The effect of food on pharmacokinetic properties of drugs is a commonly observed occurrence affecting about 40% of orally administered drugs. Within the pharmaceutical industry, significant resources are invested to predict and characterize a clinically relevant food effect. Here, the predictive performance of physiologically based pharmacokinetic (PBPK) food effect models was assessed via de novo mechanistic absorption models for 30 compounds using controlled, pre-defined in vitro, and modeling methodology. Compounds for which absorption was known to be limited by intestinal transporters were excluded in this analysis. A decision tree for model verification and optimization was followed, leading to high, moderate, or low food effect prediction confidence. High (within 0.8- to 1.25-fold) to moderate confidence (within 0.5- to 2-fold) was achieved for most of the compounds (15 and 8, respectively). While for 7 compounds, prediction confidence was found to be low (> 2-fold). There was no clear difference in prediction success for positive or negative food effects and no clear relationship to the BCS category of tested drug molecules. However, an association could be demonstrated when the food effect was mainly related to changes in the gastrointestinal luminal fluids or physiology, including fluid volume, motility, pH, micellar entrapment, and bile salts. Considering these findings, it is recommended that appropriately verified mechanistic PBPK modeling can be leveraged with high to moderate confidence as a key approach to predicting potential food effect, especially related to mechanisms highlighted here.
Minimizing in vitro and in vivo testing
in early drug discovery
with the use of physiologically based pharmacokinetic (PBPK) modeling
and machine learning (ML) approaches has the potential to reduce discovery
cycle times and animal experimentation. However, the prediction success
of such an approach has not been shown for a larger and diverse set
of compounds representative of a lead optimization pipeline. In this
study, the prediction success of the oral (PO) and intravenous (IV)
pharmacokinetics (PK) parameters in rats was assessed using a “bottom-up”
approach, combining in vitro and ML inputs with a PBPK model. More
than 240 compounds for which all of the necessary inputs and PK data
were available were used for this assessment. Different clearance
scaling approaches were assessed, using hepatocyte intrinsic clearance
and protein binding as inputs. In addition, a novel high-throughput
PBPK (HT-PBPK) approach was evaluated to assess the scalability of
PBPK predictions for a larger number of compounds in drug discovery.
The results showed that bottom-up PBPK modeling was able to predict
the rat IV and PO PK parameters for the majority of compounds within
a 2- to 3-fold error range, using both direct scaling and dilution
methods for clearance predictions. The use of only ML-predicted inputs
from the structure did not perform well when using in vitro inputs,
likely due to clearance miss predictions. The HT-PBPK approach produced
comparable results to the full PBPK modeling approach but reduced
the simulation time from hours to seconds. In conclusion, a bottom-up
PBPK and HT-PBPK approach can successfully predict the PK parameters
and guide early discovery by informing compound prioritization, provided
that good in vitro assays are in place for key parameters such as
clearance.
Controlled release (CR) formulations are usually designed to achieve similar exposure (AUC) levels as the marketed immediate release (IR) formulation. However, the AUC is often lower following CR compared to IR formulations. There are a few exceptions when the CR formulations have shown higher AUC. This study investigated the impact of CR formulations on oral drug absorption and CYP3A4-mediated gut wall metabolism. A review of the current literature on relative bioavailability (Frel) between CR and IR formulations of CYP3A substrates was conducted. This was followed by a systematic analysis to assess the impact of the release characteristics and the drug-specific factors (including metabolism and permeability) on oral bioavailability employing a physiologically-based pharmacokinetic (PBPK) modelling and simulation approach. From the literature review, only three CYP3A4 substrates showed higher Frel when formulated as CR. Several scenarios were investigated using the PBPK approach; in most of them, the oral absorption of CR formulations was lower as compared to the IR formulations. However, for highly permeable compounds that were CYP3A4 substrates the reduction in absorption was compensated by an increase in the fraction that escapes from first pass metabolism in the gut wall (FG), where the magnitude was dependent on CYP3A4 affinity. The systematic simulations of various interplays between different parameters demonstrated that BCS class 1 highly-cleared CYP3A4 substrates can display up to 220% higher relative bioavailability when formulated as CR compared to IR, in agreement with the observed data collected from the literature. The results and methodology of this study can be employed during the formulation development process in order to optimize drug absorption, especially for CYP3A4 substrates.
scite is a Brooklyn-based organization that helps researchers better discover and understand research articles through Smart Citations–citations that display the context of the citation and describe whether the article provides supporting or contrasting evidence. scite is used by students and researchers from around the world and is funded in part by the National Science Foundation and the National Institute on Drug Abuse of the National Institutes of Health.