Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Clin Pharmacol Ther ; 110(5): 1293-1301, 2021 11.
Article in English | MEDLINE | ID: mdl-34462909

ABSTRACT

We present a generic workflow combining physiology-based computational modeling and in vitro data to assess the clinical cholestatic risk of different drugs systematically. Changes in expression levels of genes involved in the enterohepatic circulation of bile acids were obtained from an in vitro assay mimicking 14 days of repeated drug administration for 10 marketed drugs. These changes in gene expression over time were contextualized in a physiology-based bile acid model of glycochenodeoxycholic acid. The simulated drug-induced response in bile acid concentrations was then scaled with the applied drug doses to calculate the cholestatic potential for each compound. A ranking of the cholestatic potential correlated very well with the clinical cholestasis risk obtained from medical literature. The proposed workflow allows benchmarking the cholestatic risk of novel drug candidates. We expect the application of our workflow to significantly contribute to the stratification of the cholestatic potential of new drugs and to support animal-free testing in future drug development.


Subject(s)
Benchmarking/methods , Cholestasis/chemically induced , Cholestasis/metabolism , Drug-Related Side Effects and Adverse Reactions/metabolism , Models, Biological , Workflow , Adult , Animals , Cholestasis/diagnosis , Drug-Related Side Effects and Adverse Reactions/diagnosis , Female , Humans , Liver/drug effects , Liver/metabolism , Male , Middle Aged , Pharmaceutical Preparations , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Young Adult
2.
Commun Biol ; 3(1): 573, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33060801

ABSTRACT

Uncovering cellular responses from heterogeneous genomic data is crucial for molecular medicine in particular for drug safety. This can be realized by integrating the molecular activities in networks of interacting proteins. As proof-of-concept we challenge network modeling with time-resolved proteome, transcriptome and methylome measurements in iPSC-derived human 3D cardiac microtissues to elucidate adverse mechanisms of anthracycline cardiotoxicity measured with four different drugs (doxorubicin, epirubicin, idarubicin and daunorubicin). Dynamic molecular analysis at in vivo drug exposure levels reveal a network of 175 disease-associated proteins and identify common modules of anthracycline cardiotoxicity in vitro, related to mitochondrial and sarcomere function as well as remodeling of extracellular matrix. These in vitro-identified modules are transferable and are evaluated with biopsies of cardiomyopathy patients. This to our knowledge most comprehensive study on anthracycline cardiotoxicity demonstrates a reproducible workflow for molecular medicine and serves as a template for detecting adverse drug responses from complex omics data.


Subject(s)
Metabolome , Models, Biological , Proteome , Transcriptome , Epigenesis, Genetic , Gene Expression Profiling/methods , Gene Expression Regulation , Gene Regulatory Networks , Humans , Metabolomics/methods , Mitochondria/genetics , Mitochondria/metabolism , Proteomics/methods , Sarcomeres/genetics , Sarcomeres/metabolism , Signal Transduction
3.
SLAS Discov ; 25(3): 265-276, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31658853

ABSTRACT

Three-dimensional (3D) cell culture models are thought to mimic the physiological and pharmacological properties of tissues in vivo more accurately than two-dimensional cultures on plastic dishes. For the development of cancer therapies, 3D spheroid models are being created to reflect the complex histology and physiology of primary tumors with the hopes that drug responses will be more similar to and as predictive as those obtained in vivo. The effect of additional cell types in tumors, such as stromal cells, and the resulting heterotypic cell-cell crosstalk can be investigated in these heterotypic 3D cell cultures. Here, a high-throughput screening-compatible drug testing platform based on 3D multicellular spheroid models is described that enables the parallel assessment of toxicity on stromal cells and efficacy on cancer cells by drug candidates. These heterotypic microtissue tumor models incorporate NIH3T3 fibroblasts as stromal cells that are engineered with a reporter gene encoding secreted NanoLUC luciferase. By tracking the NanoLUC signal in the media over time, a time-related measurement of the cytotoxic effects of drugs on stromal cells over the cancer cells was possible, thus enabling the identification of a therapeutic window. An in vitro therapeutic index parameter is proposed to help distinguish and classify those compounds with broad cytotoxic effects versus those that are more selective at targeting cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Spheroids, Cellular/drug effects , Animals , Cell Line, Tumor , Cell Survival/drug effects , Humans , Mice , NIH 3T3 Cells/drug effects , Neoplasms/drug therapy , Spheroids, Cellular/pathology , Stromal Cells/drug effects , Tumor Microenvironment/drug effects
4.
Front Physiol ; 10: 1192, 2019.
Article in English | MEDLINE | ID: mdl-31611804

ABSTRACT

Drug-induced liver injury (DILI) is a matter of concern in the course of drug development and patient safety, often leading to discontinuation of drug-development programs or early withdrawal of drugs from market. Hepatocellular toxicity or impairment of bile acid (BA) metabolism, known as cholestasis, are the two clinical forms of DILI. Whole-body physiology-based modelling allows a mechanistic investigation of the physiological processes leading to cholestasis in man. Objectives of the present study were: (1) the development of a physiology-based model of the human BA metabolism, (2) population-based model validation and characterisation, and (3) the prediction and quantification of altered BA levels in special genotype subgroups and after drug administration. The developed physiology-based bile acid (PBBA) model describes the systemic BA circulation in humans and includes mechanistically relevant active and passive processes such as the hepatic synthesis, gallbladder emptying, transition through the gastrointestinal tract, reabsorption into the liver, distribution within the whole body, and excretion via urine and faeces. The kinetics of active processes were determined for the exemplary BA glycochenodeoxycholic acid (GCDCA) based on blood plasma concentration-time profiles. The robustness of our PBBA model was verified with population simulations of healthy individuals. In addition to plasma levels, the possibility to estimate BA concentrations in relevant tissues like the intracellular space of the liver enhance the mechanistic understanding of cholestasis. We analysed BA levels in various tissues of Benign Recurrent Intrahepatic Cholestasis type 2 (BRIC2) patients and our simulations suggest a higher susceptibility of BRIC2 patients toward cholestatic DILI due to BA accumulation in the liver. The effect of drugs on systemic BA levels were simulated for cyclosporine A (CsA). Our results confirmed the higher risk of DILI after CsA administration in healthy and BRIC2 patients. The presented PBBA model enhances our mechanistic understanding underlying cholestasis and drug-induced alterations of BA levels in blood and organs. The developed PBBA model might be applied in the future to anticipate potential risk of cholestasis in patients.

5.
NPJ Syst Biol Appl ; 4: 28, 2018.
Article in English | MEDLINE | ID: mdl-30083389

ABSTRACT

A quantitative analysis of dose-response relationships is essential in preclinical and clinical drug development in order to optimize drug efficacy and safety, respectively. However, there is a lack of quantitative understanding about the dynamics of pharmacological drug-target interactions in biological systems. In this study, a quantitative systems pharmacology (QSP) approach is applied to quantify the drug efficacy of cyclooxygenase-2 (COX-2) and 5-lipoxygenase (5-LOX) inhibitors by coupling physiologically based pharmacokinetic models, at the whole-body level, with affected biological networks, at the cellular scale. Both COX-2 and 5-LOX are key enzymes in the production of inflammatory mediators and are known targets in the design of anti-inflammatory drugs. Drug efficacy is here evaluated for single and appropriate co-treatment of diclofenac, celecoxib, zileuton, and licofelone by quantitatively studying the reduction of prostaglandins and leukotrienes. The impact of rifampicin pre-treatment on prostaglandin formation is also investigated by considering pharmacokinetic drug interactions with diclofenac and celecoxib, finally suggesting optimized dose levels to compensate for the reduced drug action. Furthermore, a strong correlation was found between pain relief observed in patients as well as celecoxib- and diclofenac-induced decrease in prostaglandins after 6 h. The findings presented reveal insights about drug-induced modulation of cellular networks in a whole-body context, thereby describing complex pharmacokinetic/pharmacodynamic behavior of COX-2 and 5-LOX inhibitors in therapeutic situations. The results demonstrate the clinical benefit of using QSP to predict drug efficacy and, hence, encourage its use in future drug discovery and development programs.

6.
NPJ Syst Biol Appl ; 4: 10, 2018.
Article in English | MEDLINE | ID: mdl-29507756

ABSTRACT

Drug-induced perturbations of the endogenous metabolic network are a potential root cause of cellular toxicity. A mechanistic understanding of such unwanted side effects during drug therapy is therefore vital for patient safety. The comprehensive assessment of such drug-induced injuries requires the simultaneous consideration of both drug exposure at the whole-body and resulting biochemical responses at the cellular level. We here present a computational multi-scale workflow that combines whole-body physiologically based pharmacokinetic (PBPK) models and organ-specific genome-scale metabolic network (GSMN) models through shared reactions of the xenobiotic metabolism. The applicability of the proposed workflow is illustrated for isoniazid, a first-line antibacterial agent against Mycobacterium tuberculosis, which is known to cause idiosyncratic drug-induced liver injuries (DILI). We combined GSMN models of a human liver with N-acetyl transferase 2 (NAT2)-phenotype-specific PBPK models of isoniazid. The combined PBPK-GSMN models quantitatively describe isoniazid pharmacokinetics, as well as intracellular responses, and changes in the exometabolome in a human liver following isoniazid administration. Notably, intracellular and extracellular responses identified with the PBPK-GSMN models are in line with experimental and clinical findings. Moreover, the drug-induced metabolic perturbations are distributed and attenuated in the metabolic network in a phenotype-dependent manner. Our simulation results show that a simultaneous consideration of both drug pharmacokinetics at the whole-body and metabolism at the cellular level is mandatory to explain drug-induced injuries at the patient level. The proposed workflow extends our mechanistic understanding of the biochemistry underlying adverse events and may be used to prevent drug-induced injuries in the future.

8.
PLoS Comput Biol ; 13(2): e1005280, 2017 02.
Article in English | MEDLINE | ID: mdl-28151932

ABSTRACT

Drug-induced toxicity is a significant problem in clinical care. A key problem here is a general understanding of the molecular mechanisms accompanying the transition from desired drug effects to adverse events following administration of either therapeutic or toxic doses, in particular within a patient context. Here, a comparative toxicity analysis was performed for fifteen hepatotoxic drugs by evaluating toxic changes reflecting the transition from therapeutic drug responses to toxic reactions at the cellular level. By use of physiologically-based pharmacokinetic modeling, in vitro toxicity data were first contextualized to quantitatively describe time-resolved drug responses within a patient context. Comparatively studying toxic changes across the considered hepatotoxicants allowed the identification of subsets of drugs sharing similar perturbations on key cellular processes, functional classes of genes, and individual genes. The identified subsets of drugs were next analyzed with regard to drug-related characteristics and their physicochemical properties. Toxic changes were finally evaluated to predict both molecular biomarkers and potential drug-drug interactions. The results may facilitate the early diagnosis of adverse drug events in clinical application.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Liver/drug effects , Liver/metabolism , Models, Biological , Pharmacokinetics , Signal Transduction/drug effects , Chemical and Drug Induced Liver Injury/etiology , Computer Simulation , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Metabolic Clearance Rate
9.
Arch Toxicol ; 91(2): 865-883, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27161439

ABSTRACT

Understanding central mechanisms underlying drug-induced toxicity plays a crucial role in drug development and drug safety. However, a translation of cellular in vitro findings to an actual in vivo context remains challenging. Here, physiologically based pharmacokinetic (PBPK) modeling was used for in vivo contextualization of in vitro toxicity data (PICD) to quantitatively predict in vivo drug response over time by integrating multiple levels of biological organization. Explicitly, in vitro toxicity data at the cellular level were integrated into whole-body PBPK models at the organism level by coupling in vitro drug exposure with in vivo drug concentration-time profiles simulated in the extracellular environment within the organ. PICD was exemplarily applied on the hepatotoxicant azathioprine to quantitatively predict in vivo drug response of perturbed biological pathways and cellular processes in rats and humans. The predictive accuracy of PICD was assessed by comparing in vivo drug response predicted for rats with observed in vivo measurements. To demonstrate clinical applicability of PICD, in vivo drug responses of a critical toxicity-related pathway were predicted for eight patients following acute azathioprine overdoses. Moreover, acute liver failure after multiple dosing of azathioprine was investigated in a patient case study by use of own clinical data. Simulated pharmacokinetic profiles were therefore related to in vivo drug response predicted for genes associated with observed clinical symptoms and to clinical biomarkers measured in vivo. PICD provides a generic platform to investigate drug-induced toxicity at a patient level and thus may facilitate individualized risk assessment during drug development.


Subject(s)
Azathioprine/toxicity , Drug-Related Side Effects and Adverse Reactions , Models, Theoretical , Pharmacokinetics , Adult , Animals , Azathioprine/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Drug Overdose/etiology , Humans , Male , Rats , Reproducibility of Results , Toxicity Tests/methods , Toxicity Tests, Acute/methods
10.
Antimicrob Agents Chemother ; 60(10): 6134-45, 2016 10.
Article in English | MEDLINE | ID: mdl-27480867

ABSTRACT

Due to its high early bactericidal activity, isoniazid (INH) plays an essential role in tuberculosis treatment. Genetic polymorphisms of N-acetyltransferase type 2 (NAT2) cause a trimodal distribution of INH pharmacokinetics in slow, intermediate, and fast acetylators. The success of INH-based chemotherapy is associated with acetylator and patient health status. Still, a standard dose recommended by the FDA is administered regardless of acetylator type or immune status, even though adverse effects occur in 5 to 33% of all patients. Slow acetylators have a higher risk of development of drug-induced toxicity, while fast acetylators and immune-deficient patients face lower treatment success rates. To mechanistically assess the trade-off between toxicity and efficacy, we developed a physiologically based pharmacokinetic (PBPK) model describing the NAT2-dependent pharmacokinetics of INH and its metabolites. We combined the PBPK model with a pharmacodynamic (PD) model of antimycobacterial drug effects in the lungs. The resulting PBPK/PD model allowed the simultaneous simulation of treatment efficacies at the site of infection and exposure to toxic metabolites in off-target organs. Subsequently, we evaluated various INH dosing regimens in NAT2-specific immunocompetent and immune-deficient virtual populations. Our results suggest the need for acetylator-specific dose adjustments for optimal treatment outcomes. A reduced dose for slow acetylators substantially lowers the exposure to toxic metabolites and thereby the risk of adverse events, while it maintains sufficient treatment efficacies. Vice versa, intermediate and fast acetylators benefit from increased INH doses and a switch to a twice-daily administration schedule. Our analysis outlines how PBPK/PD modeling may be used to design and individualize treatment regimens.


Subject(s)
Antitubercular Agents/pharmacokinetics , Arylamine N-Acetyltransferase/metabolism , Immunocompromised Host , Isoniazid/pharmacokinetics , Models, Statistical , Tuberculosis, Pulmonary/drug therapy , Acetylation , Antitubercular Agents/blood , Arylamine N-Acetyltransferase/genetics , Biological Availability , Biotransformation , Computer Simulation , Drug Administration Schedule , Drug Dosage Calculations , Gene Expression , Genotype , Humans , Immunity, Innate , Isoniazid/blood , Lung/drug effects , Lung/immunology , Lung/microbiology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/immunology , Polymorphism, Genetic , Precision Medicine , Tuberculosis, Pulmonary/blood , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/microbiology
11.
J Pharm Sci ; 104(1): 191-206, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25393841

ABSTRACT

Transfer of knowledge along the different phases of drug development is a fundamental process in pharmaceutical research. In particular, cross-species extrapolation between different laboratory animals and further on to first-in-human trials is challenging because of the uncertain comparability of physiological processes. Physiologically based pharmacokinetic (PBPK) modeling allows translation of mechanistic knowledge from one species to another by specifically considering physiological and biochemical differences in between. We here evaluated different knowledge-driven approaches for cross-species extrapolation by systematically incorporating specific model parameter domains of a target species into the PBPK model of a reference species. Altogether, 15 knowledge-driven approaches were applied to murine and human PBPK models of 10 exemplary drugs resulting in 300 different extrapolations. Statistical analysis of the quality of the different extrapolations revealed not only species-specific physiology as the key determinant in cross-species extrapolation but also identified a synergistic effect when considering both kinetic rate constants and gene expression profiles of relevant enzymes and transporters. Moreover, we show that considering species-specific physiology, plasma protein binding, enzyme and transport kinetics, as well as tissue-specific gene expression profiles in PBPK modeling increases accuracy of cross-species extrapolations and thus supports first-in-human trials based on prior preclinical knowledge.


Subject(s)
Drug Evaluation, Preclinical/methods , Drugs, Investigational/pharmacokinetics , Gene Expression Regulation/drug effects , Liver/drug effects , Models, Biological , Pharmacology, Clinical/methods , Physiology, Comparative/methods , Animals , Cells, Cultured , Computational Biology , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Drugs, Investigational/metabolism , Drugs, Investigational/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Enzymologic/drug effects , Germany , Humans , Liver/cytology , Liver/enzymology , Liver/metabolism , Mice, Inbred C57BL , Organ Specificity , Species Specificity , Specific Pathogen-Free Organisms
12.
PLoS One ; 8(3): e58583, 2013.
Article in English | MEDLINE | ID: mdl-23505538

ABSTRACT

Small molecules that bind at protein-protein interfaces may either block or stabilize protein-protein interactions in cells. Thus, some of these binding interfaces may turn into prospective targets for drug design. Here, we collected 175 pairs of protein-protein (PP) complexes and protein-ligand (PL) complexes with known three-dimensional structures for which (1) one protein from the PP complex shares at least 40% sequence identity with the protein from the PL complex, and (2) the interface regions of these proteins overlap at least partially with each other. We found that those residues of the interfaces that may bind the other protein as well as the small molecule are evolutionary more conserved on average, have a higher tendency of being located in pockets and expose a smaller fraction of their surface area to the solvent than the remaining protein-protein interface region. Based on these findings we derived a statistical classifier that predicts patches at binding interfaces that have a higher tendency to bind small molecules. We applied this new prediction method to more than 10,000 interfaces from the protein data bank. For several complexes related to apoptosis the predicted binding patches were in direct contact to co-crystallized small molecules.


Subject(s)
Protein Interaction Mapping , Proteins/chemistry , Algorithms , Binding Sites , Databases, Protein , Drug Design , Ligands , Models, Molecular , Molecular Docking Simulation , Protein Binding , Protein Conformation , Protein Interaction Mapping/methods , Proteins/metabolism
13.
Psychiatry Clin Neurosci ; 56(2): 153-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11952918

ABSTRACT

The relationship between criminal behavior on the one hand and endogeneity and anxiety on the other hand was investigated in a sample of patients with unipolar depression to help elucidate factors influencing the criminality rate in this population. A lower criminality rate in patients with higher ratings of endogeneity and anxiety was predicted. Clinical records of 179 male and 99 female psychiatric inpatients were retrospectively evaluated using the Newcastle Scale II and Hamilton Anxiety Scale. A full account of conviction records served as a measure of criminal behavior. Forty per cent of male patients and 7% of female patients were criminally registered. A lower criminality rate was indeed found in male and female patients with endogenous type of depression and in male patients with higher anxiety ratings. In a multivariate evaluation, however, sociodemographic variables in terms of age and social class seem to be more important predictors of criminality and all variables we assessed contributed only marginally to the explanation of the criminality variance. Thus, in patients with unipolar depression, sociodemographic factors seem to be of a greater even though still limited importance regarding criminal behavior compared with the clinical variables of endogeneity and anxiety.


Subject(s)
Anxiety/psychology , Crime/psychology , Depressive Disorder/complications , Adolescent , Adult , Aged , Depressive Disorder/psychology , Female , Humans , Male , Middle Aged , Retrospective Studies , Risk Factors , Sex Factors , Social Class
SELECTION OF CITATIONS
SEARCH DETAIL
...