pet studies in drug development: methodological considerations

5
TECHNOLOGIES DRUGDISCOVERY TODAY PET studies in drug development: Methodological considerations Vincent J. Cunningham 1, * , Christine A. Parker 2 , Eugenii A. Rabiner 2 , Antony D. Gee 2 , Roger N. Gunn 1 1 Translational Medicine and Genetics, GlaxoSmithKline, 891-995 Greenford Road, Greenford, Middlesex, UK UB6 7HE 2 Translational Medicine and Genetics, GlaxoSmithKline, ACCI, Addenbrookes Hospital, Hills Road, Cambridge, UK CB2 2GG Positron emission tomography (PET), as an in vivo pharmacological imaging tool in experimental medi- cine, is playing an increasing role in drug development. There are two areas where PET is particularly useful in this respect, namely biodistribution and drug occu- pancy studies. Radiotracer design, the properties of the molecular targets which can be studied and the quantitative estimation of pharmacological endpoints will be discussed in relation to these applications, with particular reference to studies in brain. Section Editor: Adriaan A. Lammertsma – Department of Nuclear Medicine and PET Research, Vrije Universiteit Amsterdam, The Netherlands Introduction In addition to the study of underlying mechanisms of disease, there are currently two areas where positron emission tomo- graphy (PET) is particularly useful and can have a direct impact on the drug development process, namely biodistri- bution and drug occupancy studies. For biodistribution studies, the drug itself is radiolabelled. PET can then provide quantitative information on its deliv- ery and turnover in specific tissues following intravenous administration to the subject under study. The obvious advantage is that the concentration of the drug can be measured directly and noninvasively in the tissue of interest, rather than by biopsy or by inference from indirect or surrogate measures. The question being asked might be relatively simple: Does the drug enter the tissue or can it be part of a larger project aimed at relating the local tissue concentration time course to that in plasma or to the admi- nistered dose? Drug occupancy studies are asking a related but distinct question: What is the quantitative relationship between the dose of drug or its concentration in plasma and the occu- pancy by that drug of a specific molecular target? Here the approach is to label the target with a radioligand, that is, a radiotracer which binds specifically to that target, and to observe with PET the blocking of the binding of the trace radioligand when the cold (i.e. nonradiolabelled) drug is administered at pharmacological doses. General methodology Choice of radionuclide and labellability PET radiotracers are commonly labelled with the short-lived positron-emitting radionuclides 11 C, 18 F, 13 N and 15 O (radio- active half-lives: 20.4, 110, 10 and 2 min, respectively). For many biological applications, the time constants for uptake and turnover of small molecules in brain make 11 C and 18 F optimal choices of label with half-lives long enough to be followed during the course of the PET experiment. Typically this manifests itself in the use of a labelled autologue of the drug candidate measured over a few hours, or observing on the kinetic behaviour of a trace radioligand over a similar time Drug Discovery Today: Technologies Vol. 2, No. 4 2005 Editors-in-Chief Kelvin Lam – Pfizer, Inc., USA Henk Timmerman – Vrije Universiteit, The Netherlands Imaging technologies *Corresponding author: V.J. Cunningham ([email protected]) 1740-6749/$ ß 2005 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.ddtec.2005.11.003 www.drugdiscoverytoday.com 311

Upload: vincent-j-cunningham

Post on 24-Nov-2016

213 views

Category:

Documents


1 download

TRANSCRIPT

TECHNOLOGIES

DRUG DISCOVERY

TODAY

PET studies in drug development:Methodological considerationsVincent J. Cunningham1,*, Christine A. Parker2, Eugenii A. Rabiner2,

Antony D. Gee2, Roger N. Gunn1

1Translational Medicine and Genetics, GlaxoSmithKline, 891-995 Greenford Road, Greenford, Middlesex, UK UB6 7HE2Translational Medicine and Genetics, GlaxoSmithKline, ACCI, Addenbrookes Hospital, Hills Road, Cambridge, UK CB2 2GG

Drug Discovery Today: Technologies Vol. 2, No. 4 2005

Editors-in-Chief

Kelvin Lam – Pfizer, Inc., USA

Henk Timmerman – Vrije Universiteit, The Netherlands

Imaging technologies

*Corresponding author: V.J. Cunningham ([email protected])

1740-6749/$ � 2005 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.ddtec.2005.11.003

Section Editor:Adriaan A. Lammertsma – Department of Nuclear Medicineand PET Research, Vrije Universiteit Amsterdam,The Netherlands

Positron emission tomography (PET), as an in vivo

pharmacological imaging tool in experimental medi-

cine, is playing an increasing role in drug development.

There are two areas where PET is particularly useful in

this respect, namely biodistribution and drug occu-

pancy studies. Radiotracer design, the properties of

the molecular targets which can be studied and the

quantitative estimation of pharmacological endpoints

will be discussed in relation to these applications, with

particular reference to studies in brain.

Introduction

In addition to the study of underlyingmechanisms of disease,

there are currently two areas where positron emission tomo-

graphy (PET) is particularly useful and can have a direct

impact on the drug development process, namely biodistri-

bution and drug occupancy studies.

For biodistribution studies, the drug itself is radiolabelled.

PET can then provide quantitative information on its deliv-

ery and turnover in specific tissues following intravenous

administration to the subject under study. The obvious

advantage is that the concentration of the drug can be

measured directly andnoninvasively in the tissue of interest,

rather than by biopsy or by inference from indirect or

surrogate measures. The question being asked might be

relatively simple: Does the drug enter the tissue or can it

be part of a larger project aimed at relating the local tissue

concentration time course to that in plasma or to the admi-

nistered dose?

Drug occupancy studies are asking a related but distinct

question: What is the quantitative relationship between the

dose of drug or its concentration in plasma and the occu-

pancy by that drug of a specific molecular target? Here the

approach is to label the target with a radioligand, that is, a

radiotracer which binds specifically to that target, and to

observe with PET the blocking of the binding of the trace

radioligand when the cold (i.e. nonradiolabelled) drug is

administered at pharmacological doses.

General methodology

Choice of radionuclide and labellability

PET radiotracers are commonly labelled with the short-lived

positron-emitting radionuclides 11C, 18F, 13N and 15O (radio-

active half-lives: 20.4, 110, 10 and 2 min, respectively). For

many biological applications, the time constants for uptake

and turnover of small molecules in brain make 11C and 18F

optimal choices of label with half-lives long enough to be

followed during the course of the PET experiment. Typically

this manifests itself in the use of a labelled autologue of the

drug candidatemeasured over a fewhours, or observing on the

kinetic behaviour of a trace radioligand over a similar time

www.drugdiscoverytoday.com 311

Drug Discovery Today: Technologies | Imaging technologies Vol. 2, No. 4 2005

periodduring the course of administration of pharmacological

doses of the drug under study.

PET radionuclides are obtained directly from a cyclotron in

the form of simple molecules or ions (e.g. 11CO2,18F-fluor-

ide). These primary labelling materials are converted into

more useful synthetic reagents to enable their incorporation

into the target drug molecule. Because of the short half-lives

of these radionuclides, the repertoire of available synthetic

chemical manipulations is limited. Thus, not all drug candi-

dates can be labelled. A common example of a successful

labelling strategy for carbon-11 compounds is the use of11CO2 which is subsequently converted to 11C-methyl iodide

by a series of rapid chemical manipulations. This reagent can

be used to alkylate O, S, N and C nucleophiles to produce the

corresponding 11C-methylated product. This methodology is

often successfully utilised for the labelling of N-methyl or O-

methoxy containing compounds. For drug molecules not

containing these moieties, alternative labelling approaches

are required. Other labelling reagents available for rapid

labelling include 11C-cyanide, 11C-nitromethane, 11C-phos-

gene and 11C-carbon monoxide. Continuing development of

new synthetic reagents and reactions with radionuclides is of

crucial importance for increasing the ability to implement

successful labelling strategies for PET studies [1].

It is usually the case in drug occupancy studies that the

radioligand has a different molecular structure than the drug.

Indeed some properties of an ‘ideal’ radiotracer are different

from those of the drug, for example, the drug might interact

with several targets but the occupancy study would hope to

answer questions regarding a specific target defined by the

radioligand. Self-occupancy studies, when the drug and the

radioligand are of the same molecular species, do however

have a role to play, particularly in the development of PET

tracers.

Radiolabelled metabolites

PET measures the time course of the total concentration of

radioactivity in the tissue. In the case of CNS studies, if care is

taken in the design of the labelling position in the tracer

molecule, radioactive metabolites generated in the periphery

can be sufficiently hydrophilic to be excluded from tissue

penetration by the blood brain barrier (BBB). Knowledge of

the metabolism of the parent molecule is essential for label-

ling the PET probe in a position whereby brain penetrant

metabolites can be minimised. The fraction of radiolabelled

metabolites can be assayed directly in plasma samples to

enable a proper definition of a parent input function for

kinetic analyses, but because of short half-lives these data

might be limited. Accurate assessment of the contribution of

radiolabelled metabolites in plasma and in the tissue to the

total PET signal is crucial, both in biodistribution and occu-

pancy studies, if the data from a PET experiment are to be

interpreted accurately.

312 www.drugdiscoverytoday.com

Kinetic analysis

Mathematical modelling techniques play an essential role in

the interpretation of kinetic PET data [2]. The tissue signal is

dependent on the interrelated processes of tracer delivery in

the blood, transport across the BBB and exchange between

tissue compartments. For radioligands, it is also necessary to

distinguish the free and nonspecifically bound components

from those specifically bound to the target molecule or

receptor. For drug occupancy studies it is usually the last of

these compartments which is of interest. It is important to

emphasise that it is only under special conditions that there is

a simple linear relationship between the tissue/plasma con-

centration ratio and the size of the specifically bound pool,

and in general this is not the case. It would, for example,

require that the concentration of the radioligand is main-

tained constant and at a true equilibrium in both plasma and

tissue. Under conditions where the tracer is administered as

an intravenous bolus, a full kinetic analysis, based on a

validated model, is required to distinguish the components

involved in the total tissue signal and to obtain estimates of

the sizes of the component compartments and the flux

between them.

Labelling the drug: biodistribution studies

In the development of potentially centrally acting drugs, a

basic question is whether the drug crosses the BBB. It is well

recognised that predicting blood–brain permeability for a

given chemical structure is currently very difficult, and there

might be species differences which further confound the

extrapolation from animal studies to human. The adminis-

tration of trace quantities of a radiolabelled drug and its direct

assay in the tissue of interest, as is possible with PET, is often

referred to as ‘microdosing’ and has obvious advantages in

early drug development when the toxicology and pharma-

cology of the drug are less well defined.

Assessment of brain entry

Measurement of the concentration time courses of the radi-

olabelled drug in plasma and in brain allows quantitative

estimation of plasma-to-brain clearance (ml plasma per min

per ml of tissue, K1, equal to the product of flow and extrac-

tion). The data might also allow estimation of an equilibrium

partition coefficient between tissue and plasma. The regional

PET signal contains contributions from the tissue and from a

blood volume component (�5%, v/v). A tracer kinetic model

can be applied to the dynamic PET data, using the plasma

radiotracer concentration as the input function and includ-

ing a blood volume term to allow for the concentration of

tracer in whole blood, to estimate the uptake parameters. For

tracers exhibiting low tissue uptake it is particularly impor-

tant to account for the contribution that the blood volume

and other factors such as out-of-field scatter, make to the PET

signal. A supplementarymeasurement of blood volume based

Vol. 2, No. 4 2005 Drug Discovery Today: Technologies | Imaging technologies

on a radiolabelled carbonmonoxide scan can allow for amore

accurate correction. Interpretation of these parameters must

however take into account the limits of detection.

Nonlinear effects of dosing

In a microdosing study it might be the case that there is no

quantifiable brain uptake of the radioligand, that is, the

radiotracer might not be able to cross the BBB. It could

however be possible that the radiotracer is a substrate for

one or more of the drug efflux transporters such as P-glyco-

protein (Pgp),multidrug resistance proteins or organic anion

transporting polypeptides. These transporters are located in

the endothelial cells of the brain capillaries and keep a tight

regulation on the influx of endogenous and exogenous

agents into the CNS. If it is suspected that the radiotracer

is a substrate for oneof the efflux transporters, further studies

can be performed to assess this phenomenon, for example,

following a higher pharmacological dose of the cold drug,

when saturation of the Pgp might allow uptake of the radio-

tracer. For amore detailed review of the application of PET in

studying the drug efflux transporters in vivo see Loscher and

Potschka [3]. Conversely, if uptake of the radiotracer

involves saturable processes then the fractional uptake of

the radiotracer in a microdosing study will be higher than

that of the cold drug at pharmacological doses. Similar

considerations of nonlinearity apply to the metabolism of

the parent tracer. In certain cases, the metabolism of the

parentmolecule is rapid at tracer doses, whereas for the same

tracer, the situation can be reversed if administered at a

pharmacological dose.

Labelling the target: occupancy studies

In many studies, compounds labelled with positron-emitting

isotopes have previously been characterised for a range of

properties including their selectivity and affinity for target

proteins of interest. This information allows the design of PET

studies that aim to answer specific questions relating to the

characterisation of particular target proteins. Therefore, if a

compound has previously been demonstrated to possess a

high affinity and selectivity for one particular target, then an

occupancy study can be designed where this compound is

labelled with a positron-emitting isotope and administered at

nonpharmacological (trace) doses before and after increasing

pharmacological doses of a structurally dissimilar compound

(which also has an affinity for the target protein of interest).

In studies such as these where the radioligand only labels one

target protein, drugs with an affinity for more than one

protein can be utilised to demonstrate occupancy of the

target protein. The unlabelled compoundmust have a reason-

ably high affinity for the target protein of interest and any

affinity for other proteins must not interfere with the PET

signal, either directly or indirectly. Occupancy studies can

also be designed with radioligands that bind to more than

one target protein. In cases like these, a careful study design

needs to be adopted which will allow for the full block of the

proteins not of interest, followed by assessment of the target

proteins of interest.

Radioligands

Our understanding of small molecule–receptor/tissue inter-

actions is incomplete. Many of the assumptions we use in the

selection and design of radioligands are at best based on a

crude understanding of a very complicated in vivo system.

Successful radioligand development for in vivo imaging is at

present not an exact science, and further advances are

required in understanding small molecule-protein interac-

tions to ensure more efficient selection and design of PET

radioligands in future. In the selection and design of radi-

oligand probes for in vivo imaging, several desirable and/or

essential molecular attributes need to be considered. Two

important factors are lipophilicity and affinity.

In general, successful radioligands are characterised by a

‘window of lipophilicity’: They have sufficient lipophilicity

that they are able to diffuse across biological membranes, but

not so lipophilic that their concentration in lipid-rich tissue

obscures the binding of the radioligand at the molecular

target of interest [4]. In practical terms, several measures have

been used to establish this optimum lipophilicity window,

themost commonly used being the logarithmof the partition

of a molecule between octanol and water (often termed log P,

or log D if performed with the aqueous phase at pH 7.4).

Typically, successful radioligands for CNS applications have

measured log P values in the range 1.5–2.5.

The affinity of a radioligand should be compatible with the

known concentration of the molecular target under investi-

gation. In practical terms,molecular targets expressed in high

concentrations (e.g. dopamine transporter in striatum), can

be delineated with a radioligand with a lower affinity (10–

30 nM) than that required for a target which is present in low

concentrations in tissue (e.g. D2 receptors in cortex require a

radioligand with sub-nanomolar affinity).

Estimation of receptor availability: binding potential

The three-tissue compartment neuroreceptor binding model

described by Mintun et al. [5] has provided a basis for the

kinetic analysis of radioligand studies in PET (e.g. see Ref. [6]).

Thismodel distinguishes three states of the radioligand – free,

nonspecifically bound and specifically bound. The ratio of

the concentration of the radioligand in the specifically bound

state over that of the free state which would occur at tracer

equilibrium is termed the binding potential (BP) and can be

shown, in the simplest case, to equal the ratio of the con-

centration of available receptor sites to the dissociation con-

stant of the radioligand for the receptor (Bavail/KD). Whereas

this is an equilibrium concept, it can be estimated from

analysis of the kinetic behaviour of the radioligand in a

www.drugdiscoverytoday.com 313

Drug Discovery Today: Technologies | Imaging technologies Vol. 2, No. 4 2005

dynamic PET scan. The BP of the radioligand is a function of

the concentrations and affinities of competing cold ligands,

either endogenous or exogenous, by virtue of their effect on

Bavail. The concentrations of these cold ligands might be in or

near to a steady state and it is the kinetic behaviour of the

radioligand on this background which is of interest. It is the

changes in the BP of the radioligand which indicate occu-

pancy of the receptor sites by the cold drug. In this brief

account we will avoid the use of the loose term ‘radioligand

displacement studies’ because it is potentially misleading,

and has been used incorrectly, for example, to suggest that

a radioligand with a very high affinity is unsuitable for

estimating site occupancy by a cold drug with a relatively

lower affinity because it would not be displaced to the same

extent as a lower affinity radioligand. Consideration of the

above shows this is not the case.

Often, with typical PET data, the individual (micro) rate

constants in the standard PET neuroreceptor binding model

are not well defined andmight be numerically unidentifiable.

However, so-called macro parameters such as K1 (the unidir-

ectional clearance of tracer from plasma to tissue), and the

equilibrium partition coefficient for the total tissue/plasma

concentration ratio, are relatively well defined. (In PET lit-

erature the latter is frequently referred to as a total volume of

distribution, VDtot, in contrast to the use of this term in other

contexts.)

The existence of a reference region for the radioligand, that

is, a region of brain where there is negligible specific binding,

when this occurs, is of particular value if it can be assumed

that the size of the free and nonspecific compartments is the

same in reference and target tissues. If an arterial plasma

parent input function is available (the ideal case) then a

comparison of VDtot in reference and target allows estimates

of binding potential, BP1, to be obtained (BP1 = f1Bavail/KD,

where f1 is the fraction of the tracer in plasma which is free

and unbound to plasma protein, lipid among others; Bavail is

the concentration of available receptor sites in the tissue, and

KD the apparent equilibrium dissociation constant for the

ligand), together with BP2, the corresponding expression

incorporating the free fraction in the tissue, f2. A second

important consequence of the existence of a reference region

is the possibility of avoiding arterial blood sampling alto-

gether and expressing the target tissue kinetics as a function

of those of the reference tissue [2]. This allows estimates only

of BP2. In all these cases, however, the obvious points should

be emphasised, including the fact that in a tracer-alone

experiment, changes in Bavail and KD cannot be distinguished

and that changes in BP can also reflect changes in free

fractions. However, if these assumptions are clear and satis-

factory, the way is open for the very powerful techniques of

estimating receptor occupancy by a drug, either for the

purpose of simply demonstrating that the drug has access

to the site when direct biodistribution data cannot be

314 www.drugdiscoverytoday.com

obtained or for establishing a quantitative relationship

between dose and occupancy.

Brain penetration and receptor occupancy at pharmacological doses

As a simple example here, let us consider a case of a baseline

scan (radioligand alone) together with scans carried out with

three increasing doses of the cold drug under study. Let us

assume that these scans are carried out with no allowance for

interim analysis and adaptive design of doses. Let us also

assume a simple single-site competition model. The actual

range of the three doses will depend on the question being

asked; however, the key factor in determining the doses will

be an initial estimate of the in vivo IC50 for the compound

under investigation. This could be based on allometric scaling

[7] or extrapolation from in vitro studies. So, for the two

questions which are typically asked:

� C

NS penetration: Here the goal is to investigate the

hypothesis that the cold drug gets into the brain. Given

the focus of the question it is important to settle for a high

enough dose to avoid any false negative (Type II error).

Thus, a reasonable scheme for dose selection in our simple

example is the three doses which are D/10, D, 10D (subject

to consideration of themaximum tolerable dose). A reason-

able scaling of this range is to choose D to be the estimated

IC50. If this estimate is correct and the compound is CNS

penetrant, this should lead to occupancy values of 9%, 50%

and 91%. However, based on the need to avoid a Type II

error and one’s confidence in the predicted IC50, itmight be

appropriate to choose D to be greater than the estimated

IC50. So, themain requirement is to have an estimate of the

IC50 and some confidence interval for this estimate.

� D

ose–occupancy curve: Here the goal is to obtain a dose

occupancy curve and measure the IC50 of the compound

directly. For such studies one might employ a tighter range

of doses than for a CNS penetration study, given that an

accurate estimate of the IC50 is required. A possible strategy

is to aim for 25%, 50% and 75% occupancy based on doses

of D/3, D, 3D (subject to maximum tolerable dose). How-

ever, it might be prudent to alter the range of doses

depending on one’s confidence in the estimate of the

IC50 (=D), but again an optimal design centres on having

an initial estimate of the IC50.

It is important to note here that even if one is interested in

establishing a dose of drug with a particular target occupancy

inmind, it is better to attempt to define the whole occupancy

curve from low to high. A standard receptor-binding model

can then be used to interpolate or extrapolate to the desired

occupancy, circumventing signal-to-noise problems at high

and low occupancies. The above simple examples are for

illustrative purposes. Then, for studies involving more

scans/subjects, one can use more intelligent adaptive designs

Vol. 2, No. 4 2005 Drug Discovery Today: Technologies | Imaging technologies

in which the subsequent doses that are used can be altered

depending on values obtained from previous scans.

Thus, it is clear that PET occupancy studies can help define

an optimal dose, particularly if the target occupancy is pre-

defined. However, it goes without saying that extrapolations

from PETmeasures to optimal dose are heavily dependent on

assumptions regarding the mode of action of the drug and its

relationship to the disease process, and that it is necessary to

be very clear about what constitutes ‘optimal’. It might be

useful, when considering the contributions that PET studies

could make to the establishment of an optimal dosing regi-

men to consider the PET study as providing a ‘snapshot’, in

time, of the availability of the drug at its site of action. The

bioavailability of the drug under study, usually administered

orally, is a function of its absorption and whole body meta-

bolism and excretion. The PET tracer is usually administered

intravenously and gives a tissue response function relative to

plasma, at specific times, during this period. An early example

is the study by Bench et al. [8] looking at the clearance of a

drug from a receptor following a study in which a dose–

response curve had been established. Here we are merely

concerned with some specific and basic methodological con-

siderations to be taken into account for the type of PET

studies outlined above.

Conclusion

In this brief overview of PET methodology, we have touched

upon a few selected topics which are by no means compre-

hensive. PET is a multidisciplinary modality in Experimental

Medicine, relying on contributions from Physics, Chemistry,

Mathematics and Biology.

Advances which will impact on the field will undoubtedly

come from further developments in chemistry, in particular a

better understanding of the relationship between properties

of compounds in silico and their behaviour as tracers in vivo. In

addition to the pharmacology associated with specifically

bound state of the tracer, its behaviour in its free and non-

specifically bound states is equally important. The free frac-

tion of the tracer in tissue is amajor factor in determining the

suitability of a compound as a PET tracer. The further devel-

opment and validation of reference tissue modelling is cru-

cially dependent on a better understanding in this area. In

addition, more knowledge of factors affecting the free

fraction of the tracer in plasma and improvements in the

methodology of assaying this in individual subjects is

required, not only for tracer design but also for more accurate

quantitation of the PET signal.

We have emphasised the necessity for appropriate model-

ling to take full advantage of the quantitative PET signal.

Although there is a growing body of accepted methodologies

for quantitative analysis, it is important to carefully examine

the assumptions and limitations of these methods in each

individual case and not lose sight of the underlying biology

and pharmacology. It is the quantitative nature of the data

and their analysis that will allow for appropriate objective

decisions to be made during the drug development process.

Here we have concentrated on particular current applica-

tions, namely biodistribution and occupancy studies in brain.

There is also an increasing interest in applications of similar

techniques in other tissues. Such studies are being employed

to accelerate drug discovery and development, and are of

great value, enabling noninvasive measurements in humans

early in the drug development pipeline. There aremany other

potential applications of PET that could ultimately impact on

this area. These include techniques for imaging gene expres-

sion, characterising the downstream response to drugs and

the characterisation of novel metabolic pathways.

References1 Halldin, C. et al. (2001) Brain radioligands – state of the art and new trends.

Q. J. Nucl. Med. 45, 139–152

2 Gunn, R.N. et al. (2001) Positron emission tomography compartmental

models. J. Cereb. Blood Flow Metab. 21, 635–652

3 Loscher, W. and Potschka, H. (2005) Role of drug efflux transporters in the

brain for drug disposition and treatment of brain diseases. Prog. Neurobiol.

76, 22–76

4 Waterhouse, R.N. (2003) Determination of lipophilicity and its use as a

predictor of blood–brain barrier penetration of molecular imaging agents.

Mol. Imaging Biol. 5, 376–389

5 Mintun, M.A. et al. (1984) A quantitativemodel for the in vivo assessment of

drug binding sites with positron emission tomography. Ann. Neurol. 15,

217–227

6 Laruelle, M. (2000) Imaging synaptic neurotransmission with in vivo

binding competition techniques: a critical review. J. Cereb. Blood Flow

Metab. 20, 423–451

7 Mordenti, J. and Chapell, W. (1989) The use of interspecies scaling in

toxicokinetics. In Toxicokinetics and NewDrug Development (Yacobi, A., Kelli,

J., Batra, V., eds), pp. 42–96, Pergamon Press

8 Bench, C.J. et al. (1996) The time course of binding to striatal dopamineD2

receptors by the neuroleptic ziprasidone. Psychopharmacology 124, 141–

147

www.drugdiscoverytoday.com 315