partial agonists at the mu opioid receptor, mitragynine and 7-hydroxymitragynine, that have been subjected to the FDA's scientific and medical evaluation. However, pharmacological and toxicological data for the remaining alkaloids are limited. Therefore, we applied the Public Health Assessment via Structural Evaluation (PHASE) protocol to generate in silico binding profiles for 25 kratom alkaloids to facilitate the risk evaluation of kratom. PHASE demonstrates that kratom alkaloids share structural features with controlled opioids, indicates that several alkaloids bind to the opioid, adrenergic, and serotonin receptors, and suggests that mitragynine and 7-hydroxymitragynine are the strongest binders at the mu opioid receptor. Subsequently, the in silico binding profiles of a subset of the alkaloids were experimentally verified at the opioid, adrenergic, and serotonin receptors using radioligand binding assays. The verified binding profiles demonstrate the ability of PHASE to identify potential safety signals and provide a tool for prioritizing experimental evaluation of high-risk compounds.


Mitragyna/chemistry , Plants, Medicinal/chemistry , Secologanin Tryptamine Alkaloids/chemistry , Animals , Binding Sites , HEK293 Cells , Humans , In Vitro Techniques , Molecular Docking Simulation , Radioligand Assay , Receptors, Adrenergic/drug effects , Receptors, Adrenergic/metabolism , Receptors, Opioid/drug effects , Receptors, Opioid/metabolism , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism , Receptors, Serotonin/drug effects , Receptors, Serotonin/metabolism , Secologanin Tryptamine Alkaloids/pharmacokinetics , Secologanin Tryptamine Alkaloids/pharmacology , Structure-Activity Relationship
5.
Regul Toxicol Pharmacol ; 109: 104488, 2019 Dec.
Article En | MEDLINE | ID: mdl-31586682

The International Council on Harmonisation (ICH) M7(R1) guideline describes the use of complementary (quantitative) structure-activity relationship ((Q)SAR) models to assess the mutagenic potential of drug impurities in new and generic drugs. Historically, the CASE Ultra and Leadscope software platforms used two different statistical-based models to predict mutations at G-C (guanine-cytosine) and A-T (adenine-thymine) sites, to comprehensively assess bacterial mutagenesis. In the present study, composite bacterial mutagenicity models covering multiple mutation types were developed. These new models contain more than double the number of chemicals (n = 9,254 and n = 13,514) than the corresponding non-composite models and show better toxicophore coverage. Additionally, the use of a single composite bacterial mutagenicity model simplifies impurity analysis in an ICH M7 (Q)SAR workflow by reducing the number of model outputs requiring review. An external validation set of 388 drug impurities representing proprietary pharmaceutical chemical space showed performance statistics ranging from of 66-82% in sensitivity, 91-95% in negative predictivity and 96% in coverage. This effort represents a major enhancement to these (Q)SAR models and their use under ICH M7(R1), leading to improved patient safety through greater predictive accuracy, applicability, and efficiency when assessing the bacterial mutagenic potential of drug impurities.


Drug Contamination/prevention & control , Mutagenesis/drug effects , Mutagenicity Tests/standards , Mutagens/toxicity , Quantitative Structure-Activity Relationship , Bacteria/drug effects , Bacteria/genetics , Computer Simulation/standards , Data Accuracy , Data Analysis , Databases, Factual , Datasets as Topic , Humans , Mutagenicity Tests/methods , Mutagens/chemistry , Patient Safety , Research Design , Toxicology/methods , Toxicology/standards , Workflow
6.
Clin Pharmacol Ther ; 106(1): 116-122, 2019 07.
Article En | MEDLINE | ID: mdl-30957872

The US Food and Drug Administration's Center for Drug Evaluation and Research (CDER) developed an investigational Public Health Assessment via Structural Evaluation (PHASE) methodology to provide a structure-based evaluation of a newly identified opioid's risk to public safety. PHASE utilizes molecular structure to predict biological function. First, a similarity metric quantifies the structural similarity of a new drug relative to drugs currently controlled in the Controlled Substances Act (CSA). Next, software predictions provide the primary and secondary biological targets of the new drug. Finally, molecular docking estimates the binding affinity at the identified biological targets. The multicomponent computational approach coupled with expert review provides a rapid, systematic evaluation of a new drug in the absence of in vitro or in vivo data. The information provided by PHASE has the potential to inform law enforcement agencies with vital information regarding newly emerging illicit opioids.


Analgesics, Opioid/chemistry , Controlled Substances/chemistry , Drug and Narcotic Control/organization & administration , Molecular Docking Simulation/methods , United States Food and Drug Administration/organization & administration , Computer Simulation , Drug Design , Fentanyl/chemistry , Humans , Public Health , Structure-Activity Relationship , United States
7.
PLoS One ; 13(5): e0197734, 2018.
Article En | MEDLINE | ID: mdl-29795628

Opioids represent a highly-abused and highly potent class of drugs that have become a significant threat to public safety. Often there are little to no pharmacological and toxicological data available for new, illicitly used and abused opioids, and this has resulted in a growing number of serious adverse events, including death. The large influx of new synthetic opioids permeating the street-drug market, including fentanyl and fentanyl analogs, has generated the need for a fast and effective method to evaluate the risk a substance poses to public safety. In response, the US FDA's Center for Drug Evaluation and Research (CDER) has developed a rapidly-deployable, multi-pronged computational approach to assess a drug's risk to public health. A key component of this approach is a molecular docking model to predict the binding affinity of biologically uncharacterized fentanyl analogs to the mu opioid receptor. The model was validated by correlating the docking scores of structurally diverse opioids with experimentally determined binding affinities. Fentanyl derivatives with sub-nanomolar binding affinity at the mu receptor (e.g. carfentanil and lofentanil) have significantly lower binding scores, while less potent fentanyl derivatives have increased binding scores. The strong correlation between the binding scores and the experimental binding affinities suggests that this approach can be used to accurately predict the binding strength of newly identified fentanyl analogs at the mu receptor in the absence of in vitro data and may assist in the temporary scheduling of those substances that pose a risk to public safety.


Fentanyl/metabolism , Molecular Docking Simulation , Receptors, Opioid, mu/metabolism , Binding Sites , Fentanyl/analogs & derivatives , Fentanyl/chemistry , Humans , Kinetics , Protein Binding , Protein Structure, Tertiary , Receptors, Opioid, mu/chemistry , Thermodynamics
8.
Regul Toxicol Pharmacol ; 96: 1-17, 2018 Jul.
Article En | MEDLINE | ID: mdl-29678766

The present publication surveys several applications of in silico (i.e., computational) toxicology approaches across different industries and institutions. It highlights the need to develop standardized protocols when conducting toxicity-related predictions. This contribution articulates the information needed for protocols to support in silico predictions for major toxicological endpoints of concern (e.g., genetic toxicity, carcinogenicity, acute toxicity, reproductive toxicity, developmental toxicity) across several industries and regulatory bodies. Such novel in silico toxicology (IST) protocols, when fully developed and implemented, will ensure in silico toxicological assessments are performed and evaluated in a consistent, reproducible, and well-documented manner across industries and regulatory bodies to support wider uptake and acceptance of the approaches. The development of IST protocols is an initiative developed through a collaboration among an international consortium to reflect the state-of-the-art in in silico toxicology for hazard identification and characterization. A general outline for describing the development of such protocols is included and it is based on in silico predictions and/or available experimental data for a defined series of relevant toxicological effects or mechanisms. The publication presents a novel approach for determining the reliability of in silico predictions alongside experimental data. In addition, we discuss how to determine the level of confidence in the assessment based on the relevance and reliability of the information.


Computer Simulation , Toxicity Tests/methods , Toxicology/methods , Animals , Humans
9.
Bioinformatics ; 33(3): 464-466, 2017 02 01.
Article En | MEDLINE | ID: mdl-28172359

Summary: We have developed a public Chemical In vitro­In vivo Profiling (CIIPro) portal, which can automatically extract in vitro biological data from public resources (i.e. PubChem) for user-supplied compounds. For compounds with in vivo target activity data (e.g. animal toxicity testing results), the integrated cheminformatics algorithm will optimize the extracted biological data using in vitro­in vivo correlations. The resulting in vitro biological data for target compounds can be used for read-across risk assessment of target compounds. Additionally, the CIIPro portal can identify the most similar compounds based on their optimized bioprofiles. The CIIPro portal provides new powerful assessment capabilities to the scientific community and can be easily integrated with other cheminformatics tools. Availability and Implementation: ciipro.rutgers.edu. Contact: danrusso@scarletmail.rutgers.edu or hao.zhu99@rutgers.edu


Computational Biology/methods , Software , Toxicology/methods , Animals , Biosimilar Pharmaceuticals , Risk Assessment/methods
10.
Front Environ Sci ; 42016 Mar.
Article En | MEDLINE | ID: mdl-27642585

Estrogen receptors (ERα) are a critical target for drug design as well as a potential source of toxicity when activated unintentionally. Thus, evaluating potential ERα binding agents is critical in both drug discovery and chemical toxicity areas. Using computational tools, e.g., Quantitative Structure-Activity Relationship (QSAR) models, can predict potential ERα binding agents before chemical synthesis. The purpose of this project was to develop enhanced predictive models of ERα binding agents by utilizing advanced cheminformatics tools that can integrate publicly available bioassay data. The initial ERα binding agent data set, consisting of 446 binders and 8307 non-binders, was obtained from the Tox21 Challenge project organized by the NIH Chemical Genomics Center (NCGC). After removing the duplicates and inorganic compounds, this data set was used to create a training set (259 binders and 259 non-binders). This training set was used to develop QSAR models using chemical descriptors. The resulting models were then used to predict the binding activity of 264 external compounds, which were available to us after the models were developed. The cross-validation results of training set [Correct Classification Rate (CCR) = 0.72] were much higher than the external predictivity of the unknown compounds (CCR = 0.59). To improve the conventional QSAR models, all compounds in the training set were used to search PubChem and generate a profile of their biological responses across thousands of bioassays. The most important bioassays were prioritized to generate a similarity index that was used to calculate the biosimilarity score between each two compounds. The nearest neighbors for each compound within the set were then identified and its ERα binding potential was predicted by its nearest neighbors in the training set. The hybrid model performance (CCR = 0.94 for cross validation; CCR = 0.68 for external prediction) showed significant improvement over the original QSAR models, particularly for the activity cliffs that induce prediction errors. The results of this study indicate that the response profile of chemicals from public data provides useful information for modeling and evaluation purposes. The public big data resources should be considered along with chemical structure information when predicting new compounds, such as unknown ERα binding agents.

11.
Methods Mol Biol ; 1473: 161-72, 2016.
Article En | MEDLINE | ID: mdl-27518634

Publicly available bioassay data often contains errors. Curating massive bioassay data, especially high-throughput screening (HTS) data, for Quantitative Structure-Activity Relationship (QSAR) modeling requires the assistance of automated data curation tools. Using automated data curation tools are beneficial to users, especially ones without prior computer skills, because many platforms have been developed and optimized based on standardized requirements. As a result, the users do not need to extensively configure the curation tool prior to the application procedure. In this chapter, a freely available automatic tool to curate and prepare HTS data for QSAR modeling purposes will be described.


Algorithms , Data Curation/methods , High-Throughput Screening Assays/standards , Models, Statistical , Software , Xenobiotics/toxicity , Artifacts , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Quantitative Structure-Activity Relationship , Reproducibility of Results
12.
Pharm Res ; 32(9): 3055-65, 2015 Sep.
Article En | MEDLINE | ID: mdl-25862462

PURPOSE: Experimental Blood-Brain Barrier (BBB) permeability models for drug molecules are expensive and time-consuming. As alternative methods, several traditional Quantitative Structure-Activity Relationship (QSAR) models have been developed previously. In this study, we aimed to improve the predictivity of traditional QSAR BBB permeability models by employing relevant public bio-assay data in the modeling process. METHODS: We compiled a BBB permeability database consisting of 439 unique compounds from various resources. The database was split into a modeling set of 341 compounds and a validation set of 98 compounds. Consensus QSAR modeling workflow was employed on the modeling set to develop various QSAR models. A five-fold cross-validation approach was used to validate the developed models, and the resulting models were used to predict the external validation set compounds. Furthermore, we used previously published membrane transporter models to generate relevant transporter profiles for target compounds. The transporter profiles were used as additional biological descriptors to develop hybrid QSAR BBB models. RESULTS: The consensus QSAR models have R(2) = 0.638 for five-fold cross-validation and R(2) = 0.504 for external validation. The consensus model developed by pooling chemical and transporter descriptors showed better predictivity (R(2) = 0.646 for five-fold cross-validation and R(2) = 0.526 for external validation). Moreover, several external bio-assays that correlate with BBB permeability were identified using our automatic profiling tool. CONCLUSIONS: The BBB permeability models developed in this study can be useful for early evaluation of new compounds (e.g., new drug candidates). The combination of chemical and biological descriptors shows a promising direction to improve the current traditional QSAR models.


Blood-Brain Barrier/metabolism , Models, Theoretical , Quantitative Structure-Activity Relationship , Biological Assay/methods , Biological Transport/physiology , Databases, Factual , Humans , Permeability
13.
Chem Res Toxicol ; 27(10): 1643-51, 2014 Oct 20.
Article En | MEDLINE | ID: mdl-25195622

High-throughput screening (HTS) assays that measure the in vitro toxicity of environmental compounds have been widely applied as an alternative to in vivo animal tests of chemical toxicity. Current HTS studies provide the community with rich toxicology information that has the potential to be integrated into toxicity research. The available in vitro toxicity data is updated daily in structured formats (e.g., deposited into PubChem and other data-sharing web portals) or in an unstructured way (papers, laboratory reports, toxicity Web site updates, etc.). The information derived from the current toxicity data is so large and complex that it becomes difficult to process using available database management tools or traditional data processing applications. For this reason, it is necessary to develop a big data approach when conducting modern chemical toxicity research. In vitro data for a compound, obtained from meaningful bioassays, can be viewed as a response profile that gives detailed information about the compound's ability to affect relevant biological proteins/receptors. This information is critical for the evaluation of complex bioactivities (e.g., animal toxicities) and grows rapidly as big data in toxicology communities. This review focuses mainly on the existing structured in vitro data (e.g., PubChem data sets) as response profiles for compounds of environmental interest (e.g., potential human/animal toxicants). Potential modeling and mining tools to use the current big data pool in chemical toxicity research are also described.


High-Throughput Screening Assays , Inorganic Chemicals/analysis , Organic Chemicals/analysis , Toxicity Tests , Animals , Biological Assay , Databases, Factual , Humans , Toxicogenetics
14.
J Comput Aided Mol Des ; 28(6): 631-46, 2014 Jun.
Article En | MEDLINE | ID: mdl-24840854

Compared to the current knowledge on cancer chemotherapeutic agents, only limited information is available on the ability of organic compounds, such as drugs and/or natural products, to prevent or delay the onset of cancer. In order to evaluate chemical chemopreventive potentials and design novel chemopreventive agents with low to no toxicity, we developed predictive computational models for chemopreventive agents in this study. First, we curated a database containing over 400 organic compounds with known chemoprevention activities. Based on this database, various random forest and support vector machine binary classifiers were developed. All of the resulting models were validated by cross validation procedures. Then, the validated models were applied to virtually screen a chemical library containing around 23,000 natural products and derivatives. We selected a list of 148 novel chemopreventive compounds based on the consensus prediction of all validated models. We further analyzed the predicted active compounds by their ease of organic synthesis. Finally, 18 compounds were synthesized and experimentally validated for their chemopreventive activity. The experimental validation results paralleled the cross validation results, demonstrating the utility of the developed models. The predictive models developed in this study can be applied to virtually screen other chemical libraries to identify novel lead compounds for the chemoprevention of cancers.


Anticarcinogenic Agents/chemistry , Anticarcinogenic Agents/pharmacology , Drug Design , Support Vector Machine , Anticarcinogenic Agents/chemical synthesis , Biological Products/chemical synthesis , Biological Products/chemistry , Biological Products/pharmacology , Computer-Aided Design , Databases, Pharmaceutical , Humans , Models, Biological , Neoplasms/drug therapy , Quantitative Structure-Activity Relationship
15.
Pharm Res ; 31(4): 1002-14, 2014 Apr.
Article En | MEDLINE | ID: mdl-24306326

PURPOSE: Oral bioavailability (%F) is a key factor that determines the fate of a new drug in clinical trials. Traditionally, %F is measured using costly and time-consuming experimental tests. Developing computational models to evaluate the %F of new drugs before they are synthesized would be beneficial in the drug discovery process. METHODS: We employed Combinatorial Quantitative Structure-Activity Relationship approach to develop several computational %F models. We compiled a %F dataset of 995 drugs from public sources. After generating chemical descriptors for each compound, we used random forest, support vector machine, k nearest neighbor, and CASE Ultra to develop the relevant QSAR models. The resulting models were validated using five-fold cross-validation. RESULTS: The external predictivity of %F values was poor (R(2) = 0.28, n = 995, MAE = 24), but was improved (R(2) = 0.40, n = 362, MAE = 21) by filtering unreliable predictions that had a high probability of interacting with MDR1 and MRP2 transporters. Furthermore, classifying the compounds according to the %F values (%F < 50% as "low", %F ≥ 50% as 'high") and developing category QSAR models resulted in an external accuracy of 76%. CONCLUSIONS: In this study, we developed predictive %F QSAR models that could be used to evaluate new drug compounds, and integrating drug-transporter interactions data greatly benefits the resulting models.


Chemistry, Pharmaceutical/standards , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry , Quantitative Structure-Activity Relationship , Administration, Oral , Biological Availability , Chemistry, Pharmaceutical/methods , Databases, Factual , Humans
...