27.07.2013 Views

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

The Toxicologist - Society of Toxicology

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

852 UNDERSTANDING STRUCTURAL AND PHYSICAL<br />

CHEMICAL DRIVERS OF DRUG TOXICITY: UTILITY<br />

AND TRANSLATABLE VALUE.<br />

D. P. Hartley 1 and M. Kansy 2 . 1 Genentech, South San Francisco, CA and 2 F.<br />

H<strong>of</strong>fmann-LaRoche AG, Basel, Switzerland.<br />

Given the demands on drug discovery teams to produce effective, specific, and safe<br />

new molecules, toxicologists are now finding themselves with new responsibilities<br />

in the drug discovery setting as part <strong>of</strong> lead optimization teams. To be effective<br />

members <strong>of</strong> these teams, toxicologists are now required to gain awareness and experience<br />

in recognizing potential structural and physicochemical properties <strong>of</strong> compounds<br />

that <strong>of</strong>ten lend to certain <strong>of</strong>f-target toxicities, such as <strong>of</strong>f-target receptor activities,<br />

QT-interval prolongation, phospholipidosis, enzyme induction, liver<br />

injury, and genotoxicity. <strong>The</strong> understanding <strong>of</strong> structural attributes <strong>of</strong> compounds<br />

that may drive specific liabilities will enhance the toxicologist’s contributions by enabling<br />

active collaboration with chemists to inform synthesis <strong>of</strong> new molecules to<br />

gain a safety advantage over previous molecules. To this end, in vitro assays (e.g.,<br />

hERG channel inhibition, pharmacology ligand binding panels, enzyme induction,<br />

and transporter inhibition), coupled with specific mathematical or statistical models,<br />

and pharmacophore models (e.g., hERG channel and PXR activation) can be<br />

effective at instructing chemical modifications to attenuate the potential for activity<br />

against specific toxicological endpoints a priori. Furthermore, new in silico approaches<br />

in chemoinformatics are proving useful for predicting <strong>of</strong>f-target polypharmacology<br />

by mapping potential drug-target interactions through ligand-based similarities.<br />

Similarly, mapping adverse drug reactions to chemical space may hold<br />

promise for predicting specific features <strong>of</strong> compounds that lead to adverse drug reactions<br />

in humans. Finally, detailed assessments <strong>of</strong> pharmacokinetic or physical<br />

chemical drivers <strong>of</strong> tissue accumulation are improving our understanding <strong>of</strong> in<br />

vitro—in vivo correlations. Overall these approaches demonstrate the importance<br />

and utility <strong>of</strong> understanding structural motifs and physicochemical properties <strong>of</strong><br />

compounds that lead to <strong>of</strong>f-target activity.<br />

853 CLINICAL TRANSLATIONAL VALUE OF<br />

PHARMACOLOGICAL PROMISCUITY OBSERVED IN<br />

IN VITRO SAFETY PROFILING.<br />

L. A. Urban 1 , S. Whitebread 1 , J. Hamon 2 , K. Azzaoui 2 and D. Mikhailov 1 .<br />

1 Center for Proteomic Chemistry, NIBR, Cambridge, MA and 2 Center for Proteomic<br />

Chemistry, NIBR, Basel, Switzerland.<br />

In vitro safety pr<strong>of</strong>iling <strong>of</strong> a large number <strong>of</strong> pharmacological targets has recently<br />

gained significant contribution to drug discovery. Extensive panels <strong>of</strong> biochemical<br />

and functional assays addressing single targets in native or engineered cellular environment<br />

provide large volume <strong>of</strong> data. In vitro safety pr<strong>of</strong>iling determines whether<br />

compounds affect <strong>of</strong>f-targets associated with ADRs and their pharmacological<br />

promiscuity. In silico tools made it possible to deconvolute and interpret these data<br />

and combine the chemistry and pharmacology space. Integrated assessment <strong>of</strong> data<br />

today includes ADME, metabonomics and epigenetic considerations.<br />

We focus here on pharmacological promiscuity, an important element <strong>of</strong> early pr<strong>of</strong>iling.<br />

Excluding psychiatric drugs, marketed medicines show little promiscuity.<br />

Most <strong>of</strong> these drugs are safe in the clinic, while those with high hitrate show more<br />

serious ADRs and <strong>of</strong>ten attract black box warnings or labels.<br />

Analysis <strong>of</strong> compounds from different phases <strong>of</strong> drug discovery show on average<br />

higher pharmacological promiscuity then marketed drugs and high attrition rates<br />

associated with safety during late preclinical development and in clinical trials show<br />

that they are more likely to fail to reach the clinic.<br />

Pharmacological promiscuity can be associated with physicochemical properties<br />

(logP,PSA). In addition structural features <strong>of</strong> molecules can be recognized within<br />

SAR for promiscuity over a broad range <strong>of</strong> targets/target families. We will highlight<br />

association <strong>of</strong> SAR with <strong>of</strong>f-target activities which could be successfully mitigated<br />

during drug discovery. In vitro-in vivo correlations, determination <strong>of</strong> PD/PK-based<br />

estimates <strong>of</strong> safety index and corresponding clinical therapeutic index should be<br />

part <strong>of</strong> drug discovery decision trees and flowcharts. <strong>The</strong>se preclinical forms <strong>of</strong> evaluations<br />

place in vitro safety pharmacology data in clinical context and investigate<br />

whether a molecule within a particular structural class could satisfy requirements<br />

for a medicine.<br />

854 PREDICTING DRUG OFF-TARGETS UNDERLYING<br />

ADVERSE EVENTS.<br />

M. Keiser 1, 2 and B. Shoichet 2 . 1 SeaChange Pharmaceuticals, Inc., San Francisco, CA<br />

and 2 Pharmaceutical Chemistry, University <strong>of</strong> California, San Francisco, San<br />

Francisco, CA. Sponsor: D. Hartley.<br />

Chemically similar drugs <strong>of</strong>ten bind biologically diverse targets, yet most drugs are<br />

presumed selective at therapeutic concentrations. To investigate this assumption,<br />

we used the Similarity Ensemble Approach to compare several hundred approved<br />

drugs against a panel <strong>of</strong> over 170,000 ChEMBL ligands organized into thousands<br />

<strong>of</strong> sets according to the targets they modulate. Novel <strong>of</strong>f-target drug activities consistent<br />

with adverse events were predicted and confirmed in pharmacological assays.<br />

In several cases, the drug’s novel target may be more consistent with the adverse<br />

event than are any <strong>of</strong> its previously known targets. <strong>The</strong> chemical similarity approach<br />

used here is systematic and comprehensive, and may find use for illuminating<br />

side effects <strong>of</strong> approved drugs.<br />

855 EARLY SAFETY PROFILING IN DRUG DISCOVERY:<br />

HOW STRUCTURAL AND PHYSICOCHEMICAL<br />

COMPOUND CHARACTERISTICS INFLUENCE THE<br />

SAFETY PROFILE OF DRUG CANDIDATES.<br />

M. Kansy and H. Fischer. Nonclinical Safety, F. H<strong>of</strong>fmann-La Roche Ltd., Basel,<br />

Switzerland.<br />

<strong>The</strong> determination and optimization <strong>of</strong> drug compound characteristics in fast feedback<br />

loops is an important activity in the lead identification (LI) and lead optimization<br />

phase (LO) <strong>of</strong> drug discovery research. Usually in vitro assays are applied<br />

at these phases. With the increasing number <strong>of</strong> available safety relevant measurement<br />

results predictive (in-silico) models for safety assessments can be developed.<br />

<strong>The</strong> combined application <strong>of</strong> in silico and specific in vitro methods allows the overall<br />

reduction <strong>of</strong> wet lab experiments and the prediction <strong>of</strong> potential safety liabilities<br />

<strong>of</strong> candidate molecules before they are synthesized (virtual compounds). Thus, the<br />

influence <strong>of</strong> compound structural and property changes on the safety pr<strong>of</strong>ile can be<br />

partly evaluated. Compounds with improved overall quality can be selected and<br />

cost as well as attrition rate be reduced. <strong>The</strong> talk will focus on newest achievements<br />

in the early assessment <strong>of</strong> safety liabilities by in silico tools with focus on phospholipidosis,<br />

phototoxicity, cardiovascular toxicities. <strong>The</strong> importance <strong>of</strong> amphiphilic<br />

vector calculations for phospholipids prediction will be emphasized. New multivariate<br />

in silico models based on UV-characteristics and compound properties will<br />

be described and several computational models for prediction <strong>of</strong> cardiovascular<br />

safety liabilities discussed.<br />

856 PHARMACOKINETIC DRIVERS OF TOXICITY FOR<br />

SMALL MOLECULES: EVALUATING PLASMA-TISSUE<br />

CONCENTRATION RELATIONSHIPS.<br />

D. Diaz. Investigative Safety Assessment, Genentech, South San Francisco, CA.<br />

Plasma drug exposures are routinely used in the interpretation <strong>of</strong> animal toxicity<br />

data. In reality, target tissue exposures can be much higher than plasma exposures,<br />

which could have implications in the understanding <strong>of</strong> in vivo toxicities and in the<br />

determination <strong>of</strong> in vivo-in vitro correlations. Examples will be presented in which<br />

in vitro-in vivo correlations and differences in species sensitivity can be better understood<br />

by considering target organ exposures. Systematic analysis <strong>of</strong> tissue drug<br />

levels for a group <strong>of</strong> structurally diverse small molecules reveals large differences in<br />

tissue distribution; in addition, tissue exposure levels show improved correlation<br />

with toxicity compared to plasma exposure levels. Analysis <strong>of</strong> pharmacokinetic parameters<br />

for these compounds indicates that tissue distribution can be roughly predicted<br />

by taking into consideration the volume <strong>of</strong> distribution (Vss) and the clearance<br />

(CLp) <strong>of</strong> a particular molecule. <strong>The</strong>se findings suggest that consideration <strong>of</strong><br />

these pharmacokinetic parameters could potentially help provide more relevant and<br />

translatable safety information.<br />

857 MAPPING ADVERSE DRUG REACTIONS IN<br />

CHEMICAL SPACE.<br />

J. Scheiber. Pharmacology Research & Early Development Informatics, Disease &<br />

Translational Informatics, Pharmacology Research and Early Development (pRED),<br />

Penzberg, Germany. Sponsor: D. Hartley.<br />

This talk will introduce a novel method that links substructures <strong>of</strong> drug molecules<br />

to adverse drug reactions (ADRs) on a large scale. By leveraging the content <strong>of</strong> the<br />

Pharmapendium database we were able to generate statistical models for many<br />

ADRs based on chemical substructures. For the modeling we used the well-established<br />

Scitegic fingerprints as input for a Naïve Bayesian classifier. For each <strong>of</strong> the<br />

models we extracted the most significant chemical fragments so that a map <strong>of</strong> these<br />

could be generated. Each adverse drug reaction is represented by a node and is<br />

linked to other ADRs if they share at least one significant fragment. <strong>The</strong>se results<br />

are then visualized by using Cytoscape, the state-<strong>of</strong>-the-art network visualization<br />

SOT 2011 ANNUAL MEETING 183

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!