Vous êtes sur la page 1sur 11

Current Medicinal Chemistry, 2004, 11, 267-277 267

Schizophrenia: From Dopamine to Glutamate and Back


M.L. Carlsson1,2*, A. Carlsson1,2 and M. Nilsson1,2

1Institute of Clinical Neuroscience, Neuropsychiatric Research Unit, Göteborg University,


Sweden and 2A. Carlsson Research AB, Göteborg, Sweden
Abstract: The first part of the present review describes the exciting journey of
dopamine stabilizers, starting in the early eighties with the development of the partial
dopamine agonist (-)-3-PPP of phenylpiperidine structure, via various compounds with
aminotetraline structure with preferential autoreceptor antagonist properties, and then
back again to phenylpiperidine compounds carrying substituents on the aromatic ring
that transformed them from partial dopamine agonists to partial dopamine receptor
antagonists, such as OSU6162. OSU6162 was brought to the clinic and has in preliminary
trials showed antidyskinetic and antipsychotic efficacy. The second part of this review describes results from a
hypoglutamatergia mouse model for cognitive symptoms of schizophrenia, where we have tested traditional
neuroleptics, new generation antipsychotics with marked 5-HT2 vs dopamine D2 receptor blockade as well as a
dopamine stabilizer belonging to the partial dopamine receptor antagonist category.
Keywords: schizophrenia, dopamine stabilizer, cognition, video tracking, glutamate, MK-801, mouse

INTRODUCTION with respect to vocational and social functioning, and


quality of life. Admittedly the so-called atypical
The most important basis for currently used
antipsychotic agents, clozapine being the most convincing
antipsychotics is dopamine D2 receptor blockade but
example, can induce an antipsychotic action at a lower
unfortunately this suppression of dopaminergic tone also
dopamine receptor occupancy than the classical agents, but
entails serious motor and mental side effects. The motor side
they can still cause hypodopaminergia with resultant side
effects (extrapyramidal side effects; EPS) can be reduced by
effects. Moreover, the currently used atypical antipsychotic
careful dosage, because they occur at somewhat higher
agents have been found to have other side effects, such as
dopamine receptor occupancy than that generally needed for
weight gain and insulin resistance, which may lead to
antipsychotic action. This does not seem to be true of the
diabetes.
mental side effects, however, since they, similarly to the
antipsychotic action, seem to reside largely in the ventral Dopaminergic stabilizers are antipsychotics with much
striatum, which seems to be more sensitive to dopamine less or none of those side effects that are a consequence of
receptor blockade than the dorsal striatum, where the EPS hypodopaminergia, because they can suppress dopamine
appear to be located. Some of the mental side effects involve functions at most moderately below baseline. They can thus
an interference with the reward system and show up as alleviate abnormal motor and mental hyperactivity with but
anhedonia and dysphoria. Another mental side effect is slight, if any, risk of attaining activity levels below
impairment of cognition, which may result from D2 receptor baseline. In fact, many of them can stimulate low-baseline
blockade both in the striatum and in the cerebral cortex. The behavior. In other words, a dopamine stabilizer is a
mental side effects are experienced by many patients as a compound that can influence behavior in different directions,
high, sometimes too high, price to be paid for the depending on the baseline.
antipsychotic action. Dopaminergic stabilizers may be divided into two
There is now considerable evidence, mainly from classes: Partial dopamine receptor agonists and partial
imaging studies [1-7] that dopaminergic presynaptic tone is dopamine receptor antagonists.
elevated in schizophrenia during the active psychotic state.
However, this may not occur in all patients and may not PARTIAL DOPAMINE RECEPTOR AGONISTS
involve all dopamine systems. Moreover, it does not seem
to occur, at least not to the same extent, in schizophrenic (-)-3-PPP
patients in remission. From a therapeutic point of view this The dopamine stabilizer project was originally aimed at
is important, because many schizophrenic patients need developing dopaminergic agonists with better
antipsychotic medication even in remission in order to pharmacokinetic properties and less side effects than
prevent relapse. apomorphine. This goal was never reached, but instead a
Thus, the currently available antipsychotic agents have compound with unusual properties, i. e. 3-(3-hydroxyphe-
serious limitations, in many cases leading to a poor outcome nyl)-N-n-propylpiperidine (3-PPP), emerged [8]. It was
discovered in the seventies that the receptor-mediated
*Address correspondence to this author at the Göteborg University,
feedback mechanism regulating the activity of dopaminergic
Institute of Clinical Neuroscience, Neuropsychiatric Research Unit, neurons is, at least partly, mediated by receptors located on
Guldhedsgatan 19, SE-413 46 Göteborg, Sweden; Tel: +46 73 693 96 22; the dopaminergic neurons themselves [9]. Stimulation of
E-mail: maria.carlsson@psychiat.gu.se these receptors, which later were shown to be almost

0929-8673/04 $45.00+.00 © 2004 Bentham Science Publishers Ltd.


268 Current Medicinal Chemistry, 2004, Vol. 11, No. 3 Carlsson et al.

exclusively of the D2 subtype, causes inhibition of the case the preferential activation of the autoreceptors presents
dopaminergic neuron with respect to firing, release, as a biphasic response: in low dosage this activation leads to
synthesis and metabolism of dopamine. Sometimes these behavioral inhibition, whereas in higher dosage the
receptors are called presynaptic, but a preferable term may be postsynaptic receptor activation takes over resulting in
autoreceptors, partly because they are largely located behavioral stimulation. For a partial agonist there is enough
extrasynaptically, partly because they should be agonism to stimulate the autoreceptors and induce behavioral
distinguished from presynaptic heteroreceptors. inhibition, but not enough to stimulate the postsynaptic
In various in-vivo models 3-PPP at first seemed to receptors to the extent of causing behavioral stimulation.
behave as a selective dopamine autoreceptor agonist, The response to, for example, (-)-3-PPP will thus be
inhibiting the synthesis and metabolism of dopamine and inhibitory and monophasic. However, in contrast to pure
locomotor activity of normal animals, whereas in animals dopamine receptor antagonists, partial agonists are unable to
depleted of dopamine it was unable to stimulate behavior. In bring down the behavioral activity to zero and to induce
other words, it seemed to stimulate the autoreceptors but be catalepsy. Their behavioral inhibition will level out, the
devoid of stimulating activity on postsynaptic receptors [8]. extent of inhibition being inversely related to their intrinsic
activity.
However, when shortly afterwards the two enantiomers of
3-PPP were separated, the (+)-form was found to behave From a clinical point of view the pharmacological profile
more or less like the full classical dopamine receptor agonist of a partial agonist such as (-)-3-PPP, as it shows up in
apomorphine, whereas the (-)-form turned out to be a partial animal models, would seem attractive for two reasons: first
dopamine receptor agonist [10-12]. In low dosage both this agent is unable to induce catalepsy, suggesting a low
enantiomers were found to induce behavioral inhibition, liability for EPS as well as other signs of dopaminergic
which was largely due to autoreceptor stimulation. However, hypoactivity. Secondly, as a partial agonist it is capable of
in higher doses they differed in that only the (+)-form was antagonizing the hyperactivity induced by a full agonist such
able to stimulate behavior. When the two enantiomers were as apomorphine, (+)-3-PPP, and dopamine itself. Thus, if
combined, as in the racemate, the (-)-form blocked the dopaminergic hyperactivity is involved in schizophrenia, a
stimulation induced by the (+)-form. That is why the partial agonist should be able to alleviate psychosis as
racemate mistakenly had been interpreted as being a selective effectively as a pure dopamine receptor antagonist, and this
autoreceptor agonist, devoid of activity on the postsynaptic should be achieved without the risk of inducing severe
receptors. hypodopaminergia.

Both enantiomers of 3-PPP, and especially the (-)-form, (-)-3-PPP, also called preclamol, has undergone trials
have been extensively studied in various animal models. mainly in schizophrenic patients, and been found to possess
Electrophysiological as well as biochemical studies antipsychotic activity and to be entirely devoid of
demonstrate their agonist properties on both dopamine extrapyramidal or any other serious side effects. However, its
autoreceptors and postsynaptic receptors, the (-)- form being antipsychotic activity, although pronounced following
a partial and the (+)-form a full agonist [11, 12]. treatment for one week, did not last much longer in spite of
continuous treatment [13]. Possibly, this loss of therapeutic
In order to understand the pharmacological profile of full activity is due to a too high intrinsic activity of this
or partial dopamine agonists it is essential to recognize the compound resulting in reduced autoreceptor sensitivity. In
different responsiveness of autoreceptors and postsynaptic support of this hypothesis, Tamminga had earlier found that
receptors. The autoreceptors are to a large extent located even full dopamine receptor agonists such as apomorphine
outside the dopaminergic synapses. This is clearly the case and its congener NPA possess antipsychotic properties but
of the autoreceptors located to the dopaminergic cell bodies that in that case the antipsychotic action was lost within a
and dendrites but may well be true of a major portion of the few days [13]. A logical conclusion would thus be that a
terminal autoreceptors as well. It is therefore natural that the partial dopamine receptor agonist with an intrinsic activity
autoreceptors will have to be geared to a lower concentration lower than (-)-3-PPP should possess a more long-lasting
of dopamine than the postsynaptic receptors in order to be antipsychotic activity.
functionally active; most, but perhaps not all (see below), of
the latter are probably exposed to the high dopamine levels In rats (-)-3-PPP has been able to antagonize catalepsy
in the synaptic cleft. induced by haloperidol, which supports its partial agonistic
properties [14]. Likewise, in two different clinical studies
The varying responsiveness of receptors to agonists is this agent has been found to possess a mild anti-Parkinson
sometimes described in terms of "receptor reserve". It is true action [15, 16]. Thus, even at the clinical level, (-)-3-PPP
that a high responsiveness shows up as a high receptor appears to fulfil the criteria for a dopaminergic stabilizer,
reserve, but this term is somewhat misleading, because it that is, it will antagonize both hyper- and hypoactivity of
tends to indicate that we are dealing with a variation in the dopaminergic system.
receptor number rather than coupling of the receptor
molecule to other components in the receptor complex. The
latter alternative is probably more relevant because the Aripiprazole
supersensitivity induced by denervation develops before any The Japanese drug company Otsuka, in collaboration
increase in receptor density. with Bristol-Myers Squibb, has developed aripiprazole,
The higher responsiveness of the autoreceptors, as which is a partial dopamine receptor agonist with a lower
compared to the postsynaptic receptors, shows up somewhat intrinsic activity than (-)-3-PPP [17]. Indeed, this agent has
differently for a full than for a partial agonist. In the former been found to have antipsychotic properties that seem to be
Schizophrenia: From Dopamine to Glutamate and Back Current Medicinal Chemistry, 2004, Vol. 11, No. 3 269

longlasting, apparently at least up to one year, as far as can (-)-5-hydroxy-2-(n-dipropylamino)tetraline. Addition of a


be judged by the now available, limited data. The partial methyl group in position 1 yielded two different cis-
dopamine agonist profile of this agent has been enantiomers. The (-)-form turned out to be a partial
demonstrated both in vitro, where it has been shown to dopamine receptor agonist and the (+)-form an antagonist
possess a high affinity to dopamine D2 receptors, and in devoid of intrinsic activity. The O-methylated derivative of
animals [18]. Moreover, imaging studies in humans show the latter had a similar profile and was more metabolically
that therapeutic doses of aripiprazole can yield D2-receptor stable. This agent, called (+)-UH232 (Fig (1)), as well as its
occupancies clearly higher than 80 percent without any EPS, monopropyl (N-n-propyl) analog (+)-AJ76, were
except for a certain degree of akathisia at very high levels of characterized pharmacologically and turned out to have a
occupancy [19]. This is in contrast to dopamine receptor dopamine-stabilizing profile, that is, they antagonized the
antagonists, which are known to cause EPS at occupancies hyperactivity induced by dopaminergic agonists without
above 80 percent, and clearly supports the view that changing the behavioral activity level of normal rats
aripiprazole is a partial dopamine receptor agonist also in exploring a new environment to any marked extent.
humans. This is further supported by this agent's ability to Moreover, they stimulated the behavior of animals with a
reduce rather than increase serum prolactin levels [20]. The low activity level habituated to their environment and were
clinical pharmacology of aripiprazole has been reviewed in a devoid of cataleptogenic action. Virtual absence of intrinsic
recent paper by Keck and McElroy [21], where it was activity was shown after previous dopamine depletion and in
concluded that aripiprazole has shown antipsychotic efficacy the GBL model used for testing the activity of dopaminergic
both in short-term (4 weeks) and long-term (26 and 52 autoreceptors. Moreover, dopamine receptor antagonism
weeks) controlled trials. As indicated above the risk of showed up as a strong stimulation of dopamine synthesis,
aripiprazole causing EPS seems low and prolactin levels are turnover and release, similar to the action of typical
not elevated; in addition it does not appear to significantly dopamine receptor antagonists like haloperidol and
alter QTc interval or induce weight gain. Aripiprazole has chlorpromazine. A compound with this profile would be
recently been introduced on the U.S. and European markets expected to possess antipsychotic properties without
producing EPS [22, 23, 24].
PARTIAL DOPAMINE RECEPTOR ANTAGONISTS (+)-UH232 was brought to clinical testing on healthy
volunteers as well as schizophrenic patients, though only
In the early 1980s, the Carlsson research group after single doses. In the former group the drug had some
discovered several dopamine D2 receptor antagonists, devoid mixed actions with respect to mood and wakefulness.
of intrinsic activity, which also possess the dopaminergic However, a rather striking effect was some abnormalities in
stabilizer profile; these agents behave as partial dopamine perception observed in a number of individuals (illusions
receptor antagonists. A partial dopamine receptor antagonist with a trend to hallucinations). In some of the schizophrenic
can be defined as a pure antagonist with an affinity for patients the psychotic symptoms worsened. These effects
subpopulations of postsynaptic dopamine receptors that is so may be due to 5-HT2-receptor agonist effects of (+)-UH232
low that it even in high dosage can only partially displace a [25].
"ubiquitous" D2 receptor high-affinity ligand, leading to an
inability to cause severe hypomotility or catalepsy. The first In spite of this disappointing result it was felt that the
agents discovered belong chemically to the aminotetralines. pharmacological profile of (+)-UH232 was intriguing and
The work started out with the full dopamine receptor agonist clearly worthy of further exploration.

OH OH
HO
H
NH2 N

DOPAMINE (-)-3-PPP

OCH3 CN

H
N N
(+)-UH23 2 CH3
(-)-DS12 1

SO2CH3 SO2CH3

H
N ACR16
(-)-OSU6162 N
Fig. (1). Structures of dopamine stabilizers.
270 Current Medicinal Chemistry, 2004, Vol. 11, No. 3 Carlsson et al.

Thus, a further step was taken when the chemists of the such in the binding assays even in the absence of intrinsic
Carlsson group switched from the aminotetraline back to the activity.
phenylpiperidine structure in order to find out if also in the Although both partial dopamine receptor agonists and
latter it would be possible to convert a partial agonist such partial dopamine receptor antagonists fulfil the criteria for
as (-)-3-PPP into an antagonist, and then perhaps with dopaminergic stabilizers, they differ in one potentially
"stabilizer" properties. Initially Wikström and coworkers important respect: the partial agonists inhibit normal
[26] reported that replacing the aromatic ring hydroxyl group exploratory behavior, whereas the partial antagonists are, if
of (-)-3-PPP with CF3 resulted in a compound with weak anything, slightly activating on this behavior over a large
dopamine receptor antagonist properties judged from effects dose range; higher doses can decrease motor activity to a
on dopamine turnover. An increase in motor activity was certain degree during the early exploratory phase when the
observed concomitant with the increased dopamine neuronal base-line is high. However, this distinction is observed in
activity and the compound was suggested to be a preferential rodents after single doses; whether it remains after repeated
dopamine autoreceptor antagonist. Subsequently, the dosage and occur in other species is an open question.
hydroxyl group of (-)-3-PPP was replaced by other electron-
drawing groups, such as CN or SO2 CH 3 (Fig (1)). The It may seem remarkable that compounds with agonistic
compounds thus synthesised were (-)-DS121 and (-)- and antagonistic properties may have such similar
OSU6162 and they too behaved, biochemically and pharmacological profiles. A plausible hypothetic explanation
behaviorally, as preferential dopamine autoreceptor may be the following. In partial agonists the stabilizing
antagonists, or as we prefer to call them now partial properties are inherent in the partial agonism: at high levels
dopamine receptor antagonists. Thus, it was concluded that of endogenous agonist (dopamine) a partial agonist will
the replacement of a hydrogen bond donor, such as a antagonize the stimulant action of the agonist. At low levels
hydroxyl group, on the aromatic ring of (-)-3-PPP to an of endogenous agonist the partial agonist will add to the
electron-drawing group, such as CF3, CN or SO2CH, was agonism. Regardless of whether the endogenous agonist
important in converting a partial dopamine agonist to an level is high or low, the partial agonist will bring the
antagonist [26, 27]. It should be noted that the racemic form activity towards the level determined by the intrinsic
of DS121 had several years previously been observed to lack activity. In principle this should be true regardless of the
intrinsic activity on dopamine receptors [8]. Both (-)-DS121 responsiveness of the receptor. However, at a high receptor
1
and (-)-OSU6162, and more recently ACR16 , were responsiveness, like in the case of the autoreceptors, a partial
characterized pharmacologically and were found to fulfil the agonist is more likely to act as an agonist. Therefore, the
criteria for a dopaminergic stabilizer. In other words, they situation can occur that a partial agonist acts as an agonist
have but slight effect on the behavior of actively exploring on the autoreceptors and as an antagonist on the postsynaptic
rats but antagonize the hyperactivity induced by receptors. In such a case the effect on the autoreceptors and
dopaminergic agonists; in animals with low motor activity the postsynaptic receptors will influence behavior in the
following habituation to their environment they induce a same direction and cause inhibition.
clear-cut, though moderate stimulation, and they are devoid Since the autoreceptors are in contact with low
of cataleptogenic activity. (-)-OSU6162 was brought to endogenous agonist levels, it should be easier for an
clinical testing and was found to have antipsychotic and antagonist to compete for the autoreceptors than for the
antidyskinetic efficacy (sometimes dramatic) in short term postsynaptic receptors. Antagonists should therefore have a
studies. Long-term studies remain to be done [28]. preferential action on the autoreceptors, showing up in low
In conventional receptor binding assays, (-)-DS121 and dosage as behavioral stimulation. The reason that the
(-)-OSU6162 show very low ability to displace a dopamine- behavioral influence following interaction with the
receptor blocking ligand but a stronger ability to displace a autoreceptors is so profound, despite the low concentration
dopamine-receptor agonist [29]. This is remarkable, because of dopamine molecules at the autoreceptors, is probably a
generally agonists are more efficient in displacing agonists greater impact per dopamine molecule due to a higher
than antagonists, whereas antagonists are more efficient in responsiveness of autoreceptors, which in turn is due to
displacing antagonists than agonists. As mentioned, (-)- exposure to much lower dopamine concentrations compared
DS121 and (-)-OSU6162 are devoid of intrinsic activity and to the postsynaptic receptors of the synapse. Although this
are thus pure antagonists but structurally they are derived phenomenon has been described after treatment with full
from agonists, and this may explain why they behave as dopaminergic antagonists, it is not always obvious, possibly
because it may occur only in a narrow dose range. However,
if the affinity for D2 receptors comes down to a certain level,
1 with a Ki around a couple of hundred nM (using an agonist
F. Pettersson, N. Waters, E.S. Waters, A. Carlsson, C. Sonesson. The binding assay), such as has been found for e g (-)-DS121 and
development of a new class of dopamine stabilizers. Program No. 894.11*.
N. Waters, A. Carlsson, M.L Carlsson, P. Martin, J. Tedroff, J. Kullingsjö,
(-)-OSU6162 [29], a separation between binding to
F. Pettersson, C. Sonesson, E.S. Waters. Pharmacology of ACR16, a autoreceptors and postsynaptic receptors becomes apparent.
dopamine stabilizer. Program No 894.13*. H. Ponten, A Carlsson, J. In other words, such compounds can be called preferential
Kullingsjö, C. Sonesson, E.S. Waters, N. Waters, J. Tedroff. The dopamine dopamine autoreceptor antagonists, a term that we have
stabilizer ACR16 prevents L-DOPA induced sensitization in the 6-OHDA- previously used, even though now we prefer to call them
lesioned rat. Program No 787.6*. Abstract Viewer online, Society for
Neuroscience*, 2002. Carlsson ML (2002) E x p e r i m e n t a l partial dopamine receptor antagonists. In support of the latter
hypoglutamatergia model for cognitive deficits of schizophrenia: Effects term there are data showing that there is an ability of these
of classical neuroleptics, 5-HT2A receptor blocking antipsychotics, the agents to displace a dopaminergic agonist from striatal
ampakine CX516 and the dopaminergic stabiliser ACR16. European binding sites, but that this effect is only partial, compared to
Neuropsychopharmacology 12: (suppl 3) 167-168.
Schizophrenia: From Dopamine to Glutamate and Back Current Medicinal Chemistry, 2004, Vol. 11, No. 3 271

a typical dopamine receptor antagonist such as haloperidol receptor antagonists, although being able to block only a
[30]. portion of postsynaptic receptors, can nevertheless prevent a
sufficient recruitment of receptors to lead to behavioral
Our interpretation of the stabilizer profile of the partial
hyperactivity. In habituated rats the effect of these agents on
dopamine receptor antagonists is thus the following: In
the autoreceptors will predominate, and thus a moderate
normal rats exploring a new environment there is no striking
stimulation will ensue.
effect of these agents, because the relatively weak
antagonism on the postsynaptic receptors and the ensuing In the case of amphetamine-induced locomotor
mild behavioral inhibition, is counter-balanced by a strong stimulation, which is effectively antagonised by partial
antagonism on the autoreceptors, which acts in a stimulating dopamine receptor antagonists, postsynaptic receptors
direction. The hyperactivity induced by dopaminergic located outside the synapse may play an important role (see
agonists probably depends on the recruitment of a large Fig (2)). These receptors are normally exposed to extremely
population of postsynaptic receptors. The partial dopamine low levels of dopamine and will thus display a high

Fig. (2). Dopaminergic synapse at different activity levels. The size of the arrow indicates the degree of firing activity. Red dots
represent dopamine molecules, blue dots represent apomorphine molecules.
272 Current Medicinal Chemistry, 2004, Vol. 11, No. 3 Carlsson et al.

responsiveness, which means that each dopamine molecule autoreceptors and thus of the biphasic dose-response curve of
will have a much greater impact following binding to the a dopaminergic agonist such as apomorphine or (+)-3-PPP.
receptor than is the case with intrasynaptic receptors with a However, one would expect an array of examples
lower responsiveness. Following amphetamine demonstrating qualitative changes in the pharmacological
administration a massive dopamine release will ensue, profile of a dopaminergic agonist, depending on the dosage.
resulting in diffusion of some dopamine molecules even to For example, the emetic action of apomorphine shows up
these extrasynaptic receptors on the postsynaptic neuron. already in low dosage, which could be due to the absence of
Here the partial dopamine receptor antagonists will have a well-developed dopaminergic synapses in the emetic trigger
chance to compete, though, because the dopamine zone. Another phenomenon might be explained in a similar
concentrations will still be much lower than in the synapse way: with increasing doses of apomorphine behavioral
under normal circumstances and of course after amphetamine inhibition switches into stimulation, but the picture of
treatment. Since the interaction of dopamine with the stimulation will undergo changes. In an intermediate dose
extrasynaptic sites probably is important for the range an increase in locomotor activity predominates. In
hyperactivity response, administration of a partial dopamine higher doses stereotypic activity (compulsive gnawing etc.)
receptor antagonist will in this situation, contrary to the takes over at the expense of locomotion. Locomotion is
normal situation where intrasynaptic dopamine probably to a initiated predominantly from the nucleus accumbens, that is
larger extent determines the behavior, have a large behavioral a part of the ventral striatum, whereas stereotypies are
impact, effectively bringing motor activity down to normal probably elicited from the dorsal striatum. As mentioned,
levels. the synapses in the ventral striatum are not so well
An interesting observation with partial dopamine receptor developed as in the dorsal striatum. This will provide an
antagonists is that they appear to more effectively counteract explanation of the differences between drugs in regard to the
amphetamine- than apomorphine-induced hyperactivity [e. g. balance between actions on the "limbic" versus
23]. A reason for this may be that whereas in the "extrapyramidal" systems.
amphetamine situation the concentration of dopamine If we now turn our attention from agonist to antagonist
molecules reaching strategic extrasynaptic sites is still pharmacology, the perspective will become even more
relatively modest and thus readily counteracted by partial interesting. Let us go back to the above example with the
dopamine receptor antagonists, apomorphine administration introduction of an exogenous agonist but replace it by an
will result in a ubiquitous increase in agonist concentration antagonist. Let us also assume that following a period of
due to an increased agonist concentration in the extracellular equilibration the antagonist's concentration in the
water phase (see Fig. (2)). extracellular water phase of the brain and its affinity for the
Regarding the concentration of endogenous agonist it is receptor are the same in different locations. Under these
reasonable to assume that it will vary considerably with the assumptions the action of the antagonist will depend on its
structure of the junction, and that the highest concentration ability to compete with the endogenous neurotransmitter.
will occur in the synaptic cleft, while the lowest This competition will be least successful in locations where
concentrations will be in locations where the distance the concentration of the neurotransmitter is the highest, that
between the neurotransmitter releasing structure and the is, in the well-developed synapses of, for example, the
receptor is the largest. dorsal striatum. It will be somewhat more successful in the
ventral striatum, and thus the EPS of a dopamine receptor
The pharmacological consequences of these varying antagonist will occur after somewhat higher doses than the
distances could be dramatic. Let us assume that that an antipsychotic action, which presumably resides in the
exogenous agonist capable of getting into the brain via the ventral, "limbic" striatum. Furthermore, the competition
blood-brain barrier will be equally distributed in the will be even more successful in the non-synaptic areas, such
extracellular water phase of the brain, allowing for a period as the dendritic dopaminergic regions, and the emetic trigger
of equilibration, and that under resting conditions the degree area. Certainly the antiemetic effects of dopamine receptor
of receptor activation, expressed as percent of full activation, antagonists show up in low dosage, and signs of behavioral
for example 25 percent, is about the same in different stimulation, among other things an antidepressant effect,
locations, and that finally the affinity of either the have been described after treatment with antidopaminergic
endogenous or the exogenous agonist does not vary between agents in low dosage.
different locations. In other words, the variations in the
responsiveness of receptors at different locations are due to In this context the profile of the partial dopaminergic
factors other than affinity. If we now compare a synaptic antagonists is interesting. When this group of compounds -
with a non-synaptic location, the concentration of the aminotetralines and phenylpiperidines - are examined
endogenous agonist should, under the assumptions given together, there is a clear trend for the molecules to shift from
above, be much higher in the synapse than in the non- a typical neuroleptic to a stabilizing profile, when they are
synaptic location. Let us, for the sake of the argument, modified in the direction of diminishing the affinity to D2
assume that it is five-fold higher. If we now look at the receptors [30].
action of the exogenous agonist and assume that it will have
an action equivalent to a doubling of the endogenous agonist GLUTAMATE
at the non-synaptic site, it will then only be expected to be
equivalent to a twenty percent increase in endogenous For several years considerable interest has focused on the
agonist in the synaptic site. We may thus have a simple possible role of glutamate in schizophrenia. One reason for
explanation of the preferential action of an agonist on the this is the discovery that phencyclidine (PCP, "angel dust"),
Schizophrenia: From Dopamine to Glutamate and Back Current Medicinal Chemistry, 2004, Vol. 11, No. 3 273

which can induce a psychotic condition mimicking virtually without side effects. These clinical data underline
schizophrenia, perhaps even more faithfully than the the relevance and predictive value of our experimental MK-
amphetamines, is a powerful antagonist on one of the 801 model for schizophrenia and are remarkable in that they
glutamate receptor subtypes, namely the NMDA receptor. showed for the first time that dopamine D2 blockade is not
This receptor is equipped with an ion channel regulating the obligatory for achieving antipsychotic effect.
penetration of calcium and other cations into the neuron.
In schizophrenia, classical antipsychotic compounds
PCP binds to a specific site in this channel, thereby
often alleviate positive symptoms like hallucinations and
blocking the function of the receptor. Examples of other
delusions but are in general less effective in overcoming
NMDA antagonists that bind to this "PCP”-site are
negative and cognitive symptoms with severe consequences
ketamine and MK-801. There are also competitive NMDA
for the patients’ quality of life, hampering vocational and
antagonists available, such as D-CPPene and CGS 19755,
social rehabilitation [45, 46, 47]. Impaired cognitive
which bind to the same site as glutamate. Another way to
functioning was previously thought to be solely secondary
modulate NMDA receptor-mediated activity is via the
to the psychosis, or a side effect of antipsychotic treatment,
associated glycine site, where the mixed agonist-antagonist
but is today regarded as a core feature of the disease [46, 47,
D-cycloserine binds and where glycine and D-serine serve
48] which may even be manifested in childhood and precede
agonist functions [for refs see 31]. Another potentially useful
the onset of psychotic symptoms by many years [49].
tool to enhance NMDA receptor-associated glycine
transmission is a glycine transporter 1 inhibitor [see 32]. Administration of a glutamate antagonist like MK-801
Strengthening NMDA receptor-mediated transmission by causes a marked primitivization of the behavior. An
direct or indirect stimulation of the associated glycine interesting serendipity observation we made in the
receptor has for several years been discussed as a possible hypoglutamatergic mice treated with haloperidol and
treatment strategy in schizophrenia. M100907 respectively, was that whereas both compounds
counteracted MK-801-induced hyperactivity, only M100907
In the late eighties we set up a hypoglutamatergia model
improved the quality of the behavior. Mice receiving MK-
of schizophrenia in mice, where we as a rule used MK-801
801 in combination with haloperidol looked very similar to
to interfere with central glutamate activity [33, 34, 35], but
those animals treated with MK-801 only, i. e. they
we also observed behavioral effects of competitive NMDA
displayed mainly monotonous, forward locomotion
antagonists in this mouse model that were predictive of their
behavior, whereas the hypoglutamatergic mice treated with
psychotogenic actions in man [see 36]. Furthermore, we
M100907 displayed a more varied, control-like behavior,
observed psychotomimetic actions of the glycine antagonist
although the restoration of behavioral quality was far from
(+)-HA-966 following local administration into the nucleus
complete.
accumbens of mice, results that are in consonance with the
psychotogenic effects reported with D-cycloserine in man This observation prompted us to set up an automated
[37, 38]. Interestingly, there is some evidence that the video system for characterization and quantification of the
enhanced dopaminergic tone observed in the active psychotic behavioral pattern of hypoglutamatergic animals. We
phase of schizophrenia (see Introduction) is secondary to hypothesised that the MK-801-induced primitivization of
deficient glutamate transmission [39]. behavior might correspond to cognitive deficits of
schizophrenia and that this might be a good model to screen
Brain glutamatergic systems also interact with serotonin
for new compounds that could be developed to treat
(5-HT). NMDA receptor antagonists stimulate 5-HT turnover
cognitive symptoms of schizophrenia.
and induce increased responsiveness of 5-HT2 receptors [25,
40, 41]. This is of special interest in view of the effective
counteraction of the selective 5-HT2A antagonist M100907 Experimental Hypoglutamatergia Model for Cognitive
on the behavioral stimulation induced by NMDA receptor Deficits of Schizophrenia
antagonism [42].
NMDA receptor antagonist-induced hyperactivity in
We also compared the effects of the 5-HT2A receptor rodents has become a popular model of schizophrenia, and
antagonist M100907 in different psychosis models using there is reason to believe that this hyperactivity may
haloperidol as reference [42]. Two hyperdopaminergia and correspond to the positive symptomatology of this disorder.
two hypoglutamatergia models were used. M100907 was As mentioned above, NMDA receptor antagonist treatment
found to counteract NMDA receptor antagonist induced also causes a behavioral primitivization, which may
hyperactivity whereas spontaneous locomotion and correspond to cognitive deficits of schizophrenia. Therefore,
dopamine agonist induced hyperactivity were not we characterize and quantify changes in behavioral pattern
significantly affected. Haloperidol, on the other hand, induced by NMDA receptor antagonists, using an automated
antagonised both NMDA antagonist and dopamine agonist video tracking system. Mice are treated with a glutamate
induced hyperactivity as well as spontaneous locomotion. antagonist, e g MK-801, before being introduced into an
Based on these and previous results we concluded that 5- unfamiliar, diffusely illuminated arena and video taped from
HT2A receptor antagonists may be particularly effective in above. The video tapes are analyzed with the video tracking
hypoglutamatergic conditions [including e. g. subgroups of system EthoVision from Noldus Information Technologies.
schizophrenia, autism [see 43] and ADHD [see 44]. The What we then find is that MK-801, apart from causing
modest effect of M100907 on spontaneous behavior would hyperactivity, wipes out or markedly weakens other
predict a low risk for side effects. Indeed, clinical phase III components of normal mouse behavior. This is reflected in
trials in schizophrenia conducted by Aventis demonstrated the behavioral variables we measure as a decreased number of
antipsychotic effect of M100907; in addition, this agent was turnings per cm moved, a reduced number of switches
274 Current Medicinal Chemistry, 2004, Vol. 11, No. 3 Carlsson et al.

between the states "moving"and "stationary”, reduced rearing 49] but needless to say considerably more research is needed
(= rising on hind legs) frequency, reduced grooming before we can tell if different modern antipsychotics indeed
behavior and a reduced variation in the digitised video image target specific cognitive symptoms.
of the animals’ body size seen from above. What we see, in
In our model, the classical neuroleptics haloperidol,
short, in these hypoglutamatergic animals is a severely
chlorpromazine and trifluoperazine administered in doses
impoverished behavioral repertoire.
that reduced hyperactivity in hypoglutamatergic mice to
control levels were unable to restore behavioral diversity.
Effects of Classical Neuroleptics vs Antipsychotics with This is illustrated e. g. by their inability to restore
Strong 5-HT2A Relative to D2 Receptor Antagonism on variability in body area (Fig. (3)) as well as by their
Behavioral Quality in Hypoglutamatergic Mice inability to improve the quality of the ethograms in
In an attempt to validate our model we have compared hypoglutamatergic mice (Fig. (4)); [53]. The body area is
traditional neuroleptics with a new generation perceived as large by the camera when the animals engage in
antipsychotics, based on the view that while the old forward locomotion and as small when the animals engage
compounds are often either ineffective against or may even in rearing, grooming and digging and these latter behaviors
worsen cognitive symptoms [45], the newer agents seem to are abolished or attenuated by MK-801. In contrast to
cause partial relief [45, 46, 50]. This is obviously an conventional neuroleptics, various antipsychotic compounds
intensely debated issue, and exceedingly difficult to with pronounced 5-HT2A receptor-blocking properties, i. e.
penetrate for a number of reasons [see e. g. 51]. The compounds with strong serotonin(5-HT)2A receptor
consensus at present nevertheless seems to be that the new blockade in relation to the dopamine (DA) D2 receptor
agents with lower D2 receptor affinity often combined with blockade, increased behavioral complexity in MK-801-
substantial 5-HT2 receptor blockade have beneficial, albeit treated mice in doses that reduced MK-801-induced
modest or moderate, effects on cognitive deficits of hyperactivity to control levels, as evident from the
schizophrenia, although it should be noted that such ethograms seen in Fig. (4 ), but, like the classical
beneficial effects on cognition have also been observed neuroleptics they were unable to restore variability in body
following low dose haloperidol treatment [52]. It has been area (Fig. (3)). The compounds tested from this latter
suggested that there may be some specificity in how the category were clozapine, risperidone, olanzapine, ziprasidone
different agents affect various cognitive symptoms [e. g. 45, and the selective 5HT2A receptor antagonist M100907. The

Fig. (3). Effects of haloperidol (0.2, 0.5 mg/kg), chlorpromazine (0.3, 0.9, 1.2 mg/kg), trifluoperazine (0.33, 1, 3 mg/kg), clozapine
(0.05, 0.4, 3.2 mg/kg), risperidone (0.01, 0.03, 0.09 mg/kg), olanzapine (0.083, 0.25, 0.75 mg/kg), ziprasidone (0.33, 1, 3 mg/kg),
M100907 (0.001, 0.01, 0.1 mg/kg) and ACR16 (8, 20, 50 µmol/kg) on the MK-801 (0.2 mg/kg) induced decrease in body area
variation in mice at two different time intervals. *p<0.05 compared to MK-801, +p<0.05 compared to controls (Mann-Whitney U-
test)2 .
Schizophrenia: From Dopamine to Glutamate and Back Current Medicinal Chemistry, 2004, Vol. 11, No. 3 275

reason we chose to compare the different antipsychotic Effects of the Dopaminergic Stabilizer ACR16 on
compounds in doses that restored hyperactivity to control Behavioral Quality in Hypoglutamatergic Mice
levels, is that we believe that MK-801-induced hyperactivity The dopaminergic stabilizer ACR16, developed by
corresponds to positive symptomatology of schizophrenia. Carlsson Research, was also tested in hypoglutamatergic
In other words, we wished to see if the different mice. As described above, dopaminergic stabilizers are
antipsychotic agents tested, in addition to controlling compounds that are able to counteract both hypo- and
"positive symptoms”, could affect cognitive status as hyperdopaminergia, and, in consequence, both hypo- and
reflected by behavioral complexity level. The antipsychotic hyperactivity. In contrast to currently used antipsychotic
compounds with strong 5-HT2A receptor antagonism in agents, the risk for causing dysphoria and anhedonia is
relation to their DA D2 receptor antagonism increased probably low.
behavioral complexity in hypoglutamatergic mice with
respect to horizontal (e. g. spontaneous turning), but not ACR16 counteracted the MK-801-induced
vertical (rearing) measures. Olanzapine seemed to be primitivization of the behavior, apparently even more so
somewhat superior to the other compounds in restoring than M100907 and the other antipsychotic compounds with
behavioral quality, which is interesting in view of a recent strong 5-HT2A receptor blockade [Figs (3), (4)]. Most
report [50] showing some advantage of olanzapine over both importantly, it influenced aspects of the behavior that were
haloperidol and risperidone with respect to cognitive not restored by the 5-HT2A receptor blocking
functions in schizophrenia. antipsychotics. For instance, ACR16 partly restored rearing
in the hypoglutamatergic mice and this is the first time we
276 Current Medicinal Chemistry, 2004, Vol. 11, No. 3 Carlsson et al.

(Fig. 4). contd.....

Fig. (4). Effects of haloperidol, chlorpromazine, trifluoperazine, clozapine, risperidone, olanzapine, ziprasidone, M100907 and
ACR16 on the MK-801 (0.2 mg/kg) induced primitivization of the movement pattern in mice at two different time intervals.
Whenever possible, tracks from mice treated with the test compounds were chosen among individuals having motor activity levels
similar to controls to facilitate comparisons2.

make this observation following administration of an ACR16 has been found safe in phase I studies on healthy
established or potential antipsychotic agent [54]. Since we volunteers. In an open study involving seven patients with
consider explorative rearing as one of the most sophisticated Parkinson’s disease, ACR16 showed efficacy against L-
components of rodent behavior we deem this to be an
important finding.
role of comorbidities in the causation of neurodevelopmental disorders”,
The Novartis Foundation, London, November 2002; Carlsson, M.L.
2 Experimental hypoglutamatergia model for cognitive deficits of
These data have previously been published in/presented at: M. Nilsson, schizophrenia: Effects of classical neuroleptics, 5-HT2A receptor
S. Waters, N. Waters, A. Carlsson, M.L Carlsson: A behavioural pattern blocking antipsychotics, the ampakine CX516 and the dopaminergic
analysis of hypoglutamatergic mice - effects of four different stabiliser ACR16, European Neuropsychopharmacology, 2002, 12 (suppl
antipsychotic agents, J Neural Transm. 2001;108(10):1181-96; Stockholm 3), 167-168/15 t h Congress of the European College of
autism patient association, February 2002; 4th International Conference Neuropsychopharmacology, Barcelona, October 2002.
on Methods and Techniques in Behavioral Research, Amsterdam, August
2002; First Arvid Carlsson Symposium, Göteborg, September 2002; “The
Schizophrenia: From Dopamine to Glutamate and Back Current Medicinal Chemistry, 2004, Vol. 11, No. 3 277

DOPA-induced dyskinesias. ACR16 is currently undergoing [22] Johansson, A.M.; Arvidsson, L.E.; Hacksell, U., Nilsson, J.L.;
Svensson, K., Hjorth, S., Clark, D.; Carlsson, A.; Sanchez, D.;
clinical testing in schizophrenia. Andersson, B.; Wikström, H. J. Med. Chem., 1985, 28, 1049-53.
[23] Svensson, K.; Hjorth, S.; Clark, D.; Carlsson, A.; Wikstrom, H.;
Andersson, B.; Sanchez, D.; Johansson, A.M.; Arvidsson, L.E.;
ACKNOWLEDGEMENTS Hacksell, U.; Nilsson, J.L.G. J. Neural Transm., 1986, 65, 1-27.
[24] Svensson, K.; Johansson, A.M.; Magnusson, T.; Carlsson, A.
The present study was supported by grants from the Naunyn Schmiedebergs Arch Pharmacol., 1986, 334, 234-45.
Swedish Medical Research Council (9067). [25] Carlsson, M.L. J. Neural Transm., 1995, 100, 225-237.
[26] Wikström, H.; Andersson, B.; Elebring, T.; Svensson, K.; Carlsson,
A.; Largent, B. J. Med. Chem., 1987, 30, 2169-2174.
REFERENCES [27] Sonesson, C.; Waters, N.; Svensson, K; Carlsson, A.; Smith, M.W.;
Piercy, M.F.; Meier, E.; Wikström, H. J. Med. Chem., 1993, 36,
[1] Abi-Dargham, A.; Gil, R.; Krystal, J.; Baldwin, R.M .; Seibyl, J.P.; 3188-3196.
Bowers, M.; van Dyck, C.H.; Charney, D.S.; Innis, R.B .; Laruelle, [28] Carlsson, A.; Waters, N.; Waters, S.; Tedroff, J.; Nilsson, M.;
M. Am. J. Psychiatry, 1998, 155, 761-767. Carlsson, M.L. Interactions between monoamines, glutamate, and
[2] Breier, A.; Su, T.-P.¸ Saunders, R.; Carson, R.E.; Kolachana, B.S.; GABA in schizophrenia: new evidence. Annu. Rev. Pharmacol.
de Bartolomeis, A.; Weinberger, D.R.; Weisenfeld, N.; Malhotra, Toxicol., 2001, 41, 237-60.
A.K.; Eckelman, W.C.; Pickar, D. Proc. Natl. Acad. Sci. USA, [29] Sonesson, C.; Lin, C-H.; Hansson, L.; Waters, N.; Svensson, K;
1997, 94, 2569-2574. Carlsson, A.; Smith, M.W.; Wikström, H. J. Med. Chem., 1994, 37,
[3] Hietala, J.; Syvalahti, E.; Vuorio, K.; Rakkolainen, V.; Bergman, 2735-2753.
J.; Haaparanta, M.; Solin, O.; Kuoppamaki, M.; Kirvela, O.; [30] Waters, N.; Löfberg, L.; Haadsma-Svensson, S.; Svensson, K.;
Ruotsalainen, U.; Salokangas, R.K.R. Lancet, 1995, 346, 1130- Sonesson, C.; Carlsson, A. J. Neural Transm. Gen Sect, 1994, 98,
1131. 39-55.
[4] Laruelle, M.; Abi-Dargham, A.; Gil, R.; Kegeles, L.; Innis, R. [31] Nilsson, M.; Carlsson, A.; Carlsson, M.L. J. Neural Transm., 1997,
Biol. Psychiatry, 1999, 46, 56-72. 104, 1195-1205.
[5] Reith, J.; Benkelfat, C.; Sherwin, A.; Yasuhara, Y.; Kuwabara, H.; [32] Atkinson, B.N.; Bell, S.C.; De Vivo, M.; Kowalski, L.R.; Lechner,
Andermann, F.; Bachneff, S.; Cumming, P.; Diksic, M.; Dyve, S.M.; Ognyanov, V.I., Tham, CS.; Tsai, C.; Jia, J.; Ashton, D.;
S.E.; Etienne, P; Evans, A.C.; Lal, S.; Shevell, M; Savard, G.; Klitenick, Mol Pharmacol., 2001, 60, 1414-20.
Wong, D.F.; Chouinard, G.; Gjedde, A. Proc. Natl. Acad. Sci. [33] Carlsson, M.; Carlsson, A. TINS, 1990, 13, 272-276.
USA, 1994, 91, 11651-11654. [34] Carlsson, M.; Carlsson, A. J. Neural Transm., 1989, 77, 65-71.
[6] Laruelle, M.; Abi -Dargham, A.; van Dyck, CH.; Gil, R.; D'Souza, [35] Carlsson, M.; Carlsson, A. J. Neural Transm., 1989, 75, 221-226.
CD.; Erdos, J.; McCance, E.; Rosenblatt, W.; Fingado, C.; Zoghbi, [36] Carlsson, M.L. J. Neural Transm. [GenSect], 1993, 94, 1-10.
S.S.; Baldwin, R.M.; Seibyl, I.P.; Crystal, J.H ; Charney, D.S ; [37] Cascella N.G.; Macciardi, F.; Cavallini, C., Smeraldi, E. J. Neural
Innis, R.B. Proc. Natl. Acad. Sci. USA, 1996, 93, 9235-40. Transm. Gen Sect, 1994, 95, 105-11
[7] Lindström, L.H.; Gefvert, O.; Hagberg, G.; Hagström, P.; [38] Nilsson, M.; Carlsson, M.L. J. Neural Transm., 1997, 104, 419-
Lundberg, T.; Bergström, M.; Hartvig, P.; Långström. B. Biol. 425.
Psychiat., 1999, 46, 681-688. [39] Kegeles L.S.; Abi-Dargham, A.; Zea-Ponce, Y.; Rodenhiser-Hill,
[8] Hacksell, U.; Arvidsson, L-E.; Svensson, U.; Nilsson, L.G.; J.; Mann, J.J.; Van Heertum, R.L.; Cooper, T.B.; Carlsson, A.;
Sanchez, D.; Wikström, H.; Lindberg, P.; Hjorth, S.; Carlsson, A. Laruelle, M. Biol. Psychiat., 2000, 48, 627-40.
J. Med. Chem., 1981, 24, 1475-1482. [40] Martin, P.; Carlsson, M.L.; Hjorth, S. SNeuroReport, 1998, 9,
[9] Carlsson, A. In Chemical Tools in Catecholamine Research; 2985-2988
Almgren, O.; Carlsson, A.; Engel, J., Eds.; Amsterdam: North- [41] Waters, N.; Lundgren, C.; Hansson, L.O.; Carlsson, M.L. J.
Holland, 1975, Vol. II, pp. 219-224. Neural Transm., 1996, 103, 117-129.
[10] Wikström, H.; Sanchez, D.; Lindberg, P.; Hacksell, U.; Arvidsson, [42] Carlsson, M.L., Martin, P.; Nilsson, S.M.; Sorensen, S.M.;
L.E.; Johansson, A.M.; Thorberg, S.O.; Nilsson, J.L.; Svensson, K.; Carlsson, A.; Waters, S.; N Waters, N. J. Neural Transm., 1999,
Hjorth, S.; Clark, D.; Carlsson, A. J. Med. Chem., 1984, 27, 1030- 106, 123-129.
6. [43] Carlsson, M.L. J. Neural Transm., 1998, 105, 525-535.
[11] Clark, D.; Hjorth, S.; Carlsson, A. J. Neural Transm., 1985, 62, 1- [44] Carlsson, M.L Prog. Neuro-Psychopharmacol. & Biol. Psychiat.,
52. 2001, 25, 5-26.
[12] Clark, D.; Hjorth, S.; Carlsson, A. J. Neural Transm. 1985, 62, [45] Keefe R.S.; Silva, S.G.; Perkins, D.O.; Lieberman, J.A. Schizophr.
171-207. Bull., 1999, 25, 201-22.
[13] Tamminga, C.A. J. Neural Transm., 2002, 109, 411-20 [46] Meltzer, H.Y.; McGurk, S.R. Schizophr. Bull., 1999, 25, 233-55.
[14] Svensson, K.; Eriksson, E.; Carlsson, A. Neuropharmacology, [47] Sharma T. Curr. Med. Res. Opin., 2002, 18 (Suppl. 3), 13-7.
1993, 32, 1037-45. [48] Saykin, A.J.; Shtasel, D.L.; Gur, R.E.; Kester, D.B.; Mozley, L.H.;
[15] Metman, L.V.; Sethy, V.H.; Roberts, J.R.; Bravi, D.; Hoff, J.I.; Stafiniak, P.; Gur, R.C. Arch. Gen. Psychiatry, 1994, 51, 124-31.
Mouradian, M.M., Chase, T.N. Mov. Disord., 1994, 9, 577-81. [49] Cannon, M.; Caspi, A.; Moffitt, T.E.; Harrington, H., Taylor, A.;
[16] Pirtosek, Z.; Merello, M.; Carlsson, A.; Stern, G. C l i n . Murray, RM.; Poulton, R. Arch. Gen. Psychiatry, 2002, 59, 449-
Neuropharmacol., 1993, 16, 550-4. 56.
[17] Jordan, S.; Chen, R.; Johnson, J.; Regardie, K.; Tadori, Y.; [50] Purdon, S.E., Jones, B.D.; Stip, E.; Labelle, A.; Addington, D.,
Kikuchi, T. European Neuropsychopharmacology, 2002, 1 2 David, S.R.; Breier, A.; Tollefson, G.D. Arch. Gen. Psychiatry,
(Suppl. 3) 293-294. 2000, 57, 249-58.
[18] Kikuchi, T.; Tottori, K.; Uwahodo, Y.; Hirose, T.; Miwa, T.; [51] Harvey P.D; Keefe R.S. Am. J. Psychiatry, 2001, 158, 176-84.
Oshiro, Y., Morita, S. J. Pharmacol. Exp. Ther., 1995, 274, 329- [52] Green, M.F.; Marder, S.R.; Glynn, S.M.; McGurk, S.R.; Wirshing,
36. W.C.; Wirshing, D.A.; Liberman, R.P.; Mintz, J. Biol. Psychiat.,
[19] Yokoi, F.; Grunder, G; Biziere, K.; Stephane, M.; Dogan, A.S.; 2002, 51, 972-8.
Dannals, R.F.; Ravert, H.; Suri, A.; Bramer, S.; Wong, D.F. [53] Nilsson, M.; Waters, S.; Waters, Carlsson, A.; Carlsson, M.L. J.
Neuropsychopharmacology 2002, 27, 248-59. Neural Transm., 2001, 108, 1181-96.
[20] Goodnick, P.J.: Rodriguez, L.; Santana, O. Expert Opin. [54] Carlsson, M.L. European Neuropsychopharmacology, 2002, 12
Pharmacother., 2002, 3, 1381-91. (Suppl. 3), 167-168.
[21] Keck, P.E. Jr; McElroy, S.L. Expert Opin. Investig. Drugs, 2003,
12, 655-62.

Vous aimerez peut-être aussi