Вы находитесь на странице: 1из 4

Editorial

From in vivo to in vitro/in silico


ADME: progress and challenges
Han Van de Waterbeemd
Pharmaceutical Consultant in In Vitro and In Silico ADME Technologies,
1. Evaluation of research so far: han.vdwaterbeemd@btinternet.com
ADME-Tox screening in drug
High-throughput screening technologies in biological sciences of large
discovery
Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Memorial University of Newfoundland on 08/01/14

libraries of compounds obtained via combinatorial or parallel chemistry


2. Future prospects: challenges in
approaches, as well as the application of design rules for drug-likeness, have
drug metabolism,
resulted in more hits to be evaluated with respect to their ADME or drug
pharmacokinetics and
metabolism and pharmacokinetic properties. The traditional in vivo meth-
toxicology
ods using preclinical species, such as rat, dog or monkey, are no longer suffi-
3. Expert opinion: ADME studies in
cient to cope with this demand. This editorial discusses the changes towards
the future
medium- to high-throughput in vitro and in silico ADME screening. In addi-
tion, much more attention is now put on early safety and risk assessment of
promising lead series and potential clinical candidates.

Keywords: ADME, DMPK, high-throughput screening, in combo screening, in silico ADME,


lead profiling, toxicology

Expert Opin. Drug Metab. Toxicol. (2005) 1(1):1-4


For personal use only.

1.Evaluation of research so far: ADME-Tox screening in


drug discovery

Only recently have pharmacokinetics, drug metabolism and toxicology of selected


clinical candidates been studied during clinical development. Analysing the root
cases of attrition demonstrated that lack of efficacy and toxicity, along with inappro-
priate ADME, are among the major determinants for failure [1,2]. Lack of efficacy, in
addition to poor target binding may be caused by poor absorption and distribution,
and rapid metabolism, leading to drug concentrations at the target that are too low
[3]. As toxicity is a major factor in causing attrition, the development of compounds
is often halted even before a detailed human pharmacokinetics or efficacy study is
performed. Hence, the real impact of ADME processes remains somewhat hidden in
(incomplete) attrition data. However, it became good practice in the 1990s to
collect ADME and toxicity data during the drug discovery stage so that it can be
used in decision making to select the best possible clinical candidates [4]. However,
the success of this move to early involvement has been questioned [5]. The main
contribution of a discovery (research) department of drug metabolism and pharma-
cokinetics has been to enable the design of pharmacokinetically adequate rather
than optimal molecules and, thus, to make it possible to work on difficult targets [5].
Despite all of the preclinical efforts, there will be a need for extensive clinical
pharmacokinetics to provide a safe prescription to patients [5].
The number of compounds synthesised has increased dramatically through
combinatorial library strategies in chemistry, and is driven by high-throughput screen-
ing (HTS) in biology. Much attention is also paid to the design criteria for lead- and
drug-like compounds [6], which further increased the output of compounds with
appropriate ADME properties. The concept of property-based design [7] is now com-
monly used to address ADME issues as early as possible. Thus, the former in vivo ani-
mal ADME evaluation could no longer cope with the demand, and in vitro ADME
Ashley Publications screens became widely introduced. Despite rapid advances in the use of automation
www.ashley-pub.com and robotics to increase the throughput of the in vitro ADME screens, it became clear
that screening all available compounds is not necessarily most efficient and

10.1517/17425255.1.1.1 © 2005 Ashley Publications Ltd ISSN 1742-5255 1


From in vivo to in vitro/in silico ADME: progress and challenges

cost-effective. It is, therefore, sensible and vital to develop (P-gp) substrate or inhibitor. It is believed that CYP3A4 and
in silico tools to predict and simulate various ADME properties, P-gp in the gastrointestinal tract work in concert to limit
and use these in decision-making in a well-balanced fashion uptake of xenobiotics, including drugs [16]. Current inhibi-
together with in vivo and in vitro approaches. tion studies are based on Ki or mean inhibitory concentra-
ADME studies are aimed at getting an early estimate of tion values, but more quantitative approaches would be
human pharmacokinetics (PK) and metabolism [8]. These stud- beneficial [17]. Great progress has been made in the reliable
ies include: estimates of dose, dose interval, absorption, bioa- simulation of DDIs, even taking into account the variability
vailability, clearance, volume of distribution, half-life, clearance in the population [18]. A further question is how metabolites
mechanism, potential for drug–drug interactions, involvement contribute to DDIs and a better understanding of allosteric
Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Memorial University of Newfoundland on 08/01/14

of major metabolising enzymes and metabolite formation. kinetics of P450s may be required [19].
Early toxicology and safety studies should eliminate
compounds before they enter lengthy and costly clinical trials. 2.3 Enzyme induction
As a result of the recent withdrawal of a number of marketed Although clinically less important than enzyme inhibition,
drugs, more pressure is now put on pharmaceutical companies protocols are being developed to estimate enzyme induction.
regarding safety evaluations, including pharmacological and It needs to be seen where to place such assay or screen in the
toxicological safety. Investigation of the potential to cause QT discovery process [20].
prolongation is now routine. However, the interpretation of Transporter proteins constitute a significant fraction of
the data in not straightforward, as many marketed drugs can membrane-bound proteins [21,22]. They are typically expressed
prolong the QT interval [9,101]. in all organs involved in uptake and elimination of drugs,
including the gastrointestinal tract, blood–brain barrier, liver
2. Future
prospects: challenges in drug and kidney. It is hoped that in the near future three-dimen-
metabolism, pharmacokinetics and toxicology sional structures of the key transporters might be obtained.
Interaction of drugs with transporters can alter their behav-
2.1 In vitro screening iour in membrane transport, which may result in active
For personal use only.

Based on the knowledge gained from high-throughput screen- uptake, efflux and rapid elimination, for example. In other
ing in biology, many in vitro ADME screens can now run in words, the pharmacokinetics of a drug may be influenced by
medium- or high-throughput using automation, robotics and transporters. Drug–drug interactions may have their cause
miniaturisation [10-12]. However, due to the nature of the apart from a metabolic component, in the involvement of
assays, the typical analytical end point is often liquid chroma- transporters [20,23]. However, although the basic knowledge of
tography/mass spectrometry. Cell-based assays, such as the transporters is growing rapidly [21], the clinical significance is
Caco-2 screen for permeability/absorption are, therefore, open to much debate, but some examples have been presented
quite expensive, particularly when run in screening mode on [24]. A number of P-gp assays have been developed, as well as
many compounds. The trend is to investigate either cheaper some double and triple transfected assays. More transporter
in vitro alternatives such as the parallel artificial membrane assays will soon be available. The challenge will be to translate
permeability assay (PAMPA) method or to move to in silico the flood of experimental data into relevant information for
approaches. A proper synergistic hybrid combination of drug design projects.
in vitro and in silico methods [13], which has been called the
in combo approach [14], may be the most cost-effective 2.4 In silico ADME
approach to ADME screening in drug discovery. Prediction and simulation of various ADME properties is con-
siderably less expensive than in vitro screening. Therefore, great
2.2 Drug–drug interactions efforts have been made to turn all available data into predictive
Regulatory authorities require information to be submitted computational models using quantitative structure–activity
on the potential for interactions to cause adverse effects. relationship methods and molecular modelling [14,25]. In vitro
With the availability of in vitro systems this aspect is, there- data are now generated for many ADME and physicochemical
fore, often considered at early stages of discovery, including end points and can be used to build more robust models.
hit evaluation. Oxidative metabolism by cytochrome P450 Model updating will need to be automated and fitted in the
(CYP) enzymes is, for most drugs, the major route of elimi- data generation cycle. There is a need for both local (project-
nation. As P450s are also capable to metabolise multiple specific) as well as global (general) models. Unfortunately, there
substrates, their inhibition is the major focus of drug–drug is still a paucity of human in vivo data, and, thus, models based
interaction (DDI) studies. CYP3A4 is not only the most on these will need to be handled with care. Interindividual vari-
abundant in the liver, but is also present in the gut wall, and ability within the population is another key factor to take into
is responsible for the metabolism of 50 – 60% of all drugs. account for human predictions. Predictions will be ranges
This enzyme is, therefore, highly susceptible to both reversi- rather than hard numbers. For the development of potential
ble and irreversible (mechanism-based) inhibition [15]. Most drugs, predictions may contribute to high-throughput
CYP3A4 substrates or inhibitors are also P-glycoprotein pharmaceutics and rational drug delivery [26].

2 Expert Opin. Drug Metab. Toxicol. (2005) 1(1)


Van de Waterbeemd

2.5 Simulations: PBPK and PK/PD 2.7 Metabolite identification


Physiologically-based pharmacokinetic (PBPK) models are With increasing resolution of mass spectrometry and nuclear
based on well-known principles in chemical engineering, magnetic resonance it becomes feasible to detect small fractions
and describe the human or animal body as a series of pipes of metabolites. Debate is ongoing to define major versus minor
and tanks [27,28]. Convenient software has now become metabolites. Some metabolites may be pharmacologically active
available and makes PBPK modelling more accessible to and contribute to the overall PK. Reactive metabolites may
drug discovery and development to predict various PK bind to apoproteins and cause idiosyncratic reactions. The chal-
parameters and concentration–time profiles in the body. lenge remains to detect these as early as possible. It has been
Population variability [29] and specific groups such as chil- suggested that time-dependent inhibition should be in standard
Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Memorial University of Newfoundland on 08/01/14

dren and the elderly can also be taken into account. The in vitro screening protocols [20]. Good progress has been made
more experimental data are available, the better the simula- in metabolite prediction [33].
tions, making these approaches of interest in the discovery
and clinical development settings. Some commercial 3. Expert opinion: ADME studies in the future
programmes have put considerable effort in simulating the
absorption process, which is of interest in optimising The pharmaceutical world is rapidly changing. The output of
pharmaceutical formulations. new chemical entities is low despite the enormous investments
Pharmacokinetic/pharmacodynamic (PK/PD) modelling in screening technology. The increasing demands of the regula-
links dose–concentration relationships (PK) to concentra- tory bodies have put more pressure on the delivery of high qual-
tion–effect relationships (PD). This approach helps to simu- ity clinical candidates and ultimately at the end of the pipeline,
late the time course of drug effects depending on the dose thus resulting in safe and efficacious new drugs. As in most
regimen [30]. Specialised software and better understanding other industries, the pharmaceutical industry will invest more
of the applications will help to speed up the clinical in computer predictions and simulations. This seems to be
development. taking off only slowly, and much is still driven on experimental
data. The future no doubt will bring about a move from ‘wet’ to
For personal use only.

2.6 Toxicology and safety prediction ‘web’ profiling. This pharmaceutical ‘model’ may change, with
Drug safety is a great concern to patients and, therefore, to the large pharmaceutical companies increasingly outsourcing to
regulatory bodies. Early prediction gets more and more atten- smaller biotechnology companies. With a better understanding
tion in any drug discovery programme. Simple in vitro screen- of the underlying principles in drug metabolism and pharma-
ing assays for, for example, hERG and other cardiac ion cokinetics, the field will evolve from an empirical one to a truly
channels, genetic toxicology and cytotoxicity, are now rational, in cerebro approach to drug design [4]. In this future
routinely added to the growing battery of biology, ADME new world based on automated screening, model building and
and toxicity/safety screens. Predictive toxicology its still in its decision-making, the acronym ADME may well be redefined as
early days [31]. A promising tool is the integration of ADME, ‘automated decision-making engine’. In any case, the field of
toxicology and pharmacology data to predict side effects, as in ADME (DMPK) is more crucial than ever to drug discovery
the BioPrint approach [32]. and development.

Bibliography 5. SMITH D, SCHMID E, JONES B: • Introduction to the concept of property-


Papers of special note have been highlighted as of Do drug metabolism and pharmacokinetic based drug design.
interest (•) to readers. departments make any contributions to 8. PALMER AM: New horizons in drug
drug discovery? Clin. Pharmacokinet. (2002) metabolism, pharmacokinetics and drug
1. KENNEDY T: Managing the drug
41:1005-1019. discovery. Drug News Perspect. (2003)
discovery/development interface. DDT
(1997) 2:436-444. 6. LIPINSKI CA, LOMBARDO F, 16:57-62.
DOMINY BW, FEENEY PJ: Experimental 9. ABRIEL H, SCHLAEPFER J, KELLER DI
2. KOLA I, LANDIS J: Can the
and computational approaches to estimate et al.: Molecular and clinical deteriminants
pharmaceutical industry reduce attrition
solubility and permeability in drug of drug-induced long QT syndrome: an
rates? Nature Revs. Drug Disc. (2004)
discovery and development settings. iatrogenic channelopathy. Swiss Med. Wkly
3:711-713.
Adv. Drug Del. Revs. (1997) 23:3-25. (2004) 134:685-694.
3. KUBINYI H: Drug research: myths, hype • Original rule-of-five paper. • Good list of marketed drugs with QT
and reality. Nature Revs. Drug Disc. (2003)
7. VAN DE WATERBEEMD H, prolongation.
2:665-668.
SMITH DA, BEAUMONT K, 10. SAUNDERS KC: Automation and robotics
4. EDDERSHAW P, BERESFORD AP, WALKER DK: Property-based design: in ADME screening. DDT Technologies
BAYLISS MK: ADME/PK as part of a optimization of drug absorption and (2004) 1:373-380.
rational approach to drug discovery. DDT pharmacokinetics.
(2000) 5:409-414. 11. JENKINS KM, ANGELES R,
J. Med. Chem. (2001) 44:1313-1333.
QUINTOS MT et al.: Automated high-

Expert Opin. Drug Metab. Toxicol. (2005) 1(1) 3


From in vivo to in vitro/in silico ADME: progress and challenges

throughput ADME assays for metabolic 19. ATKINS WM: Implications of the allosteric 28. POGGESI I: Predicting human
stability and cytochrome P450 inhibition kinetics of cytochrome P450s. DDT (2004) pharmacokinetics from preclinical data.
profiling of combinatorial libraries. 9:478-484. Curr. Opin. Drug Disc. Dev. (2004)
J. Pharm. Biomed. Anal. (2004) 20. TUCKER G, HOUSTON JB, 7:100-111.
34:989-1004. HUANG S-M: Optimising drug 29. YANG J, ROSTAMI-HODJEGAN A,
12. WANG J, URBAN L: The impact of early development: strategies to assess drug TUCKER G: Prediction of fluconazole
ADME profiling on drug discovery and metabolism/transporter interaction interaction with midazolam and triazolam:
development strategy. Drug Disc. World potential – towards a consensus. Eur. J. incorporating population variability. J. Clin.
(2004):73-86. Pharm. Sci. (2001) 13:417-428. Pharmacol. (2001) 52:465-486.
• List of probe substrates and inhibitors for
Expert Opin. Drug Metab. Toxicol. Downloaded from informahealthcare.com by Memorial University of Newfoundland on 08/01/14

13. YU H, ADEDOYIN A: ADME-Tox in 30. ZUIDEVELD KP, VAN DER GRAAF PH,
drug discovery: integration of experimental CYPs. NEWGREEN D et al: Mechanism-based
and computational technologies. DDT 21. HO RJ, SHEN DD: Research on the role of pharmacokinetic–pharmcodynamic
(2003) 8:852-860. membrane transporters in drug disposition: modelling of 5-HT1A receptor agonists:
14. DICKINS M, VAN DE present challenges and future directions. estimation of in vivo affinity and intrinsic
WATERBEEMD H: Simulation models for Curr. Drug Metab. (2004) 5:i-ii. efficacy on body temperature in rats.
drug disposition and drug interactions. 22. DANTZIG AH, HILLGREN KM, J. Pharmacol. Exp. Ther. (2004)
DDT Biosilico (2004) 2:38-45. DE ALWIS DP: Drug transporters and 308:1012-1020.

15. ZHOU S, CHAN E, LIM LY et al.: their role in tissue distribution. Ann. Rep. 31. EGAN WJ, ZLOKARNIK G,
Therapeutic drugs that behave as Med. Chem. (2004) 39:279-291. GROOTENHUIS PD: In silico prediction
mechanism-based inhibitors of cytochrome 23. BALAYSSAC D, AUTHIER N, of drug safety: despite progress there is
P450 3A4. Curr. Drug Metab. (2004) COUDORE F: Drug–drug interactions abundant room for improvement. DDT
5:415-442. linked to a P-glycoprotein inhibition. Technologies (2004) 1:381-387.
• List of CYP3A4 mechanism-based Lett. Pharmacol. (2004) 18:76-80. 32. KREJSA CM, HORVATH D,
inhibitors. 24. AYRTON A, MORGAN P: The role of ROGALSKI SL et al.: Predicting ADME
16. CUMMINS CL, JACOBSEN W, transport proteins in drug absorption, properties and side effects: the BioPrint
For personal use only.

BENET LZ: Unmasking the dynamic distribution and disposition. Xenobiotica approach. Curr. Opin. Drug Disc. Dev.
interplay between intestinal P-glycoprotein (2001) 31:469-497. (2003) 6:470-480.
and CYP3A4. J. Pharmacol. Exp. Ther. 25. VAN DE WATERBEEMD H, 33. LANGOWSKI J, LONG A: Computer
(2002) 300:1036-1045. GIFFORD E: ADMET in silico modelling: systems for the prediction of xenobiotic
17. BLANCHARD N, RICHERT L, towards prediction paradise? Nature Revs. metabolism. Adv. Drug Deliv. Revs. (2002)
COASSOLO P, LAVE T: Qualitative and Drug Disc. (2003) 2:192-204. 54:407-415.
quantitative assessment of drug–drug • Recent review of ADME/toxicity
interaction potential in man, based on Ki, predictions. Websites
IC50 and inhibitor concentration. Curr. 26. VARMA MVS, KHANDAVILLI S,
Drug Metab. (2004) 5:147-156. 101. http://www.torsades.org
ASHOKRAJ Y et al.: Biopharmaceutic
18. ROSTAMI-HODJEGAN A, TUCKER G: classification system: a scientific framework
In silico simulations to assess the in vivo for pharmacokinetic optimization in drug
Affiliation
Han Van de Waterbeemd
consequences of in vitro metabolic drug– research. Curr. Drug Metab. (2004)
Pharmaceutical Consultant in In Vitro and
drug interactions. DDT Technologies (2004) 5:375-388.
In Silico ADME Technologies, UK
1:441-448. 27. LEAHY DE: Progress in simulation Tel: +44 (0) 1622 686 630;
modelling for pharmacokinetics. Curr. E-mail: han.vdwaterbeemd@btinternet.com
Topics Med. Chem. (2003) 3:1257-1268.

4 Expert Opin. Drug Metab. Toxicol. (2005) 1(1)

Вам также может понравиться