3 research outputs found

    Physiologically Based Pharmacokinetic Modeling in Lead Optimization. 1. Evaluation and Adaptation of GastroPlus To Predict Bioavailability of Medchem Series

    No full text
    When medicinal chemists need to improve bioavailability (%F) within a chemical series during lead optimization, they synthesize new series members with systematically modified properties mainly by following experience and general rules of thumb. More quantitative models that predict %F of proposed compounds from chemical structure alone have proven elusive. Global empirical %F quantitative structureā€“property (QSPR) models perform poorly, and projects have too little data to train local %F QSPR models. Mechanistic oral absorption and physiologically based pharmacokinetic (PBPK) models simulate the dissolution, absorption, systemic distribution, and clearance of a drug in preclinical species and humans. Attempts to build global PBPK models based purely on calculated inputs have not achieved the <2-fold average error needed to guide lead optimization. In this work, local GastroPlus PBPK models are instead customized for individual medchem series. The key innovation was building a local QSPR for a numerically fitted effective intrinsic clearance (CL<sub>loc</sub>). All inputs are subsequently computed from structure alone, so the models can be applied in advance of synthesis. Training CL<sub>loc</sub> on the first 15ā€“18 rat %F measurements gave adequate predictions, with clear improvements up to about 30 measurements, and incremental improvements beyond that

    Physiologically Based Pharmacokinetic Modeling in Lead Optimization. 2. Rational Bioavailability Design by Global Sensitivity Analysis To Identify Properties Affecting Bioavailability

    No full text
    When medicinal chemists need to improve oral bioavailability (%F) during lead optimization, they systematically modify compound properties mainly based on their own experience and general rules of thumb. However, at least a dozen properties can influence %F, and the difficulty of multiparameter optimization for such complex nonlinear processes grows combinatorially with the number of variables. Furthermore, strategies can be in conflict. For example, adding a polar or charged group will generally increase solubility but decrease permeability. Identifying the 2 or 3 properties that most influence %F for a given compound series would make %F optimization much more efficient. We previously reported an adaptation of physiologically based pharmacokinetic (PBPK) simulations to predict %F for lead series from purely computational inputs within a 2-fold average error. Here, we run thousands of such simulations to generate a comprehensive ā€œbioavailability landscapeā€ for each series. A key innovation was recognition that the large and variable number of p<i>K</i><sub>a</sub>ā€™s in drug molecules could be replaced by just the two straddling the isoelectric point. Another was use of the ZINC database to cull out chemically inaccessible regions of property space. A quadratic partial least squares regression (PLS) accurately fits a continuous surface to these thousands of bioavailability predictions. The PLS coefficients indicate the globally sensitive compound properties. The PLS surface also displays the %F landscape in these sensitive properties locally around compounds of particular interest. Finally, being quick to calculate, the PLS equation can be combined with models for activity and other properties for multiobjective lead optimization

    Best of Both Worlds: Combining Pharma Data and State of the Art Modeling Technology To Improve <i>in Silico</i> p<i>K</i><sub>a</sub> Prediction

    No full text
    In a unique collaboration between a software company and a pharmaceutical company, we were able to develop a new <i>in silico</i> p<i>K</i><sub>a</sub> prediction tool with outstanding prediction quality. An existing p<i>K</i><sub>a</sub> prediction method from Simulations Plus based on artificial neural network ensembles (ANNE), microstates analysis, and literature data was retrained with a large homogeneous data set of drug-like molecules from Bayer. The new model was thus built with curated sets of āˆ¼14,000 literature p<i>K</i><sub>a</sub> values (āˆ¼11,000 compounds, representing literature chemical space) and āˆ¼19,500 p<i>K</i><sub>a</sub> values experimentally determined at Bayer Pharma (āˆ¼16,000 compounds, representing industry chemical space). Model validation was performed with several test sets consisting of a total of āˆ¼31,000 new p<i>K</i><sub>a</sub> values measured at Bayer. For the largest and most difficult test set with >16,000 p<i>K</i><sub>a</sub> values that were not used for training, the original model achieved a mean absolute error (MAE) of 0.72, root-mean-square error (RMSE) of 0.94, and squared correlation coefficient (<i>R</i><sup>2</sup>) of 0.87. The new model achieves significantly improved prediction statistics, with MAE = 0.50, RMSE = 0.67, and <i>R</i><sup>2</sup> = 0.93. It is commercially available as part of the Simulations Plus ADMET Predictor release 7.0. Good predictions are only of value when delivered effectively to those who can use them. The new p<i>K</i><sub>a</sub> prediction model has been integrated into Pipeline Pilot and the PharmacophorInformatics (PIx) platform used by scientists at Bayer Pharma. Different output formats allow customized application by medicinal chemists, physical chemists, and computational chemists
    corecore