throbber
R E V I E W S
`
`ADMET IN SILICO MODELLING:
`TOWARDS PREDICTION PARADISE?
`
`Han van de Waterbeemd* and Eric Gifford‡
`
`Following studies in the late 1990s that indicated that poor pharmacokinetics and toxicity were
`important causes of costly late-stage failures in drug development, it has become widely
`appreciated that these areas should be considered as early as possible in the drug discovery
`process. However, in recent years, combinatorial chemistry and high-throughput screening have
`significantly increased the number of compounds for which early data on absorption, distribution,
`metabolism, excretion (ADME) and toxicity (T) are needed, which has in turn driven the develop-
`ment of a variety of medium and high-throughput in vitro ADMET screens. Here, we describe how
`in silico approaches will further increase our ability to predict and model the most relevant pharma-
`cokinetic, metabolic and toxicity endpoints, thereby accelerating the drug discovery process.
`
`Why is in silico ADMET needed?
`Traditionally, drugs were discovered by testing com-
`pounds synthesized in time-consuming multi-step
`processes against a battery of in vivo biological screens.
`Promising compounds were then further studied in
`development, where their pharmacokinetic properties,
`metabolism and potential toxicity were investigated.
`Adverse findings were often made at this stage1 (FIG. 1),
`with the result that the project would be halted or re-
`started to find another clinical candidate — an unac-
`ceptable burden on the research and development bud-
`get of any pharmaceutical company.
`Today, this paradigm has been re-worked in several
`ways. The testing of drug metabolism, pharmaco-
`kinetics and toxicity is today done much earlier; that is,
`before a decision is taken to evaluate a compound in the
`clinic. However, the rate at which biological screening
`data are obtained has dramatically increased, and
`(ultra)high-throughput screening (HTS) facilities are
`now common at large pharmaceutical companies and
`at specialized biotechs2. In response to these develop-
`ments, a new approach to chemistry — combinatorial
`chemistry — has been adopted to feed these highly
`efficient hit-finding machines. Combinatorial chem-
`istry makes it possible to synthesize large series of
`closely related libraries of chemicals using the same
`chemical reaction and appropriate reagents. Such
`
`libraries are then run through the HTS to find hits
`around which further, more focused, series are designed
`and synthesized in a next round.
`As the capacity for biological screening and chemical
`synthesis have dramatically increased, so have the
`demands for large quantities of early information on
`absorption, distribution, metabolism, excretion
`(ADME) and toxicity data (together called ADMET
`data). Various medium and high-throughput in vitro
`ADMET screens are therefore now in use. In addition,
`there is an increasing need for good tools for predicting
`these properties to serve two key aims — first, at the
`design stage of new compounds and compound
`libraries so as to reduce the risk of late-stage attrition;
`and second, to optimize the screening and testing by
`looking at only the most promising compounds.
`
`Drug-like properties. Which properties make drugs dif-
`ferent from other chemicals? A number of studies have
`been performed with the aim of answering this question
`(for examples, see REFS 3–6). A particularly influential
`example — the analysis of the World Drug Index
`(WDI)5, which lead to Lipinski’s ‘rule-of-five’ — identi-
`fies several critical properties that should be considered
`for compounds with oral delivery in mind. These prop-
`erties, which are usually viewed more as guidelines rather
`than absolute cutoffs, are molecular mass <500 daltons
`
`*Pfizer Global Research
`& Development, PDM,
`Sandwich, Kent CT13 9NJ,
`UK.
`‡Pfizer Global Research &
`Development, Discovery
`Research Informatics,
`2,800 Plymouth Road,
`Ann Arbor, Michigan
`48105, USA.
`Correspondence to H.v.d.W.
`e-mail: han_waterbeemd@
`sandwich.pfizer.com
`doi:10.1038/nrd1032
`
`192 | MARCH 2003 | VOLUME 2
`
`www.nature.com/reviews/drugdisc
`
`© 2003 Nature Publishing Group
`
`1 of 15
`
`PENN EX. 2061
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`R E V I E W S
`
`When is ADMET data needed? The need for ADMET
`information starts with the design of new compounds.
`This information can influence the decision to proceed
`with synthesis either via traditional medicinal chemistry
`or combinatorial chemistry strategies. Obviously, at this
`stage, computational approaches are the only option for
`getting this information, but it is also acceptable that the
`predictions are not perfect at this point. Once a series of
`molecules is focused around a lead and is further opti-
`mized towards a clinical candidate, more robust mecha-
`nistic models will be required.
`
`What ADME properties do we want to predict? A deeper
`understanding of the relationships between important
`ADME parameters and molecular structure and proper-
`ties has been used to develop in silico models that allow
`the early estimation of several ADME properties10–17.
`Among other important issues, we want to predict
`properties that provide information about dose size and
`dose frequency (BOX 1), such as oral absorption, bioavail-
`ability, brain penetration, clearance (for exposure) and
`volume of distribution (for frequency).
`As a result of the availability of experimental data in
`the literature, considerable effort has gone into the
`development of models to predict physicochemical
`properties relevant to ADME, such as lipophilicity.
`However, despite its importance, the prediction of phar-
`macokinetic properties such as clearance, volume of dis-
`tribution and half-life directly from molecular structure
`is making slower progress owing to a lack of published
`data. Similarly, the prediction of various aspects of
`metabolism and toxicity is also underdeveloped.
`
`What computational tools are used? Here, there are two
`aspects to consider: data modelling and molecular
`modelling, which have different toolboxes. Molecular
`modelling includes approaches such as protein
`modelling18, which uses quantum mechanical methods
`to assess the potential for interaction between the small
`molecules under consideration and proteins known to
`be involved in ADME processes, such as cytochrome
`P450s. This requires three-dimensional structural
`information on the protein, which can be built by
`homology modelling of related structures if the human
`protein structure is not available. If no structural infor-
`mation on the protein is available, an alternative way of
`assessing the potential of a small molecule to interact
`with a particular protein is to use PHARMACOPHORE models,
`which are built from a superposition of known sub-
`strates of the protein.
`For data modelling, quantitative structure–activity
`relationship (QSAR) approaches19 are typically applied.
`QSAR and quantitative structure–property relationship
`(QSPR) studies have been performed since the 1960s
`with a variety of biological and physicochemical data.
`These studies use statistical tools to search for correla-
`tions between a given property and a set of molecular
`and structural descriptors of the molecules in question.
`Once such a QSAR model has been ‘TRAINED’ using a set
`of molecules for which experimental data on the prop-
`erty in question are available, it can be used to make
`
`30%
`
`39%
`
`5%
`
`10%
`
`5%
`
`11%
`
`Pharmacokinetics
`
`Adverse effects in man
`
`Animal toxicity
`
`Miscellaneous
`
`Commercial reasons
`
`Lack of efficacy
`
`Figure 1 | An analysis of the main reasons for attrition in
`drug development1. In this analysis, published five years ago,
`half of all failures were attributed to poor pharmacokinetics
`(39%) and animal toxicity (11%). Such analyses clearly
`indicated that these two areas should be focused on as early
`as possible in the drug-discovery process (although it should
`be noted that the interpretation of such data is often hampered
`by the fact that compounds may have more than one flaw and,
`as the project was halted, these might not always have been
`identified). An even better approach would be to use predictive
`tools in the design phase of the synthesis of compounds and
`compound libraries.
`
`(Da), calculated octanol/water partition coefficient
`(CLOGP) <5, number of hydrogen-bond donors <5 and
`number of hydrogen-bond acceptors <10. In general,
`such studies, and others not cited here, point to the most
`important physicochemical and structural properties
`characteristic of a good drug in the context of our current
`knowledge. These properties are then typically used to
`construct predictive ADME models and form the basis
`for what has been called property-based design7. To a
`certain extent, similar molecules can be expected to have
`similar ADME properties8. This concept is the basis of
`software called SLIPPER-2001, in which physicochemical
`DESCRIPTORS and molecular similarity are used for the
`prediction of properties such as lipophilicity, solubility
`and fraction absorbed in humans9.
`
`How are ADMET data obtained? The quest for early,
`fast and relevant ADMET data is tackled in three ways.
`First, a variety of in vitro assays have been further auto-
`mated through the use of robotics and miniaturization.
`Second, in silico models are being used to assist in the
`selection of both appropriate assays, as well as in the
`selection of subsets of compounds to go through these
`screens. Third, predictive models have been developed
`that might ultimately become sophisticated enough to
`replace in vitro assays and/or in vivo experiments.
`
`DESCRIPTOR
`A structural or physicochemical
`property of a molecule or part
`of a molecule. Examples include
`log P, molecular mass and polar
`surface area.
`
`PHARMACOPHORE
`A pharmacophore is the
`ensemble of steric and electronic
`features that are necessary to
`ensure the optimal
`supramolecular interactions
`with a specific biological target
`structure and to trigger (or to
`block) its biological response.
`
`TRAINING
`The building of a model using
`part of the data (that is, the
`training set), followed by
`validation of the model using
`the rest of the data (that is, the
`validation set). Finally, the
`model is tested using
`compounds (the test set) not
`used for training and validation.
`
`NATURE REVIEWS | DRUG DISCOVERY
`
`VOLUME 2 | MARCH 2003 | 1 9 3
`
`© 2003 Nature Publishing Group
`
`2 of 15
`
`PENN EX. 2061
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`R E V I E W S
`
`Box 1 | Pharmacokinetics
`
`Pharmacokinetics is the study of the time course of a drug within the body and
`incorporates the processes of absorption, distribution, metabolism and excretion
`(ADME)76. Pharmacokinetic parameters are derived from the measurement of drug
`concentrations in blood or plasma. The key pharmacokinetic parameters and their
`importance for the dose regimen and dose size are shown in the figure80.
`Most drugs are given orally for reasons of convenience and compliance. Typically, a drug
`dissolves in the gastro-intestinal tract, is absorbed through the gut wall and then passes the
`liver to get in to the blood circulation. The percentage of the dose reaching the circulation
`is called the bioavailability. From there, the drug will be distributed to various tissues and
`organs in the body. The extent of distribution will depend on the structural and
`physicochemical properties of the compound. Some drugs can enter the brain and central
`nervous system by crossing the blood–brain barrier. Finally, the drug will bind to its
`molecular target, for example, a receptor or ion channel, and exert its desired action.
`• Volume of distribution (Vd) is a theoretical concept that connects the administered
`dose with the actual initial concentration (C0) present in the circulation:
`Vd = Dose/C0
`Most drugs will bind to various tissues and in particular to proteins in the blood, such
`as albumin. As only the free (unbound) drug will bind to the molecular target, the
`concept of unbound volume of distribution is used:
`Vdu = Vd/fu, where fu is the fraction unbound.
`• Clearance (Cl) of the drug from the body mainly takes place via the liver (hepatic
`clearance or metabolism, and biliary excretion) and the kidney (renal excretion).
`By plotting the plasma concentration against time, the area under the curve (AUC)
`relates to dose, bioavailability and clearance.
`AUC = F x Dose/Cl
`• Half-life (t1/2) — the time taken for a drug concentration in the plasma to reduce by
`50% — is a function of the clearance and volume of distribution, and determines how
`often a drug needs to be administered.
`t1/2 = 0.693 Vd/Cl
`
`Volume of
`distribution
`
`Clearance
`
`Absorption
`
`Half-life
`
`Oral
`bioavailability
`
`Dosing regimen:
`How often?
`
`Dosing regimen:
`How much?
`
`predictions on molecules not in the training set,
`although, in general, reliable predictions are only possible
`for molecules similar to those in the training set.
`A wide variety of descriptors for use in QSAR
`studies have been developed over the last 40 years20
`(for example, those available in the program Dragon).
`A subset of these descriptors is potentially useful for
`predicting ADME properties. Indeed, with the
`increased interest in the prediction of ADME proper-
`ties, specifically tailored descriptors have already been
`reported, for example, those in the VolSurf program21.
`Some of the descriptors used are close to the chemist’s
`
`MULTIVARIATE ANALYSIS
`A subset of statistical techniques
`that can deal with larger sets of
`molecular descriptors that is
`aimed at finding relationships or
`patterns in data sets. Examples
`include multiple linear
`regression (MLR) and partial
`least squares (PLS).
`
`intuition, such as molecular size and hydrogen bonding.
`Other descriptors are merely topological or quantum
`chemical concepts, but can produce highly predictive
`models, although these might be ‘black boxes’ for
`most people.
`Using appropriate descriptors, QSAR approaches —
`ranging from simple multiple linear regression to
`modern MULTIVARIATE ANALYSIS techniques, such as partial
`least squares (PLS) — are now being applied to the
`analysis of ADME data22. Data-mining and machine-
`learning methods originally developed and used in other
`fields are now also successfully being used for this pur-
`pose. Examples of such methods include NEURAL NETWORKS
`(NN), self-organizing maps (SOM; also called Kohonen
`networks), RECURSIVE PARTITIONING (RP) and support
`vector machines (SVM).
`Good predictive models for ADMET parameters
`depend crucially on selecting the right mathematical
`approach, the right molecular descriptors for the particu-
`lar ADMET endpoint, and a sufficiently large set of
`experimental data relating to this endpoint for the valida-
`tion of the model (BOX 2). Insight is growing as to which
`of the available descriptors and QSAR tools are most
`appropriate, although there often seems to be different
`options with similar predictive power. In particular, more
`needs to be learnt about how the size of the training set
`influences the choice of the most capable model.
`
`Prediction of physicochemical properties
`The physicochemical properties of a drug have an
`important impact on its pharmacokinetic (BOX 1) and
`metabolic (BOX 3) fate in the body, and so a good under-
`standing of these properties, coupled with their mea-
`surement and prediction, are crucial for a successful
`drug discovery programme.
`
`Lipophilicity. Poor biopharmaceutical properties — in
`particular, poor aqueous solubility and slow dissolu-
`tion rate — can lead to poor oral absorption and
`hence low oral bioavailability. In general, poor solubility
`is related to high lipophilicity, whereas hydrophilic
`compounds generally show poor permeability and
`hence low absorption. Therefore, the measurement of
`solubility and lipophilicity, as well as ionization con-
`stants affecting these two properties, has been auto-
`mated and integrated in the high-throughput drug
`discovery paradigm.
`The relationship between lipophilicity and phar-
`macokinetic properties has been discussed by various
`workers in the field23–25. Lipophilicity is the key
`physicochemical parameter linking membrane per-
`meability — and hence drug absorption and distribu-
`tion — with the route of clearance (metabolic or
`renal). Measuring the lipophilicity of a compound is
`readily amenable to automation. The gold standard
`for expressing lipophilicity is the partition coefficient
`P (or log P to have a more convenient scale) in an
`octanol/water system; alternatives include applica-
`tions of immobilized artificial membranes (IAM),
`immobilized liposome chromatography (ILC) and
`liposome/water partitioning.
`
`194 | MARCH 2003 | VOLUME 2
`
`www.nature.com/reviews/drugdisc
`
`© 2003 Nature Publishing Group
`
`3 of 15
`
`PENN EX. 2061
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`Box 2 | The need for good data
`
`Clearly, larger databases of marketed drugs are required to establish more robust models
`to predict various ADME properties, including drug–drug interactions. Several
`published ADME data sets are available for data modelling13,36,47,105–107, but the quality of
`the data and the number of available training examples remain important issues. In the
`future, service providers such as Cerep, Novascreen and Cyprotex will be able to offer
`larger data sets with which to build more robust models.
`In a recent symposium, the question of whether the Internet can help as a resource
`to collect relevant ADME data was addressed108. The current opinion is that there are
`some good and well-maintained websites available, but unfortunately also many of
`questionable use in research owing to a lack of control of data quality or reference to
`the original data.
`
`There is continued interest in developing and
`improving log P calculation programs, and there are
`many such programs available. Most calculation
`approaches rely on fragment values, although simple
`methods based on molecular size and hydrogen-bonding
`indicators for functional groups to calculate log P values
`have also been shown to be extremely versatile22.
`However, log P values can only be a first estimate of
`the lipophilicity of a compound in a biological environ-
`ment. For partition processes in the body, the distribu-
`tion coefficient D (log D) — for which an aqueous
`buffer at pH 7.4 (blood pH) or 6.5 (intestinal pH) is
`used in the experimental determination — often pro-
`vides a more meaningful description of lipophilicity,
`especially for ionizable compounds. However, in our
`experience, programs that can reliably predict log D are
`scarce at present.
`
`Solubility. The first step in the drug absorption process
`is the disintegration of the tablet or capsule, followed by
`the dissolution of the active drug. Obviously, low solu-
`bility is detrimental to good and complete oral absorp-
`tion, and so the early measurement of this property is of
`great importance in drug discovery. Reflecting this need,
`rapid, robust methods reliant on turbidimetry and
`nephelometry have been developed to efficiently measure
`the solubility of large numbers of compounds6,26.
`Ideally, only soluble compounds would be synthe-
`sized in a drug-discovery programme. Predictive solu-
`bility methods — for example, neural networks —
`might assist in this effort. However, at present, no
`approaches are robust enough to accurately predict low
`solubility. Many current predictive solubility programs27
`use training data from different laboratories with varying
`quality and different experimental conditions. Hopefully,
`by measuring many compounds under standardized
`conditions, current predictive models can be improved28.
`
`pKa. As ionization can also affect the solubility,
`lipophilicity (log D), permeability and absorption of a
`compound, approaches have been developed for the
`rapid measurement of pKa values of sparingly soluble
`drug compounds. Using experimental data reported in
`the literature, several approaches have been used to
`develop pKa calculators. Programs include ACD/pKa
`(ACD), Pallas/pKa (Compudrug) and SPARC29.
`
`NEURAL NETWORKS
`Neural networks are
`computational models that are
`based on the principles of the
`functioning of the brain. They
`can be used to model nonlinear
`relationships between dependent
`(biological endpoint to be
`predicted) and independent
`(molecular and structural
`descriptors) variables. Examples
`include back-propagation and
`self-organising maps (SOM;
`also called Kohonen neural
`networks).
`
`RECURSIVE PARTITIONING
`OR DECISION TREES
`A supervised learning method
`producing a tree-structured
`series of rules to predict a
`particular property using a set of
`molecular descriptors as input.
`
`R E V I E W S
`
`Hydrogen bonding. The hydrogen-bonding capacity of a
`drug solute is now recognized as an important determi-
`nant of permeability. In order to cross a membrane, a
`drug molecule needs to break hydrogen bonds with its
`aqueous environment. The more potential hydrogen
`bonds a molecule can make, the more energy this bond-
`breaking costs, and so high hydrogen-bonding potential
`is an unfavourable property that is often related to low
`permeability and absorption.
`Initially, ∆logP — the difference between octanol/
`water and alkane/water partitioning — was used as a
`measure for solute hydrogen-bonding, but this tech-
`nique is limited by the poor solubility of many com-
`pounds in the alkane phase. A variety of computational
`approaches have addressed the problem of estimating
`hydrogen-bonding capacity, ranging from simple hetero-
`atom (O and N) counts, the consideration of molecules
`in terms of the number of hydrogen-bond acceptors
`and donors, and more sophisticated measures that take
`into account such parameters as free-energy factors30
`and (dynamic) polar surface area (PSA)31. The latter
`are easily calculated, and it is now believed that a
`single minimum-energy conformation is sufficient to
`compute the PSA, instead of the more computation-
`ally demanding and time-consuming dynamic polar-
`surface-area calculation31. A fast fragment-based
`algorithm for PSA has been reported32, which allows
`PSA calculations to be implemented in virtual screen-
`ing approaches.
`
`Permeability. Efforts have been undertaken to predict
`the permeability of compounds through Caco-2 cells,
`which serve as a model for human intestinal absorption,
`in an approach called membrane-interaction quantita-
`tive structure–activity relationships (MI-QSAR)33. But
`one could ask the question,“Why model the model of
`human absorption?”. A more direct approach is to
`model processes that would address ‘pure’ measures of
`permeability. These include octanol/water partitioning,
`liposome partitioning, retention on immobilized arti-
`ficial membranes (IAM), the parallel artificial mem-
`brane-permeability assay (PAMPA) and binding to
`liposomes measured by surface-plasmon-resonance
`(SPR) biosensors.
`
`Prediction of ADME and related properties
`Absorption. For a compound crossing a membrane by
`purely passive diffusion, a reasonable permeability
`estimate can be made using single molecular properties,
`such as log D or hydrogen-bonding capacity. However,
`besides the purely physicochemical component con-
`tributing to membrane transport, many compounds are
`affected by biological events, including the influence of
`transporters and metabolism (further discussed in later
`sections). Many drugs seem to be substrates for trans-
`porter proteins, which can either promote or hinder
`permeability. In particular, the combined role of
`cytochrome P450 3A4 (CYP3A4) and P-glycoprotein
`(P-gp) in the gut as a barrier to drug absorption has
`been well studied34. Currently, no theoretical SAR basis
`exists to account for these effects.
`
`NATURE REVIEWS | DRUG DISCOVERY
`
`VOLUME 2 | MARCH 2003 | 1 9 5
`
`© 2003 Nature Publishing Group
`
`4 of 15
`
`PENN EX. 2061
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`R E V I E W S
`
`Box 3 | Metabolism
`
`The body will eventually try to eliminate xenobiotics, including drugs. For many drugs,
`this first requires metabolism or biotransformation, which takes place partly in the gut
`wall during uptake, but primarily in the liver. The figure shows where metabolism occurs
`during the absorption process. The fraction of the initial dose appearing in the portal
`vein is the fraction absorbed, and the fraction reaching the blood circulation after the
`first-pass through the liver defines the bioavailability of the drug.
`Traditionally, a distinction is made between phase I and phase II metabolism, although
`these do not necessarily occur sequentially. In phase I metabolism, a molecule is
`functionalized, for example, through oxidation, reduction or hydrolysis. The most
`important enzymes involved are the cytochrome P450s. In particular, CYP3A4, CYP2D6,
`CYP2C9 and CYP2C19 are important for the metabolism of drugs in humans. In phase II
`metabolism, the functionalized drug molecule is further transformed in so-called
`conjugation reactions. These include for example, glucuronidation and sulfation, as well as
`conjugation with glutathione. It should be noted that the metabolism in animals might be
`different from that in humans, and therefore the prediction of human pharmacokinetics
`and metabolism from animal data might not be straightforward.
`
`Dose
`
`Absorption
`
`Portal
`vein
`
`Gut
`wall
`
`Liver
`
`Bioavailability
`
`To faeces
`
`Metabolism
`
`Metabolism
`
`In vitro methods, such as Caco-2 or Madin-Darby
`canine kidney (MDCK) monolayers, are widely used to
`make oral absorption estimates. These cells also express
`transporter proteins, but only express very low levels of
`metabolizing enzymes. Similarly, there is a continued
`interest in finding a relevant in vitro screen for estimating
`the permeability of drugs for diseases of the central
`nervous system (CNS). The bovine microvessel endo-
`thelial cell (BMEC) model has been explored as a possible
`in vitro model of the blood–brain barrier35.
`Considerable effort has also gone into the develop-
`ment of in silico models for the prediction of oral
`absorption36–42. The simplest models are based on a single
`descriptor, such as log P or log D, or polar surface area,
`which is a descriptor of hydrogen-bonding potential31.
`Different multivariate approaches, such as multiple linear
`regression, partial least squares and artificial neural
`networks41, have been used to develop quantitative
`structure–human-intestinal-absorption relationships. In
`all approaches, hydrogen bonding is considered to be a
`property with an important effect on oral absorption.
`Absorption-simulation programs, such as Gastro-
`Plus43 and Idea44, might eventually become a valuable
`tool in lead optimization and compound selection.
`These programs, which have recently been compared45,
`are computer simulation models developed and vali-
`dated to predict ADME outcomes, such as rate of
`absorption and extent of absorption, using a limited
`
`number of in vitro data inputs. They are based on
`advanced compartmental absorption and transit
`(ACAT) models, in which physicochemical concepts,
`such as solubility and lipophilicity, are more readily
`incorporated than physiological aspects involving trans-
`porters and metabolism. In more recent versions,
`attempts are being made to model the influence of
`transporters, in addition to gut-wall metabolism, on
`gastrointestinal uptake. For example, the oral bioavail-
`ability of ganciclovir in dogs and humans was simulated
`using a physiologically based model that utilized many
`biopharmaceutically relevant parameters, such as the
`concentration of ganciclovir in the duodenum,
`jejunum, ileum and colon at a variety of dose levels and
`solubility values. The simulation results demonstrated
`that the low bioavailability of ganciclovir is limited by
`compound solubility rather than permeability due to
`partitioning, as previously speculated44.
`
`Bioavailability. Recently, the first attempts to predict
`bioavailability directly from molecular structure have
`been published. However, this is not an easy task, as
`bioavailability depends on a superposition of two
`processes: absorption and liver first-pass metabolism.
`Absorption in turn depends on the solubility and per-
`meability of the compound, as well as interactions with
`transporters and metabolizing enzymes in the gut wall.
`Important properties for determining permeability
`seem to be the size of the molecule, as well as its capacity
`to make hydrogen bonds, its overall lipophilicity and
`possibly its shape and flexibility. Molecular flexibility,
`for example, as evaluated by counting the number of
`rotatable bonds, has been identified as a factor influenc-
`ing bioavailability in rats46.
`Yoshida and Topliss47 trained a QSAR model with log
`D at pH 7.4 and 6.5 as inputs for the physicochemical
`properties and the presence/absence of typical functional
`groups most likely to be involved in metabolic reactions
`as the structural input. This approach used ‘fuzzy adap-
`tive least squares’, and drugs could be classified into one of
`four predefined bioavailability ranges. Using this
`approach, a new drug can be assigned to the correct class
`with an accuracy of 60%.An unpublished effort based on
`classification using the SIMCA approach and which
`seems to achieve similar success has also been reported12.
`In another approach, regression and recursive parti-
`tioning have been used48. In this study, 591 compounds
`were included and a set of 85 structural descriptors was
`generated. The authors noted that the mean error in the
`experimental data used to generate the model is ~12%,
`with an increase in error for well-absorbed drugs.
`Therefore, the models should not be expected to gener-
`ate predictions that are more accurate than the variabil-
`ity inherent in the biological measurements.
`Genetic programming, which is a specific form of
`evolutionary programming, has recently been used for
`predicting bioavailability49. The results show a slight
`improvement compared with the Yoshida-Topliss
`approach, although a direct comparison is difficult
`owing to a different selection of the bioavailability
`ranges of the four classes.
`
`196 | MARCH 2003 | VOLUME 2
`
`www.nature.com/reviews/drugdisc
`
`© 2003 Nature Publishing Group
`
`5 of 15
`
`PENN EX. 2061
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`A method for predicting bioavailability using adap-
`tive fuzzy partitioning (AFP) has recently been pre-
`sented at conferences50. The best molecular descriptors
`were selected with a genetic algorithm, and in the next
`step SOMs were used for the classification, which cor-
`rectly classified the molecule in the right bioavailability
`class in 64% of cases.
`The methods described above demonstrate that at
`least qualitative (binned) predictions of oral bioavail-
`ability seem tractable directly from molecular structure.
`Approaches using in vitro data are also under continual
`development. For example, a graphical approach for
`bioavailability prediction based on the combined mea-
`surement of Caco-2 flux and microsomal stability was
`recently presented51 that uses a reference plot to make a
`prediction of bioavailability for a new compound.
`Typically, the prediction will classify a compound as
`0–20%, 20–50% or 50–100% bioavailable. Extending
`this approach to include solubility, for example, might
`increase its predictive power.
`
`Blood–brain barrier penetration. Drugs that act in the
`CNS need to cross the blood–brain barrier (BBB) to
`reach their molecular target. By contrast, for drugs
`with a peripheral target, little or no BBB penetration
`might be required in order to avoid CNS side effects. A
`key issue in the development of models to predict BBB
`penetration is the use of appropriate data to describe
`brain uptake of compounds. There is an ongoing dis-
`cussion about the use of total-brain data versus extra-
`cellular fluid (ECF) or cerebro-spinal fluid (CSF) data
`or data generated by microdialysis52. Another point of
`debate relates to the time point of measurement, which
`is clearly crucial. Overall, data in the literature are
`rather limited in number, and are also generated from
`different experimental protocols. All of these factors
`limit the development of highly predictive models of
`BBB penetration.
`Nevertheless, a variety of models for the prediction
`of uptake into the brain have been developed53–59.
`‘Rule-of-five’-like recommendations regarding the
`molecular parameters that contribute to the ability of
`molecules to cross the BBB have been made to aid
`BBB-penetration predictions53; for example, molecules
`with a molecular mass of <450 Da or with PSA <100 Å2
`are more likely to penetrate the BBB. Most of the early
`predictive models are based on a multiple linear
`regression approach and many use physicochemical
`properties60. One example of such a model is based on
`the combination of only three descriptors, namely the
`calculated octanol/water partition coefficient, the
`number of hydrogen-bond acceptors in an aqueous
`medium and the polar surface area55. More recently,
`other multivariate techniques have been tried using new
`ADME-tailored properties, such as the Volsurf approach,
`in which

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket