Admet Properties: Presented By, Adarsha Govinda K 2 SEM. Ph. Chemistry Ngsmips
Admet Properties: Presented By, Adarsha Govinda K 2 SEM. Ph. Chemistry Ngsmips
Admet Properties: Presented By, Adarsha Govinda K 2 SEM. Ph. Chemistry Ngsmips
ADMET PROPERTIES
PRESENTED BY,
ADARSHA GOVINDA K
2ND SEM. Ph. Chemistry
NGSMIPS
CONTENTS
• Introduction
• Importance
• In silico ADMET models
Introduction
• ADMET refers to absorption,distribution, metabolism, excretion and
toxicity.
• Analysing ADMET properties before producing or synthesizing a new
drug is very important.
• It will minimize the risk of drug being incompetent.
• These ADMET properties have important role in designing of drug.
• Around 60% of failure in newer drugs are related to ADMET.
• Earlier days, ADMET tools were applied at the end of drug design.
• But, now these are considered from early stages itself, in order to
remove any risk of drug being rendered useless due to its ADMET.
Importance of determining ADMET
• Drug design is a multi step, time consuming and costly process
which requires large manpower.
• Undesired pharmacokinetic properties are analysed during
developmental stage.
• Early decision.
• Quick.
• Attrition in drug development.
• There are certain properties that make a drug different from other
compounds.
• Several pharmacokinetics properties are involved in this mechanism.
• Bioavailability depends on absorption and liver first-pass metabolism.
• The volume of its distribution, together with its clearance rate,
determines the half-life of a drug and therefore its dosage.
• To identify compounds that have good pharmacokinetic and toxicological profiles,
ADMET studies should be started as early as possible in the discovery process.
• New formats of in vitro ADMET assays serving early discovery have been
developed from traditional assays through protocol simplification and making use
of technological advancements.
• These models are promising as early screening tools for drug candidates and for
designing more successful combinatorial or in-silico libraries, where there is a very
small amount of the compound and a computational approach is then the only
option for collecting this information.
Insilico ADMET Model
1 • Data-based approach
• Structure based
2
approach
Data-based approaches
The distribution will depend on the structural and physicochemical
properties of the compound.
Most drugs exhibit a non-uniform distribution in the body with variations that
are largely determined by their ability to pass through membranes and their
lipid/water solubility.
Proteins exert a large influence on the distribution process because drugs can
bind to a variety of them such as
albumin (acidic drugs)
alpha1-acid glycoprotein (basic drugs)
lipoproteins (neutral and basic drugs)
Drug distribution can be estimated using tissue: plasma ratios or
the volume of a steady state distribution ( Vss ).
These methods are based on the assumption that there is a
passive diffusion between tissue compartments.
The identification of the tissue: plasma partition coefficients
( Kp ) needed for an initial prediction of the volume of steady state
distribution ( Vss ) of a drug in humans.
The initial estimation of the Vss of a new drug can normally be
based on only two Kp values, those of muscle and fat. The muscle
Kp can be used to represent all lean tissues, including the residual
and fat Kp can be used for the distribution of basic drugs.
Physiological information on tissue composition (lipid/water/protein
fraction), the blood composition (lipid/water/protein) and the blood
flowing into the tissues are often used to develop a partitioning
model.
Plasma protein binding is also taken into consideration, using
three simple measurements to predict the distribution volume of a
new compound:
human plasma protein binding,
experimental logD and
pKa.
QSPR techniques for the prediction of volume of distribution have
also been used.
Structural descriptors consisted of partitioning, quantum
mechanics, molecular mechanics, and connectivity parameters and
genetic algorithm or step-wise regression analyses were used in
the selection of the variable and model development.
The blood-brain barrier (BBB) is an important element in the
regulation of the internal environment of the brain. Drugs that
act in the CNS need to cross the BBB, in contrast, drugs with
a peripheral target may not cross the BBB to avoid CNS side
effects.
Early predictions logBB (logarithm value of brain to plasma
concentration ratio) involved classical QSAR approaches
using various physicochemical parameters such as
the octanol-water partition coefficient (logP) ,
molecular size descriptors and
solvation parameters.
• A combination of molecular descriptors was used to predict log BB
using lipophilicity, polarity, polarizability, and hydrogen bonding
parameters and partial least-squares statistics, and it was
demonstrated that topological and constitutional descriptors used in
partial least squares correlate well with experimental logBB data.
• There is a model using PSA as the only descriptor, calculating the
PSA from all low-energy conformations of a particular compound.
A typical drug metabolism pathway is the oxidation of the
drug (phase I oxidation), followed by conjugation (phase II
conjugation).
Several methods have been developed mostly:
based on the knowledge of the structure and mechanism of the enzymes (protein structure; 3D-structure and accessibility of the
OR
based on the physicochemical properties of the compound (molecular sites sensitive to oxidation or conjugation, structure of the
chemical, molecular surface properties, electronic structure, quantum mechanics properties, etc)
Due to the importance of cytochrome P450 in drug metabolism
many models have been developed to predict both substrates
and inhibitors.
3D models of cytochrome P450 using the x-ray structure of
bacterial cytochromes P450 as templates, were developed.
By docking some substrates into the active site of their
metabolizing enzymes it was found that substrates binding into the
active site of isoform CYP2D6 are mainly favoured by hydrogen
bonding and electrostatic interactions between the substrate
and two residues of the enzyme.
In contrast, van der Waals attraction interactions mainly
contribute to stabilize the complex involving both CYP1A2 and
CYP3A4.
Computer systems to predict xenobiotic metabolism are
commercially available. These softwares mainly predict phase I
metabolism, although some of them also predict phase II
reactions.
COMPACT (Computer-Optimized Molecular Parametric Analysis
of Chemical Toxicity) [ has modules that assess the ability of
xenobiotics to form enzyme substrate complexes and undergo
metabolic activation by the CYP1A and CYP2E subfamilies of
cytochromes P450.
The COMPACT model uses sterical and electronic parameters.
Improved criteria for CYP1A and CYP2E1 substrate specificity
have recently been reported .
META is an expert system that can predict the sites of potential
enzymatic attack and the nature of the chemicals formed by such
metabolic transformations.
The program uses dictionaries of biotransformation operators
created by experts in the field of xenobiotic metabolism to represent
known metabolic pathways .
Activation of a biotransformation operator within the program is
based on the recognition of key functional groups within the
complete chemical structure.
The program evaluates stable metabolites and transform them
further build up a complete metabolic tree.
Processing continues in this way until the program is able to
generate water soluble metabolites that are to be excreted.
MetabolExpert predicts the metabolism of exogenous compounds in
plants and mammals. The system is composed of biotransformation
databases, a knowledge-base and prediction tools .
There are two types of predictions in MetabolExpert.
In the first, the system tries to match basic biotransformations to the
compound structure.
The second type of analysis is an extended prediction model in
which metabolites generated from basic transformations are
compared to transformations in ‘learned’ trees.
Analysis of ADMET properties
Swiss ADME
ADMET Predictor
QikProp-Schrӧdinger
QikProp predicts the widest variety of pharmaceutically relevant
properties - octanol/water and water/gas log Ps, log S, log BB,
overall CNS activity, Caco-2 and MDCK cell permeabilities, log
Khsa for human serum albumin binding, and log IC50 for HERG
K+-channel blockage - so that decisions about a molecule's
suitability can be made based on a thorough analysis.
Accurate ADME properties:
QikProp bases its predictions on the full 3D molecular structure;
unlike fragment-based approaches, QikProp can provide equally
accurate results in predicting properties for molecules with novel
scaffolds as for analogs of well-known drugs.
Lead generation:
QikProp rapidly screens compound libraries for hits.
QikProp identifies molecules with computed properties that fall
outside the normal range of known drugs, making it simple to
filter out candidates with unsuitable ADME properties.
Lead optimization:
QikProp can play an important role during lead optimization by
analyzing similarity within a class of compounds as well as by
identifying compounds to avoid because they exhibit extreme
values of predicted properties.