Treatments For Schizophrenia: A Critical Review of Pharmacology and Mechanisms of Action of Antipsychotic Drugs

Download as pdf or txt
Download as pdf or txt
You are on page 1of 26

Molecular Psychiatry (2005) 10, 79–104

& 2005 Nature Publishing Group All rights reserved 1359-4184/05 $30.00
www.nature.com/mp

FEATURE REVIEW

Treatments for schizophrenia: a critical review of


pharmacology and mechanisms of action of antipsychotic
drugs
S Miyamoto1, GE Duncan2, CE Marx3 and JA Lieberman2
1
Department of Neuropsychiatry, St. Marianna University School of Medicine, Kawasaki, Japan; 2Department of Psychiatry and
Mental Health and Neuroscience Clinical Research Center, School of Medicine, The University of North Carolina at Chapel Hill,
NC, USA; 3Department of Psychiatry and Behavioral Sciences, Duke University Medical Center and Durham VA Medical
Center, NC, USA

The treatment of schizophrenia has evolved over the past half century primarily in the context
of antipsychotic drug development. Although there has been significant progress resulting in
the availability and use of numerous medications, these reflect three basic classes of
medications (conventional (typical), atypical and dopamine partial agonist antipsychotics) all
of which, despite working by varying mechanisms of actions, act principally on dopamine
systems. Many of the second-generation (atypical and dopamine partial agonist) antipsycho-
tics are believed to offer advantages over first-generation agents in the treatment for
schizophrenia. However, the pharmacological properties that confer the different therapeutic
effects of the new generation of antipsychotic drugs have remained elusive, and certain side
effects can still impact patient health and quality of life. Moreover, the efficacy of antipsychotic
drugs is limited prompting the clinical use of adjunctive pharmacy to augment the effects of
treatment. In addition, the search for novel and nondopaminergic antipsychotic drugs has not
been successful to date, though numerous development strategies continue to be pursued,
guided by various pathophysiologic hypotheses. This article provides a brief review and
critique of the current therapeutic armamentarium for treating schizophrenia and drug
development strategies and theories of mechanisms of action of antipsychotics, and focuses
on novel targets for therapeutic agents for future drug development.
Molecular Psychiatry (2005) 10, 79–104. doi:10.1038/sj.mp.4001556
Published online 3 August 2004
Keywords: antipsychotic; dopamine partial agonist; NMDA receptor; metabotropic glutamate
receptor; schizophrenia

The therapeutic armamentarium for the treatment of improvement in negative symptoms, cognitive impair-
schizophrenia has grown and diversified in the half ment, relapse prevention, functional capacity, and
century since the advent of chlorpromazine and the quality of life with fewer extrapyramidal symptoms
beginning of the pharmacologic era in psychiatry. Over (EPS), and less tardive dyskinesia (TD) (for a review,
the past decade, much of our attention regarding the see Miyamoto et al1). Accordingly, many clinicians are
treatment for schizophrenia and related psychotic prescribing these new antipsychotics as first-line
disorders has focused on a new class of antipsychotic agents for acute and maintenance therapy for schizo-
medications. The reintroduction of clozapine repre- phrenia.2–5 However, these advantages, thus far, have
sented a major step forward, and led to the prolifera- been regarded as incremental and not necessarily
tion of ‘atypical’ or second-generation antipsychotics substantial. In addition, concerns about side effects
(SGAs), including risperidone, olanzapine, quetiapine, such as EPS have been replaced by other distressing
ziprasidone, sertindole and zotepine. In fact, there is side effects, including weight gain, hyperglycemia and
growing evidence that most of the new medications dyslipidemia. At present, we are still in the process
can offer some advantages over ‘typical’ or first- of defining fully the clinical profiles of new agents
generation antipsychotics (FGAs) such as greater in terms of the extent of their therapeutic efficacy and
adverse effects, on a variety of other outcomes includ-
ing cognition, affect, suicide, subjective response,
Correspondence: Dr J Lieberman, Department of Psychiatry and social and vocational function, cost effectiveness, etc.6
Mental Health and Neuroscience Clinical Research Center, Although intensive research on the new antipsy-
University of North Carolina School of Medicine, CB #7160, chotic drugs has led to a greater understanding of the
7025 Neurosciences hospital, Chapel Hill, NC 27599-7160, USA. biochemical effects of these drugs, the pharmacologi-
E-mail: [email protected]
Received 04 February 2004; revised 11 June 2004; accepted 17 cal mechanisms underlying their various therapeutic
June 2004 properties remain to be identified. Moreover, an agent
Treatments for schizophrenia
S Miyamoto et al

80
like clozapine that was clinically superior to other psychotic symptoms either fail to respond to these
highly selective dopamine D2 antagonists demanded drugs or respond inadequately or partially.12 In
reconsideration of the principal mechanisms of action addition, they cause significant rates of undesirable
of antipsychotics as well as the pathophysiological acute and chronic adverse effects (for a review, see
mechanisms in schizophrenia.6,7 In addition, the Miyamoto et al9). At this time, the only groups of
limitations of existing antipsychotic drugs to alle- patients in which the FGAs are clearly preferable are
viate all of the pathologic dimensions of the illness those for whom there is a clear indication for short- or
(ie negative symptoms, cognitive deficits and social long-acting injectable preparations (this will quickly
disabilities) have produced an awareness that no change as injectable SGAs become more available), or
single treatment may be sufficient and prompted the who have a history of excellent response to a FGA
search for compounds that can be used adjunctively with minimal side effects.13,14
with antipsychotic drugs. Various adjunctive treat- Despite the superior clinical effectiveness and EPS
ments, including benzodiazepines, lithium, antic- profile of clozapine, its clinical utility is restricted by
onvulsants, antidepressants, beta-blockers and the propensity to cause agranulocytosis and manda-
dopamine agonists, have been used to enhance the tory hematological monitoring of patients. Newly
response to antipsychotic medications or to treat developed SGAs in addition to the benzamide
residual symptoms of chronic schizophrenia and antipsychotic drugs (sulpirides and amisulprides)
comorbid conditions with schizophrenia (for reviews, and aripiprazole, appear to provide important ad-
see Miyamoto et al5,8). However, despite the preva- vances in side effect profile and efficacy for this drug
lence of the use of adjunctive therapies in clinical class. However, a variety of side effects associated
practice, there are very few empirical data and with individual SGAs and the substituted benza-
theoretical rationale to support this practice. The mides still affect patient health and quality of life in
development of additional novel strategies to obtain addition to their limitations in efficacy. Consequently,
potentially new antipsychotic compounds and their there is a continuing need for new and better drugs.
adjuncts possessing unique pharmacological profiles Over the last few years, new routes of administra-
with few side effects is being pursued based on tion for SGAs have been developed. As of December
specific hypotheses, and actually more agents with 2003, besides oral tablet form of medication, risper-
antipsychotic efficacy are being developed. (for re- idone is available in a liquid form and long-acting
views, see Miyamoto et al5,9). This article provides a microsphere preparations in the US and some
brief review and critique of the current theory of European countries. Olanzapine is available as a
mechanisms of action of antipsychotic drugs, and rapidly disintegrating tablet (zydis) form and an
focuses on novel targets for therapeutic intervention intramuscular form in some countries. A short-acting
and potential strategies for future drug development. intramuscular parenteral form of ziprasidone is
available in the US and many European countries.15
These newly available routes of administration could
Current forms of treatment: first-generation anti-
enhance the usefulness of the SGAs significantly (for
psychotics and second-generation antipsychotics
a review, see Fleischacker16).
The strategies and forms of treatment for schizophre-
nia vary according to the phase and severity of the
Theories of mechanisms of action of antipsychotic
illness. Pharmacologic treatment is the cornerstone
drugs
and essential component of treatment for schizophre-
nia and its clinical management through the different First-generation antipsychotic agents
stages of the illness. Although various psychosocial The effect that is common to all FGAs is a high
therapies, such as cognitive behavior therapy, psy- affinity for dopamine D2 receptors,17 and there is a
choeducation and supported employment, are useful strong correlation between the therapeutic doses of
adjuncts to drug treatment,8,10,11 they all require these drugs and their binding affinity for the D2
pharmacologic treatment to be maximally effective. receptor.18–21 In vitro data show that FGAs such as
In particular, adequate pharmacotherapy during the haloperidol bind ‘tightly’ to the D2 receptor and
acute stage of the illness could set the stage for dissociate slowly.22 In vivo positron emission tomo-
subsequent long-term treatment. Although all avail- graphy (PET) and single photon emission computed
able pharmacological treatments have limitations in tomography (SPECT) studies have further demon-
their effectiveness and are associated with uncomfor- strated the importance of dopamine receptor occu-
table side effects, it is an established fact that pancy as a predictor of antipsychotic response and
antipsychotics can improve the psychotic symptoms adverse effects (for a review, see Remignton and
of schizophrenia and prevent their recurrence.10,11 At Kapur23). Such studies have demonstrated that anti-
present, a total of 11 different classes of antipsychotic psychotic effects are associated with a striatal D2
medications are available in the US. Among them are receptor occupancy of 65–70%,24–27 and D2 occu-
the currently available FGAs (phenothiazines, butyr- pancy greater than 80% significantly increases the
ophenones and thioxanthenes) which, although effec- risk of EPS.24 Recent imaging studies have also shown
tive, are far from being optimal treatments. Between that therapeutic doses of FGAs produce high blockade
30 and 60% of patients with acutely exacerbated of D2-like receptors equally in limbic cortical areas

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

81
and the striatum.28,29 Thus, a threshold between 65 and whether it would occur in the intact nonanesthe-
and 80% D2 occupancy appears to represent the tized unrestrained animals.43–45 Nevertheless, a num-
therapeutic window to minimize the risk of EPS for ber of studies have demonstrated that FGA-induced
FGAs.23,27,30 However, this is not absolute as some dopamine cell depolarization block does occur in
patients can respond below this threshold, and nonanesthetized animals40,41,46,47 (for a review, see
nonresponders can be seen in spite of adequate D2 Grace et al48).
receptor blockade reflecting the limitations of the
receptor occupancy model.27,31 Interestingly, low Benzamides
doses of haloperidol (2–5 mg/day) would be expected Amisulpride, a substituted benzamide analogue of
to induce 60–80% dopamine D2 receptor occu- sulpiride, is a highly selective anatagonist of D2 and
pancy,25,32 while dosages five to 20 times as high are D3 receptors with little affinity for D1-like or non-
often prescribed in current clinical practice.33 This dopaminergic receptors (Table 1).49,50 Its congener,
may be partly accounted for by the fact that long-term sulpiride, demonstrates a generally similar pharma-
treatment with FGAs induces upregulation in D2 cological profile. Preclinical studies suggest that low
receptors in both animals34,35 and humans,36,37 which doses of amisulpride (and probably sulpiride) pre-
appears to be associated with dopamine D2-mediated ferentially block presynaptic D2-like autoreceptors,
supersensitivity,38,39 thus theoretically, increments in and thus lead to an increase in dopaminergic release
dose may be needed to produce the same effect on and neurotransmission, while higher doses reduce
dopaminergic transmission for chronic patients.27,31 certain postsynaptic dopamine receptor-mediated
It is important to acknowledge that the gradual and behaviors that predict antipsychotic efficacy, but with
time-dependant onset of therapeutic efficacy is not little or no induction of catalepsy that predicts low
consistent with the rapid striatal D2 receptor blockade EPS liability.31,50,51 Several PET and SPECT studies in
induced by antipsychotics. Preclinical studies de- schizophrenia demonstrated that amisulpride selec-
monstrating that chronic treatment of rodents with tively binds to temporal cortical D2/D3 receptors in a
FGAs can decrease the number of spontaneously dose-dependent fashion, but this extra-striatal selec-
active dopamine neurons in both the substantia nigra tivity is lost at higher doses as striatal D2/D3 receptor
pars compacta (A9) and the ventral tegmental area occupancy increases.52–54 Another PET study found
(A10) have given rise to the ‘depolarization inactiva- no significant binding to 5-HT2A receptors in amisul-
tion (or block) hypothesis’.40–42 Mereu et al,43–45 pride-treated patients.55 It is also characterized by the
however, have suggested that the depolarization rapid dissociation from the D2 receptor similar to
inactivation of dopamine neurons may be an artifact clozapine.56 Amisulpride is essentially devoid of 5-
produced by the use of general anesthetics, and HT2A antagonism, thus its moderate affinity for
thereby questioned the validity of this phenomenon striatal D2 receptors and preferential occupancy of

Table 1 Relative neurotransmitter receptor affinities for antipsychotics at therapeutic doses (adapted from Miyamoto et al1 and
modified)

Receptor Cloza- Risperi- Olanza- Quetia- Ziprasi- Sertin- Sulpi- Amisul- Zote- Aripipra- Halope-
pine done pine pine done dole ride pride pine zole ridol

D1 þ þ þþ  þ þþ   þ  þ
D2 þ þþþ þþ þ þþþ þþþ þþþþ þþþþ þþ þþþþ þþþþ
D3 þ þþ þ  þþ þþ þþ þþ þþ þþ þþþ
D4 þþ  þþ  þþ þ   þ þ þþþ
5-HT1A     þþþ þþ þþ 
5-HT1D  þ   þþþ þ 
5-HT2A þþþ þþþþ þþþ þþ þþþþ þþþþ   þþþ þþþ þ
5-HT2C þþ þþ þþ  þþþþ þþ   þþ þ 
5-HT6 þþ  þþ  þ þþ þ 
5-HT7 þþ þþþ   þþ þþ þþ 
a1 þþþ þþþ þþ þþþ þþ þþ   þþ þ þþþ
a2 þ þþ þ   þ   þþ þ 
H1 þþþ  þþþ þþ  þ   þþ þ 
m1 þþþþ  þþþ þþ     þ  
DA þþ þþ 
transporter
NA þ þþ þþ þþ 
transporter
5-HT þþ 
transporter

 ¼ minimal to none; þ ¼ low; þ þ ¼ moderate; þ þ þ ¼ high; þ þ þ þ ¼ very high.

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

82
limbic cortical D2/D3 receptors may be reasons for its binding can act as an effective antipsychotic.6 The
therapeutic efficacy and low liability to induce EPS.54 question has been do pharmacologic effects on
dopamine-mediated pathways account for all of the
Second-generation antipsychotic agents clinical therapeutic effects of antipsychotic drugs.
Recent in vitro studies have demonstrated that
The serotonin–dopamine antagonism theory The antipsychotics dissociate from the D2 receptor at
‘serotonin–dopamine (S2/D2) antagonism theory’ very different rates, expressed as a koff value.22,74
promulgated by Meltzer et al57 suggests that a higher The SGAs have higher koff values as a group, that is
ratio of a drug’s affinity for serotonin 5-HT2A receptor faster dissociation rates, than the FGAs, but they
relative to dopamine D2 receptor affinity can predict differ among themselves on this dimension as well
‘atypicality’ and will explain the enhanced efficacy (e.g., quetiapine4clozapine4olanzapine4ziprasi-
and reduced EPS liability of SGAs (for reviews, see done4risperidone).6,56,75 Kapur and Seeman hypothe-
Miyamato et al,1 Lieberman,58 Duncan et al 59). sized that dissociation from the D2 receptor quickly
PET studies showing that therapeutic doses of makes an antipsychotic agent more accommodating of
risperidone, olanzapine and ziprasidone produce physiological dopamine transmission, permitting an
greater than 70% occupancy of D2 receptors suggest antipsychotic effect without EPS, hyperprolac-
that a specific threshold of D2 receptor antagonism tinemia, as well as conferring benefits along a
could be important in producing antipsychotic effects variety of clinical dimensions such as cognitive, affec-
of these drugs.60,61,391 Clozapine and quetiapine, tive and secondary negative symptoms.74 Accor-
however, exhibit lower levels of D2 receptor occu- dingly, they suggest that sustained D2 occupancy is
pancy (less than 70%) at therapeutically effective not necessary for antipsychotic action. However, this
doses (Table 1),24,61–63 suggesting that a threshold level theory cannot explain the greater therapeutic efficacy
of D2 receptor occupancy (and possibly antagonism) of clozapine compared with other SGAs, particularly
alone cannot fully explain the greater therapeutic in the management of treatment-resistant schizo-
efficacy of clozapine59 or for that matter serve as a phrenia. The rapid dissociation of clozapine and
model to predict antipsychotic efficacy. The low quetiapine from D2 receptors by endogenous
occupancy of striatal D2 receptors by clozapine dopamine may lead to more rapid clinical relapse
and quetiapine could account for its low EPS after discontinuation of these medications.75 At
liability.30,62–64 Interestingly, ziprasidone exhibits present, it remains unclear how long an antipsy-
high levels of D2 occupancy at doses of 20– chotic agent must bind to the D2 receptor to maximize
40 mg,65,66 doses that are substantially below the therapeutic efficacy while minimizing the risk of
therapeutically effective dose range (120–200 mg/ D2-related side effects.31 Another limitation of this
day).67,68 Thus, pharmacological properties other than model is that all antipsychotics have not been
a threshold level of D2 receptor antagonism (at least as studied with it, including the benzamides, low-
reflected by receptor occupancy levels) may account potency FGAs and partial dopamine agonists (e.g.
for the clinical efficacy of ziprasidone. aripiprazole).
Clozapine, risperidone, olanzapine and ziprasidone
occupy more than 80% of cortical 5-HT2A receptors Potential therapeutic significance of targeting other
in the therapeutic dose range in humans (Table neuroreceptors The SGAs, particularly clozapine,
1).24,60,61,63,69,70 Although 5-HT2A receptor antagonism have multiple sites of action other than dopamine D2
is likely to be associated with the low EPS liability of receptors, including dopamine (D1, D3, D4), serotonin
SGAs, risperidone at higher doses produces EPS,71 (5-HT1A, 5-HT2C, 5-HT6, 5-HT7), muscarinic
indicating that high levels of D2. antagonism cannot cholinergic and histamine receptor (Table 1). Among
be completely ameliorated by even maximal 5-HT2A them, it has been hypothesized that the partial agonist
receptor antagonism. Moreover, at this point, it is activity of clozapine at serotonin 5-HT1A receptors
unclear what clinical effects 5-HT2A antagonism may contribute to its efficacy against anxiety, depres-
confers, beyond mitigating the adverse effect of sion, cognitive and negative symptoms of schizo-
striatal D2 antagonism, and propensity to cause phrenia (Table 2).76–79 Preclinical studies have also
EPS.72 In particular, the role of 5-HT2A antagonism suggested that 5-HT1A agonists may potentiate the
in the superior therapeutic responses to clozapine antipsychotic activity of dopaminergic antagonists,80
awaits further clarification.59 The apparent lack of and activation of inhibitory 5-HT1A autoreceptors may
efficacy of monotherapy with the selective potential counteract the induction of EPS due to striatal D2
role of the 5-HT2A receptor antagonist M-10090773 receptor blockade.81 Furthermore, 5-HT1A agonism
indicates that 5HT2A antagonism alone cannot explain has been suggested to contribute to enhancement of
the efficacy of SGAs. Further studies examining prefrontal dopamine release.82 Indeed, clozapine,
combination therapy with D2 antagonist and M- and olanzapine and ziprasidone, but not haloperidol
100907 are necessary to evaluate the potential role or risperidone, can preferentially augment dopamine
of 5HT2A antagonism. and norepinephrine release in the prefrontal cortex
relative to the subcortical areas, which may be rela-
The ‘fast-off-D2’ theory To date, there is no evidence ted to their potential efficacy for negative symptoms
showing that an agent without some degree of D2 and cognitive dysfunction of schizophrenia.83 The

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

83
Table 2 Potential clinical efficacy and benefits related to the mechanisms of action of antipsychotics (adapted from Richelson6
in part and modified)

Mechanisms Potential clinical efficacy Potential consequences

D2R antagonism k positive symptoms EPS


mNegative symptoms
mCognitive symptoms
D2R partial agonism k positive symptoms Little or no EPS
k negative symptoms Behavioral activation
k cognitive symptoms
mDA and NE release in the PFC k negative symptoms Behavioral activation
k cognitive symptoms
k depressive symptoms
ACh release in the PFC k cognitive symptoms
5-HT2A antagonism k negative symptoms k EPS

5-HT1A partial agonism k negative symptoms


k cognitive symptoms
k anxiety symptoms
k depressive symptoms
Muscarinic R antagonism k EPS m Anticholinergic symptoms
e.g. dry mouth, constipation tachycardia
Muscarinic R agonism k psychotic symptoms
k cognitive symptoms
Glutamate modulation k positive symptoms
k negative symptoms
k cognitive symptoms
k illness progression

NE, norepinephrine; Ach, acetylcholine; PFC, prefrontal cortex; EPS, extrapyramidal symptoms.

prefrontal cortex contains high densities of 5-HT1A experimentally induced NMDA receptor hypo-
and 5-HT2A receptors located on affrents to and on function at the cellular and behavioral levels,96–100
pyramidal neurons.84 It has been suggested that which provides support for the NMDA receptor
activation of 5-HT2A receptors increases the release hypofunction hypothesis. For example, clozapine
of glutamate onto pyramidal cells,85 whereas sero- and olanzapine, but not haloperidol or raclopride,
tonin, possibly via activation of 5-HT1A receptors, inhibit the electrophysiological effects of PCP in brain
inhibits the release of glutamate.86 Thus, compounds slices,99,101,102 and attenuate NMDA antagonist-
with 5-HT2A antagonism and/or 5-HT1A agonism like induced deficits in prepulse inhibition (PPI)96,103
clozapine could regulate the physiological balance and social behavior.97 In addition, ketamine-induced
between excitatory and inhibitory inputs onto brain metabolic activation is blocked by acute
prefrontal pyramidal neurons.78,84 Some SGAs, parti- administration of clozapine and olanzapine, but not
cularly ziprasidone, can also increase serotonin haloperidol in rats.98,100 The well-documented
activity in the frontal cortex by virtue of their effects of the SGAs on responses to NMDA
affinity for the serotonin transporter.87,88 In addition, antagonists raise the possibility that the therapeutic
some of the SGAs, but not FGAs, can increase the mechanisms of action of these agents may be
release of acetylcholine in the prefrontal cortex, which associated with counteracting the effects of NMDA
could be a possible factor contributing to improve receptor hypofunction.104 However, since none of
cognition in schizophrenia.89 the SGAs have direct affinity for any of the
glutamate receptors including the NMDA receptor,
Antipsychotic interactions with the glutamate the mechanism by which these effects are mediated
system The ability of noncompetitive N-methyl- is poorly understood. A recent report by Sur et al.105
D-aspartate (NMDA) receptor antagonists, such as could help to elucidate this question. They have
phencyclidine (PCP) and ketamine, to induce a shown that clozapine’s biologically active metabolite,
spectrum of positive, negative and cognitive schizo- N-desmethylclozapine, which is a potent, allosteric
phrenia-like symptoms has led to the hypothesis that agonist at muscarinic M1 receptors, can potentiate
hypofunction of NMDA receptors is involved in the hippocampal NMDA receptor currents through M1
pathophysiology of schizophrenia.90–95 In a wide receptor activation.105 Thus, clozapine’s unique thera-
range of preclinical paradigms, some of the SGAs, peutic profile may be, at least in part, attributed to
but not the FGAs, selectively antagonize the effects of N-desmethylclozapine through potentiation of NMDA

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

84
receptor function mediated by M1 receptors. Further pancies appear to be due to different treatment
studies are needed to determine whether the regimens, brain regions examined and the method of
inhibition of the effects of NMDA antagonists by the assessment. Thus, it is unclear whether such changes
SGAs involves molecular modifications in glutamate reflect increased or decreased function of different
receptors or altered other neurotransmitter–glutamate glutamate receptors after chronic antipsychotic treat-
interactions. ments.
In contrast to the acute effects, chronic administra-
tion of haloperidol can block not only PCP-induced Microarray analysis of gene expression induced by
deficits in PPI,106,107 but also ketamine-induced brain antipsychotic drug treatment Microarray expression
metabolic activation.108 Thus, adaptive changes eli- profiling appears to be a useful strategy to iden-
cited by chronic treatment with both the FGAs and tify candidate target genes that may be relevant to
the SGAs appear to attenuate the effects of NMDA the mechanisms of action of antipsychotic drugs.
antagonists.108 To date, numerous animal studies have Kontkanen et al124 have found that acute clozapine
reported increases, decreases, or no change in binding treatment induces the gene expression of chromo-
sites of glutamate receptors in various brain regions granin A and calcineurin A, and decreases synapto-
after chronic administration of antipsychotics109–115 tagmin V mRNA in the rat frontal cortex. In addition,
(Table 3). At the molecular level, inconsistent and chronic administration of clozapine induced the
often opposite findings have also been reported in the differential mRNA expression patterns of chromo-
gene expression of subunits composing different granin A, son of sevenless, and Sec-1 in the cortex.
glutamate receptors following long-term treatment In contrast, chronic exposure to haloperidol regula-
with both the FGAs and SGAs.116–123 These discre- ted the gene expression of inhibitor of DNA-binding

Table 3 Major findings in studies examining effects of chronic antipsychotics treatment on glutamate receptor binding and
mRNA expression (adapted from109123)

Antipsychotics NMDA-R AMPA-R KA-R Metabotropic-R

Haloperidol MK-801binding - k m (cortex) 3H- AMPA binding - m (cortex) KA binding - Group II mRNA
CGP-39653 binding m (striatum, AMPA mRNA m (GluB & (cortex) KA2 - (cortex)
cortex) MK-801binding - m GluC) (hippocampus) mRNA m
(striatum, hippocampus) MK- Subunits mRNA - (Acb, (cortex,
801binding m (Acb) striatum) GluR1 protein striatum,
m(cortex) GluR2 mRNA m k hippocampus)
(cortex, striatum) GluR4
mRNA k (cortex, striatum)
NR1 protein m (striatum) NR1
mRNA m k (striatum, cortex) NR1
mRNA m (hippocampus) NR2A-C
mRNA m (striatum) NR2A mRNA k
(cortex, hippocampus)

Clozapine MK-801binding - k (cortex) 3H- AMPA binding - (cortex) KA binding - Group II mRNA
CGP-39653 binding m (cortex) MK- AMPA mRNA m (GluB & (cortex) GluR7 m (cortex)
801binding k (striatum) MK- GluC) (hippocampus) GluC mRNA m
801binding m (Acb) mRNA k (Acb) GluR1 protein (cortex,
m (cortex) GluR2 mRNA - striatum) KA2
(striatum) GluR3 mRNA k mRNA m
(cortex, striatum) GluR4 (striatum)
mRNA k (striatum)
NR1 mRNA m k (Acb) NR1 mRNA
- (cortex, striatum, hippocampus)
NR1 protein - (striatum) NR2A
mRNA k (cortex, hippocampus)
NR2C mRNA k (cortex, Acb)

Olanzapine MK-801binding - (cortex) AMPA mRNA m (GluB & KA binding - Subunits mRNA
GluC) (hippocampus) AMPA (cortex) - (striatum,
binding m Subunits mRNA - hippocampus)
(cortex, striatum)
MK-801binding k Subunits mRNA
- (cortex, striatum, hippocampus)

Acb, nucleus accumbens.

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

85
2 (ID-2) and Rab-12. Moreover, the expression of Aripiprazole neither conforms to the standard 5-
visinin-like proteins was regulated by chronic HT2A/D2 antagonist nor the fast dissociation theories
treatments with both agents in various brain regions. of atypicality. It has a very high affinity for the D2
Chromogranin A and synaptotagmin V have been receptor (greater than its 5-HT2A affinity) and this is
suggested to play roles in presynaptic vesicle forma- unlikely to have a fast koff. Similarly, the compound
tion and secretion.125,126 Calcineurin A, a Ca2 þ / has a long half-life and is therefore unlikely to show
calmodulin-dependent protein phosphatase, and transient receptor occupancy. PET studies in normal
visinin-like proteins are involved in the regulation humans indicate that although aripiprazole occupies
of intracellular Ca2 þ metabolism (for reviews, see up to 90% of striatal D2-like dopamine receptors at
Bultynck et al127 and Braunewell and Gundefinger128). clinical doses, it does not cause EPS, suggesting that
Thus, it is possible that both antipsychotics may its inherent agonism may provide a mechanism that
modulate neurotransmitter vesicle release and protects against excessive blockade of the D2 sys-
presynaptic organization as well as the regulation tem.139 This underlines aripiprazole’s unique me-
of intracellular Ca2 þ in the cortex.124 Interestingly, chanism of action as a partial dopamine receptor
altered expression of genes involved in presynaptic agonist,79,134 and possibly a novel form of treatment
function has been observed in the prefrontal cortex of for schizophrenia.
the postmortem brains of schizophrenics.129 However,
Bauer et al.130 failed to detect any significant changes
Clinical pharmacological profiles of antipsychotic
in cortical synaptic protein levels or their enco-
drugs
ding mRNAs after chronic haloperidol adminis-
tration in rats. Furthermore, several genes involved First-generation antipsychotic agents
in the presynaptic function were not altered after Although the FGAs (eg, chlorpromazine and haloper-
chronic haloperidol treatment in the prefrontal idol) vary in potency, their pharmacological proper-
cortex of monkeys.129 The effects of chronic anti- ties, and their propensity to induce side effects, they
psychotic administration on presynaptic function are equally effective in the treatment of positive
at the molecular and cellular levels require further symptoms of schizophrenia and in preventing their
study. recurrence (for reviews, see Miyamoto et al8,9, Davis
et al140 and American Psychiatric Assocoation141).
However, approximately 30% of patients with acutely
Partial dopamine agonists exacerbated psychotic symptoms, however, have little
Aripiprazole (OPC-14597), approved for clinical use or no response to FGAs, and up to 60% of patients
in the US and more recently in Europe, is the first of a have only a partial response to medication.12,142 FGAs
possible ‘next- generation antipsychotics’ with a are generally less effective against negative than
mechanism of action that differs from currently positive symptoms of schizophrenia (for a review,
marketed FGAs and SGAs.79 It is a partial dopamine see Miyamoto et al8). They also produce small and
agonist with a high affinity for D2 and D3 receptors,131–133 inconsistent effects on cognitive functioning.143–145
and demonstrates properties of a functional agonist Some studies report worsening, improvement, or no
and antagonist in animal models of dopaminergic change in cognitive function with FGAs treatment.144
hypoactivity and hyperactivity, respectively.131,134 The discrepancies may be due, in part, to differences
Aripiprazole acts on both postsynaptic D2 receptors in patient populations, specific tests used, and the
and presynaptic autoreceptors. Partial agonist activity differential response of psychopathology to antipsy-
at D2 receptors could stabilize the dopamine system chotics.144 Cognitive impairment may also be wor-
while avoiding the hypodopaminergia that may limit sened by adjunctive anticholinergic medications,
the efficacy and tolerability of FGAs.135 In addition, which are frequently required to treat EPS caused by
aripiprazole displays 5-HT1A partial agonism and 5- FGAs.146
HT2A antagonism.136 The distinction, pharmacologi- In addition, the prophylactic efficacy of FGAs for
cally, between aripiprazole and the SGAs in this relapse prevention is limited by poor treatment
regard is that aripiprazole’s affinity for the D2 compliance and the fact that even with full compli-
receptors exceeds that for serotonin by an order of ance approximately 20% of patients may re-
magnitude.6,137 It also has very modest affinity lapse.147,148 Other dimensions of treatment
for alpha1-adrenergic, histamine (H1), 5-HT6, and limitations are functional capacity, quality of life,
5-HT7 receptors, and no appreciable affinity for D1, prevention of illness progression and improvement of
histaminergic or cholinergic muscarinic receptors long-term outcome.
(Table 1).132,137 The clinical significance of these in In terms of adverse effects, all of the FGAs can
vitro receptor-binding affinities as well as its partial 5- produce EPS at therapeutic doses, including parkin-
HT1A agonism has not been determined apart from sonism, dystonia, akathisia and tardive dyskinesia to
their obvious association with side effects.138 It has a varying degree, and increase serum prolactin
also been proposed that aripiprazole induces ‘func- concentration in the usual clinical dose range.149
tionally selective’ activation of D2 receptors coupled When present, these EPS side efffects can be
to diverse G proteins (and hence different functions), unpleasant for the patient and frequently an impor-
thereby explaining its unique clinical effects.132,137 tant reason for noncompliance with medication.150

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

86
Benzamides clozapine, risperidone, and olanzapine were 0.49,
A meta-analysis of 11 randomized controlled trials of 0.25 and 0.21, respectively, and each was signifi-
acutely ill schizophrenic patients comparing amisul- cantly greater than those of FGAs. Importantly, there
pride with FGAs or placebo found amisulpride to be was no evidence that the FGA dose affected the
consistently more effective than FGAs for global results. With respect to comparisons of efficacy
schizophrenic symptoms and negative symptoms.151 among SGAs, Geddes et al155 asserted that the SGAs
However, the mean effect size of amisulpride for are equally efficacious as a homogeneous group, but
change in the Brief Psychiatric Rating Scale (BPRS) Davis et al157 concluded that some SGAs (clozapine,
score was relatively small (0.11). In four studies of risperidone and olanzapine) are superior to other
patients with primary or predominantly negative SGAs (sertindole, quetiapine, ziprasidone and remox-
symptoms, amisulpride was more effective than ipride). It should be noted that these three meta-
placebo, but not more effective than FGAs.151 The analyses did not include data currently available to
agent also demonstrated its therapeutic benefit with evaluate the other clinical dimensions (eg, cognition,
little or no EPS, lower use of antiparkinsonian affect, quality of life) now receiving more attention.6
medication and fewer dropout rates due to adverse Moreover, the fact that most studies included in these
effects than FGAs.151 Its main side effect is substantial reviews were pharmaceutical industry-sponsored
elevations of prolactin. Additional research is neces- trials,158 used limited types of assessment measures
sary for clarifying whether amisulpride is really more and methods of data analysis (eg, last-observation-
effective for primary negative symptoms. carried-forward analyses)155 and were relatively short
in duration could limit the ability to fully evaluate the
Second-generation antipsychotic agents comparative effects of the two drug classes.158
Investigations of SGAs in very large samples and
Efficacy There have been numerous double-blind with head-to-head comparisons to each other, such as
studies comparing the efficacy and tolerability of the Clinical Antipsychotic Trials of Intervention
SGAs with FGAs for acute and maintenance therapy Effectiveness (CATIE), could provide adequate infor-
for schizophrenia. Such an expansive review is, mation regarding the role of SGAs in efficacy and
however, beyond the scope of this paper; thus, the effectiveness.158
reader is referred to other sources.1,5,152 In general,
although the proportion of patients who improve and Efficacy on negative symptoms Although SGAs have
the magnitude of therapeutic effects vary greatly, been shown to be more effective than FGAs in treating
SGAs appear to be at least as effective for psychotic negative symptoms, there is a continuing debate as to
symptoms as FGAs (for reviews, see Markowitz whether these ostensible therapeutic effects are
et al153 and Remington and Kapur154). However, secondary to a reduction in EPS or other symp-
there has been considerable debate with regard to toms, or to a direct effect on primary negative
the clinical superiority of SGAs over FGAs. symptoms.71,154,159–162 Secondary negative symptoms
Geddes et al conducted a systematic review and may be associated with positive symptoms, EPS,
meta-analyses of 52 randomized trials comparing new depression and environmental deprivation,163,164 but
antipsychotics (clozapine, olanzapine, risperidone, most clinical studies of SGAs do not distinguish
quetiapine, sertindole and amisulpride) with FGAs between primary and secondary negative symptoms
(haloperidol or chlorpromazine).155 There was no and generally involve acutely symptomatic patients
difference in efficacy between FGAs and SGAs, in undergoing treatment for their psychosis.165
trials that used a dose of an SGA in haloperidol Moreover, the effect sizes of improvement on
equivalents of 12 mg/day or less. For example, the negative symptoms for SGAs are usually moderate
advantages of SGAs in terms of efficacy and dropout to small in comparison with placebo or FGAs.156,166,167
rates were not seen if haloperidol is used at doses of Path analyses, however, have suggested that both
12 mg/day or less, although SGAs still caused fewer risperidone and olanzapine exert direct effects on
EPS.155 They concluded that the observed superior (primary) negative symptoms independent of diffe-
efficacy of some SGAs may be due to the negative rences in psychotic, depressive or extrapyramidal
effects on efficacy of the excessively high dose of the symptoms.168,169 Nevertheless, these path analytical
FGA comparator. statistical approaches were performed post hoc, and
Leucht et al156 also performed a meta-analysis and only the most effective doses of the SGAs were
concluded that SGAs have efficacy and tolerability chosen. Thus, prospective studies on stable patients
advantages over FGAs, although the benefits of new with predominant primary negative symptoms are
agents in terms of total, positive and negative necessary to draw conclusions on this issue.170 A
symptoms are modest at best. However, they did not collaborative working group concluded that SGAs are
attempt to address dose in the same fashion as Geddes superior in terms of the ‘totality’ of negative
et al.155 symptoms, but their impact on specific components
A third meta-analysis was performed by Davis is still under investigation.164
et al157 on 124 randomized controlled studies with
efficacy data on 10 SGAs vs FGAs, and 18 trials of Efficacy on cognition Studies of the effect of SGAs
comparisons between SGAs. The effect sizes of on cognition have been limited, and the findings have

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

87
been inconsistent.171–173 It is unclear whether this FGAs in terms of overall psychopathology, EPS, and
effect is dependent on or independent of treatment compliance rate. There are several controlled, double-
effects on psychotic symptoms.2 Anticholinergic and blind trials comparing SGAs with FGAs in treatment-
sedative effects as well as the propensity to induce refractory schizophrenia, but the relative efficacy of
EPS associated with antipsychotic medications may other new agents is modest or less clear (for a review,
have detrimental effects on some (but not all) areas of see Miyamoto et al1). Volavka et al389 in a double-
cognition.174–176 Improvement in global cognitive blind PET found clozapine and olanzapine but not
functioning with SGAs may be secondary to less risperidone superior to haloperidol. Sequential
EPS liability and greater efficacy in the treatment of controlled trials of the newer agents in treatment-
negative symptoms.173 In general, the SGAs have resistant patients will be necessary to fully examine
demonstrated superior efficacy compared to FGAs on this issue.
tests of verbal fluency, digit-symbol substitution, fine
motor function, and executive function.171,172,177 Safety The major difference of the SGAs compared
Measures of learning and memory were least to the FGAs is their lower incidence of EPS and TD.
affected by SGAs.171 Because these tests all measure Most of the SGAs have little or no EPS, while
performance during a timed trial, enhanced risperidone has less at low doses but at higher doses
performance with SGAs could result, in part, from can cause EPS comparable to that of FGAs.189
reduced parkinsonian side effects 171. In a double- However, the individual SGAs have unique adverse
blind trial in the treatment of cognitive impairment in effects that were less of an issue with FGAs, are of
early-phase schizophrenia, risperidone (mean dose potential concern and undermine claims of safety
6 mg/day) and olanzapine (mean dose 11 mg/day) advantages for the SGAs. There is mounting evidence
produced significantly greater improvement in of the increased risk of weight gain, diabetes mellitus,
verbal fluency compared to haloperidol (mean dose prolonged QTc interval and possible secondary
10 mg/day), and olanzapine was superior to both cardiovascular complications.6 These side effects are
haloperidol and risperidone in effects on motor skills, associated with potential long-term health risks of
nonverbal fluency and immediate recall.178 This patients as well as decreased adherence to treatment
finding is, however, complicated by the high regimens, and eventually may lead to relapse.189
incidence of anticholinergic administration prior to Among SGAs, clozapine and quetiapine have been
the final cognitive assessment, and other problems in shown to carry minimal to no risk for EPS or
methodology.179,180 In a recent double-blind 14-week hyperprolactinemia within the therapeutic dosage
trial in chronic schizophrenia,181 olanzapine and range.185,190,191 Risperidone, however, can produce
risperidone were superior to haloperidol on global dose-related EPS (Z6 mg/day).192,193 With the excep-
cognitive function, but not different from each other tion of akathisia,194 the incidence of EPS or hyper-
or from clozapine. Average effect sizes for prolactinemia with olanzapine and ziprasidone is not
improvement with the SGAs were in the small to significantly different from that with placebo.195,196
medium range. Importantly, their results did not The relative liability of the individual SGAs to
appear to be mediated by changes in symptoms, side produce EPS will become apparent only when they
effects, or blood levels of medications. In contrast, have been directly compared with each other in
Green et al182 have found no cognitive advantage prospective clinical trials such as the NIMH ¼ CATIE
for risperidone (mean dose 6 mg/day) over low doses trials.390
of haloperidol (mean dose 5 mg/day) in stable schizo- Marked differences in liability for weight gain,
phrenic outpatients over a 2-year period.182 There is diabetes, or hyperlipidemia are seen between the
continued debate as to whether SGAs have pro-cogni- different SGAs. Analyses consistently report the
tive efficacy or have reduced cognitive liability.183,184 largest increases in weight gain with clozapine and
As in efficacy studies for negative symptoms, dose olanzapine, intermediate increases with risperidone
equivalence is an important factor in trials comparing and quetiapine, and minimal weight gain with
cognitive effects of SGAs, particularly since excessive ziprasidone.197–199
doses can impair performance on time-sensitive tasks
and can increase anticholinergic exposure. Keefe et Aripiprazole: efficacy and safety
al171 reported a meta-analysis which found a Several short-term double-blind, placebo-controlled
significant advantage for SGAs on cognitive test trials of aripiprazole (2–30 mg/day) demonstrated
performance. efficacy greater than placebo, and similar to haloper-
idol (10 mg/day) and risperidone (6 mg/day) against
Efficacy for treatment-resistant schizophrenia positive and negative symptoms in patients with
Clozapine has been consistently shown to have acute exacerbations of schizophrenia or schizoaffec-
efficacy against psychotic symptoms in well-defined tive disorder200,201 (for a review, see Bowles and
treatment refractory patients over FGAs and as Levin138). These studies suggest that aripiprazole
compared to other SGAs.185–187 Chakos et al,188 in a doses in the range of 15–30 mg/day are effective. It
review and meta-analysis of seven controlled trials can be started at a full dose without an initial titration
comparing clozapine to FGAs in treatment-resistant period. Two long-term double-blind studies showed
schizophrenia, found that clozapine is superior to that aripiprazole (15 or 30 mg/day) is superior to

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

88
placebo, and comparable or superior to haloperidol ments for negative and cognitive symptoms of
(10 mg/day) in maintaining antipsychotic response schizophrenia.9,21,218–221
and relapse prevention.202,203 All of the short- and
long-term studies have shown that aripiprazole has a Dopamine D4 receptor antagonist There are several
favorable safety and tolerability profile, with low lines of evidence suggesting that selective dopamine
liability for EPS, TD, weight gain, sedation, hyper- D4 receptor antagonists may be potential novel
prolactinemia, or QTc prolongation, and a lack of antipsychotic drugs. For example, not only
adverse effects on glucose and lipid metabolism.204 clozapine but also a number of clinically efficacious
However, it should be noted that the agent exhibits a antipsychotics have a relatively high affinity for the
lack of a predictable dose–response relationship for D4 receptors222 (Table 1). In addition, an increase in D4
efficacy and adverse events.201,204 Further clinical receptors has been reported in the schizophrenic
studies are needed to determine the efficacy and brains.223,224 The selective D4 antagonist,
safety in special populations, including suicidal and sonepiprazole (U-101387 or PNU-101387G)
treatment-resistant patients with schizophrenia, el- attenuates apomorphine-induced impairment of
derly and children, dementia, agitation, autism and prepulse inhibition,225 and antagonizes the decrease
other disease status in which antipsychotic use is in c-fos expression in the medial prefrontal cortex and
helpful.138 Long-term effectiveness studies are also neurotensin mRNA in the nucleus accumbens
necessary, particularly those examining the relative produced by repetitive amphetamine administration
effects of aripiprazole and different SGAs on negative in rats.226 However, most of the in vivo pharma-
symptoms, cognitive function, relapse prevention, cological studies of sonepiprazole indicates the
treatment adherence, disease progression, function, lack of effects in at least traditional preclinical
quality of life and use of health services.205 models of antipsychotic activity.227,228 Nevertheless,
it entered Phase II clinical trials in patients with
schizophrenia, but no further data are currently
Future strategies of drug development available.228
Dopaminergic agents An initial clinical trial with another highly selec-
tive D4 antagonist, L-745,870 failed to demonstrate
Dopamine D1 receptor antagonist or agonist Evidence any antipsychotic activity in the treatment of schizo-
suggests an important role for D1-like dopamine phrenia.229,230 While the single dose tested and the
receptors in the pathophysiology of small number of patients make it difficult to draw
schizophrenia.206,207 Earlier preclinical studies firm conclusions regarding the potential efficacy of D4
demonstrated that selective D1-like antagonists were antagonists as antipsychotic agents,231 this drug
active in most traditional functional models held to seemed to cause a worsening of symptoms.230 Simi-
predict antipsychotic activity (for a review, see larly, NGD-94-1 and the D4/5-HT2A antagonist finan-
Waddington208). A clinical trial of the selective D1- serin (RP62203) also did not show clinical efficacy in
like antagonist SCH39166,209,210 and NNC 01-0687,211 limited trials in schizophrenics.228,232 Therefore, these
however, demonstrated no antipsychotic activity, and data cast doubt as to whether D4 antagonism alone is
instead may have aggravated psychoses in some responsible for the antipsychotic efficacy of cloza-
patients.209 pine, and that selective D4 antagonists could indeed
In contrast to the ineffectiveness of D1-like antago- have therapeutic potential in schizophrenia (for
nists in the treatment of schizophrenia, low doses of reviews, see Miyamoto et al5, Waddington et al31
selective full D1-like receptor agonists, such as and Rowley et al233).
dihydrexidine, A77636 and SKF81297, have been
reported to have cognitive-enhancing actions in non- Dopamine D3 antagonist or partial agonist Dopamine
human primates.212–214 In drug-naı̈ve schizophrenics, D3 receptor is a D2-like dopamine receptor that is
Okubo et al215 found decreased D1-like receptor localized in the mesolimbic areas of the brain, and for
binding using PET in the frontal cortex and basal which most antipsychotics have relatively high
ganglia, and correlation between the reduction in affinity (for a review, see Schwartz et al234). In
prefrontal D1-like receptors and the severity of addition, a post-mortem study demonstrated
negative symptoms and cognitive disturbance. Such elevation of D3 receptor levels in the limbic striatum
data are consistent with the fact that pyramidal of drug-free patients with schizophrenia, whereas D3
neurons in the prefrontal cortex postulated to be receptor expression was normal in subjects treated
involved in working memory express a high degree of with antipsychotic drugs.235 These findings have
D1-like dopamine receptors.207,216 It is postulated that prompted much interest in the D3 receptor as a
either insufficient or excessive D1-like receptor potential novel therapeutic target for antipsychotic
stimulation is deleterious to cognitive function of activity.31 A dopamine D3 receptor agonist, ( þ )-PD
the prefrontal cortex, thus an ‘optimal’ level of D1-like 128,907, can block stereotypy produced by NMDA
receptor activation is necessary for normal cognitive antagonists in mice, suggesting its antipsychotic
function.216,217 The finding that full D1-like receptor profile.236 So far, partial agonists at D3 receptors are,
agonists can improve working memory suggests that however, supposed to be beneficial only when
such class of drugs might be novel potential treat- administered to drug abusers or in Parkinson’s

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

89
disease (for review, see Hacking and Stark237). The control the extracellular glycine concentration.252
novel selective dopamine D3 antagonists such as Thus, blockade of the GLYT-1 transporter would
S33084, SB-277011-A, and AVE5997 have been deve- increase NMDA receptor-mediated transmission.
loped for the treatment of psychosis like schizo- Although there is some controversy as to whether
phrenia. While S33084 was not active in traditional the glycine regulatory site on the NMDA receptor is
models of antipsychotic activity in the manner of D2 saturated under physiological conditions, preclinical
antagonists,238 SB-277011-A can produce an increase data demonstrate that N-(3-(40 -fluorophenyl)-3-
in extracellular levels of dopamine, norepinephrine (40 -phenylphenoxy)propyl) sarcosine, a selective and
and acetylcholine in the rat anterior cingulate cortex, potent GLYT-1 reuptake inhibitor, can potentiate
similar to the effects of SGAs, clozapine and electrophysiological effects of NMDA.252,253 Further-
olanzapine, but not haloperidol.239 At present, the more, the glycine reuptake inhibitor glycyldo-
role of D3 antagonism in antipsychotic activity decylamide attenuated PCP-induced hyperactivity
remains unclear, and only controlled clinical trials more potently than glycine.254,255 These preclinical
with selective D3 antagonists in schizophrenia will data suggest that inhibition of glycine reuptake could
eventually clarify this issue.31 represent a feasible approach to potentiate NMDA
receptor-mediated neurotransmission and, possibly,
Glutamatergic agents treat schizophrenic patients.

NMDA receptor positive allosteric modulators If Glutamate reuptake inhibitors Glutamate trans-
reduced NMDA receptor function is involved in the porters (excitatory amino-acid transporters (EAATs)),
pathophysiology of schizophrenia, then drugs that normally expressed in both glia (EAAT1 and EAAT2)
enhance NMDA receptor function could be and neurons (EAAT3 and EAAT4), can control
therapeutic agents and potentially improve upon, or glutamatergic neurotransmission by removal of
supplement, current antipsychotic treatments glutamate from the synaptic cleft (for a review, see
(for reviews, see Miyamoto et al8, Duncan et al59, Danbolt256). EAAT3 (called EAAC1 in the rodent) is
Abi-Saab et al240 and Goff and Coyle241). Direct predominantly expressed in the cerebral cortex, basal
agonists of the NMDA receptor, however, may not be ganglia and hippocampus.257 Post-mortem studies in
feasible candidates in this regard, because of the schizophrenic patients revealed alterations in gene
propensity of such drugs to produce excessive expression of glutamate transporters.258,259 In
excitation and seizures. addition, preclinical studies demonstrated that
Glycine is a positive allosteric modulator and chronic treatment with clozapine or haloperidol can
obligatory co-agonist at the NMDA receptor.242 This downregulate EAAT3 in the infralimbic cortex and
allosteric regulatory site represents a potential target hippocampal CA2.260 Thus, glutamate reuptake
for drugs to augment NMDA-mediated neurotransmis- inhibitors such as EAAT3 antagonists could increase
sion. The glycine site agonists, including glycine, the synaptic availability of glutamate, and increase
D-cycloserine and D-serine, appear to be effective in glutamatergic action at the postsynaptic neuron, and
reducing negative symptoms and cognitive impair- thereby might produce therapeutic effects on some
ment in patients with schizophrenia when they are symptom dimensions following the model of
added to ongoing antipsychotic treatment, with the diminished glutamate activity in schizophrenia.
exception of clozapine.243–249 Their beneficial effects
on positive and depressive symptoms are less robust. Metabotropic glutamate receptors agonists
The poor penetration of the blood–brain barrier by Metabotropic glutamate (mGlu) receptors (mGluR),
glycine, and the partial agonistic properties of D- of which there are eight subtypes (mGluR1–8), are
cycloserine, appear to make these agents less than categorized into three groups according to their
optimal for providing pharmacological agonism of the agonist pharmacology, sequence similarity and signal
glycine regulatory site on the NMDA receptor.59 Of transduction pathways (for a review, see Rowley et
these glycine agonists, D-serine appears to be the most al233). NMDA antagonists induce hyperlocomotion
promising agent. It is a full agonist on the strychnine- and stereotypy, accompanied by an increase in
insensitive glycine site of NMDA receptor250 and is glutamate release in several brain regions of rats,261
more permeable than glycine at the blood–brain suggesting that pharmacological agents that decrease
barrier, thus requiring a lower dosage. In an 8-week glutamate release should block the effects of the
clinical trial, D-serine added to neuroleptic treatment drugs. Group II mGluR (mGluR2/3) are located
in treatment-resistant patients with schizophrenia presynaptically on glutamate terminals where they
demonstrated significant improvements not only in may act as autoreceptors regulating glutamate release
negative and cognitive symptoms but also an in in vivo (for reviews, see Rowley et al233 and Chavez-
positive symptoms, which is different from glycine.251 Noriega et al262). Administration of a group II mGluR
agonist, LY-354740, blocks both behavioral activation
Glycine reuptake inhibitors Glycine transporters, and increased glutamate (but not dopamine) release
GLYT-1 and GLYT-2, have been identified on both provoked by PCP in rats.263 Thus, group II mGluR
neuronal and glial cells in the central nervous system. agonists could be beneficial in the treatment of
A function of these transporters has been suggested to schizophrenia, although LY-354740 cannot attenuate

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

90
PCP-induced disruptions in prepulse inhibition (PPI) Glutathione prodrugs Glutathione is the principal
of acoustic startle responses. On the other hand, nonenzymatic endogenous antioxidant, and plays a
one of the effects induced by activation of group I critical role in protecting cells from damage by
mGluR (mGluR1/5), particularly of mGluR5, is a reactive oxygen species generated by dopamine
significant potentiation of NMDA receptor func- metabolism.274,275 A glutathione deficit can leave the
tion, suggesting that mGluR5 agonists may display brain susceptible to oxidation, and oxidative stress-
antipsychotic activity (for review, see Chavez-Noriega mediated cell damage has been considered in one of
et al262). the pathophysiologies of schizophrenia. Indeed, a
decrease of glutathione levels was observed in the
cerebrospinal fluid and the medial prefrontal cortex
AMPA/kainate receptor antagonists The increased in drug-naı̈ve schizophrenic patients.274 Its deficit
release of glutamate observed in response to NMDA would lead to degenerative processes in the
antagonists could mediate some of the behavioral surrounding of dopaminergic terminals, resulting in
actions of the drugs by activation of non-NMDA loss of connectivity.274 Glutathione also potentiates
receptors, including a-amino-3-hydroxy-5-methy-iso- the NMDA receptor response to glutamate via its
xazole-4-propionic acid (AMPA) and kainate recep- redox modulatory site.276 Taken together, although
tors.261 In support of the hypothesis that behavioral speculative, glutathione supplementation by gluta-
effects of NMDA antagonists relate to increased thione prodrugs could be an interesting treatment
glutamate release, administration of an AMPA/ strategy for schizophrenia in terms of preventing
kainate receptor antagonist, LY-293558, partially re- oxidative stress and enhancing neurotransmission at
versed impairment of working memory induced by NMDA receptors in the brain.
subanesthetic doses of ketamine in rats.261.Furthermore,
AMPA/kainate receptor antagonists reduce NMDA Noradrenergic agents
antagonist-induced hyperlocomotion264–266 and neuro-
degeneration.267 Systemic administration of other Alpha-2 adrenergic receptor agonist or anta-
AMPA receptor antagonists GYKI52466 and LY- gonist Norepinephrine plays an important role in
326325 can suppress conditioned avoidance response cognitive function of the prefrontal cortex (PFC) by its
in rats.268 These data suggest that AMPA/kainate actions at alpha-2 adrenergic receptors (Ars) located
receptor antagonists may possess an antipsychotic in the principal sulcus of the PFC (for reviews, see
effect, and have utility for treatment of cognitive Goldman-Rakic et al277, Arnsten et al278 and Friedman
deficits in which NMDA receptor hypofunction is et al279,280). Indeed, the alpha-2 agonist clonidine has
suspected.261 been shown to improve performance on working
memory tasks in young monkeys with noradrenergic
depleting lesions of the PFC, presumably through its
Ampakines (CX-516) In apparent contrast to the drug actions at post-synaptic alpha-2 Ars in the
postulated utility of AMPA/kainate receptor anta- PFC.281 In schizophrenia, clonidine also improves
gonists as antipsychotics, ampakines, a class of com- PFC-mediated cognitive dysfunction.282 In addition,
pounds that allosterically enhance AMPA receptor guanfacine, a selective alpha-2A agonist,283 improves
function, have also been suggested to represent poten- PFC-mediated working memory in aged non-human
tial adjunctive treatments for schizophrenia. primates, but without the significant adverse effects
Ampakines and AMPA potentiators enhance excita- associated with clonidine (eg, sedation, hypoten-
tory (glutamatergic) transmission, facilitate long-term sion).284 A 4-week, placebo-controlled, double-blind
potentiation, learning and memory in rodents,269,270 study demonstrated the efficacy and safety of guan-
and have synergistic effects with FGAs and SGAs on facine as adjunctive treatment of cognitive impair-
blocking behavioral effects of methamphetamine.271 In ment in schizophrenia.285 Those patients receiving
addition, preliminary results suggest that chronic guanfacine plus risperidone showed significant
administration of an ampakine (CX-516) can improve improvement on tasks of working memory and atten-
negative and cognitive symptoms in schizophrenia tion compared with patients receiving FGAs plus
patients that also receive clozapine.272 Thus, such guanfacine.285 The potential ability of alpha-2
findings are paradoxical with regard to the hypo- agonists to improve cognitive performance on tasks
thesis that excessive glutamate release may be dependent on PFC function appears to be of great
involved in behavioral effects of reduced NMDA importance in the search for a new pharmacologic
receptor function. In a recent double-blind placebo- approach for schizophrenia.
controlled small study of patients with schizophrenia Clozapine and risperidone have potent antagonist
who were partially refractory to treatment with the properties at alpha-2 Ars. Millan et al286 have
FGAs, CX516 as a sole agent did not produce drama- postulated the significance of the alpha-2 Ars antag-
tic effects on positive symptoms and cognitive impair- onistic activity for the antipsychotic effects of neuro-
ment.273 In the case of AMPA ligands, it seems at leptics. Blockade of inhibitory alpha-2-AR heterocep-
present unclear if agonists, antagonists or partial tors on terminals of dopaminergic fibers can enhance
agonists/modulators have potential therapeutic frontocortical dopaminergic transmission compared
application. with subcortical dopaminergic pathways.287 Litman

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

91
et al288 reported that combined treatment with as 3-2,4-dimethoxybenzylidene anabaseine (DMXB-A
idazoxan, a highly selective alpha-2-AR antago- or GTS-21) can normalize the auditory gating deficits
nist, and the FGA fluphenazine can produce a in rodents.303 Alpha-7 nAChR agonists are currently
‘clozapine-like’ profile of antipsychotic activity. under development for clinical trial in schizophrenia,
Antagonist properties of alpha-2-Ars appear to be although it is unclear whether such agonists have
implicated in the functional actions of clozapine in beneficial effects on symptoms other than the
humans,289 and contribute to an improvement in auditory gating deficit. Moreover, long-term use of
mood.286 such agents might induce desensitization of nAChRs,
leading to tolerance and therefore limiting the
duration of efficacy.
COMT inhibitors Considerable data suggest that
catechol-O-methyl transferase (COMT), a postsynap- Alpha4–beta2 nicotinic receptor agonist It has been
tic methylation enzyme that metabolizes released suggested that alpha4–beta2 nAChRs affect auditory
dopamine, is primarily responsible for synaptic sensorimotor gating.304 They are considered to
dopamine inactivation in the prefrontal cortex, and represent more than 90% of the high-affinity
that variation in COMT activity may affect prefrontal nicotine-binding sites in rat brain,305 and appear to
cortical activity, especially during working memory play an important role in many of the behavioral
tasks.290,291 Interestingly, studies of COMT-deficient actions of nicotine. SIB-1553A, an alpha4–beta2
mice have demonstrated that dopamine levels are subtype-selective nicotinic receptor agonist, appears
increased in the prefrontal cortex but not in the to produce enhanced performance in a variety of
striatum, and that memory performance is en- models of cognitive impairment in areas such as
hanced.292 Abnormalities of prefrontal dopamine spatial and nonspatial working and reference memory
function associated with working memory appear to in aged rodents and monkeys.306,307 Interestingly, SIB-
be prominent features of schizophrenia, and certain 1553A has also been shown to stimulate the release of
alleles of the COMT gene run in families with a high dopamine, norepinephrine and acetylcholine in the
incidence of the illness.293 Tolcapone, a reversible, frontal cortex and hippocampus in rats.306 These data
selective inhibitor of COMT has been reported to suggest that the use of alpha4–beta2 nicotinic
improve working memory in rodents,294 and receptor agonists could produce therapeutic benefit
tolcapone as adjunct to L-dopa therapy has been for the treatment of cognitive deficits in schizo-
shown to improve cognitive dysfunction in patients phrenia.
with advanced Parkinson’s disease.295 At present,
Egan and Weinberger are conducting a trial of COMT Allosteric modulators of nicotinic receptor and
inhibitors in schizophrenia and healthy controls with acetylcholinesterase inhibitor Galantamine is a
and without the high-risk combination of COMT positive allosteric modulator of nAChRs and an
alleles.296 However, tolcapone was withdrawn from inhibitor of acetylcholinesterase (AchE), the enzyme
the market in Europe and Canada due to the risk of responsible for catabolizing acetylcholine (Ach) (for a
serious hepatic dysfunction,297 and in the US review, see Maelicke et al308). The allosteric inter-
restrictive liver enzyme monitoring measures are action amplifies the actions of Ach at pre- and post-
necessary, which severely limits the use of the synaptic nAChR.309 Presynaptic nAChRs are capable
agent.298 of modulating the release of Ach, and other neuro-
transmitters, such as glutamate, serotonin, and
Cholinergic agents GABA, which may contribute to the symptoms of
schizophrenia.310 Galantamine has been shown to
Alpha-7 nicotinic receptor agonist Cognitive improve cognitive and global function in placebo-
impairments are cardinal features of schizophrenia. controlled trials in Alzheimer’s disease patients (for a
Nicotinic acetylcholine receptors (nAChRs) have review, see Coyle and Kershaw311). Results from case
been implicated in cognitive function and reports suggest that adjuvant galantamine adminis-
formation of sensory processing (for a review, see tration improves negative symptoms in patients with
Rezvani and Levin299). In particular, auditory gating treatment-refractory schizophrenia.312,313 The extent
is modulated by the alpha-7 nAChR subtype, which to which the clinical benefits of galantamine are
is a rapidly desensitizing low-affinity nAChR (for attributable specifically to its nicotinic effects is
a review, see Simosky et al300). Genetic studies linking unclear, and prospective double-blind data are
the alpha-7 nAChR gene to sensory processing defi- required.
cits in schizophrenia, together with reductions of this Several case studies and an open-label trial of
receptor in discrete regions of the brains of schizo- adjunctive donepezil, a reversible AchE inhibitor,
phrenia patients, suggest that the alpha-7 nAChR may demonstrate some of its beneficial effects on cogni-
be a relevant therapeutic target in schizophrenia (for tive impairment in schizophrenia.314–316 However, a
review, see Adler et al301). Interestingly, clozapine, recent double-blind controlled trial of donepezil
but not haloperidol, can improve deficient inhibitory added to risperidone did not show any positive
auditory processing through stimulation of alpha-7 effects on cognitive deficit associated with schizo-
nAChRs in mice.302 Agonists at alpha-7 nAChRs such phrenia.317

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

92
Muscarinic receptor agonist There is a large body of tor antagonist, SR141716, can reduce hyperactivity
anatomical and pharmacological evidence for induced in gerbils by various stimulant drugs,
potential modulation of dopamine and glutamatergic including cocaine, D-amphetamine, morphine and
neurons by cholinergic muscarinic receptors (for Win 55212-2, known to produce or exacerbate
a review, see Bymaster318). Recent findings that schizophrenic symptoms.329 In addition, SR141716
partial agonists of muscarinic receptors are active in dose-dependently alters Fos protein and neurotensin
animal models are predictive of antipsychotic expression in a manner comparable to that observed
activity, and the SGAs clozapine and olanzapine are with the SGAs.330 These findings suggest that
partial agonists for cholinergic M1, M2 and M4 selective CB1 receptor antagonists may be effective
receptors. Recently, the N-desmethyl metabolite of in the pharmacological treatment of schizophrenia.
clozapine was reported to preferentially bind to M1
muscarinic receptors with an IC50 of 55 nM, and was Neurokinin 3 antagonist Neurokinin 3 (NK3)
a more potent partial agonist (EC50, 115 nM and 50% tachykinin receptors appear to regulate midbrain
of acetylcholine response) at this receptor than dopamine neuronal activity.331 Preclinical studies
clozapine.105 Furthermore, pharmacological and site- have shown that a potent and selective nonpeptide
directed mutagenesis studies suggested that N- NK3 antagonist, osanetant (SR-142801), selectively
desmethylclozapine preferentially activated M1 inhibits dopamine release in certain brain regions (for
receptors by interacting with a site that does not a review, see Kamali332). Several NK3 compounds are
fully overlap with the acetylcholine orthosteric site. currently in development (eg, osanetant (Sanofi-
Moreover, N-desmethylclozapine is able to potentiate Synthélabo) and talnetant (Glaxo Smith Kline)) as
hippocampal NMDA receptor currents through M1 potential treatments for schizophrenia. Preliminary
receptor activation. In addition, muscarinic agonists clinical trials have demonstrated that osanetant is
have activity in animal models of negative symptoms, superior to placebo on global assessment of efficacy
cognitive dysfunction and affective disorders, and measures of positive symptoms in
suggesting the potential usefulness of muscarinic schizophrenia.333 Whether NK3 antagonist adminis-
agonists in the treatment of schizophrenia (for tration in schizophrenia may serve as novel anti-
reviews, see Rowly et al233 and Bymaster et al319). psychotics merits further investigation.
Examples of these agents are the muscarinic M1/M4
agonist xanomeline, and the muscarinic M2/M4 Neurotensin agonist Neurotensin (NT) is a
agonists PTAC, and BuTAC (for a review, see neuropeptide that regulates the function of
Bymaster318). Xanomeline has been demonstrated to mesolimbic dopamine neurons,334 and has been
have positive effects on cognitive and psychotic-like implicated in the pathophysiology of schizophrenia
symptoms (eg, hallucinations and delusions) in (for a review, see Binder et al335). The central
Alzheimer’s disease.320 Accumulating data suggest administration of NT induces behavioral and
that muscarinic partial agonists might be efficacious biochemical effects that are very similar to the
in treating not only positive, but also negative and effects of antipsychotic drugs.336 Thus, there is
cognitive symptoms in schizophrenia.319,321,322 much interest in the potential use of NT agonists
as novel antipsychotics. In vitro studies indicate
Other agents that NT behaves as an agonist at NT1 receptors and
as an antagonist at NT2 receptors.337 Systematic
Cannabinoid CB1 antagonist Acute cannabis administration of metabolically stable NT analogues
intoxication can produce schizophrenia-like such as PD-149163 and NT69L can attenuate
symptoms, including hallucinations, altered amphetamine- or NMDA antagonist-induced
judgment, false beliefs and cognitive dysfunction,323 hyperactivity and PPI without inducing catalepsy
and long-term cannabis use often induces negative or affecting baseline startle responses.338,339 NT1
schizophrenia-like symptoms.324 In addition, canna- receptor agonists, however, may have problems in
bis can precipitate psychotic symptoms in schizo- clinical use, since NT can produce autonomic adverse
phrenia, and may increase the risk of developing the effects. Chronic administration of SR-48692, which
illness.325 Several reports also demonstrated elevated is a nonpeptide and a high-affinity NT1 receptor
levels of the endogenous cannabinoids (anandamide antagonist with agonist activity at NT2 receptors,
and palmitylethanolamide) in both the cerebrospinal can decrease methamphetamine-induced dopamine
fluid and the blood of schizophrenic patients when release in the nucleus accumbens,340 suggesting this
compared to normal controls.326,327 These accu- compound could also have antipsychotic-like effects.
mulating evidences have led to ‘a cannabinoid
hypothesis of schizophrenia’ in which cannabinoid MAO B inhibitors It has been suggested that negative
receptors, the pharmacological target of cannabis- symptoms of schizophrenia may be manifestations
derived drugs, and their accompanying system of of regionally deficient dopaminergic activity in the
endogenous activators may be dysregulated in brain, thus augmentation of dopaminergic neurotrans-
schizophrenia.328 The endogenous cannabinoid sys- mission could be a beneficial treatment strategy.
tem comprises at least two cannabinoid receptors, Selegiline (deprenyl) is a monoamine oxidase
the CB1 and CB2 receptors. A selective CB1 recep- (MAO)-B inhibitor that selectively enhances

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

93
dopaminergic activity. Selectivity for inhibition of Neurosteroids Dehydroepiandrosterone: Dehydro-
MAO-B without inhibition of MAO-A is clinically epiandrosterone (DHEA) and its sulfate derivative
important, since MAO-A inhibition is responsible for (DHEA-S) are neuroactive neurosteroids that repre-
most of the side effects of MAO inhibitors.341 sent steroid hormones synthesized de novo in the
Although several case series reported the beneficial brain and acting locally on nerve cells.356 Although
effects of selegiline on negative symptoms of DHEA and DHEA-S are the most abundant circulating
schizophrenia,342–344 one double-blind, controlled steroid hormones in humans, their precise physio-
study of the agent as adjunct to antipsychotic logical roles remain uncertain. In vitro data suggest
treatment failed to offer therapeutic benefit.345 The that DHEA and DHEA-S enhance neuronal and glial
selective irreversible MAO-B inhibitors, selegiline survival and differentiation.357–359 In addition, DHEA-
and rasagiline, have been shown to possess S shows marked neuroprotective ability against the
neuroprotective activities in cell culture and in vivo glutamate-induced toxicity360 and oxidative stress.361
models of Parkinson’s disease (for a review, see Interestingly, DHEA has been demonstrated to
Maruyama et al346). For example, these agents can potentiate neuronal responses at the NMDA
prevent experimentally induced apoptotoic DNA receptor.359,362,363 The enhancement of physiological
damage, and induce pro-survival genes.347 Thus, the response to NMDA by DHEA has also been suggested
MAO-B inhibitors may rescue degenerating dopamine to result from agonistic actions at ó1 receptors in the
neurons through inhibiting death signal transduction, brain.363 Consistent with a positive modulatory action
but clinical trials failed to confirm it.346 So far, no of DHEA at the NMDA receptor, the neurosteroid
solid conclusions could be drawn from the data enhances memory,364–367 and DHEA-S attenuates
regarding the effects of the MAO-B inhibitors on NMDA receptor antagonist MK-801-induced learning
schizophrenia. impairment in mice.368 In chronic schizophrenics,
significantly lower morning levels of plasma
DHEA were observed.369 Further, there are a number
PDE10 inhibitors PDE10A is a recently identified of earlier case reports suggesting that DHEA may
cyclic nucleotide phosphodiesterase expressed at be useful in the treatment of schizophrenia, espe-
high levels in the brain and more specifically in the cially for negative symptoms.370–372 A recent double-
medium spiny neurons of the striatum and associated blind study of DHEA as adjunct to antipsychotic
nucleus accumbens and olfactory tubercle.348 treatment in chronic schizophrenic patients with
Papaverine, a potent and selective PDE10A prominent negative symptoms suggests that it can
inhibitor, can dose-dependently attenuate hyper- improve negative, depressive and anxiety symp-
activity induced by both amphetamine and PCP in toms of the illness, especially in women.373
rats.349 The agent does not affect extracellular Although the mechanism of action of DHEA and
dopamine in the striatum nor alter PCP-induced DHEA-S has still to be further characterized, the
dopamine release in the nucleus accumbens. possibility that these compounds may have efficiency
Papaverine can also produce a dose-dependent in schizophrenia should be further investigated.
reduction in conditioned avoidance responding in Pregnenolone: Intensive studies in animals estab-
rodents. These data suggest the possibility that lished that neuroactive steroids such as pregnenolone
selective inhibitors of PDE10 may provide a target (PREG) and pregnenolone sulfate (PREGS) display
for the development of a new class of antipsychotic neuronal actions and influence behavioral func-
drugs.349 tions.374 For example, PREG and PREGS exhibit
memory-enhancing properties in aged rodents (for a
review, see Vallee et al375). It has also been suggested
NNOS inhibitor Nitric oxide (NO) is a diffusible gas that PREGS can ameliorate MK-801-induced memory
and an important inter- and intracellular messenger in impairment by acting as ó1 receptor agonists.376 In
the central nervous system. NO activates guanylyl addition, PREGS can attenuate the conditioned fear
cyclase and increases the synthesis of cyclic stress response via ó1 receptors.377 Albeit speculative,
GMP. Nitric oxide synthase (NOS) converts arginine these findings suggest that PREG and PREGS may
into NO and citrulline in response to increased intra- have therapeutic potential for improving cognitive
cellular calcium levels (for a review, see Rowley et deficit observed in schizophrenia.
al233). Preclinical studies demonstrated that inhi-
bition of NOS activity with methylene blue, L- Neurotrophic factors The role of neurotrophic
NOARG, L-NAME, or 7-nitroindazole can attenuate factors in the pathophysiology of schizophrenia is
hyperactivity and disruption of PPI produced by rapidly becoming an important and exciting focus of
NMDA antagonists but not amphetamine.350–354 In investigation. A more recent pathophysiologic theory
treatment-refractory schizophrenia, methylene blue of schizophrenia suggests that it involves a limited
moderately improved symptoms.355 However, it neurodegenerative process reflected by the
should be noted that all available NOS inhibitors progressive and deteriorating clinical course of the
can produce serious side effects including hyper- illness.378 Recent longitudinal neuroimaging studies
tension and cognitive dysfunction, which may be of first-episode schizophrenia have demonstrated
due to poor selectivity for different NOS isoforms.233 morphological changes in cortical gray matter and

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

94
ventricular volumes that have been suggested to reviews, see Basile et al385 and Arranz and Kerwin388).
reflect pathological processes of developmental In addition, a more long-term approach may be to take
maturation and/or illness progression.379–381 The advantage of information from clinical imaging (sMRI,
neurotrophic factors such as nerve growth factor structural magnetic resonance imaging; MRS, mag-
(NGF), brain-derived neurotrophic factor (BDNF) netic resonance spectroscopy; fMRI, functional MRI)
and neurotrophin (NT)-3/4/5 play a decisive role in and electrophysiologic (ERP, event-related potential)
a neurodevelopmental process, including neuronal studies that are becoming increasingly sophisticated,
and glial differentiation, migration, proliferation providing more detailed information of the individual
and regeneration.382 They are not only active during brain morphologic changes, pathways and circuits
embryo- and organogenesis but also influence the involved in various aspects and stages of schizophre-
synaptic organization and the synthesis of neuro- nia.233 These measures can also serve to define bio or
transmitters in the adult brain, and are therefore surrogate markers of treatment effects.
involved in the maintenance of neural plasticity.382 As yet no drug that did not have some affinity
Thus, pathological alterations of the neurotrophic for and activity at the dopamine D2 receptor has
factor system may lead to neural maldevelopment, been proven to have antipsychotic efficacy. Thus,
migration deficits and dysconnections, which are the development of novel compounds targeting
proposed to be the characteristic pathogenetic other systems than dopamine will likely be employed
features of the neurodevelopmental hypothesis of as adjunctive or combined treatments in addition
schizophrenia.382,383 If neurotrophic factors salvage to whatever monotherapeutic applications they may
degenerating cells, facilitate desirable synaptic have.
connections, and hence, halt the progression of With the improvements in pharmacological treat-
neurodegenerative process of schizophrenia, drugs ment seen over the last decade, the focus of therapy
that selectively stimulate the production of neuro- for schizophrenia has shifted from the relief of
trophic factors could represent a new approach to psychotic symptoms to other pathological domains
forestall the progression of schizophrenia and prevent including negative and mood symptoms, cognitive
morbidity from increasing.384 Although neurotrophic deficits and the functional impairment that under-
factors are unable to cross the blood–brain barrier, mines patients’ capacities for daily functioning,
potential strategies for the administration of these reintegration into society, and recovery. At present,
factors are transplantation of neurotrophic factor- much remains to be done in terms of developing
producing cells, direct transfection of neurotrophic treatment strategies and the determination of its
factor gene and development of compounds which optimal use in conjunction with psychosocial and
modulate endogenous neurotrophic factor homeo- adjunctive therapies.
stasis and/or the influence their signal transduction
mechanisms.382 The augmentation therapy with
Conclusion
neurotrophic factors suggests novel and innovative
pharmacotherapeutic, but as yet unproven, strategies Psychopharmacological research efforts have focused
for schizophrenia. on developing compounds with unique combinations
of effects at different perisynaptic neurotransmitter
sites as described above.189 Future efforts must move
Future directions
beyond strategies that develop drugs which solely
Although a recent meta-analysis suggests that some target the modulation of chemical neurotransmission
SGAs are more efficacious than FGAs,157 one cannot at synapses to the development of agents that can
reliably predict which patient will respond best to a affect other cellular functions including signal trans-
particular antipsychotic medication.205 Significant duction, signaling pathways and gene expression,
differences between the new antipsychotics are and that underlie mechanisms of cell develop-
emerging so that drug choice needs to be tailored for ment, plasticity and resilience. In addition, efforts
individual patients.152 Recent developments in si- to identify genetic mechanisms that underlie mental
multaneous profiling of gene transcripts (gene chips) illnesses will reveal new targets for drug develop-
and gene products (proteomics) will allow definition ment. Such efforts could offer a more powerful
of the genes and proteins that are affected by method for identifying the neural and molecular
antipsychotic medication. It is very likely that mechanisms causing schizophrenia, and for develop-
individual genetic differences are important deter- ing animal models and novel therapeutic approaches.
mining factors in the efficacy and side effect profiles One of the important goals of pharmacological
of antipsychotic medication.385 Therefore, it is possi- research should be to develop new ideal antipsycho-
ble to improve drug response at the level of the tic drugs with low associated risk, rapid onset of
individual patient by detecting single-nucleotide action, a more effective treatment for negative,
polymorphism in patients’ DNA.386,387 Knowledge of cognitive and affective symptoms, in better efficacy
the relationship between specific genetic polymorph- against positive symptoms, and improved relapse
isms of genes involved in a drug’s pharmacokinetics rates and reduction or even reversal of cumulative
and pharmacodynamics may lead to better drug morbidity.189 The hope for further progress relies
design and to individualized pharmacotherapy (for upon development of a number of different basic and

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

95
clinical neuroscience strategies, but with these in- 14 Schulz C, McGorry P. Traditional antipsychotic medications:
novations it is likely that future progress will be contemporary clinical use. In: Buckley PF, Waddington JL (eds).
relatively quick to come. Schizophrenia and Mood Disorders: The New Drug Therapies in
Clinical Practice. Butterworth-Heinemann: Woburn, MA, pp
2000; 14–20.
15 Breier A, Wright P, Birkett M, Meehan K, David, Brook S. A
Competing interests double-blind dose response study comparing intramuscular
olanzapine, haloperidol and placebo in acutely agitated schizo-
The following financial interests by coauthors in-
phrenic patients, ACNP 39th Annual Meeting Abstract. American
clude consultant fees, honoraria and/or research College of Neuropsychopharmacology: Puerto Rico, 2000.
funds. 16 Fleischhacker WW. New developments in the pharmaco-
Seiya Miyamoto: Eli Lilly Japan therapy of schizophrenia. J Neural Transm 2003; 64(Suppl):
Gary E Duncan: Eli Lilly 105–117.
17 Marder SR, Van Putten T. Antipsychotic medications. In:
Christine E Marx: Eli Lilly Schatzberg AF, Nemeroff CB (eds). The American Psychiatric
Jeffrey A Lieberman: Abbott, Astra Zeneca, Bristol- Press Textbook of Psychopharmacology. American Psychiatric
Myers Squibb, Eli Lilly, GlaxoSmithKline, Janssen, Press, Inc.: Washington, DC, pp 1995; 247–261.
Novartis, Johnson and Johnson, Organon, Pfizer and 18 Creese I, Burt DR, Snyder SH. Dopamine receptor binding
Sanofi-Synthelabo Solvay. predicts clinical and pharmacological potencies of antischizo-
phrenic drugs. Science 1976; 192: 480–483.
19 Seeman P, Lee T, Chau-Wong M, Wong K. Antipsychotic drug
doses and neuroleptic/dopamine receptors. Nature 1976; 261:
Acknowledgements 717–719.
20 Seeman P. Dopamine receptors and the dopamine hypothesis of
This work was supported in part by the UNC schizophrenia. Synapse 1987; 1: 133–152.
Schizophrenia Research Center, an NIMH Silvio O. 21 Miyamoto S, Mailman RB, Lieberman JA, Duncan GE. Blunted
Conte Center for the Neuroscience of Mental Dis- brain metabolic response to ketamine in mice lacking D1A
orders (MH064065)(JL), and the Foundation of Hope dopamine receptors. Brain Res 2001; 894: 167–180.
of Raleigh North Carolina (JL, GD). We thank Mr Kenji 22 Kapur S, Seeman P. Antipsychotic agents differ in how fast
they come off the dopamine D2 receptors. Implications for
Seo and Ms Yoshiko Suzuki for preparing some atypical antipsychotic action. J Psychiatry Neurosci 2000; 25:
references. No support was provided by any pharma- 161–166.
ceutical company. 23 Remington G, Kapur S. D2 and 5-HT2 receptor effects of
antipsychotics: bridging basic and clinical findings using PET.
J Clin Psychiatry 1999; 60(Suppl 10): 15–19.
References 24 Farde L, Nordstrom AL, Wiesel FA, Pauli S, Halldin C,
Sedvall G. Positron emission tomographic analysis of
1 Miyamoto S, Lieberman JA, Fleischhacker WW, Aoba A, Marder central D1 and D2 dopamine receptor occupancy in patients
SR. Antipsychotic drugs. In: Tasman A, Kay J, Lieberman JA treated with classical neuroleptics and clozapine. Relation
(eds). Psychiatry, 2nd edn. John Wiley & Sons, Ltd: Chichester, to extrapyramidal side effects. Arch Gen Psychiatry 1992; 49:
2003, pp 1928–1964. 538–544.
2 Buckley PF. Broad therapeutic uses of atypical antipsychotic 25 Kapur S, Remington G, Jones C, Wilson A, DaSilva J, Houle S et
medications. Biol Psychiatry 2001; 50: 912–924. al. High levels of dopamine D2 receptor occupancy with low-
3 Kane JM, Leucht S, Carpenter D, Docherty JP. Expert consensus dose haloperidol treatment: a PET study. Am J Psychiatry 1996;
guideline series. Optimizing pharmacologic treatment of psycho- 153: 948–950.
tic disorders. Introduction: methods, commentary, and summary. 26 Nordstrom AL, Farde L, Wiesel FA, Forslund K, Pauli S, Halldin
J Clin Psychiatry 2003; 64(Suppl 12): 5–19. C et al. Central D2-dopamine receptor occupancy in relation to
4 McEvoy JP, Scheifler PL, Frances A. The expert consensus antipsychotic drug effects: a double-blind PET study of schizo-
guideline series: treatment of schizophrenia 1999. J Clin Psy- phrenic patients. Biol Psychiatry 1993; 33: 227–235.
chiatry 1999; 60: 1–80. 27 Kapur S, Zipursky R, Jones C, Remington G, Houle S. Relation-
5 Miyamoto S, Stroup TS, Duncan GE, Aoba A, Lieberman JA. ship between dopamine D2 occupancy, clinical response, and
Acute pharmacologic treatment of schizophrenia. In: Hirsch SR, side effects: a double-blind PET study of first-episode schizo-
Weinberger DR (eds). Schizophrenia, 2nd edn. Blackwell Science, phrenia. Am J Psychiatry 2000; 157: 514–520.
Oxford, 2003, pp 442–473. 28 Bigliani V, Mulligan RS, Acton PD, Visvikis D, Ell PJ, Stephenson
6 Remington G. Understanding antipsychotic ‘atypicality’: a clin- C et al. In vivo occupancy of striatal and temporal cortical D2/D3
ical and pharmacological moving target. J Psychiatry Neurosci dopamine receptors by typical antipsychotic drugs. epidepride
2003; 28: 275–284. single photon emission tomography (SPET) study. Br J Psychiatry
7 Richelson E. Receptor pharmacology of neuroleptics: relation to 1999; 175: 231–238.
clinical effects. J Clin Psychiatry 1999; 60(Suppl 10): 5–14. 29 Xiberas X, Martinot JL, Mallet L, Artiges E, Loc’h C, Maziere B et
8 Miyamoto S, Duncan GE, Goff DC, Lieberman JA. Therapeutics of al. Extrastriatal and striatal D(2) dopamine receptor blockade
schizophrenia. In: Davis KL, Charney D, Coyle JT, Nemeroff C with haloperidol or new antipsychotic drugs in patients with
(eds). Neuropsychopharmacology: The Fifth Generation of schizophrenia. Br J Psychiatry 2001; 179: 503–508.
Progress. Lippincott Williams & Wilkins, Philadelphia, 2002, 30 Nyberg S, Farde L. Non-equipotent doses partly explain differ-
pp 775–807. ences among antipsychotics—implications of PET studies.
9 Miyamoto S, Duncan GE, Mailman RB, Lieberman JA. Developing Psychopharmacology 2000; 148: 22–23.
novel antipsychotic drugs:strategies and goals. Curr Opin CPNS 31 Waddington JL, Kapur S, Remington GJ. The neuroscience and
Invest Drugs 2000; 2: 25–39. clinical psychopharmacology of first- and second-generation
10 Kane JM. Schizophrenia. N Engl J Med 1996; 334: 34–41. antipsychotic drugs. In: Hirsch SR, Weinberger DR (eds).
11 Lewis DA, Lieberman JA. Catching up on schizophrenia: natural Schizophrenia, 2nd edn. Blackwell Science: Oxford, 2003, pp
history and neurobiology. Neuron 2000; 28: 325–334. 421–441.
12 Kane JM. The current status of neuroleptic therapy. J Clin 32 Kapur S, Zipursky R, Roy P, Jones C, Remington G, Reed K et al.
Psychiatry 1989; 50: 322–328. The relationship between D2 receptor occupancy and plasma
13 Sharif ZA. Common treatment goals of antipsychotics: acute levels on low dose oral haloperidol: a PET study. Psychophar-
treatment. J Clin Psychiatry 1998; 59(Suppl. 19): 5–8. macology 1997; 131: 148–152.

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

96
33 Stip E. Novel antipsychotics: issues and controversies. Typicality 53 Xiberas X, Martinot JL, Mallet L, Artiges E, Canal M, Loc’h C et al.
of atypical antipsychotics. J Psychiatry Neurosci 2000; 25: In vivo extrastriatal and striatal D2 dopamine receptor blockade
137–153. by amisulpride in schizophrenia. J Clin Psychopharmacol 2001;
34 Burt DR, Creese I, Snyder SH. Antischizophrenic drugs: chronic 21: 207–214.
treatment elevates dopamine receptor binding in brain. Science 54 Bressan RA, Erlandsson K, Jones HM, Mulligan R, Flanagan RJ,
1977; 196: 326–328. Ell PJ et al. Is regionally selective D2/D3 dopamine occupancy
35 Florijn WJ, Tarazi FI, Creese I. Dopamine receptor subtypes: sufficient for atypical antipsychotic effect? An in vivo quantita-
differential regulation after 8 months treatment with antipsycho- tive epidepride SPET study of amisulpride-treated patients. Am J
tic drugs. J Pharmacol Exp Ther 1997; 280: 561–569. Psychiatry 2003; 160: 1413–1420.
36 Lee T, Seeman P, Tourtellotte WW, Farley IJ, Hornykeiwicz O. 55 Trichard C, Paillere-Martinot ML, Attar-Levy D, Recassens C,
Binding of 3H-neuroleptics and 3H-apomorphine in schizophre- Monnet F, Martinot JL. Binding of antipsychotic drugs to cortical
nic brains. Nature 1978; 274: 897–900. 5-HT2A receptors: a PET study of chlorpromazine, clozapine, and
37 Silvestri S, Seeman MV, Negrete JC, Houle S, Shammi CM, amisulpride in schizophrenic patients. Am J Psychiatry 1998;
Remington GJ et al. Increased dopamine D2 receptor binding after 155: 505–508.
long-term treatment with antipsychotics in humans: a clinical 56 Seeman P. Atypical antipsychotics: mechanism of action. Can J
PET study. Psychopharmacology 2000; 152: 174–180. Psychiatry 2002; 47: 27–38.
38 Lai H, Carino MA, Horita A. Chronic treatments with zotepine, 57 Meltzer HY, Matsubara S, Lee JC. Classification of typical and
thioridazine, and haloperidol affect apomorphine-elicited stereo- atypical antipsychotic drugs on the basis of dopamine D1, D2
typic behavior and striatal 3H-spiroperidol binding sites in the and Serotonin2 pKi values. J Pharmacol Exp Ther 1989; 251:
rat. Psychopharmacology 1981; 75: 388–390. 238–246.
39 Fleminger S, Rupniak NM, Hall MD, Jenner P, Marsden CD. 58 Lieberman JA. Understanding the mechanism of action of
Changes in apomorphine-induced stereotypy as a result of atypical antipsychotic drugs: a review of compounds in use and
subacute neuroleptic treatment correlates with increased D-2 development. Br J Psychiatry 1993; 163: 7–18.
receptors, but not with increases in D-1 receptors. Biochem 59 Duncan GE, Zorn S, Lieberman JA. Mechanisms of typical and
Pharmacol 1983; 32: 2921–2927. atypical antipsychotic drug action in relation to dopamine and
40 Bunney BS, Grace AA. Acute and chronic haloperidol treatment: NMDA receptor hypofunction hypotheses of schizophrenia. Mol
comparison of effects on nigral dopaminergic cell activity. Life Psychiatry 1999; 4: 418–428.
Sci 1978; 23: 1715–1728. 60 Kapur S, Zipursky RB, Remington G, Jones C, DaSilva J, Wilson
41 Chiodo LA, Bunney BS. Typical and atypical neuroleptics: AA et al. 5-HT2 and D2 receptor occupancy of olanzapine in
differential effects of chronic administration on the activity of schizophrenia: a PET investigation. Am J Psychiatry 1998; 155:
A9 and A10 midbrain dopaminergic neurons. J Neurosci 1983; 3: 921–928.
1607–1619. 61 Kapur S, Zipursky RB, Remington G. Clinical and theoretical
42 White FJ, Wang RY. Differential effects of classical and atypical implications of 5-HT2 and D2 receptor occupancy of clozapine,
antipsychotic drugs on A9 and A10 dopamine neurons. Science risperidone, and olanzapine in schizophrenia. Am J Psychiatry
1983; 221: 1054–1057. 1999; 156: 286–293.
43 Mereu G, Lilliu V, Vargiu P, Muntoni AL, Diana M, Gessa GL. 62 Kapur S, Zipursky R, Jones C, Shammi CS, Remington G, Seeman
Failure of chronic haloperidol to induce depolarization inactiva- P. A positron emission tomography study of quetiapine in
tion of dopamine neurons in unanesthetized rats. Eur J schizophrenia: a preliminary finding of an antipsychotic effect
Pharmacol 1994; 264: 449–453. with only transiently high dopamine D2 receptor occupancy.
44 Mereu G, Lilliu V, Vargiu P, Muntoni AL, Diana M, Gessa GL. Arch Gen Psychiatry 2000; 57: 553–559.
Depolarization inactivation of dopamine neurons: an artifact? 63 Nordstrom AL, Farde L, Nyberg S, Karlsson P, Halldin C, Sedvall
J Neurosci 1995; 15: 1144–1149. G. D1, D2, and 5-HT2 receptor occupancy in relation to clozapine
45 Melis M, Mereu G, Lilliu V, Quartu M, Diana M, Gessa GL. serum concentration: a PET study of schizophrenic patients. Am J
Haloperidol does not produce dopamine cell depolarization- Psychiatry 1995; 152: 1444–1449.
block in paralyzed, unanesthetized rats. Brain Res 1998; 783: 64 Seeman P, Tallerico T. Antipsychotic drugs which elicit little or
127–132. no parkinsonism bind more loosely than dopamine to brain D2
46 Moore H, Todd CL, Grace AA. Striatal extracellular dopamine receptors, yet occupy high levels of these receptors. Mol
levels in rats with haloperidol-induced depolarization block Psychiatry 1998; 3: 123–134.
of substantia nigra dopamine neurons. J Neurosci 1998; 18: 65 Bench CJ, Lammertsma AA, Dolan RJ, Grasby PM, Warrington SJ,
5068–5077. Gunn K et al. Dose dependent occupancy of central dopamine D2
47 Boye SM, Rompre PP. Behavioral evidence of depolarization receptors by the novel neuroleptic CP-88,059-01: a study using
block of dopamine neurons after chronic treatment with positron emission tomography and 11C-raclopride. Psychophar-
haloperidol and clozapine. J Neurosci 2000; 20: 1229–1239. macology 1993; 112: 308–314.
48 Grace AA, Bunney BS, Moore H, Todd CL. Dopamine-cell 66 Bench CJ, Lammertsma AA, Grasby PM, Dolan RJ, Warrington SJ,
depolarization block as a model for the therapeutic actions of Boyce M et al. The time course of binding to striatal dopamine D2
antipsychotic drugs. Trends Neurosci 1997; 20: 31–37. receptors by the neuroleptic ziprasidone (CP-88,059-01) deter-
49 Schoemaker H, Claustre Y, Fage D, Rouquier L, Chergui K, Curet mined by positron emission tomography. Psychopharmacology
O et al. Neurochemical characteristics of amisulpride, an atypical 1996; 124: 141–147.
dopamine D2/D3 receptor antagonist with both presynaptic and 67 Keck PJ, Buffenstein A, Ferguson J, Feighner J, Jaffe W, Harrigan
limbic selectivity. J Pharmacol Exp Ther 1997; 280: 83–97. EP et al. Ziprasidone 40 and 120 mg/day in the acute exacerbation
50 Waddington J, Casey D. Comparative pharmacology of classical of schizophrenia and schizoaffective disorder: a 4-week placebo-
and novel (second-generation) antipsychotics. In: Buckley PF, controlled trial. Psychopharmacology 1998; 140: 173–184.
Waddington JL (eds). Schizophrenia and Mood Disorders: The 68 Goff DC, Posever T, Herz L, Simmons J, Kletti N, Lapierre K et al.
New Drug Therapies in Clinical Practice. Butterworth-Heine- An exploratory haloperidol-controlled dose-finding study of
mann: Woburn, MA, 2000, pp 3–13. ziprasidone in hospitalized patients with schizophrenia
51 Perrault G, Depoortere R, Morel E, Sanger DJ, Scatton B. or schizoaffective disorder. J Clin Psychopharmacol 1998; 18:
Psychopharmacological profile of amisulpride: an antipsychotic 296–304.
drug with presynaptic D2/D3 dopamine receptor antagonist 69 Farde L, Nyberg S, Oxenstierna G, Nakashima Y, Halldin C,
activity and limbic selectivity. J Pharmacol Exp Ther 1997; 280: Ericsson B. Positron emission tomography studies on D2 and 5-
73–82. HT2 receptor binding in risperidone-treated schizophrenic
52 Martinot JL, Paillere-Martinot ML, Poirier MF, Dao-Castellana patients. J Clin Psychopharmacol 1995; 15: 19S–23S.
MH, Loc’h C, Maziere B. In vivo characteristics of dopamine D2 70 Fischman AJ, Bonab AA, Babich JW, Alpert NM, Rauch SL,
receptor occupancy by amisulpride in schizophrenia. Psycho- Elmaleh DR et al. Positron emission tomographic analysis of
pharmacology 1996; 124: 154–158. central 5-hydroxytryptamine2 receptor occupancy in healthy

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

97
volunteers treated with the novel antipsychotic agent, ziprasi- 93 Coyle JT. The glutamatergic dysfunction hypothesis for schizo-
done. J Pharmacol Exp Ther 1996; 279: 939–947. phrenia. Harv Rev Psychiatry 1996; 3: 241–253.
71 Marder SR, Meibach RC. Risperidone in the treatment of 94 Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner
schizophrenia. Am J Psychiatry 1994; 151: 825–835. JD et al. Subanesthetic effects of the noncompetitive NMDA
72 Lieberman JA, Mailman RB, Duncan G, Sikich L, Chakos M, antagonist, ketamine, in humans. Psychotomimetic, perceptual,
Nichols DE et al. Serotonergic basis of antipsychotic drug effects cognitive, and neuroendocrine responses. Arch Gen Psychiatry
in schizophrenia. Biol Psychiatry 1998; 44: 1099–1117. 1994; 51: 199–214.
73 Carlsson A. Focusing on dopaminergic stabilizers and 5-HT2A 95 Malhotra AK, Pinals DA, Weingartner H, Sirocco K, Missar CD,
receptor antagonists. Curr Opin CPNS Invest Drugs 2000; 2: Pickar D et al. NMDA receptor function and human cognition—
22–24. The effects of ketamine in healthy volunteers. Neuropsychophar-
74 Kapur S, Seeman P. Does fast dissociation from the dopamine macology 1996; 14: 301–307.
d(2) receptor explain the action of atypical antipsychotics? a new 96 Bakshi VP, Geyer MA. Antagonism of phencyclidine-induced
hypothesis. Am J Psychiatry 2001; 158: 360–369. deficits in prepulse inhibition by the putative atypical anti-
75 Seeman P, Tallerico T. Rapid release of antipsychotic drugs psychotic olanzapine. Psychopharmacology 1995; 122: 198–201.
from dopamine D2 receptors: an explanation for low 97 Corbett R, Camacho F, Woods AT, Kerman LL, Fishkin RJ, Brooks
receptor occupancy and early clinical relapse upon withdrawal K et al. Antipsychotic agents antagonize non-competitive N-
of clozapine or quetiapine. Am J Psychiatry 1999; 156: methyl-D-aspartate antagonist-induced behaviors. Psychophar-
876–884. macology 1995; 120: 67–74.
76 Meltzer HY. Pre-clinical pharmacology of atypical antipsychotic 98 Duncan GE, Leipzig JN, Mailman RB, Lieberman JA. Differential
drugs: a selective review. Br J Psychiatry 1996; 29(Suppl): 23–31. effects of clozapine and haloperidol on ketamine-induced brain
77 Newman-Tancredi A, Chaput C, Verriele L, Millan MJ. Clozapine metabolic activation. Brain Res 1998; 812: 65–75.
is a partial agonist at cloned, human serotonin 5-HT1A receptors. 99 Wang RY, Liang X. M100907 and clozapine, but not haloperidol
Neuropharmacology 1996; 35: 119–121. or raclopride, prevent phencyclidine-induced blockade of NMDA
78 Millan MJ. Improving the treatment of schizophrenia: focus on responses in pyramidal neurons of the rat medial prefrontal
serotonin (5-HT)(1A) receptors. J Pharmacol Exp Ther 2000; 295: cortical slice. Neuropsychopharmacology 1998; 19: 74–85.
853–861. 100 Duncan GE, Miyamoto S, Leipzig JN, Lieberman JA. Comparison
79 Keltner NL, Johnson V. Biological perspectives. Aripiprazole: a of the effects of clozapine, risperidone, and olanzapine on
third generation of antipsychotics begins? Perspect Psychiatr ketamine-induced alterations in regional brain metabolism.
Care 2002; 38: 157–159. J Pharmacol Exp Ther 2000; 293: 8–14.
80 Evenden JL. Effects of 8-hydroxy-2-(di-n-propylamino)tetralin (8- 101 Arvanov VL, Liang X, Schwartz J, Grossman S, Wang RY.
OH-DPAT) after repeated administration on a conditioned Clozapine and haloperidol modulate N-methyl-D-aspartate- and
avoidance response (CAR) in the rat. Psychopharmacology non-N-methyl-D-aspartate receptor-mediated neurotransmission
1992; 109: 134–144. in rat prefrontal cortical neurons in vitro. J Pharmacol Exp Ther
81 Lucas G, Bonhomme N, De Deurwaerdere P, Le Moal M, 1997; 283: 226–234.
Spampinato U. 8-OH-DPAT, a 5-HT1A agonist and ritanserin, a 102 Arvanov VL, Wang RY. Clozapine, but not haloperidol, prevents
5-HT2A/C antagonist, reverse haloperidol-induced catalepsy in the functional hyperactivity of N-methyl-D-aspartate receptors in
rats independently of striatal dopamine release. Psychopharma- rat cortical neurons induced by subchronic administration of
cology 1997; 131: 57–63. phencyclidine. J Pharmacol Exp Ther 1999; 289: 1000–1006.
82 Ichikawa J, Ishii H, Bonaccorso S, Fowler WL, O’Laughlin IA, 103 Bakshi VP, Swerdlow NR, Geyer MA. Clozapine antagonizes
Meltzer HY. 5-HT(2A) and D(2) receptor blockade increases phencyclidine-induced deficits in sensorimotor gating of the
cortical DA release via 5-HT(1A) receptor activation: a possible startle response. J Pharmacol Exp Ther 1994; 271: 787–794.
mechanism of atypical antipsychotic-induced cortical dopamine 104 Duncan GE, Sheitman BB, Lieberman JA. An integrated view of
release. J Neurochem 2001; 76: 1521–1531. pathophysiological models of schizophrenia. Brain Res Rev 1999;
83 Li X-M, Perry KW, Wong DT, Bymaster FP. Olanzapine increases 29: 250–264.
in vivo dopamine and norepinephrine release in rat prefrontal 105 Sur C, Mallorga PJ, Wittmann M, Jacobson MA, Pascarella D,
cortex, nucleus accumbens and striatum. Psychopharmacology Williams JB et al. N-desmethylclozapine, an allosteric agonist at
1998; 136: 153–161. muscarinic 1 receptor, potentiates N-methyl-D-aspartate receptor
84 Martin-Ruiz R, Puig MV, Celada P, Shapiro DA, Roth BL, Mengod activity. Proc Natl Acad Sci U S A 2003; 100: 13674–13679.
G et al. Control of serotonergic function in medial prefrontal 106 Pietraszek M, Ossowska K. Chronic treatment with haloperidol
cortex by serotonin-2A receptors through a glutamate-dependent diminishes the phencyclidine-induced sensorimotor gating def-
mechanism. J Neurosci 2001; 21: 9856–9866. icit in rats. Naunyn-Schmiedeberg’s Arch Pharmacol 1998; 357:
85 Aghajanian GK, Marek GJ. Serotonin model of schizophrenia: 466–471.
emerging role of glutamate mechanisms. Brain Res Brain Res Rev 107 Ossowska K, Pietraszek M, Wardas J, Nowak G, Zajaczkowski W,
2000; 31: 302–312. Wolfarth S et al. The role of glutamate receptors in antipsychotic
86 Tanaka E, North RA. Actions of 5-hydroxytryptamine on neurons drug action. Amino Acids 2000; 19: 87–94.
of the rat cingulate cortex. J Neurophysiol 1993; 69: 1749–1757. 108 Duncan GE, Miyamoto S, Lieberman JA. Chronic administration
87 Ase AR, Amdiss F, Hebert C, Huang N, van Gelder NM, Reader of haloperidol and olanzapine attenuates ketamine-induced
TA. Effects of antipsychotic drugs on dopamine and serotonin brain metabolic activation. J Pharmacol Exp Ther 2003; 305:
contents and metabolites, dopamine and serotonin transporters, 999–1005.
and serotonin1A receptors. J Neural Transm 1999; 106: 75–105. 109 Giardino L, Bortolotti F, Orazzo C, Pozza M, Monteleone P, Calza
88 Tarazi FI, Zhang K, Baldessarini RJ. Olanzapine, quetiapine, and L et al. Effect of chronic clozapine administration on MK801-
risperidone: long-term effects on monoamine transporters in rat binding sites in the rat brain: a side-preference action in cortical
forebrain. Neurosci Lett 2000; 287: 81–84. areas. Brain Res 1997; 762: 216–218.
89 Ichikawa J, Dai J, O’Laughlin IA, Fowler WL, Meltzer HY. 110 McCoy L, Cox C, Richfield EK. Antipsychotic drug regulation of
Atypical, but not typical, antipsychotic drugs increase cortical AMPA receptor affinity states and GluR1, GluR2 splice variant
acetylcholine release without an effect in the nucleus accumbens expression. Synapse 1998; 28: 195–207.
or striatum. Neuropsychopharmacology 2002; 26: 325–339. 111 Ossowska K, Pietraszek M, Wardas J, Nowak G, Wolfarth S.
90 Deutsch SI, Mastropaolo J, Schwartx BL, Rosse R, Morihisa JM. A Chronic haloperidol and clozapine administration increases the
‘glutamatergic hypothesis’ of schizophrenia.Rationale for phar- number of cortical NMDA receptors in rats. Naunyn-Schmiede-
macotherapy with glycine. Clin Neuropharmacol 1989; 12: 1–13. berg’s Arch Pharmacol 1999; 359: 280–287.
91 Javitt DC, Zukin SR. Recent advances in the phencyclidine model 112 Spurney CF, Baca SM, Murray AM, Jaskiw GE, Kleinman JE,
of schizophrenia. Am J Psychiatry 1991; 148: 1301–1308. Hyde TM. Differential effects of haloperidol and clozapine
92 Olney JW, Farber NB. Glutamate receptor dysfunction and on ionotropic glutamate receptors in rats. Synapse 1999; 34:
schizophrenia. Arch Gen Psychiatry 1995; 52: 998–1007. 266–276.

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

98
113 Tarazi FI, Florijn WJ, Creese I. Regulation of ionotropic glutamate 132 Lawler CP, Prioleau C, Lewis MM, Mak C, Jiang D, Schetz JA et al.
receptors following subchronic and chronic treatment with Interactions of the novel antipsychotic aripiprazole (OPC-14597)
typical and atypical antipsychotics. Psychopharmacology 1996; with dopamine and serotonin receptor subtypes. Neuropsycho-
128: 371–379. pharmacology 1999; 20: 612–627.
114 Tascedda F, Lovati E, Blom JM, Muzzioli P, Brunello N, Racagni G 133 Semba J, Watanabe A, Kito S, Toru M. Behavioural and
et al. Regulation of ionotropic glutamate receptors in the rat brain neurochemical effects of OPC-14597, a novel antipsychotic drug,
in response to the atypical antipsychotic seroquel (quetiapine on dopaminergic mechanisms in rat brain. Neuropharmacology
fumarate). Neuropsychopharmacology 1999; 21: 211–217. 1995; 34: 785–791.
115 Tarazi FI, Baldessarini RJ, Kula NS, Zhang K. Long-term effects of 134 Burris KD, Molski TF, Xu C, Ryan E, Tottori K, Kikuchi T et al.
olanzapine, risperidone, and quetiapine on ionotropic glutamate Aripiprazole, a novel antipsychotic, is a high-affinity partial
receptor types: implications for antipsychotic drug treatment. agonist at human dopamine D2 receptors. J Pharmacol Exp Ther
J Pharmacol Exp Ther 2003; 306: 1145–1151. 2002; 302: 381–389.
116 Meador-Woodruff JH, King RE, Damask SP, Bovenkerk KA. 135 Carlsson A, Waters N, Waters S, Carlsson ML. Network interac-
Differential regulation of hippocampal AMPA and kainate tions in schizophrenia—therapeutic implications. Brain Res
receptor subunit expression by haloperidol and clozapine. Mol Brain Res Rev 2000; 31: 342–349.
Psychiatry 1996; 1: 41–53. 136 Jordan S, Koprivica V, Chen R, Tottori K, Kikuchi T, Altar CA.
117 Eastwood SL, Porter RH, Harrison PJ. The effect of chronic The antipsychotic aripiprazole is a potent, partial agonist at the
haloperidol treatment on glutamate receptor subunit (GluR1, human 5-HT1A receptor. Eur J Pharmacol 2002; 441: 137–140.
GluR2, KA1, KA2, NR1) mRNAs and glutamate binding protein 137 Shapiro DA, Renock S, Arrington E, Chiodo LA, Liu LX, Sibley
mRNA in rat forebrain. Neurosci Lett 1996; 212: 163–166. DR et al. Aripiprazole, a novel atypical antipsychotic drug with a
118 Fitzgerald LW, Deutch AY, Gasic G, Heinemann SF, Nestler EJ. unique and robust pharmacology. Neuropsychopharmacology
Regulation of cortical and subcortical glutamate receptor 2003; 28: 1400–1411.
subunit expression by antipsychotic drugs. J Neurosci 1995; 15: 138 Bowles TM, Levin GM. Aripiprazole: a new atypical antipsycho-
2453–2461. tic drug. Ann Pharmacother 2003; 37: 687–694.
119 Riva MA, Tascedda F, Lovati E, Racagni G. Regulation of NMDA 139 Yokoi F, Grunder G, Biziere K, Stephane M, Dogan AS, Dannals
receptor subunit messenger RNA levels in the rat brain following RF et al. Dopamine D2 and D3 receptor occupancy in normal
acute and chronic exposure to antipsychotic drugs. Brain Res Mol humans treated with the antipsychotic drug aripiprazole (OPC
Brain Res 1997; 50: 136–142. 14597): a study using positron emission tomography and
120 Healy DJ, Meador-Woodruff JH. Clozapine and haloperidol raclopride. Neuropsychopharmacology 2002; 27: 248–259.
differentially affect AMPA and kainate receptor subunit mRNA 140 Davis JM, Schaffer CB, Killian GA, Kinard C, Chan C. Important
levels in rat cortex and striatum. Brain Res Mol Brain Res 1997; issues in the drug treatment of schizophrenia. Schizophr Bull
47: 331–338. 1980; 6: 70–87.
121 Tascedda F, Blom JM, Brunello N, Zolin K, Gennarelli M, Colzi A 141 American Psychiatric Association. Practice Guideline for the
et al. Modulation of glutamate receptors in response to the Treatment of Patients with Schizophrenia. Practice Guidelines
novel antipsychotic olanzapine in rats. Biol Psychiatry 2001; 50: for the Treatment of Psychiatric Disorders. American Psychiatric
117–122. Association, Washington, D.C, 2000, pp 299–412.
122 Ossowska K, Pietraszek M, Wardas J, Dziedzicka-Wasylewska M, 142 Fleischhacker WW. New drugs for the treatment of schizophrenic
Nowicka D, Wolfarth S. Chronic treatments with haloperidol and patients. Acta Psychiatr Scand 1995; 388(Suppl): 24–30.
clozapine alter the level of NMDA-R1 mRNA in the rat brain: an 143 Spohn HE, Strauss ME. Relation of neuroleptic and antic-
in situ hybridization study. Pol J Pharmacol 2002; 54: 1–9. holinergic medication to cognitive functions in schizophrenia.
123 Schmitt A, Zink M, Muller B, May B, Herb A, Jatzko A et al. J Abnorm Psychol 1989; 98: 367–380.
Effects of long-term antipsychotic treatment on NMDA receptor 144 Meltzer HY, Thompson PA, Lee MA, Ranjan R. Neuropsychologic
binding and gene expression of subunits. Neurochem Res 2003; deficits in schizophrenia: relation to social function and effect of
28: 235–241. antipsychotic drug treatment. Neuropsychopharmacology 1996;
124 Kontkanen O, Toronen P, Lakso M, Wong G, Castren E. 14: 27S–33S.
Antipsychotic drug treatment induces differential gene expres- 145 Collaborative Working Group on Clinical Trial Evaluations.
sion in the rat cortex. J Neurochem 2002; 83: 1043–1053. Evaluating the effects of antipsychotics on cognition in schizo-
125 Kim T, Tao-Cheng JH, Eiden LE, Loh YP. Chromogranin A, an ‘on/ phrenia. Collaborative Working Group on Clinical Trial Evalua-
off’ switch controlling dense-core secretory granule biogenesis. tions. J Clin Psychiatry 1998; 59(Suppl 12): 35–40.
Cell 2001; 106: 499–509. 146 Tollefson GD. Cognitive function in schizophrenic patients. J Clin
126 Sudhof TC, Rizo J. Synaptotagmins: C2-domain proteins that Psychiatry 1996; 57(Suppl 11): 31–39.
regulate membrane traffic. Neuron 1996; 17: 379–388. 147 Ayuso-Gutierrez JL, del RV. Factors influencing relapse in
127 Bultynck G, Vermassen E, Szlufcik K, De Smet P, Fissore RA, the long-term course of schizophrenia. Schizophr Res 1997; 28:
Callewaert G et al. Calcineurin and intracellular Ca2 þ -release 199–206.
channels: regulation or association? Biochem Biophys Res 148 Kane JM. Pharmacologic treatment of schizophrenia. Biol
Commun 2003; 311: 1181–1193. Psychiatry 1999; 46: 1396–1408.
128 Braunewell KH, Gundelfinger ED. Intracellular neuronal calcium 149 Meltzer HY. Long-term effects of neuroleptic drugs on the
sensor proteins: a family of EF-hand calcium-binding proteins in neuroendocrine system. Adv Biochem Psychopharmacol 1985;
search of a function. Cell Tissue Res 1999; 295: 1–12. 40: 59–68.
129 Mirnics K, Middleton FA, Marquez A, Lewis DA, Levitt P. 150 Gaebel W. Towards the improvement of compliance: the
Molecular characterization of schizophrenia viewed by micro- significance of psycho-education and new antipsychotic drugs.
array analysis of gene expression in prefrontal cortex. Neuron Int Clin Psychopharmacol 1997; 12(Suppl 1): S37–S42.
2000; 28: 53–67. 151 Leucht S, Pitschel-Walz G, Engel RR, Kissling W. Amisulpride, an
130 Bauer R, Mayr A, Lederer W, Needham PL, Kilpatrick IC, unusual ‘atypical’ antipsychotic: a meta-analysis of randomized
Fleischhacker WW et al. Further evidence that behavioral tests controlled trials. Am J Psychiatry 2002; 159: 180–190.
and neuropeptide mRNA and tissue level alterations can 152 Emsley R, Oosthuizen P. The new and evolving pharmacotherapy
differentiate between typical and atypical antipsychotic drugs. of schizophrenia. Psychiatr Clin North Am 2003; 26: 141–163.
Neuropsychopharmacology 2000; 23: 46–55. 153 Markowitz JS, Brown CS, Moore TR. Atypical antipsychotics Part
131 Kikuchi T, Tottori K, Uwahodo Y, Hirose T, Miwa T, Oshiro Y I: pharmacology, pharmacokinetics, and efficacy. Ann Pharmac-
et al. 7-(4-butyloxy)-3,4-dihydro-2(1H)-quino linone (OPC- other 1999; 33: 73–85.
14597), a new putative antipsychotic drug with both presynaptic 154 Remington G, Kapur S. Atypical antipsychotics:are some more
dopamine autoreceptor agonistic activity and postsynaptic D2 atypical than others? Psychopharmacology 2000; 148: 3–15.
receptor antagonistic activity. J Pharmacol Exp Ther 1995; 274: 155 Geddes J, Freemantle N, Harrison P, Bebbington P. Atypical
329–336. antipsychotics in the treatment of schizophrenia: systematic

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

99
overview and meta-regression analysis. BMJ 2000; 321: 175 Velligan DI, Miller AL. Cognitive dysfunction in schizo
1371–1376. phrenia and its importance to outcome: the place of atypical
156 Leucht S, Pitschel-Walz G, Abraham D, Kissling W. Efficacy and antipsychotics in treatment. J Clin Psychiatry 1999; 60(Suppl 23):
extrapyramidal side-effects of the new antipsychotics olanza- 25–28.
pine, quetiapine, risperidone, and sertindole compared to 176 Green MF, Braff DL. Translating the basic and clinical cognitive
conventional antipsychotics and placebo. A meta-analysis of neuroscience of schizophrenia to drug development and clinical
randomized controlled trials. Schizophr Res 1999; 35: 51–68. trials of antipsychotic medications. Biol Psychiatry 2001; 49:
157 Davis JM, Chen N, Glick ID. A meta-analysis of the efficacy of 374–384.
second-generation antipsychotics. Arch Gen Psychiatry 2003; 60: 177 Green MF, Marshall BDJ, Wirshing WC, Ames D, Marder SR,
553–564. McGurk S et al. Does risperidone improve verbal working
158 Stroup TS, McEvoy JP, Swartz MS, Byerly MJ, Glick ID, Canive memory in treatment-resistant schizophrenia? Am J Psychiatry
JM et al. The National Institute of Mental Health Clinical 1997; 154: 799–804.
Antipsychotic Trials of Intervention Effectiveness (CATIE) pro- 178 Purdon SE, Jones BD, Stip E, Labelle A, Addington D, David SR et
ject: schizophrenia trial design and protocol development. al. Neuropsychological change in early phase schizophrenia
Schizophr Bull 2003; 29: 15–31. during 12 months of treatment with olanzapine, risperidone, or
159 Kane JM, Gunduz H, Malhortra AK. Second generation anti- haloperidol. The Canadian Collaborative Group for research in
psychotics in the treatment of schizophrenia: clozapine. In: schizophrenia.. Arch Gen Psychiatry 2000; 57: 249–258.
Breier A, Tran PV, Herrera JM, Tollefson GD, Bymaster FP (eds). 179 Weiss E, Kemmler G, Fleischhacker WW. Improvement of
Current Issues in the Psychopharmacology of Schizophrenia. cognitive dysfunction after treatment with second-generation
Lippincott Williams & Wilkins Healthcare: Philadelphia, 2001, antipsychotics. Arch Gen Psychiatry 2002; 59: 572–573.
pp 209–223. 180 Harvey PD, Keefe RS. Studies of cognitive change in patients
160 Carpenter WTJ, Conley RR, Buchanan RW, Breier A, Tamminga with schizophrenia following novel antipsychotic treatment. Am
CA. Patient response and resource management: another view of J Psychiatry 2001; 158: 176–184.
clozapine treatment of schizophrenia. Am J Psychiatry 1995; 152: 181 Bilder RM, Goldman RS, Volavka J, Czobor P, Hoptman M,
827–832. Sheitman B et al. Neurocognitive effects of clozapine, olanza-
161 Conley R, Gounaris C, Tamminga C. Clozapine response varies in pine, risperidone, and haloperidol in patients with chronic
deficit versus non-deficit schizophrenic subjects. Biol Psychiatry schizophrenia or schizoaffective disorder. Am J Psychiatry 2002;
1994; 35: 746–747. 159: 1018–1028.
162 Meltzer HY. Clozapine: is another view valid? Am J Psychiatry 182 Green MF, Marder SR, Glynn SM, McGurk SR, Wirshing WC,
1995; 152: 821–825. Wirshing DA et al. The neurocognitive effects of low-dose
163 Buchanan RW, Gold JM. Negative symptoms: diagnosis, treat- haloperidol: a two-year comparison with risperidone. Biol
ment and prognosis. Int Clin Psychopharmacol 1996; 11(Suppl Psychiatry 2002; 51: 972–978.
2): 3–11. 183 Carpenter WT, Gold JM. Another view of therapy for cognition in
164 Collaborative Working Group on Clinical Trial Evaluations. schizophrenia. Biol Psychiatry 2002; 51: 969–971.
Assessing the effects of atypical antipsychotics on negative 184 Meltzer HY, Sumiyoshi T. Atypical antipsychotic drugs improve
symptoms. Collaborative Working Group on Clinical Trial cognition in schizophrenia. Biol Psychiatry 2003; 53: 265–267.
Evaluations. J Clin Psychiatry 1998; 59(Suppl 12): 28–34. 185 Kane J, Honigfeld G, Singer J, Meltzer H. Clozapine for the
165 Lehman AF, Lieberman JA, Dixon LB, McGlashan TH, Miller AL, treatment-resistant schizophrenic. A double-blind comparison
Perkins DO et al. Practice guideline for the treatment of patients with chlorpromazine. Arch Gen Psychiatry 1988; 45: 789–796.
with schizophrenia, second edition. Am J Psychiatry 2004; 161: 186 Fleischhacker WW. Clozapine: a comparison with other novel
1–56. antipsychotics. J Clin Psychiatry 1999; 60: 30–34.
166 Marder SR, Davis JM, Chouinard G. The effects of risperidone on 187 Conley RR, Kelly DL. Management of treatment resistance in
the five dimensions of schizophrenia derived by factor analysis: schizophrenia. Biol Psychiatry 2001; 50: 898–911.
combined results of the North American trials. J Clin Psychiatry 188 Chakos M, Lieberman J, Hoffman E, Bradford D, Sheitman B.
1997; 58: 538–546. Effectiveness of second-generation antipsychotics in patients
167 Goff DC, Evins AE. Negative symptoms in schizophrenia: with treatment-resistant schizophrenia: a review and
neurobiological models and treatment response. Harv Rev meta-analysis of randomized trials. Am J Psychiatry 2001; 158:
Psychiatry 1998; 6: 59–77. 518–526.
168 Moller HJ, Muller H, Borison RL, Schooler NR, Chouinard G. A 189 Patel JK, Pinals DA, Breier A. Schizophrenia and other
path-analytical approach to differentiate between direct psychoses. In: Tasman A, Kay J, Lieberman JA (eds). PSYCHIA-
and indirect drug effects on negative symptoms in schizo- TRY, 2nd edn. John Wiley & Sons, Ltd: Chichester, 2003,
phrenic patients. A re-evaluation of the North American pp 1131–1206.
risperidone study. Eur Arch Psychiatry Clin Neurosci 1995; 190 Small JG, Hirsch SR, Arvanitis LA, Miller BG, Link CG.
245: 45–49. Quetiapine in patients with schizophrenia. A high- and low-
169 Tollefson GD, Sanger TM. Negative symptoms: a path analytic dose double-blind comparison with placebo. Seroquel Study
approach to a double-blind, placebo- and haloperidol-controlled Group. Arch Gen Psychiatry 1997; 54: 549–557.
clinical trial with olanzapine. Am J Psychiatry 1997; 154: 191 Dev V, Raniwalla J. Quetiapine: a review of its safety in the
466–474. management of schizophrenia. Drug Saf 2000; 23: 295–307.
170 Moller HJ. Neuroleptic treatment of negative symptoms in 192 Chouinard G. Effects of risperidone in tardive dyskinesia: an
schizophrenic patients. Efficacy problems and methodological analysis of the Canadian multicenter risperidone study. J Clin
difficulties. Eur Neuropsychopharmacol 1993; 3: 1–11. Psychopharmacol 1995; 15: 36S–44S.
171 Keefe RSE, Silva SG, Perkins DO, Lieberman JA. The effects of 193 Csernansky J, Okamoto A. Risperidone vs haloperidol for
atypical antipsychotic drugs on neurocognitive impairment in prevention of relapse in schizophrenia and schizoaffective
schizophrenia: a review and meta-analysis. Schizophr Bull 1999; disorders: a long-term double-blind comparison. The 10th
25: 201–222. Biennial Winter Workshop on Schizophrenia. Davos, Switzer-
172 Meltzer HY, McGurk SR. The effects of clozapine, risperidone, land, 2000.
and olanzapine on cognitive function in schizophrenia. Schi- 194 Tran PV, Dellva MA, Tollefson GD, Beasley CMJ, Potvin JH,
zophr Bull 1999; 25: 233–255. Kiesler GM. Extrapyramidal symptoms and tolerability of
173 Worrel JA, Marken PA, Beckman SE, Ruehter VL. Atypical olanzapine versus haloperidol in the acute treatment of schizo-
antipsychotic agents: a critical review. Am J Health Syst Pharm phrenia. J Clin Psychiatry 1997; 58: 205–211.
2000; 57: 238–255. 195 Tollefson GD, Beasley CMJ, Tamura RN, Tran PV, Potvin JH.
174 Mortimer AM. Cognitive function in schizophrenia—do neuro- Blind, controlled, long-term study of the comparative incidence
leptics make a difference? Pharmacol Biochem Behav 1997; 56: of treatment-emergent tardive dyskinesia with olanzapine or
789–795. haloperidol. Am J Psychiatry 1997; 154: 1248–1254.

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

100
196 Ferris P. Ziprasidone. Curr Opin CPNS Invest Drugs 2000; 2: 217 Williams GV, Goldman-Rakic PS. Modulation of memory fields
58–70. by dopamine D1 receptors in prefrontal cortex. Nature 1995; 376:
197 Allison DB, Mentore JL, Heo M, Chandler LP, Cappelleri JC, 572–575.
Infante MC et al. Antipsychotic-induced weight gain: a compre- 218 Ghosh D, Snyder SE, Watts VJ, Mailman RB, Nichols DE. 9-
hensive research synthesis. Am J Psychiatry 1999; 156: 1686– Dihydroxy-2,3,7,11b-tetrahydro-1H-naphisoquinoline: a potent
1696. full dopamine D1 agonist containing a rigid-beta-phenyldopa-
198 Taylor DM, McAskill R. Atypical antipsychotics and mine pharmacophore. J Med Chem 1996; 39: 549–555.
weight gain—a systematic review. Acta Psychiatr Scand 2000; 219 Castner SA, Williams GV, Goldman-Rakic PS. Reversal
101: 416–432. of antipsychotic-induced working memory deficits by short-
199 Sussman N. Review of atypical antipsychotics and weight gain. term dopamine D1 receptor stimulation. Science 2000; 287:
J Clin Psychiatry 2001; 62(Suppl 23): 5–12. 2020–2022.
200 Kane JM, Carson WH, Saha AR, McQuade RD, Ingenito GG, 220 Nichols DE, Mailman RB. Substituted hexa-hydro[a]phenanthri-
Zimbroff DL et al. Efficacy and safety of aripiprazole and dines. US Patent 1995; 5: 134.
haloperidol versus placebo in patients with schizophrenia and 221 Nichols DE, Mailman RB. Fused isoquinolines as dopamine
schizoaffective disorder. J Clin Psychiatry 2002; 63: 763–771. receptor ligands. US Patent 1999; 5: 110.
201 Potkin SG, Saha AR, Kujawa MJ, Carson WH, Ali M, Stock E et al. 222 Van Tol HH, Bunzow JR, Guan HC, Sunahara RK, Seeman P,
Aripiprazole, an antipsychotic with a novel mechanism of action, Niznik HB et al. Cloning of the gene for a human dopamine D4
and risperidone vs placebo in patients with schizophrenia and receptor with high affinity for the antipsychotic clozapine.
schizoaffective disorder. Arch Gen Psychiatry 2003; 60: 681–690. Nature 1991; 350: 610–614.
202 Pigott TA, Carson WH, Saha AR, Torbeyns AF, Stock EG, Ingenito 223 Seeman P, Guan HC, Van Tol HH. Dopamine D4 receptors
GG. Aripiprazole for the prevention of relapse in stabilized elevated in schizophrenia. Nature 1993; 365: 441–445.
patients with chronic schizophrenia: a placebo-controlled 26- 224 Lahti RA, Roberts RC, Cochrane EV, Primus RJ, Gallager DW,
week study. J Clin Psychiatry 2003; 64: 1048–1056. Conley RR et al. Direct determination of dopamine D4 receptors
203 Kasper S, Lerman MN, McQuade RD, Saha A, Carson WH, Ali M in normal and schizophrenic postmortem brain tissue: a NGD-94-
et al. Efficacy and safety of aripiprazole vs. haloperidol for long- 1 study. Mol Psychiatry 1998; 3: 528–533.
term maintenance treatment following acute relapse of schizo- 225 Mansbach RS, Brooks EW, Sanner MA, Zorn SH. Selective
phrenia. Int J Neuropsychopharmacol 2003; 6: 325–337. dopamine D4 receptor antagonists reverse apomorphine-induced
204 Marder SR, McQuade RD, Stock E, Kaplita S, Marcus R, blockade of prepulse inhibition. Psychopharmacology 1998; 135:
Safferman AZ et al. Aripiprazole in the treatment of schizo- 194–200.
phrenia: safety and tolerability in short-term, placebo-controlled 226 Feldpausch DL, Needham LM, Stone MP, Althaus JS, Yamamoto
trials. Schizophr Res 2003; 61: 123–136. BK, Svensson KA et al. The role of dopamine D4 receptor in the
205 Crismon ML, DeLeon A, Miller AL. Aripiprazole: does partial induction of behavioral sensitization to amphetamine and
dopaminergic agonism translate into clinical benefits? Ann accompanying biochemical and molecular adaptations. J Phar-
Pharmacother 2003; 37: 738–740. macol Exp Ther 1998; 286: 497–508.
206 Sedvall GC, Karlsson P. Pharmacological manipulation of D1- 227 Merchant KM, Gill GS, Harris DW, Huff RM, Eaton MJ, Looking-
dopamine receptor function in schizophrenia. Neuropsychophar- land K et al. Pharmacological characterization of U-101387, a
macology 1999; 22: S181–S188. dopamine D4 receptor selective antagonist. J Pharmacol Exp Ther
207 Goldman-Rakic PS. The relevance of the dopamine-D1 receptor in 1996; 279: 1392–1403.
the cognitive symptoms of schizophrenia. Neuropsychopharma- 228 Danysz W. Sonepiprazole. Curr Opin CPNS Invest Drugs 2000; 2:
cology 1999; 21: S170–S180. 97–104.
208 Waddington JL. Pre- and postsynaptic D1 to D5 dopamine receptor 229 Kramer MS, Last B, Getson A, Reines SA. The effects of a
mechanisms in relation to antipsychotic activity. In: Barnes TRE selective D4 dopamine receptor antagonist (L-745,870) in acutely
(ed). Antipsychotic Drugs and Their Side Effects. Academic psychotic inpatients with schizophrenia. D4 Dopamine Antago-
Press, London, 1993, pp 65–85. nist Group. Arch Gen Psychiatry 1997; 54: 567–572.
209 Karlsson P, Smith L, Farde L, Harnryd C, Sedvall G, Wiesel FA. 230 Bristow LJ, Kramer MS, Kulagowski J, Patel S, Ragan CI, Seabrook
Lack of apparent antipsychotic effect of the D1-dopamine GR. Schizophrenia and L-745,870, a novel dopamine D4 receptor
receptor antagonist SCH39166 in acutely ill schizophrenic antagonist. Trends Pharmacol Sci 1997; 18: 186–188.
patients. Psychopharmacology 1995; 121: 309–316. 231 Mansbach RS, Brooks EW, Sanner MA, Zorn SH. Selective
210 Den Boer JA, van Megen HJ, Fleischhacker WW, Louwerens JW, dopamine D4 receptor antagonists reverse apomorphine-induced
Slaap BR, Westenberg HG et al. Differential effects of the D1-DA blockade of prepulse inhibition. Psychopharmacology 1998; 135:
receptor antagonist SCH39166 on positive and negative 194–200.
symptoms of schizophrenia. Psychopharmacology 1995; 121: 232 Truffinet P, Tamminga CA, Fabre LF, Meltzer HY, Riviere ME,
317–322. Papillon-Downey C. Placebo-controlled study of the D4/5-HT2A
211 Karle J, Clemmesen L, Hansen L, Andersen M, Andersen J, antagonist fananserin in the treatment of schizophrenia. Am J
Fensbo C et al. NNC 01-0687, a selective dopamine D1 receptor Psychiatry 1999; 156: 419–425.
antagonist, in the treatment of schizophrenia. Psychopharmacol- 233 Rowley M, Bristow LJ, Hutson PH. Current and novel approaches
ogy 1995; 121: 328–329. to the drug treatment of schizophrenia. J Med Chem 2001; 44:
212 Arnsten AF, Cai JX, Murphy BL, Goldman-Rakic PS. Dopamine 477–501.
D1 receptor mechanisms in the cognitive performance of 234 Schwartz JC, Diaz J, Pilon C, Sokoloff P. Possible implications of
young adult and aged monkeys. Psychopharmacology 1994; the dopamine D(3) receptor in schizophrenia and in antipsycho-
116: 143–151. tic drug actions. Brain Res Brain Res Rev 2000; 31: 277–287.
213 Cai JX, Arnsten AF. Dose-dependent effects of the dopamine D1 235 Gurevich EV, Bordelon Y, Shapiro RM, Arnold SE, Gur RE, Joyce
receptor agonists A77636 or SKF81297 on spatial working JN. Mesolimbic dopamine D3 receptors and use of antipsychotics
memory in aged monkeys. J Pharmacol Exp Ther 1997; 283: in patients with schizophrenia. A postmortem study. Arch Gen
183–189. Psychiatry 1997; 54: 225–232.
214 Schneider JS, Sun ZQ, Roeltgen DP. Effects of dihydrexidine, a 236 Witkin J, Gasior M, Acri J, Beekman M, Thurkauf A, Yuan J
full dopamine D-1 receptor agonist, on delayed response et al. Atypical antipsychotic-like effects of the dopamine D3
performance in chronic low dose MPTP-treated monkeys. Brain receptor agonist, ( þ )-PD 128,907. Eur J Pharmacol 1998; 347:
Res 1994; 663: 140–144. R1–R3.
215 Okubo Y, Suhara T, Suzuki K, Kobayashi K, Inoue O, Terasaki O 237 Hackling AE, Stark H. Dopamine D3 receptor ligands with
et al. Decreased prefrontal dopamine D1 receptors in schizo- antagonist properties. Chembiochem 2002; 3: 946–961.
phrenia revealed by PET. Nature 1997; 385: 634–636. 238 Millan MJ, Dekeyne A, Rivet JM, Dubuffet T, Lavielle G, Brocco
216 Goldman-Rakic PS, Muly III EC, Williams GV. D1 receptors in M. S33084, a novel, potent, selective, and competitive antagonist
prefrontal cells and circuits. Brain Res Rev 2000; 31: 295–301. at dopamine D(3)-receptors: II. Functional and behavioral profile

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

101
compared with GR218,231 and L741,626. J Pharmacol Exp Ther 260 Schmitt A, Zink M, Petroianu G, May B, Braus DF, Henn FA.
2000; 293: 1063–1073. Decreased gene expression of glial and neuronal glutamate
239 Lacroix LP, Hows ME, Shah AJ, Hagan JJ, Heidbreder CA. transporters after chronic antipsychotic treatment in rat brain.
Selective antagonism at dopamine D3 receptors enhances mono- Neurosci Lett 2003; 347: 81–84.
aminergic and cholinergic neurotransmission in the rat anterior 261 Moghaddam B, Adams B, Verman A, Daly D. Activation of
cingulate cortex. Neuropsychopharmacology 2003; 28: 839–849. glutamatergic neurotransmission by ketamine—a novel step in
240 Abi-Saab WM, D’Souza DC, Madonick SH, Krystal JH. Targeting the pathway from NMDA receptor blockade to dopaminergic and
the glutamate system. In: Breier A, Tran PV, Herrera JM, Tollefson cognitive disruptions associated with the prefrontal cortex.
GD, Bymaster FP (eds). Current Issues in the Psychopharmacol- J Neurosci 1997; 17: 2921–2927.
ogy of Schizophrenia. Lippincott Williams & Wilkins Healthcare, 262 Chavez-Noriega LE, Schaffhauser H, Campbell UC. Metabotropic
Philadelphia, 2001, pp 304–332. glutamate receptors: potential drug targets for the treatment
241 Goff DC, Coyle JT. The emerging role of glutamate in the of schizophrenia. Curr Drug Target CNS Neurol Disord 2002; 1:
pathophysiology and treatment of schizophrenia. Am J Psychia- 261–281.
try 2001; 158: 1367–1377. 263 Moghaddam B, Adams BW. Reversal of phencyclidine effects by
242 Leeson PD, Iversen LL. The glycine site on the NMDA receptor: a group II metabotrophic glutamate receptor agonist in rats.
structure–activity relationships and therapeutic potential. J Med Science 1998; 281: 1349–1352.
Chem 1994; 37: 4053–4067. 264 Bubser M, Keseberg U, Notz PK, Schmidt WJ. Differential
243 Goff DC, Tsai G, Levitt J, Amico E, Manoach D, Schoenfeld DA behavioral and neurochemical effects of competitive and non-
et al. A placebo-controlled trial of D-cycloserine added to competitive NMDA receptor antagonists in rats. Eur J Pharmacol
conventional neuroleptics in patients with schizophrenia. Arch 1992; 229: 75–82.
Gen Psychiatry 1999; 56: 21–27. 265 Hauber W, Andersen R. The non-NMDA glutamate receptor
244 Goff DC, Tsai G, Manoach DS, Coyle JT. Dose-finding trial of D- antagonist GYKI 52466 counteracts locomotor stimulation and
cycloserine added to neuroleptics for negative symptoms in anticataleptic activity induced by the NMDA antagonist dizocilpine.
schizophrenia. Am J Psychiatry 1995; 152: 1213–1215. Naunyn Schmiedeberg’s Arch Pharmacol 1993; 348: 486–490.
245 Heresco-Levy U, Javitt DC, Ermilov M, Mordel C, Horowitz A, 266 Willins DL, Narayanan S, Wallace LJ, Uretsky NJ. The role of
Kelly D. Double-blind, placebo-controlled, crossover trial of dopamine and AMPA/kainate receptors in the nucleus accum-
glycine adjuvant therapy for treatment-resistant schizophrenia. bens in the hypermotility response to MK801. Pharmacol
Br J Psychiatry 1996; 169: 610–617. Biochem Behav 1993; 46: 881–887.
246 Javitt DC, Zylberman I, Zukin SR, Heresco-Levy U, Lindenmayer 267 Sharp JW, Petersen DL, Langford MT. DNQX inhibits phencycli-
JP. Amelioration of negative symptoms in schizophrenia by dine (PCP) and ketamine induction of the hsp 70 heat shock gene
glycine. Am J Psychiatry 1994; 151: 1234–1236. in the rat cingulate and retrosplenial cortex. Brain Res 1995; 687:
247 Heresco-Levy U, Javitt DC, Ermilov M, Mordel C, Silipo G, 114–124.
Lichtenstein M. Efficacy of high-dose glycine in the treatment of 268 Svensson TH, Mathe JM. Atypical antipsychotic-like effect of
enduring negative symptoms of schizophrenia. Arch Gen AMPA receptor antagonists in the rat. Amino Acids 2000; 19:
Psychiatry 1999; 56: 29–36. 221–226.
248 Goff DC, Tsai G, Manoach DS, Flood J, Darby DG, Coyle JT. D- 269 Hampson RE, Rogers G, Lynch G, Deadwyler SA. Facilitative
cycloserine added to clozapine for patients with schizophrenia. effects of the ampakine CX516 on short-term memory in rats:
Am J Psychiatry 1996; 153: 1628–1630. correlations with hippocampal neuronal activity. J Neurosci
249 Goff DC, Henderson DC, Evins AE, Amico E. A placebo- 1998; 18: 2748–2763.
controlled crossover trial of D-cycloserine added to clozapine 270 Hampson RE, Rogers G, Lynch G, Deadwyler SA. Facilitative
in patients with schizophrenia. Biol Psychiatry 1999; 45: effects of the ampakine CX516 on short-term memory in rats:
512–514. enhancement of delayed-nonmatch-to-sample performance.
250 Hashimoto A, Oka T. Free D-aspartate and D-serine in J Neurosci 1998; 18: 2740–2747.
the mammalian brain and periphery. Prog Neurobiol 1997; 52: 271 Johnson SA, Luu NT, Herbst TA, Knapp R, Lutz D, Arai A et al.
325–353. Synergistic interactions between ampakines and antipsychotic
251 Tsai G, Yang P, Chung LC, Lange N, Coyle JT. D-serine added to drugs. J Pharmacol Exp Ther 1999; 289: 392–397.
antipsychotics for the treatment of schizophrenia. Biol Psychiatry 272 Goff D, Berman I, Posever T, Leahy L, Lynch G. A preliminary
1998; 44: 1081–1089. dose-escalation trial of CX 516 (ampakine) added to clozapine in
252 Bergeron R, Meyer TM, Coyle JT, Greene RW. Modulation of schizophrenia. Schizophr Res 1999; 36: 280.
N-methyl-D-aspartate receptor function by glycine transport. Proc 273 Marenco S, Egan MF, Goldberg TE, Knable MB, McClure RK,
Natl Acad Sci USA 1998; 95: 15730–15734. Winterer G et al. Preliminary experience with an ampakine
253 Berger AJ, Dieudonne S, Ascher P. Glycine uptake governs (CX516) as a single agent for the treatment of schizophrenia: a
glycine site occupancy at NMDA receptors of excitatory case series. Schizophr Res 2002; 57: 221–226.
synapses. J Neurophysiol 1998; 80: 3336–3340. 274 Do KQ, Trabesinger AH, Kirsten-Kruger M, Lauer CJ, Dydak U,
254 Javitt DC, Sershen H, Hashim A, Lajtha A. Reversal of Hell D et al. Schizophrenia: glutathione deficit in cerebrospinal
phencyclidine-induced hyperactivity by glycine and the glycine fluid and prefrontal cortex in vivo. Eur J Neurosci 2000; 12:
uptake inhibitor glycyldodecylamide. Neuropsychopharmacol- 3721–3728.
ogy 1997; 17: 202–204. 275 Grima G, Benz B, Parpura V, Cuenod M, Do KQ. Dopamine-
255 Javitt DC, Frusciante M. Glycyldodecylamide, a phencyclidine induced oxidative stress in neurons with glutathione deficit:
behavioral antagonist, blocks cortical glycine uptake: implica- implication for schizophrenia. Schizophr Res 2003; 62: 213–224.
tions for schizophrenia and substance abuse. Psychopharmacol- 276 Janaky R, Ogita K, Pasqualotto BA, Bains JS, Oja SS, Yoneda Y
ogy 1997; 129: 96–98. et al. Glutathione and signal transduction in the mammalian
256 Danbolt NC. Glutamate uptake. Prog Neurobiol 2001; 65: 1–105. CNS. J Neurochem 1999; 73: 889–902.
257 Gadea A, Lopez-Colome AM. Glial transporters for glutamate, 277 Goldman-Rakic PS, Lidow MS, Gallager DW. Overlap of dopa-
glycine and GABA I. Glutamate transporters. J Neurosci Res 2001; minergic, adrenergic, and serotoninergic receptors and comple-
63: 453–460. mentarity of their subtypes in primate prefrontal cortex.
258 Smith RE, Haroutunian V, Davis KL, Meador-Woodruff JH. J Neurosci 1990; 10: 2125–2138.
Expression of excitatory amino acid transporter transcripts in 278 Arnsten AF, Steere JC, Hunt RD. The contribution of alpha 2-
the thalamus of subjects with schizophrenia. Am J Psychiatry noradrenergic mechanisms of prefrontal cortical cognitive func-
2001; 158: 1393–1399. tion. Potential significance for attention-deficit hyperactivity
259 McCullumsmith RE, Meador-Woodruff JH. Striatal excitatory disorder. Arch Gen Psychiatry 1996; 53: 448–455.
amino acid transporter transcript expression in schizophrenia, 279 Friedman JI, Temporini H, Davis KL. Pharmacologic strategies for
bipolar disorder, and major depressive disorder. Neuropsycho- augmenting cognitive performance in schizophrenia. Biol Psy-
pharmacology 2002; 26: 368–375. chiatry 1999; 45: 1–16.

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

102
280 Friedman JI, Adler DN, Davis KL. The role of norepinephrine in 301 Adler LE, Olincy A, Waldo M, Harris JG, Griffith J, Stevens K et
the pathophysiology of cognitive disorders: potential applica- al. Schizophrenia, sensory gating, and nicotinic receptors.
tions to the treatment of cognitive dysfunction in schizophrenia Schizophr Bull 1998; 24: 189–202.
and Alzheimer’s disease. Biol Psychiatry 1999; 46: 1243–1252. 302 Simosky JK, Stevens KE, Adler LE, Freedman R. Clozapine
281 Arnsten AF, Goldman-Rakic PS. Alpha 2-adrenergic mechanisms improves deficient inhibitory auditory processing in DBA/2
in prefrontal cortex associated with cognitive decline in aged mice, via a nicotinic cholinergic mechanism. Psychopharmacol-
nonhuman primates. Science 1985; 230: 1273–1276. ogy 2003; 165: 386–396.
282 Fields RB, Van Kammen DP, Peters JL, Rosen J, Van Kammen WB, 303 Simosky JK, Stevens KE, Kem WR, Freedman R. Intragastric
Nugent A et al. Clonidine improves memory function in DMXB-A, an alpha7 nicotinic agonist, improves deficient
schizophrenia independently from change in psychosis. Preli- sensory inhibition in DBA/2 mice. Biol Psychiatry 2001; 50:
minary findings. Schizophr Res 1988; 1: 417–423. 493–500.
283 Uhlen S, Muceniece R, Rangel N, Tiger G, Wikberg JE. 304 Schreiber R, Dalmus M, De Vry J. Effects of alpha 4/beta 2- and
Comparison of the binding activities of some drugs on alpha alpha 7-nicotine acetylcholine receptor agonists on prepulse
2A, alpha 2B and alpha 2C-adrenoceptors and non-adrenergic inhibition of the acoustic startle response in rats and mice.
imidazoline sites in the guinea pig. Pharmacol Toxicol 1995; 76: Psychopharmacology 2002; 159: 248–257.
353–364. 305 Flores CM, Rogers SW, Pabreza LA, Wolfe BB, Kellar KJ. A
284 Arnsten AF, Cai JX, Goldman-Rakic PS. The alpha-2 adrenergic subtype of nicotinic cholinergic receptor in rat brain is composed
agonist guanfacine improves memory in aged monkeys without of alpha 4 and beta 2 subunits and is up-regulated by chronic
sedative or hypotensive side effects: evidence for alpha-2 nicotine treatment. Mol Pharmacol 1992; 41: 31–37.
receptor subtypes. J Neurosci 1988; 8: 4287–4298. 306 Bontempi B, Whelan KT, Risbrough VB, Rao TS, Buccafusco JJ,
285 Friedman JI, Adler DN, Temporini HD, Kemether E, Harvey PD, Lloyd GK et al. SIB-1553A, ( þ /-)-4-[[2-(1-methyl-2-pyrrolidiny-
White L et al. Guanfacine treatment of cognitive impairment in l)ethyl]thio]phenol hydrochloride, a subtype-selective ligand for
schizophrenia. Neuropsychopharmacology 2001; 25: 402–409. nicotinic acetylcholine receptors with putative cognitive-enhan-
286 Millan MJ, Gobert A, Newman-Tancredi A, Lejeune F, Cussac D, cing properties: effects on working and reference memory
Rivet JM et al. S18327 (1-ethyl]3-phenyl imidazolin-2-one), a performances in aged rodents and nonhuman primates.
novel, potential antipsychotic displaying marked antagonist J Pharmacol Exp Ther 2001; 299: 297–306.
properties at alpha(1)- and alpha(2)-adrenergic receptors: I. 307 Lloyd GK, Menzaghi F, Bontempi B, Suto C, Siegel R, Akong M et
Receptorial, neurochemical, and electrophysiological profile. al. The potential of subtype-selective neuronal nicotinic acet-
J Pharmacol Exp Ther 2000; 292: 38–53. ylcholine receptor agonists as therapeutic agents. Life Sci 1998;
287 Gobert A, Rivet JM, Audinot V, Newman-Tancredi A, Cistarelli L, 62: 1601–1606.
Millan MJ. Simultaneous quantification of serotonin, dopamine 308 Maelicke A, Samochocki M, Jostock R, Fehrenbacher A, Ludwig
and noradrenaline levels in single frontal cortex dialysates of J, Albuquerque EX et al. Allosteric sensitization of nicotinic
freely-moving rats reveals a complex pattern of reciprocal auto- receptors by galantamine, a new treatment strategy for Alzhei-
and heteroreceptor-mediated control of release. Neuroscience mer’s disease. Biol Psychiatry 2001; 49: 279–288.
1998; 84: 413–429. 309 Albuquerque EX, Santos MD, Alkondon M, Pereira EF, Maelicke
288 Litman RE, Su TP, Potter WZ, Hong WW, Pickar D. Idazoxan and A. Modulation of nicotinic receptor activity in the central
response to typical neuroleptics in treatment-resistant schizo- nervous system: a novel approach to the treatment of Alzheimer
phrenia. Comparison with the atypical neuroleptic, clozapine. Br disease. Alzheimer Dis Assoc Disord 2001; 15(Suppl 1): S19–S25.
J Psychiatry 1996; 168: 571–579. 310 Maelicke A. Allosteric modulation of nicotinic receptors as a
289 Elman I, Goldstein DS, Eisenhofer G, Folio J, Malhotra AK, Adler treatment strategy for Alzheimer’s disease. Dement Geriatr Cogn
CM et al. Mechanism of peripheral noradrenergic stimulation by Disord 2000; 11(Suppl 1): 11–18.
clozapine. Neuropsychopharmacology 1999; 20: 29–34. 311 Coyle J, Kershaw P. Galantamine, a cholinesterase inhibitor that
290 Egan MF, Goldberg TE, Kolachana BS, Callicott JH, Mazzanti CM, allosterically modulates nicotinic receptors: effects on the course
Straub RE et al. Effect of COMT Val108/158 Met genotype on of Alzheimer’s disease. Biol Psychiatry 2001; 49: 289–299.
frontal lobe function and risk for schizophrenia. Proc Natl Acad 312 Allen TB, McEvoy JP. Galantamine for treatment-resistant
Sci USA 2001; 98: 6917–6922. schizophrenia. Am J Psychiatry 2002; 159: 1244–1245.
291 Weinberger DR, Egan MF, Bertolino A, Callicott JH, Mattay VS, 313 Rosse RB, Deutsch SI. Adjuvant galantamine administration
Lipska BK et al. Prefrontal neurons and the genetics of improves negative symptoms in a patient with treatment-
schizophrenia. Biol Psychiatry 2001; 50: 825–844. refractory schizophrenia. Clin Neuropharmacol 2002; 25:
292 Gogos JA, Morgan M, Luine V, Santha M, Ogawa S, Pfaff D et al. 272–275.
Catechol-O-methyltransferase-deficient mice exhibit sexually 314 MacEwan GW, Ehmann TS, Khanbhai I, Wrixon C. Donepezil in
dimorphic changes in catecholamine levels and behavior. Proc schizophrenia—is it helpful? An experimental design case study.
Natl Acad Sci USA 1998; 95: 9991–9996. Acta Psychiatr Scand 2001; 104: 469–472.
293 Goldberg TE, Egan MF, Gscheidle T, Coppola R, Weickert T, 315 Howard AK, Thornton AE, Altman S, Honer WG. Donepezil for
Kolachana BS et al. Executive subprocesses in working memory: memory dysfunction in schizophrenia. J Psychopharmacol 2002;
relationship to catechol-O-methyltransferase Val158Met geno- 16: 267–270.
type and schizophrenia. Arch Gen Psychiatry 2003; 60: 889–896. 316 Buchanan RW, Summerfelt A, Tek C, Gold J. An open-labeled
294 Liljequist R, Haapalinna A, Ahlander M, Li YH, Mannisto PT. trial of adjunctive donepezil for cognitive impairments in
Catechol O-methyltransferase inhibitor tolcapone has minor patients with schizophrenia. Schizophr Res 2003; 59: 29–33.
influence on performance in experimental memory models in 317 Friedman JI, Adler DN, Howanitz E, Harvey PD, Brenner G,
rats. Behav Brain Res 1997; 82: 195–202. Temporini H et al. A double blind placebo controlled trial
295 Gasparini M, Fabrizio E, Bonifati V, Meco G. Cognitive improve- of donepezil adjunctive treatment to risperidone for the
ment during Tolcapone treatment in Parkinson’s disease. J Neural cognitive impairment of schizophrenia. Biol Psychiatry 2002;
Transm 1997; 104: 887–894. 51: 349–357.
296 Holden C. Neuroscience. Deconstructing schizophrenia. Science 318 Bymaster FP. Possible role of muscarinic receptor agonists as
2003; 299: 333–335. therapeutic agents for psychosis. In: Breier A, Tran PV, Herrera
297 Watkins P. COMT inhibitors and liver toxicity. Neurology 2000; JM, Tollefson GD, Bymaster FP (eds). Current Issues in the
55: S51–S52. Psychopharmacology of Schizophrenia. Lippincott Williams &
298 Borges N. Tolcapone-related liver dysfunction: implications for Wilkins Healthcare, Philadelphia, 2001, pp 333–348.
use in Parkinson’s disease therapy. Drug Saf 2003; 26: 743–747. 319 Bymaster FP, Felder C, Ahmed S, McKinzie D. Muscarinic
299 Rezvani AH, Levin ED. Cognitive effects of nicotine. Biol receptors as a target for drugs treating schizophrenia. Curr Drug
Psychiatry 2001; 49: 258–267. Target CNS Neurol Disord 2002; 1: 163–181.
300 Simosky JK, Stevens KE, Freedman R. Nicotinic agonists and 320 Bodick NC, Offen WW, Levey AI, Cutler NR, Gauthier SG, Satlin
psychosis. Curr Drug Target CNS Neurol Disord 2002; 1: 149–162. A et al. Effects of xanomeline, a selective muscarinic receptor

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

103
agonist, on cognitive function and behavioral symptoms in 342 Perenyi A, Goswami U, Frecska E, Arato M, Bela A. L-deprenyl in
Alzheimer disease. Arch Neurol 1997; 54: 465–473. treating negative symptoms of schizophrenia. Psychiatry Res
321 Perry KW, Bymaster FP, Shannon HE, Rasmussen K, DeLapp NW, 1992; 42: 189–191.
Zhang W et al. The muscarinic agonist xanomeline has 343 Bodkin JA, Cohen BM, Salomon MS, Cannon SE, Zornberg GL,
antipsychotic-like activity in animals and in man. Schizophr Cole JO. Treatment of negative symptoms in schizophrenia and
Res 1999; 36: 117–118. schizoaffective disorder by selegiline augmentation of antipsy-
322 Shannon HE, Rasmussen K, Bymaster FP, Hart JC, Peters SC, chotic medication. A pilot study examining the role of dopamine.
Swedberg MD et al. Xanomeline, an M(1)/M(4) preferring J Nerv Ment Dis 1996; 184: 295–301.
muscarinic cholinergic receptor agonist, produces antipsycho- 344 Gupta S, Droney T, Kyser A, Keller P. Selegiline augmentation of
tic-like activity in rats and mice. Schizophr Res 2000; 42: antipsychotics for the treatment of negative symptoms in
249–259. schizophrenia. Compr Psychiatry 1999; 40: 148–150.
323 Chopra GS, Smith JW. Psychotic reactions following cannabis use 345 Jungerman T, Rabinowitz D, Klein E. Deprenyl augmentation for
in East Indians. Arch Gen Psychiatry 1974; 30: 24–27. treating negative symptoms of schizophrenia: a double-blind,
324 Andreasson S, Allebeck P, Engstrom A, Rydberg U. Cannabis and controlled study. J Clin Psychopharmacol 1999; 19: 522–525.
schizophrenia. A longitudinal study of Swedish conscripts. 346 Maruyama W, Akao Y, Carrillo MC, Kitani K, Youdium MB, Naoi
Lancet 1987; 2: 1483–1486. M. Neuroprotection by propargylamines in Parkinson’s disease:
325 Voruganti LN, Slomka P, Zabel P, Mattar A, Awad AG. Cannabis suppression of apoptosis and induction of prosurvival genes.
induced dopamine release: an in-vivo SPECT study. Psychiatry Neurotoxicol Teratol 2002; 24: 675–682.
Res 2001; 107: 173–177. 347 Naoi M, Maruyama W, Youdim MB, Yu P, Boulton AA. Anti-
326 Leweke FM, Giuffrida A, Wurster U, Emrich HM, Piomelli D. apoptotic function of propargylamine inhibitors of type-B
Elevated endogenous cannabinoids in schizophrenia. Neuro- monoamine oxidase. Inflammopharmacology 2003; 11: 175–181.
report 1999; 10: 1665–1669. 348 Seeger TF, Bartlett B, Coskran TM, Culp JS, James LC, Krull DL et
327 De Marchi N, De Petrocellis L, Orlando P, Daniele F, Fezza F, Di al. Immunohistochemical localization of PDE10A in the rat brain.
M, V. Endocannabinoid signalling in the blood of patients with Brain Res 2003; 985: 113–126.
schizophrenia. Lipids Health Dis 2003; 2: 5. 349 Schmidt CJ, Chapin DS, McCarthy SA, Fujiwara RA, Harms JF,
328 Emrich HM, Leweke FM, Schneider U. Towards a cannabinoid Shrikhande A et al. The neurochemical and behavioral effects of
hypothesis of schizophrenia: cognitive impairments due to papaverine in vivo suggest PDE10 inhibition is ‘antipsychotic’.
dysregulation of the endogenous cannabinoid system. Pharmacol Schizophr Res 2003; 60: 114.
Biochem Behav 1997; 56: 803–807. 350 Noda Y, Yamada K, Furukawa H, Nabeshima T. Involvement of
329 Poncelet M, Barnouin MC, Breliere JC, Le Fur G, Soubrie P. nitric oxide in phencyclidine-induced hyperlocomotion in mice.
Blockade of cannabinoid (CB1) receptors by 141716 selectively Eur J Pharmacol 1995; 286: 291–297.
antagonizes drug-induced reinstatement of exploratory beha- 351 Deutsch SI, Rosse RB, Paul SM, Tomasino V, Koetzner L, Morn
viour in gerbils. Psychopharmacology 1999; 144: 144–150. CB et al. 7-Nitroindazole and methylene blue, inhibitors of
330 Alonso R, Voutsinos B, Fournier M, Labie C, Steinberg R, neuronal nitric oxide synthase and NO-stimulated guanylate
Souilhac J et al. Blockade of cannabinoid receptors by cyclase, block MK-801-elicited behaviors in mice. Neuropsycho-
SR141716 selectively increases Fos expression in rat mesocorti- pharmacology 1996; 15: 37–43.
colimbic areas via reduced dopamine D2 function. Neuroscience 352 Johansson C, Jackson DM, Svensson L. Nitric oxide synthase
1999; 91: 607–620. inhibition blocks phencyclidine-induced behavioural effects on
331 Gueudet C, Santucci V, Soubrie P, Le Fur G. Blockade of prepulse inhibition and locomotor activity in the rat. Psycho-
neurokinin3 receptors antagonizes drug-induced population pharmacology 1997; 131: 167–173.
response and depolarization block of midbrain dopamine 353 Wiley JL, Golden KM, Bowen SE. Effects of modulation of nitric
neurons in guinea pigs. Synapse 1999; 33: 71–79. oxide on acoustic startle responding and prepulse inhibition in
332 Kamali F. Osanetant Sanofi-Synthelabo. Curr Opin Investig Drugs rats. Eur J Pharmacol 1997; 328: 125–130.
2001; 2: 950–956. 354 Wiley JL. Nitric oxide synthase inhibitors attenuate phencycli-
333 Rein W, Arvanitis L. Antipsychotic effect of four different dine-induced disruption of prepulse inhibition. Neuropsycho-
compounds—results of the metatrial. Eur Neuropsychopharma- pharmacology 1998; 19: 86–94.
col 2003; 13: S95. 355 Deutsch SI, Rosse RB, Schwartz BL, Fay-McCarthy M, Rosenberg
334 Nemeroff CB. Neurotensin: perchance an endogenous neurolep- PB, Fearing K. Methylene blue adjuvant therapy of schizophre-
tic? Biol Psychiatry 1980; 15: 283–302. nia. Clin Neuropharmacol 1997; 20: 357–363.
335 Binder EB, Kinkead B, Owens MJ, Nemeroff CB. The role of 356 Maurice T, Phan VL, Urani A, Kamei H, Noda Y, Nabeshima T.
neurotensin in the pathophysiology of schizophrenia and the Neuroactive neurosteroids as endogenous effectors for the sigma1
mechanism of action of antipsychotic drugs. Biol Psychiatry (sigma1) receptor: pharmacological evidence and therapeutic
2001; 50: 856–872. opportunities. Jpn J Pharmacol 1999; 81: 125–155.
336 Kinkead B, Nemeroff CB. Neurotensin: an endogenous antipsy- 357 Roberts E, Bologa L, Flood JF, Smith GE. Effects of dehydroe-
chotic? Curr Opin Pharmacol 2002; 2: 99–103. piandrosterone and its sulfate on brain tissue in culture and on
337 Vita N, Oury-Donat F, Chalon P, Guillemot M, Kaghad M, Bachy memory in mice. Brain Res 1987; 406: 357–362.
A et al. Neurotensin is an antagonist of the human neurotensin 358 Bologa L, Sharma J, Roberts E. Dehydroepiandrosterone and its
NT2 receptor expressed in Chinese hamster ovary cells. Eur J sulfated derivative reduce neuronal death and enhance astrocytic
Pharmacol 1998; 360: 265–272. differentiation in brain cell cultures. J Neurosci Res 1987; 17:
338 Feifel D, Reza TL, Wustrow DJ, Davis MD. Novel antipsycho- 225–234.
tic-like effects on prepulse inhibition of startle produced 359 Compagnone NA, Mellon SH. Dehydroepiandrosterone: a poten-
by a neurotensin agonist. J Pharmacol Exp Ther 1999; 288: tial signalling molecule for neocortical organization during
710–713. development. Proc Natl Acad Sci USA 1998; 95: 4678–4683.
339 Shilling PD, Richelson E, Feifel D. The effects of systemic NT69L, 360 Mao X, Barger SW. Neuroprotection by dehydroepiandrosterone-
a neurotensin agonist, on baseline and drug-disrupted prepulse sulfate: role of an NFkappaB-like factor. Neuroreport 1998; 9:
inhibition. Behav Brain Res 2003; 143: 7–14. 759–763.
340 Azzi M, Betancur C, Sillaber I, Spangel R, Rostene W, Berod A. 361 Bastianetto S, Ramassamy C, Poirier J, Quirion R. Dehydroepian-
Repeated administration of the neurotensin receptor antagonist drosterone (DHEA) protects hippocampal cells from oxidative
SR 48692 differentially regulates mesocortical and mesolimbic stress-induced damage. Brain Res Mol Brain Res 1999; 66:
dopaminergic systems. J Neurochem 1998; 71: 1158–1167. 35–41.
341 Finberg JP, Youdim MB. Pharmacological properties of the anti- 362 Bergeron R, de Montigny C, Debonnel G. Potentiation of neuronal
Parkinson drug rasagiline; modification of endogenous brain NMDA response induced by dehydroepiandrosterone and its
amines, reserpine reversal, serotonergic and dopaminergic suppression by progesterone: effects mediated via sigma recep-
behaviours. Neuropharmacology 2002; 43: 1110–1118. tors. J Neurosci 1996; 16: 1193–1202.

Molecular Psychiatry
Treatments for schizophrenia
S Miyamoto et al

104
363 Debonnel G, Bergeron R, de Montigny C. Potentiation by 379 Lieberman J, Chakos M, Wu H, Alvir J, Hoffman E, Robinson D
dehydroepiandrosterone of the neuronal response to N-methyl- et al. Longitudinal study of brain morphology in first episode
D-aspartate in the CA3 region of the rat dorsal hippocampus: an schizophrenia. Biol Psychiatry 2001; 49: 487–499.
effect mediated via sigma receptors. J Endocrinol 1996; 380 Kasai K, Shenton ME, Salisbury DF, Hirayasu Y, Lee CU,
150(Suppl): S33–S42. Ciszewski AA et al. Progressive decrease of left superior temporal
364 Flood JF, Smith GE, Roberts E. Dehydroepiandrosterone and its gyrus gray matter volume in patients with first-episode schizo-
sulfate enhance memory retention in mice. Brain Res 1988; 447: phrenia. Am J Psychiatry 2003; 160: 156–164.
269–278. 381 Kasai K, Shenton ME, Salisbury DF, Hirayasu Y, Onitsuka T,
365 Flood JF, Roberts E. Dehydroepiandrosterone sulfate improves Spencer MH et al. Progressive decrease of left Heschl gyrus and
memory in aging mice. Brain Res 1988; 448: 178–181. planum temporale gray matter volume in first-episode schizo-
366 Flood JF, Morley JE, Roberts E. Memory-enhancing effects in male phrenia: a longitudinal magnetic resonance imaging study. Arch
mice of pregnenolone and steroids metabolically derived from it. Gen Psychiatry 2003; 60: 766–775.
Proc Natl Acad Sci USA 1992; 89: 1567–1571. 382 Thome J, Foley P, Riederer P. Neurotrophic factors and the
367 Reddy DS, Kulkarni SK. The effects of neurosteroids on maldevelopmental hypothesis of schizophrenic psychoses.
acquisition and retention of a modified passive-avoidance Review article. J Neural Transm 1998; 105: 85–100.
learning task in mice. Brain Res 1998; 791: 108–116. 383 Weinberger DR. Implications of normal brain development for the
368 Maurice T, Junien JL, Privat A. Dehydroepiandrosterone sulfate pathogenesis of schizophrenia. Arch Gen Psychiatry 1987; 44:
attenuates dizocilpine-induced learning impairment in mice via 660–669.
sigma 1-receptors. Behav Brain Res 1997; 83: 159–164. 384 Stahl SM. When neurotrophic factors get on your nerves:
369 Tourney G, Erb JL. Temporal variations in androgens and stress therapy for neurodegenerative disorders. J Clin Psychiatry 1998;
hormones in control and schizophrenic subjects. Biol Psychiatry 59: 277–278.
1979; 14: 395–404. 385 Basile VS, Masellis M, Ozdemir V, Meltzer HY, Macciardi FM,
370 Strauss EB, Sands DE, Robibson AM, Tindall WJ, Stevenson Kennedy JL. Application of pharmacogenetics to schizophrenia:
WAH. Use of dehydroisoandrosterone in psychiatric treatment: a Emerging insights from studies of clozapine response and tardive
preliminary survey. Br Med J 1952; 2: 64–66. dyskinesia. In: Breier A, Tran PV, Herrera JM, Tollefson GD,
371 Sands DE. Further studies on endocrine treatment in adolescence Bymaster FP (eds). Current Issues in the Psychopharmacology of
and early adult life. J Ment Sci 1954; 100: 211–219. Schizophrenia. Lippincott Williams & Wilkins Healthcare,
372 Strauss EB, Stevenson WAH. Use of dehydroisoandrosterone in Philadelphia, 2001, pp 85–110.
psychiatric practice. J Neurol Neurosurg Psychiatry 1955; 18: 386 Arranz MJ, Munro J, Birkett J, Bolonna A, Mancama D, Sodhi M
137–144. et al. Pharmacogenetic prediction of clozapine response. Lancet
373 Strous RD, Maayan R, Lapidus R, Stryjer R, Lustig M, Kotler M et 2000; 355: 1615–1616.
al. Dehydroepiandrosterone augmentation in the management of 387 Shastry BS. Schizophrenia: a genetic perspective (review). Int J
negative, depressive, and anxiety symptoms in schizophrenia. Mol Med 2002; 9: 207–212.
Arch Gen Psychiatry 2003; 60: 133–141. 388 Arranz MJ, Kerwin RW. Advances in the pharmacogenetic
374 Vallee M, Purdy RH, Mayo W, Koob GF, Le Moal M. Neuroactive prediction of antipsychotic response. Toxicology 2003; 192:
steroids: new biomarkers of cognitive aging. J Steroid Biochem 33–35.
Mol Biol 2003; 85: 329–335. 389 Volavka J, Czobor P, Sheitman B, Lindenmayer JP, Citrome L,
375 Vallee M, Mayo W, Le Moal M. Role of pregnenolone, dehydroe- McEvoy J et al. Cozapine, olanzapine, risperidone, and haloper-
piandrosterone and their sulfate esters on learning and memory in idol in patiens with chronic schizophrenia and schizoaffective
cognitive aging. Brain Res Brain Res Rev 2001; 37: 301–312. disorder. Am J Psychiatry 2002; 159: 255–262.
376 Zou LB, Yamada K, Sasa M, Nakata Y, Nabeshima T. Effects of 390 Stroup TS, McEvoy JP, Swartz MS, Byerly MJ, Glick ID, Canive
sigma(1) receptor agonist SA4503 and neuroactive steroids on JM et al. The National Institute of Mental Health Clinical
performance in a radial arm maze task in rats. Neuropharmacol- Antipsychotic Trials in Intervention Effectiveness (CATIE)
ogy 2000; 39: 1617–1627. Project: Schizophrenia trial design and protocol development.
377 Noda Y, Kamei H, Kamei Y, Nagai T, Nishida M, Nabeshima T. Schizophrenia Bull 2003; 29: 15–31.
Neurosteroids ameliorate conditioned fear stress: an association with 391 Mamo D, Kapur S, Shammi CM, Papatheodorou G, Mann S,
sigma receptors. Neuropsychopharmacology 2000; 23: 276–284. Therrien F et al. A PET study of dopamine D2 and serotonin 5-
378 Lieberman JA. Is schizophrenia a neurodegenerative disorder?: HT2 receptor occupancy in patients with schizophrenia treated
a clinical and pathophysiological perspective. Biol Psychiatry with therapeutic doses of ziprasidone. Am J Psychiatry 2004; 161:
1999; 46: 729–739. 818–825.

Molecular Psychiatry

You might also like