Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
PLoS One ; 14(2): e0209587, 2019.
Article in English | MEDLINE | ID: mdl-30759154

ABSTRACT

The therapeutic effect of a drug is governed by its pharmacokinetics which determine the downstream pharmacodynamic response within the cellular network. A complete understanding of the drug-effect relationship therefore requires multi-scale models which integrate the properties of the different physiological scales. Computational modelling of these individual scales has been successfully established in the past. However, coupling of the scales remains challenging, although it will provide a unique possibility of mechanistic and holistic analyses of therapeutic outcomes for varied treatment scenarios. We present a methodology to combine whole-body physiologically-based pharmacokinetic (PBPK) models with mechanistic intracellular models of signal transduction in the liver for therapeutic proteins. To this end, we developed a whole-body distribution model of IFN-α in human and a detailed intracellular model of the JAK/STAT signalling cascade in hepatocytes and coupled them at the liver of the whole-body human model. This integrated model infers the time-resolved concentration of IFN-α arriving at the liver after intravenous injection while simultaneously estimates the effect of this dose on the intracellular signalling behaviour in the liver. In our multi-scale physiologically-based pharmacokinetic/pharmacodynamic (PBPK/PD) model, receptor saturation is seen at low doses, thus giving mechanistic insights into the pharmacodynamic (PD) response. This model suggests a fourfold lower intracellular response after administration of a typical IFN-α dose to an individual as compared to the experimentally observed responses in in vitro setups. In conclusion, this work highlights clear differences between the observed in vitro and in vivo drug effects and provides important suggestions for future model-based study design.


Subject(s)
Immunologic Factors/pharmacology , Interferon-alpha/pharmacology , Cells, Cultured , Computer Simulation , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Immunologic Factors/pharmacokinetics , Interferon-alpha/pharmacokinetics , Janus Kinases/metabolism , Liver/drug effects , Liver/metabolism , Models, Biological , STAT Transcription Factors/metabolism , Signal Transduction
3.
Annu Int Conf IEEE Eng Med Biol Soc ; 2016: 1417-1421, 2016 Aug.
Article in English | MEDLINE | ID: mdl-28268592

ABSTRACT

A multiscale model for blood glucose regulation in diabetes type I patients is constructed by integrating detailed metabolic network models for fat, liver and muscle cells into a whole body physiologically-based pharmacokinetic/pharmacodynamic (pBPK/PD) model. The blood glucose regulation PBPK/PD model simulates the distribution and metabolization of glucose, insulin and glucagon on an organ and whole body level. The genome-scale metabolic networks in contrast describe intracellular reactions. The developed multiscale model is fitted to insulin, glucagon and glucose measurements of a 48h clinical trial featuring 6 subjects and is subsequently used to simulate (in silico) the influence of geneknockouts and drug-induced enzyme inhibitions on whole body blood glucose levels. Simulations of diabetes associated gene knockouts and impaired cellular glucose metabolism, resulted in elevated whole body blood-glucose levels, but also in a metabolic shift within the cell's reaction network. Such multiscale models have the potential to be employed in the exploration of novel drug-targets or to be integrated into control algorithms for artificial pancreas systems.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 1/blood , Insulin/blood , Carbohydrate Metabolism , Glucagon , Glucose/metabolism , Hepatocytes/metabolism , Humans , Liver/metabolism , Metabolic Networks and Pathways , Models, Biological , Muscles/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL