Abstract
Current antipsychotic drugs (APDs) act on D2 receptors, and preclinical studies demonstrate that repeated D2 antagonist administration downregulates spontaneously active DA neurons by producing overexcitation-induced inactivation of firing (depolarization block). Animal models of schizophrenia based on the gestational MAM administration produces offspring with adult phenotypes consistent with schizophrenia, including ventral hippocampal hyperactivity and a DA neuron overactivity. The MAM model reveals that APDs act differently in a hyperdopamineregic system compared to a normal one, including rapid onset of depolarization block in response to acute D2 antagonist administration and downregulation of DA neuron population activity following acute and repeated D2 partial agonist administration, none of which are observed in normal rats. Novel target compounds have been developed based on the theory that glutamatergic dysfunction is central to schizophrenia pathology. Despite showing promise in preclinical research, none of the novel drugs succeeded in clinical trials. However, preclinical research is generally performed in normal, drug-naïve rats, whereas models with disease-relevant pathology and prior APD exposure may improve the predictive validity of preclinical research. Indeed, in MAM rats, chronic D2 antagonist treatment leads to persistent DA supersensitivity that interferes with the response to drugs that target upstream pathology. Moreover, MAM rats revealed that the peri-pubertal period is a stress-sensitive window that can be targeted to prevent the development of MAM pathology in adulthood. Neurodevelopmental models, such as the MAM model, can thus be used to test potential pharmacotherapies that may be able to treat schizophrenia in early stages of the disease.
1. Introduction
Animal models are essential for generating predictions and confirming causal relationships about the development, pathophysiology, and treatment of psychiatric disorders. The value of a given model is traditionally based on validity criteria, which consist of face validity (phenotypes that resemble the human condition), construct validity (similar theoretical rationale, such as a common etiology), and predictive validity (the ability to extrapolate the effects of a manipulation between species). An inherent challenge to validity criteria in translational schizophrenia research is the inability to reproduce all dimensions of a uniquely human condition. Current models are thus limited to a subset of phenotypes, with no effective means to directly measure subjective mental states, such as hallucinations and delusions (Arguello and Gogos, 2006; Powell and Miyakawa, 2006).
Despite their limitations, animal models are necessary tools for preclinical research into the mechanism of action and targeting of antipsychotic drugs (APDs). APDs are the primary treatment for schizophrenia and they dramatically reduce psychotic symptoms in many patients (Leucht et al., 2009). However, their therapeutic value is hindered by numerous side effects, limited efficacy across symptom domains, and failure to treat the disease pathology, which can lead to a lifetime of maintenance treatment (Lieberman et al., 2005; Leucht et al., 2012). These factors negatively impact quality of life and contribute to a high level of treatment nonadherence that typically results in relapse (Robinson et al., 2002; Lieberman et al., 2005). Current APDs are also unable to address heterogeneity, both between individuals with distinct pathologies (Demjaha et al., 2014; Howes and Kapur, 2014) and within an individual through different phases of the disease (Krystal and Anticevic, 2015). Novel pharmacological targets have been investigated based on theories largely generated from animal research, yet clinical trial results have been less than promising (Dunlop and Brandon, 2015). The apparent lack of predictive validity, as evidenced by the failed clinical trials, necessitates reevaluation of the rigor and design of preclinical research for the future development of novel therapies. Similarly, clinical trial design should be re-evaluated based on data emerging from preclinical studies. Current efforts must be aimed at investigating potential variables that contribute to this disconnect to improve the translation of animal models and the therapeutic value of pharmaceutical treatments.
2. Modeling dopamine dysfunction in schizophrenia
2.1. Clinical evidence of dopamine dysfunction in schizophrenia
The success of the first APD chlorpromazine in the 1950s was a breakthrough in the current era of pharmacotherapy and represented a shift in the conceptualization of psychiatric symptoms as biological constructs that can be targeted and treated (López-Munoz et al., 2005). In the decade following their established efficacy, it was suggested that APDs may work by acting on dopamine (DA) receptors (Carlsson and Lindqvist, 1963; Van Rossum, 1967). This was later confirmed by the finding that all APDs block D2 receptors, which is strongly linked to their antipsychotic effect (Seeman and Lee, 1975; Creese et al., 1976). Action at D2 receptors has since been proposed to be both necessary and sufficient for the therapeutic effect of current APDs on positive symptoms (Kapur and Remington, 2001).
The DA hypothesis of schizophrenia postulates that hyperactivity of the DA system drives psychotic symptoms. Patients with schizophrenia who respond to current treatments consistently demonstrate measures of increased presynaptic DA function compared to healthy controls (Howes et al., 2012). Early pivotal studies in patients used positron emission tomography (PET) and single photon emission computed tomography (SPECT) imaging to examine radioligand displacement from DA receptors as a measure of DA release. Compared to healthy controls, patients with schizophrenia show increased release of DA in the striatum at basal conditions (Abi-Dargham et al., 2000) and in response to amphetamine (Laruelle et al., 1996; Abi-Dargham et al., 1998). Furthermore, the increased DA signal is correlated with positive symptom severity (Laruelle et al., 1999). Patients with schizophrenia also show elevated striatal DA synthesis capacity, measured by fluorodopa uptake into DA terminals. The increased response capacity of the DA system is present in individuals at ultrahigh risk for psychosis (UHR), with further increase associated with the onset of psychosis (Howes et al., 2009; Howes et al., 2011a; Howes et al., 2011b). Although measures of DA function are reliable markers of psychosis in patients, there are challenges associated with choosing appropriate animal models to best study DA dysfunction relevant to schizophrenia.
2.2. Animal models of dopamine dysfunction
2.2.1. Pharmacological models of schizophrenia
Pharmacological models of schizophrenia are based on studying the action of drugs that induce psychotic states. For example, NMDA receptor antagonists, including phencyclidine (PCP) and ketamine, produce psychotomimetic effects in healthy subjects (Krystal et al., 1994) and exacerbate symptoms in patients with schizophrenia (Lahti et al., 1995). Acute administration of ketamine and PCP increases striatal DA levels in rats (Deutch et al., 1987; Moghaddam et al., 1997) and healthy human subjects (Kegeles et al., 2000; Vollenweider et al., 2000). In contrast, chronic administration of an NMDA receptor antagonist has been shown to have no effect on striatal DA levels (Lannes et al., 1991). Amphetamine has also been examined as a schizophrenia model based on clinical evidence that acute administration can increase DA efflux and exacerbate symptoms in patients (Snyder, 1973; Robinson and Becker, 1986). Prolonged amphetamine use in the normal population can lead to paranoid psychosis, but by the time an individual is addicted to amphetamine, the drug elicits a relatively small DA release compared to the initial dose (Castner and Williams, 2007; Nutt et al., 2015). Both NMDA receptor antagonists and amphetamine demonstrate significant differences between their acute and chronic effects that impact the interpretations that can be drawn from their use in animal models.
Pharmacological models are useful for studying the consequences of a manipulation that may have relevance to schizophrenia, but they provide limited value in understanding the etiology of the disease, which must be considered in the discovery of novel APDs. For example, the ability of NMDA receptor antagonists to mimic psychosis does not necessarily mean that NMDA receptors are impaired in schizophrenia, but they have provided insight into pyramidal neuron disinhibition relevant to the glutamatergic dysfunction observed in schizophrenia (Homayoun and Moghaddam, 2007; O’Donnell, 2013). However, the circuit and system-level abnormalities that underlie psychotic symptoms develop over a lifetime in response to genetic and environmental factors (Lewis and Levitt, 2002).
2.2.2. Neurodevelopmental models of schizophrenia
Neurodevelopmental animal models are based on the hypothesis that deviations from normal maturation can produce long-term changes in the brain. The structural and functional abnormalities that result from early-life insults often only fully emerge in adulthood. This makes developmental models valuable for identifying processes through which a triggering event can progress into a pathological state and evaluating preventative interventions with potential use in the prodromal stage (Meyer and Feldon, 2010).
Developmental consequences are commonly observed from environmental manipulations that involve early-life stress, such as social isolation or maternal separation (Harlow et al., 1965; Meaney et al., 1996). Adversity during the juvenile and adolescent period can shape the maturation of circuits that underlie emotional function (Cohen et al., 2013) and the resulting hyperresponsivity to stress is a core component of many psychiatric disorders (Heim and Nemeroff, 2001; Lupien et al., 2009). A large body of work highlights the importance of stress as a risk factor in the development of schizophrenia (Corcoran et al., 2003; van Os et al., 2010; Holtzman et al., 2013; Gomes and Grace, 2017). The correlation between early life stress and severity of positive symptoms (Ruby et al., 2017) may partially be due to the interaction between stress and the DA system (Belujon and Grace, 2015; Howes et al., 2017). Both UHR individuals and schizophrenia patients demonstrate elevated DA release in response to stress compared to healthy controls (Pruessner et al., 2004; Mizrahi et al., 2012). Furthermore, peripubertal stress exposure in rats has been shown to increase DA neuron activity in adulthood, suggesting that stress before or during puberty is particularly impactful to the responsivity of the DA system (Gomes and Grace, 2016).
The neonatal ventral hippocampal lesion (NVHL) model was the first to test the hypothesis that altered development of the ventral hippocampus (vHPC), a critical site of pathology in schizophrenia, can mimic the developmental course of schizophrenia. The model showed that a vHPC lesion in the early postnatal period results in the adult onset of behavioral abnormalities and DA dysfunction relevant to schizophrenia (Lipska et al., 1992; Lipska and Weinberger, 2000). Inspired by findings from the NVHL model and studies demonstrating the importance of immune activation during pregnancy as a risk factor for schizophrenia (Zuckerman et al., 2003; Canetta and Brown, 2012), another prominent neurodevelopmental model of schizophrenia was developed, the methylazoxymethanol acetate (MAM) model (Johnston et al., 1981; Moore et al., 2006). The MAM model involves administration of the mitotoxin MAM to pregnant dams on gestational day 17, which correlates with the vulnerable timepoint of the 2nd trimester in humans to adverse events such as maternal infection (Brown, 2006). The offspring of MAM-treated dams (“MAM rats”) develop region-specific disruption of neuronal maturation that results in adult phenotypes relevant to schizophrenia, compared to the offspring of dams that receive a saline injection, (“SAL rats”) (Flagstad et al., 2004; Moore et al., 2006; Modinos et al., 2015). Despite their different approaches, both the NVHL model and the MAM model display enhanced responsivity to stress (Lipska et al., 1993; Zimmerman et al., 2013) in addition to similar structural and functional abnormalities in adulthood.
One of the characteristic phenotypes observed in MAM rats is a hyperdopaminergic system. MAM rats demonstrate an increased number of spontaneously active DA neurons in the VTA, referred to as population activity, consistent with the hyperactive presynaptic DA function observed in schizophrenia (Laruelle and Abi-Dargham, 1999; Lodge and Grace, 2007; Howes et al., 2011a). Spontaneously active DA neurons fire in a tonic, irregular pattern in their basal state. When exposed to a behaviorally salient signal, DA neurons respond by firing bursts of action potentials, resulting in phasic DA release. However, only spontaneously active DA neurons can produce burst firing in response to glutamatergic afferents acting on NMDA receptors (Grace and Bunney, 1984; Legault and Wise, 1999; Floresco et al., 2003; Lodge and Grace, 2006b). Regulatory inputs to the VTA normally modulate DA neuron population activity to vary the phasic signal based on environmental context, such as situations that are threatening or rewarding (Floresco et al., 2003; Grace et al., 2007). We propose that pathologically increased DA neuron population activity observed in MAM rats causes stimuli to produce a maximal DA response independent of context (Fig. 1; Lodge and Grace, 2011; Grace and Gomes, 2018), consistent with the “aberrant salience” framework of psychosis (Kapur, 2003). We also propose that the MAM rats are not necessarily “doomed from the womb”, but instead fit a “two hit model” where the early disruption in brain development leads to heightened vulnerability to stress, with the potential for intervention prior to the second hit (Gomes et al., 2016).
Figure 1.

Hippocampal-striatal regulation of DA neuron activity in normal and schizophrenia. A) In a normal system, the limbic hippocampus (HPC), regulates dopamine (DA) neuron population activity in the ventral tegmental area (VTA), with variable gain based on context. The HPC sends glutamatergic input to the nucleus accumbens in the ventral striatum, which sends inhibitory projections to the ventral pallidum. The ventral pallidum holds a proportion of DA neurons in an inhibited state. In response to relevant stimuli, only spontaneously active DA neurons can respond to glutamatergic input from the pedunculopontine tegmentum (PPTg), gated by the laterodorsal tegmentum (LDT), with burst firing. The DA signal to the striatum has topographic organization with DA neurons in the substantia nigra pars compacta (SNc) projections to the dorsal striatum, lateral VTA projections to the associative striatum, and medial VTA projections to the ventral striatum. B) In schizophrenia, the limbic HPC is in a pathologically hyperactive state and no longer responds appropriately to context. Increased excitatory input from HPC to the ventral striatum results in less inhibition of the ventral pallidum and fewer DA neurons held in a silent state. The increased DA signal is most prevalent in the associative striatum.
2.3. Using the MAM model to study hippocampal drive of DA pathology
The vHPC, the rodent analog of the anterior HPC in humans (Grace, 2016; Heilbronner et al., 2016), plays a critical role in the pathophysiology of schizophrenia. Its involvement in context-based control of DA neuron activity is thought to contribute to the aberrant salience proposed to underlie psychosis (Kapur, 2003; Lisman et al., 2008). Deficits in the structure and function of the HPC are consistently observed in imaging and post-mortem studies of schizophrenia patients (Tamminga et al., 2010). Patients exhibit difficulty recruiting the HPC during tasks (Heckers et al., 1998; Weiss et al., 2003) and increased HPC activity at rest and during cognitive tasks involving processing of salient information (Medoff et al., 2001; Holt et al., 2006; Tregellas et al., 2014). Most studies report increased HPC glutamate levels in both first-episode and chronic patients, independent of medication status (Poels et al., 2014). HPC activity is also linked to disease progression; increased cerebral blood volume (CBV) has been reported in the anterior HPC during the prodromal stage and predicts conversion to psychosis (Harrison, 2004; Schobel et al., 2009) and atrophy of the anterior HPC (Schobel et al., 2013). Cross-sectional comparison of UHR subjects to healthy controls generally does not reveal differences in HPC volume between the two populations (Velakoulis et al., 2006; Wood et al., 2008). However, longitudinal studies have demonstrated that UHR patients that convert to psychosis show a greater progression of grey matter reduction of the anterior CA1 and subiculum compared to those who do not convert to psychosis (Pantelis et al., 2003; Borgwardt et al., 2007; Schobel et al., 2013). Reduced volume of the anterior HPC has also been associated with illness duration (Velakoulis et al., 2006), although not all studies have found significant reduction over time (Lieberman et al., 2001) and chronic APD treatment or other external factors may contribute to progressive volume loss (Ho et al., 2011; Zipursky et al., 2012). Excitotoxicity from sustained oxidative stress may explain the correlation between increased regional glutamate levels and corresponding reduction in volume found in neuroimaging studies (Benes, 2000; Kraguljac et al., 2013; Do et al., 2015). Together, these findings suggest a strong link between HPC pathology and the onset of psychosis.
The MAM model has provided critical insights into how HPC pathology leads to increased DA neuron activity (Fig. 1). Silent DA neurons are held in a hyperpolarized state by the ventral pallidum, which is controlled by a circuit originating with the vHPC (Floresco et al., 2001; Lodge and Grace, 2006b). Both schizophrenia patients (Benes and Berretta, 2001) and MAM rats (Gill and Grace, 2014) show loss of parvalbumin (PV+) interneurons in the vHPC that provide inhibitory control of pyramidal cell activity, resulting in a baseline hyperactive state. Increased vHPC drive indirectly releases DA neurons from inhibition, which increases the population of spontaneously active DA neurons available for phasic burst firing in response to glutamatergic input from the pedunculopontine tegmentum (PPTg) (Floresco et al., 2001), which itself is enabled by the laterodorsal tegmentum (LDT) (Lodge and Grace, 2006a). Activation of the vHPC in normal rats dramatically increases DA neuron population activity in the VTA that is correlated with increased DA efflux in the associative areas of the ventral striatum and NAc, whereas inactivation of vHPC in MAM rats can normalize the DA neuron activity and related aberrant behavior (Floresco et al., 2003; Lodge and Grace, 2007). Current APDs reduce DA neuron hyperactivity through their action on D2 receptors, but they do not target or resolve the HPC pathology, instead working as many as 5 synapses downstream from the apparent site of pathology.
3. Antipsychotic drug regulation of dopamine system dysfunction
3.1. D2 receptor antagonists
Much of the research on APDs has focused on their mechanisms of action to understand how they achieve their therapeutic effect. APDs are traditionally categorized as either first-generation antipsychotics (FGAs), such as haloperidol, or second-generation antipsychotics (SGAs), such as clozapine, which act on a larger spectrum of receptor types, including serotonergic, cholinergic, and adrenergic receptors (Miyamoto et al., 2005). Due to the high level of heterogeneity within these broad designations, some argue that the current nomenclature is uninformative and should be replaced with a neuroscience-based naming system (Zohar et al., 2015). Like many psychoactive drugs, APDs have numerous physiological effects due to their rich pharmacology, although the antipsychotic effect of both FGAs and SGAs remains dependent on the D2 receptor and are given at doses that match D2 occupancy (Kapur and Remington, 2001). Most current APDs act as D2 receptor antagonists (Seeman, 1987), but to understand how FGAs and SGAs regulate the DA system, it is necessary to look beyond the receptor at downstream consequences on DA neuron activity.
D2 antagonists block both postsynaptic and presynaptic D2 receptors. Acute administration of D2 antagonists in anesthetized rats increases DA neuron firing rate and bursting activity, primarily through blockade of somatodendritic D2 autoreceptors that normally provide negative feedback for the neuron (Grace and Bunney, 1986; Pucak and Grace, 1994). However, with repeated treatment there is a substantially depolarized membrane potential that culminates in inactivation such that the DA neuron is no longer able to generate action potentials. The inactivation can be reversed by compounds that inhibit DA neurons, such as DA agonists, whereas excitatory compounds are no longer capable of stimulating activity (White and Wang, 1983; Grace and Bunney, 1986). This phenomenon, referred to as depolarization block, is proposed to underlie the regulatory action of D2 antagonists on DA neuron hyperactivity (Fig. 2A). This review will discuss some of the consequences of depolarization block, but for more detailed reviews on APD regulation of the DA system see (Grace et al., 1997; Arnt and Skarsfeldt, 1998).
Figure 2.

Antipsychotic drug modulation of DA neuron activity states. A) First generation D2 antagonist antipsychotic drugs (APDs) act on postsynaptic and presynaptic D2 receptors in the striatum to produce depolarization block of DA neurons in the ventral tegmental area (VTA) and substantia nigra pars compacta (SNc) and reduces DA neuron population activity in both regions. The DA signal is diminished to all domains of the striatum, resulting in both the antipsychotic effect and extrapyramidal side effects, including motor dysfunction. Following prolonged treatment, the system adapts by upregulating the number of D2 receptors, which can produce DA supersensitivity. B) Second generation D2 antagonist APDs only produce depolarization block of DA neurons in the ventral tegmental area (VTA), resulting in a reduced DA signal to the associative and ventral striatum without effect on the DA neurons that project to the dorsal striatum. Thus, SGAs produce an antipsychotic effect without motor side effects. However, like first generation APDs, following prolonged treatment, there is an upregulation of D2 receptors, which can produce DA supersensitivity. C) D2 partial agonists normalize DA neuron population activity by reducing hyperdopaminergic activity via DA neuron inhibition and increasing hypodopamineregic activity via postsynaptic stimulation. The downregulation of DA neuron activity does not occur through depolarization block and does not produce an upregulation of D2 receptors.
FGAs and SGAs have similar efficacy on positive symptoms and they are primarily distinguished in clinical use by differing side effect profiles (Leucht et al., 2013). One hypothesis is that APD efficacy is related to inhibition of DA function in the VTA, whereas extrapyramidal symptoms (EPS) are mediated by inhibition of DA function in the substantia nigra (SNc) (Deutch et al., 1991). Therefore, differences in specificity of depolarization block between FGAs and SGAs may explain their comparable efficacy and difference in propensity to cause EPS (Fig. 2; Haro and Salvador-Carulla, 2006; Correll and Schenk, 2008). Acute administration of FGAs activates DA neurons in both the VTA and substantia nigra (SNc), whereas SGAs only activate DA neurons in the VTA (Bunney and Grace, 1978; Chiodo and Bunney, 1985; Goldstein et al., 1993; Stockton and Rasmussen, 1996). Accordingly, FGAs produce depolarization block in both the VTA and SNc, whereas SGAs selectively produce depolarization block in the VTA (Chiodo and Bunney, 1983; White and Wang, 1983; Grace and Bunney, 1986; Lane and Blaha, 1987; Goldstein et al., 1993). It has been suggested that differing pharmacokinetics may also contribute to their side effect profiles: SGAs typically dissociate from the D2 receptor more readily than FGAs, which may make them more accommodating to physiological DA transmission (Kapur and Seeman, 2001). In support of this hypothesis, quetiapine and clozapine, the SGAs with the fastest dissociation rates, show no greater risk of EPS than placebo across their dosage range, whereas risperidone and olanzapine, SGAs with slower dissociation rates from the D2 receptor, are more likely to cause EPS at high doses (Kapur and Seeman, 2001). Of course, this depends on how EPS are defined. Whereas the FGAs cause motor-related EPS like Parkinson’s symptoms and tardive dyskinesia, akathisia is a dose-dependent adverse effect of all APDs, including SGAs and some D2 partial agonists (Berman et al., 2007; Kane et al., 2009). Although akathisia was initially classified as EPS, the fact that the motor activation is driven more by agitation or anxiety suggest a limbic origin of this symptom.
The onset of depolarization block may relate to the range of latencies in clinical improvement of psychosis, which can occur as fast as 24 h following the initial dose of an APD, with full effect after several weeks of treatment (Agid et al., 2003; Kapur et al., 2005; Leucht et al., 2005). In normal rats, 21 d repeated administration of FGAs and SGAs is necessary to reduce the number of spontaneously active DA neurons (Chiodo and Bunney, 1983; Grace and Bunney, 1986), whereas MAM rats undergo rapid onset of depolarization block following acute administration (Valenti et al., 2011). Enhanced DA system excitability in MAM rats likely underlies the rapid onset of depolarization block. This is consistent with clinical studies showing that patients with the highest level of psychosis, implicating a more hyperactive DA system, tend to show a rapid response to APD treatment (Robinson et al., 1999).
Depolarization block has been observed across numerous studies in rats, but questions remain about the consistency and consequences of the phenomenon. Some have argued that it may be an artifact of anesthesia (Mereu et al., 1995; Melis et al., 1998). However, chloral hydrate, the anesthesia used for DA neuron recordings, normally blunts the excitatory response of DA neurons to APDs (Bunney et al., 1973) and depolarization block has been observed in nonanesthetized, paralyzed rats (Bunney and Grace, 1978; Chiodo and Bunney, 1985). There have also been inconsistencies on the effect of depolarization block on striatal DA release. Some have reported a reduction in basal DA release that corresponds with depolarization block (Blaha and Lane, 1987; Lane and Blaha, 1987; Moore et al., 1998), whereas others have not found a difference in striatal DA levels (Ichikawa and Meltzer, 1991; Moghaddam and Bunney, 1993). However, the lesion produced from microdialysis probe insertion was later found to disrupt depolarization block, and the issue can be avoided by implanting the microdialysis probe prior to APD administration (Moore et al., 1998). The long-term changes to DA neuron activity following APD withdrawal are also unclear and require further investigation. We previously found that DA neuron activity remains reduced for at least a week following repeated D2 antagonist administration in both MAM and SAL rats (Gill et al., 2014). However, population activity is no longer restored with administration of a DA agonist, indicating that the reduction following withdrawal may no longer be caused by depolarization block and may be due to other compensatory mechanisms (Gill et al., 2014).
It has long been suggested that neuroadaptations to chronically administered psychiatric drugs can produce discontinuation syndromes and facilitate relapse, with rapid withdrawal inducing greater relapse (Viguera et al., 1997). One of the consequences of chronic D2 antagonist treatment is D2 receptor upregulation (Silvestri et al., 2000; Samaha et al., 2007), which can produce a state referred to as DA supersensitivity (Fig. 2; Chouinard et al., 2017). Development of DA supersensitivity has been attributed to breakthrough symptoms and the rapid rate of relapse following APD discontinuation or dose reduction, which often results in new and more severe symptoms (Chouinard and Jones, 1980; Chouinard et al., 2017). Studies in rats have demonstrated that DA supersensitivity occurs both during and after treatment, potentially accounting for breakthrough symptoms, although the consequences are more apparent following withdrawal (Samaha et al., 2007). Some have argued that breakthrough symptoms are more likely due to treatment noncompliance and can be circumvented by continuous blood levels using long acting injectables (LAIs) (Correll et al., 2018). However, long-term treatment with LAIs has been shown to provide modest, if any, reduction in the rate of relapse compared to oral administration in randomized controlled trials (Kishimoto et al., 2012) and roughly 20% of patients relapse within a year (Leucht et al., 2011). Further research on the interrelated topics of APD tolerance, withdrawal, and relapse is critical for clear guidelines on switching between APDs and determining which patients would benefit from dose reduction following stabilization (Murray et al., 2016). Indeed, one would predict that after the onset of depolarization block, the resultant decrease in DA release would require a lower APD dose to maintain this state. Despite the increase in D2 receptors, less therapeutic tolerance occurs to APDs in a clinical setting than what would be expected from antagonism of a receptor, which may be due to the indirect method of DA neuron regulation (i.e. depolarization block); however, the reduction of DA neuron activity is far from returning the system to a normal state (Fig. 2B).
3.2. D2 partial agonists
D2 partial agonists, such as aripiprazole, activate the receptor to a lower degree than endogenous DA and are sometimes referred to as third generation antipsychotics. They are thought to stabilize DA neurotransmission by reducing excessive striatal D2 stimulation and restoring deficient D2 stimulation in regions including the prefrontal cortex (Grace, 1991; Burris et al., 2002). . In contrast to D2 antagonists, which are associated with a therapeutic effect at striatal D2 receptor occupancy of 65-70% and increased risk of EPS at D2 receptor occupancy exceeding 80% (Farde et al., 1992; Kapur et al., 2000), aripiprazole has a therapeutic effect with 85-95% striatal D2 receptor occupation, yet does not produce EPS (Yokoi et al., 2002; Mamo et al., 2007). Furthermore, aripiprazole has been shown to not produce D2 receptor upregulation or behavioral DA supersensitivity in rats (Fig. 2; Inoue et al., 1997; Tadokoro et al., 2011), and there is preliminary evidence that it may alleviate DA supersensitivity in humans (Tadokoro et al., 2017). D2 receptor partial agonists may thus be less likely to induce adverse effects related to DA supersensitivity, as observed following chronic D2 antagonist treatment (Silvestri et al., 2000; Chouinard et al., 2017). These differences between D2 receptor antagonists and D2 partial agonists may be related to their distinct methods of DA neuron regulation.
Like the rapid reduction of DA neuron population activity in MAM rats in response to D2 receptor antagonist administration, acute aripiprazole administration also reduces the number of spontaneously active DA neurons in MAM rats (Valenti et al., 2011; Sonnenschein et al., 2019). The reduction in DA neuron activity in MAM rats remains stabilized following 1-week withdrawal from repeated aripiprazole administration, as previously observed following withdrawal from repeated haloperidol treatment (Gill et al., 2014; Sonnenschein et al., 2019). However, unlike D2 receptor antagonists (Valenti et al., 2011; Gill et al., 2014), neither acute nor repeated aripiprazole administration shows an effect on DA neuron population activity in normal rats (Sonnenschein et al., 2019). This is consistent with studies showing, in contrast to MAM rats, no effect of acute or repeated D2 partial agonist p.o. administration on extracellular DA levels in normal rats across a range of dosages (Jordan et al., 2004; Maeda et al., 2014) and suggests distinct functional effects dependent on the state of the DA system, in line with its action as a partial agonist. Other studies, using i.p. administration, have reported regional, dose dependent changes in DA levels in response to aripiprazole, which may relate to the different administration method (Li et al., 2004).
Despite aripiprazole’s antagonist-like reduction of hyperdopaminergic activity in MAM rats, it does not act via depolarization block (Fig. 2). Low doses of apomorphine either failed to restore, or further reduced, DA neuron population activity in MAM rats (Sonnenschein et al., 2019). No significant differences were observed in average firing rate and bursting activity across the total population of DA neurons recorded between aripiprazole and vehicle-treated MAM rats, p.o. (Sonnenschein et al., 2019), but acute i.p. aripiprazole administration has been shown to reduce DA neuron firing rate and bursting activity (Bortolozzi et al., 2007; Dahan et al., 2009; Sonnenschein et al., 2019), suggesting that it may reduce DA neuron population activity through action on presynaptic D2 receptors to downregulate DA neuron activity (Fig. 3). The greater number of presynaptic spare receptors may underlie aripiprazole’s agonist-like action on D2 autoreceptors to inhibit DA neuron activity (Kikuchi et al., 1995; Burris et al., 2002), while its lower intrinsic activity at the receptor than a full agonist may simultaneously explain its antagonist-like effect at postsynaptic D2 receptors in states of DA hyperactivity, such as blockade of apomorphine-induced hyperlocomotion and stereotypy (Kikuchi et al., 1995). Indeed, aripiprazole has been shown to reverse depolarization block in haloperidol-treated MAM rats (Sonnenschein et al., 2019), resembling depolarization block reversal following low-dose DA agonist administration (Valenti et al., 2011). This phenomenon may relate to the temporary worsening of psychotic symptoms reported in patients transitioning to aripiprazole from D2 antagonist treatment (Takase et al., 2015; Tadokoro et al., 2017).
Figure 3.

A proposed model for ntipsychotic drug effects on symptom domains of schizophrenia. A) In schizophrenia, a hyperactive and dysrhythmic limbic hippocampus (HPC) contributes to positive symptoms through its indirect regulation of DA activity in the ventral tegmental area (VTA), which projects to the associative striatum. It also contributes to negative and cognitive symptoms through its disruption of activity patterns with regions including the prefrontal cortex (PFC) and amygdala. All current antipsychotic drugs (APDs) act on D2 receptors in the striatum, which results in a reduction in positive symptoms, but produce minimal benefit on cognitive symptoms or negative symptoms. B) Compounds that target the site of pathology in the limbic HPC by either increasing function of parvalbumin positive interneurons (PVI) or reducing the activity of pyramidal neurons (Pyr) should normalize hippocampal output and thereby may be more beneficial in treating all symptom domains of schizophrenia.
It is difficult to conclude whether receptor reserve entirely accounts for aripiprazole’s complex effects across a range of systems. It has been suggested from in vitro studies that aripiprazole possesses functional selectivity dependent on the cellular environment of the receptor and the G protein-coupled receptor (GPCR) signaling pathway activated, which may contribute to its state-dependent effects (Lawler et al., 1999; Shapiro et al., 2003; Mailman and Gay, 2004; Urban et al., 2007). Aripiprazole may lack biased signaling in vivo, but it has served as a template for β-arrestin-biased D2 receptor ligands that have recently been studied as potential APD compounds to more effectively target both cortical and striatal DA dysfunction (Urs et al., 2017). Additional in vivo studies are needed to determine the mechanisms underlying aripiprazole’s selective downregulation of DA neuron activity in MAM rats and whether they can be generalized to all D2 receptor partial agonists.
4. Use of animal models in predicting efficacy of novel compounds
4.1. The necessity of novel antipsychotic mechanisms
A key goal of APD research is the development of personalized medication, which requires reliable biomarkers to determine which treatment is most suitable for a patient’s individual needs. Approximately 20-30% of patients with schizophrenia display treatment resistance with little or no response to standard medication (Conley and Buchanan, 1997) and a higher rate, up to 60%, only partially respond to treatment (Kane, 1989). Even clozapine, the treatment of choice for refractory symptoms, can be ineffective in approximately 40-70% of patients who take it (Kane et al., 1988; Meltzer et al., 1989; Lieberman et al., 1994; Remington et al., 2005). In the event of inadequate response, clinicians often resort to the combination of multiple APDs, with a wide range of approximately 10-50% of patients treated by polypharmacy, depending on the clinical setting (Stahl and Grady, 2004; Faries et al., 2005; Fleischhacker and Uchida, 2014). Despite its prevalence, there is limited evidence that long-term APD polypharmacy benefits patients (Stahl and Grady, 2004), and may instead be detrimental due to increased frequency of adverse effects (Correll et al., 2007; De Hert et al., 2012).
Treatment resistance can occur in patients who once responded to APDs but no longer do, which can occur following relapse and may be related to DA supersensitivity (Emsley et al., 2013). There are also patients who never responded to APDs whose psychotic symptoms have been proposed to not involve DA dysregulation (Howes and Kapur, 2014). Studies have shown that schizophrenia patients with low striatal DA synthesis capacity, and thus fewer DA neurons active, are more likely to be treatment non-responders (Demjaha et al., 2012; Demjaha et al., 2014). Given that a more active DA system responds faster to D2 blockade, this subset of patients may still have a pathology involving the DA system, but the low responsivity of their DA system would not be a pathology conducive to induction of depolarization block or other means of DA neuron downregulation (Grace and Gomes, 2018). However, these patients still show signs of glutamatergic dysfunction, including higher glutamate levels in the anterior cingulate compared to treatment responders (Demjaha et al., 2014), suggesting that targeting the glutamatergic pathology may alleviate symptoms for both treatment responders and non-responders. Current APDs exclusively target the DA system, necessitating further development of novel target options for treatment resistant patients.
Among both treatment responders and nonresponders, there remain unmet needs in tolerability and safety of current APDs. In recent years, concern has been raised about the risk-to-benefit ratio of maintenance APD treatment, due to issues including DA supersensitivity and potential deleterious effects on brain structure and functioning (Murray et al., 2016; Chouinard et al., 2017). Treating patients with the minimum necessary dose and using D2 partial agonists may minimize side effects and the risks associated with long term APD treatment, but treating the disease closer to the site of pathology may circumvent the issues associated with D2-targeting drugs and address additional symptoms. To improve functional outcome, an ideal treatment would also address negative symptoms and cognitive deficits (Green et al., 2000; Milev et al., 2005), but this may be more readily accomplished through the use of adjunctive treatments that are not necessarily disease specific (Carpenter and Koenig, 2008). However, given that the vHPC is dysrhythmic and hyperactive, and projects not only to the DA controlling regions but also to the prefrontal cortex and amygdala, normalizing vHPC activity may provide symptomatic relief across all symptom classes (Fig. 3; Grace and Gomes, 2018).
4.2. The rise and fall of novel target compounds
A massive investment has gone into the development of novel target compounds with the hope that they would be more effective in treating the positive symptoms of schizophrenia, in addition to negative and cognitive symptoms. A common theme among them was promising performance in preclinical research, but failure to show sufficient antipsychotic efficacy in larger clinical trials (Dunlop and Brandon, 2015). However, a common caveat is that in preclinical trials, all drugs were tested on APD-naïve animals, whereas in clinical trials the drugs were all tested on schizophrenia patients that had received long courses of APD therapy. This review focuses on two of the novel target compounds that were aimed at regulating glutamatergic abnormalities in schizophrenia.
4.2.1. Glycine Transport Inhibitors
A number of potential APD compounds were developed based on findings that NMDA receptor antagonists can produce effects similar to the symptoms of schizophrenia. Blockade of NMDA receptors on PV+ interneurons results in disinhibition of pyramidal cell activity similar to the dysregulated pyramidal cell activity observed in schizophrenia (Homayoun and Moghaddam, 2007; Schobel et al., 2013), which led to efforts to enhance NMDA receptor function in patients (Coyle et al., 2003). Both glutamate and glycine sites need to be occupied on NMDA receptors for the channel to open, but direct drug action at the glutamate site can be toxic (Thomson et al., 1989). There was initial promise in initial clinical studies using glycine or D-serine, the endogenous co-agonist of NMDA receptors (Mothet et al., 2000), as an adjunct to existing APDs to improve positive, negative, and cognitive symptoms in schizophrenia (Heresco-Levy et al., 1996; Tsai et al., 1998; Heresco-Levy et al., 1999; Evins et al., 2000; Javitt et al., 2001; Evins et al., 2002; Heresco-Levy et al., 2002; Heresco-Levy et al., 2004; Heresco-Levy et al., 2005), though larger, longer studies did not yield positive results (Goff et al., 2005; Buchanan et al., 2007; Weiser et al., 2012). A selective glycine transporter 1 (GlyT1) inhibitor was studied as an alternative strategy with greater brain penetrance to increase the availability of glycine to bind to NMDA receptors (Javitt, 2012). The most extensively studied GlyT1 inhibitor was bitopertin from Roche. Bitopertin showed promise in preclinical research, including reversing amphetamine-induced hyperlocomotion in both naïve mice and mice chronically treated with PCP (Alberati et al., 2012). Despite some initial success of a similar compound as an adjunct therapy (Tsai et al., 2004), biopertin failed to demonstrate efficacy in both phase 2 clinical trials (Umbricht et al., 2014) and phase 3 clinical trials as either a monotherapy or adjunct therapy compared to standard treatment (Bugarski-Kirola et al., 2014; Bugarski-Kirola et al., 2016; Bugarski-Kirola et al., 2017). Biopertin was grounded in decades of research on the potential of targeting glutamatergic dysfunction, yet, like other novel target compounds tested at the time, followed the trend of initial success leading to failure to replicate in larger clinical trials, potentially due to patient heterogeneity (Goff, 2014).
4.2.2. mGluR2/3 agonists
Group II metabotropic glutamate receptor (mGluR2/3) agonists were another promising novel target compound investigated for the treatment of schizophrenia. They were designed to regulate glutamate release by acting on the presynaptic mGlu2/3 receptors located on glutamatergic terminals in cortical and limbic brain regions (Nicoletti et al., 2011). Following extensive preclinical support for mGluR2/3 as an APD target (Moghaddam and Adams, 1998; Cartmell et al., 1999, 2000; Lorrain et al., 2003; Greenslade and Mitchell, 2004; Homayoun et al., 2005; Pehrson and Moghaddam, 2010), an mGluR2/3 agonist from Eli Lilly, pomaglumetad methionil, was advanced to clinical trials. It showed promise in preclinical research, including reversing effects caused by NMDA receptor antagonists, blocking amphetamine-induced hyperlocomotion, and showing anxiolytic effects in models of anxiety (Rorick-Kehn et al., 2007; Mezler et al., 2010). It demonstrated potential as a monotherapy in its first double-blind, randomized, placebo-controlled phase 2 clinical trial, and it was found to be well tolerated with no propensity to cause weight gain or EPS (Patil et al., 2007). This success was followed by a long line of disappointments in subsequent phase 2 trials as a monotherapy or adjunct therapy (Adams et al., 2013; Stauffer et al., 2013; Downing et al., 2014) and failure to meet the primary endpoint as a monotherapy in a large phase 3 trial (Adams et al., 2014; J Marek, 2015). Later analyses of the clinical trials found that certain populations responded better to pomaglumetad (Kinon et al., 2015), leaving questions about why those groups responded better and whether glutamatergic-targeting therapies may be best suited for certain populations. For example, early-in-disease patients responded better to pomaglumetad treatment than chronic patients (Kinon et al., 2015), which could potentially be due to factors including treatment history and disease progression. Additionally, a study in mice showed that SGA administration can downregulate mGluR2 expression, thought to be due to action on 5-HT2A receptors, which may have contributed to the failure (Kurita et al., 2012). There were also efforts to develop mGluR2 positive allosteric modulators (PAMs) that would induce a leftward shift in the dose-response curve for glutamate without directly acting at the glutamate site (Conn et al., 2009; Fraley, 2009; Fell et al., 2012). They similarly showed promising results in APD screening assays (Lavreysen et al., 2013), though a compound tested by AstraZeneca once again did not show efficacy in patients with schizophrenia as a monotherapy (Litman et al., 2016). Despite the failure of the clinical trials, they are suggestive that there may be sub-populations of patients who may benefit most from novel drugs and they also highlight the need to study the effects of prior APD treatment.
4.3. Future directions in preclinical research for novel target antipsychotic drugs
The mechanisms of action tested in clinical trials were derived from decades of human and animal model research showing excitatory-inhibitory imbalance as a core component schizophrenia and a promising target across symptom domains. Their failure created uncertainty in the current theoretical framework of schizophrenia regarding the potential to treat glutamatergic dysfunction. Moving forward, one option is to leave these mechanisms of action in the “pharmaceutical graveyard” and forgo pursuit of similar novel target compounds for the treatment of schizophrenia. Alternatively, we propose that these drugs, or others based on similar theoretical concepts, still hold promise, but require additional preclinical screening to better conduct clinical trials. A particular caveat is that the prior clinical trial protocol was based on finding more effective D2 antagonist APDs, such that switching from one D2 antagonist to another may not be as critical an issue. However, when testing novel target compounds, prior treatment history may significantly impact the results.
4.3.1. Animal Model Selection and Interpretation
The brain of a patient with schizophrenia who has had years of APD exposure is distinct from that of a drug naïve patient or a healthy volunteer. Consequently, normal animals are useful as a blank slate to examine the effects of a drug, but they are not well-suited for APD screening compared to approaches that incorporate disease-relevant abnormalities. For example, D2 antagonists and D2 partial agonists function differently in a hyperdopaminergic state, as observed in the MAM model of schizophrenia, which may be more representative of how they act in a patient (Valenti et al., 2011; Sonnenschein et al., 2019). The choice of proper animal models and assays to study APDs is particularly important when testing novel mechanisms.
There has been much debate over whether current preclinical assays of APD efficacy are useful for screening novel mechanisms of action (Carpenter and Koenig, 2008; Nestler and Hyman, 2010). For example, blockade of amphetamine-induced hyperlocomotion is a behavioral task in rodents commonly used to predict APD efficacy to treat positive symptoms. However, amphetamine-induced hyperlocomotion is believed to arise from a ventromedial limbic striatal action (Creese and Iversen, 1975), whereas imaging studies show that increased raclopride displacement and increased f-DOPA uptake that correlate with psychosis is present primarily in the associative striatum (Kegeles et al., 2010; Kesby et al., 2018). Hypodopaminergic signaling has been observed in extra-striatal regions, and DA deficits in the dorsolateral prefrontal cortex in particular is thought to contribute to cognitive deficits in schizophrenia (Slifstein et al., 2015). It is currently unknown what accounts for these coexisting differences in DA regulation, which have yet to be demonstrated in an animal model. Given the current models available, a more effective screen should selectively target DA release in the associative striatum, such as a task that addresses the role of DA function in salience attribution and selective attention, which may be a more reliable measure of APD efficacy. Additionally, behavioral tasks that do not result in obvious face validity should not be considered irrelevant to the disease, as common neurobiological effects could underlie different behavioral phenotypes across species.
Drug dosage and administration methods are critical factors in demonstrating consistent effects, both when comparing between animal studies and translating between animal and human research. Metabolic differences must be considered to determine corresponding receptor occupancy (Kapur et al., 2003) in addition to the administration method, particularly when looking at the effects of repeated treatment. Transient APD delivery was found to be more effective than continuous D2 blockade, which was more likely to result in D2 receptor upregulation and behavioral tolerance. It has been reported that continuous administration may be more likely to result in DA supersensitivity (Samaha et al., 2008), whereas intermittent dosing with “drug holidays” is more likely to maintain efficacy (Remington and Kapur, 2010). Given that patients typically receive long-term APD treatment, animal research would benefit from implementing more chronic treatment studies to examine long-term responses and identify any compensatory mechanisms, including changes in receptor response and more widescale changes in circuit plasticity.
4.3.2. Treatment History
Preclinical modeling can be used to determine the contribution of chronic APD treatment in clinical findings. We previously found that 1 week withdrawal from repeated haloperidol treatment produces persistent DA supersensitivity in MAM rats, interfering with the ability of a novel target compound to reduce amphetamine-induced hyperlocomotion, in contrast to its ability to regulate DA hyperresponsivity in drug-naïve rats (Gill et al., 2011; Gill et al., 2014). While the washout period of 1-2 weeks typically used in clinical studies may be adequate to decrease blood levels of the APD, the study demonstrates that a short withdrawal period is not sufficient to reverse the effects of the drug on the DA system. It also raises the possibility that DA supersensitivity following prior D2 antagonist treatment in patients with schizophrenia may similarly mask potential effects of novel target compounds in clinical trials, despite their promise in screening assays performed in normal, drug-naïve rodents. The results of the trial could potentially be confounded by the long-lasting changes in the system made by the presence of the APD, which may contribute to why the novel drugs performed better in early-in-disease patients (Kinon et al., 2015). In the presence of D2 receptor upregulation following chronic D2 receptor antagonist treatment (Silvestri et al., 2000; Samaha et al., 2007), it is possible that only another D2 receptor-targeting drug may be effective in reversing hyperresponsivity to DA.
4.3.3. Targeting Upstream Pathology
Future development of APDs should aim to provide normal regulation of DA neuron activity rather than general suppression of DA neurotransmission. Given the large amount of evidence that loss of PV+ interneuron regulation of pyramidal cell activity underlies many aspects of schizophrenia, including its role in DA system hyperactivity, it still holds promise as an APD target and may avoid consequences associated with D2 antagonist treatment. Targeting excitatory-inhibitory processes may also alleviate more symptoms of the disorder, including negative and cognitive symptoms, which often precede the first psychotic episode and persist after treatment of psychotic symptoms (Harvey et al., 2005). Several studies from our lab and others have found success with using strategies to target vHPC dysfunction to normalize DA neuron activity and behavior in MAM rats (Gill et al., 2011; Gastambide et al., 2012; Perez and Lodge, 2013; Perez et al., 2013).
Targeting upstream pathology has the potential to be more effective in treating schizophrenia in patients who have already developed the disease, but an ideal approach would involve intervention prior to the transition to psychosis, which may lead to a better long-term outcome (Perkins et al., 2005; Gomes et al., 2016; Grace and Gomes, 2018). Treatment of glutamatergic dysfunction during the pre or peri-pubertal period can produce long-lasting effects on DA function based on studies in MAM rats. Prior to puberty, rats do not display the full expression of MAM-associated phenotypes (Moore et al., 2006). Peripubertal administration of the benzodiazepine diazepam has been shown to attenuate the increased anxiety-like behavior and basolateral amygdala hyperactivity normally present in MAM rats. The reduced stress response also likely underlies the decreased PV+ interneuron loss in the vHPC and prevention of hyperdopamineregic activity in adult MAM rats observed following peripubertal diazepam administration (Du and Grace, 2013, 2016b, a). Benzodiazepines are not a realistic prophylactic option clinically due to a number of issues including dependence and tolerance, but it demonstrates the potential long-term benefits of treatment during the peripubertal period. It has been reported that increased HPC activity, local reduction in PV+ interneurons, and volume loss following repeated ketamine administration in mice can be prevented by coadministration with an mGluR2/3 agonist. However, the same drug may lose its efficacy for patients with long-standing illness (Kinon et al., 2015). Together, these studies suggest that decreasing stress or other means of reducing HPC activity prior to the first episode (Schobel et al., 2013) could circumvent the damage that leads to the emergence of the DA dysregulation and psychotic symptoms (Gomes et al., 2016; Grace and Gomes, 2018). They also suggest the potential for stage-specific pharmacotherapies whereby patients may have options for optimal treatment strategies based on the neurobiological course of the disease (Krystal and Anticevic, 2015).
5. Conclusion
Animal research has provided critical insight into the mechanisms of action of current APDs and a framework to test hypotheses about the neurobiology underlying schizophrenia. The failure of recent novel target drugs does not invalidate the theory behind their development, but highlights the gaps that exist in their translation. In particular, studies performed in the MAM model have demonstrated the importance of studying the effects of APDs on a system representative of patients. Their hyperdopaminergic pathology shows rapid onset of depolarization block to D2 antagonists and responds to D2 partial agonists through a manner distinct from depolarization block (Valenti et al., 2011; Sonnenschein et al., 2019). These findings are consistent with clinical results and not observed in normal rats. We also propose that therapeutic strategies should be directed on upstream factors that control presynaptic release of DA. However, the effects of prior APD treatment must be considered in the development of novel drugs. Most patients receive chronic treatment with APDs, resulting in long-lasting changes to the brain that are not represented in normal or drug-naïve rats. Results from the MAM model have suggested that this may be a significant factor in novel-target response, which was prevented due to persistent DA supersensitivity from prior D2 antagonist treatment (Gill et al., 2014). In contrast, evidence suggests that the partial agonist aripiprazole does not induce DA supersensitivity (Tadokoro et al., 2011). Thus, testing novel compounds on patients who are withdrawn from aripiprazole, drug-naïve, or confirmed to be non-compliant with their prior medication may be more effective strategies. Additionally, it is important to determine whether novel target compounds are effective in patients that are treatment resistant to current APDs. The MAM model also has shown that pharmacological methods can be used to prevent full emergence the pathology in adulthood (Du and Grace, 2013, 2016b, a). These findings suggest the potential of targeting glutamatergic dysfunction early in the disease to alleviate symptoms and potentially prevent conversion of psychosis. Animal models are best applied in collaboration with studies performed in patients to find parallels and test predictions. Representative animal models, informed by clinical studies, can then be used to investigate the contribution of variables that underlie patient heterogeneity. These findings must then be incorporated into the design of clinical trials of novel target compounds, or else promising targets may be abandoned despite great potential to improve treatment options for schizophrenia.
Failure of recent novel target antipsychotics reveals a critical gap in translational research.
Antipsychotics can have different effects depending on the state of the dopamine system.
Representative models may provide better predictive validity than normal rodents.
Acknowledgments
AAG receives funds from Lundbeck, Pfizer, Otsuka, Lilly, Roche, Asubio, Abbott, Autofony, Janssen, Alkermes, Newron.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
CONFLICT OF INTEREST
SFS declares no conflicts of interest.
REFERENCES
- Abi-Dargham A, Gil R, Krystal J, Baldwin RM, Seibyl JP, Bowers M, van Dyck CH, Charney DS, Innis RB, Laruelle M (1998) Increased striatal dopamine transmission in schizophrenia: confirmation in a second cohort. American Journal of Psychiatry 155:761–767. [DOI] [PubMed] [Google Scholar]
- Abi-Dargham A, Rodenhiser J, Printz D, Zea-Ponce Y, Gil R, Kegeles LS, Weiss R, Cooper TB, Mann JJ, Van Heertum RL (2000) Increased baseline occupancy of D2 receptors by dopamine in schizophrenia. Proceedings of the National Academy of Sciences 97:8104–8109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adams DH, Zhang L, Millen BA, Kinon BJ, Gomez J-C (2014) Pomaglumetad methionil (LY2140023 monohydrate) and aripiprazole in patients with schizophrenia: a phase 3, multicenter, double-blind comparison. Schizophrenia research and treatment 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adams DH, Kinon BJ, Baygani S, Millen BA, Velona I, Kollack-Walker S, Walling DP (2013) A long-term, phase 2, multicenter, randomized, open-label, comparative safety study of pomaglumetad methionil (LY2140023 monohydrate) versus atypical antipsychotic standard of care in patients with schizophrenia. BMC psychiatry 13:143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Agid O, Kapur S, Arenovich T, Zipursky RB (2003) Delayed-onset hypothesis of antipsychotic action: a hypothesis tested and rejected. Archives of general psychiatry 60:1228–1235. [DOI] [PubMed] [Google Scholar]
- Alberati D, Moreau J-L, Lengyel J, Hauser N, Mory R, Borroni E, Pinard E, Knoflach F, Schlotterbeck G, Hainzl D (2012) Glycine reuptake inhibitor RG1678: a pharmacologic characterization of an investigational agent for the treatment of schizophrenia. Neuropharmacology 62:1152–1161. [DOI] [PubMed] [Google Scholar]
- Arguello PA, Gogos JA (2006) Modeling madness in mice: one piece at a time. Neuron 52:179–196. [DOI] [PubMed] [Google Scholar]
- Arnt J, Skarsfeldt T (1998) Do novel antipsychotics have similar pharmacological characteristics? A review of the evidence. Neuropsychopharmacology 18:63. [DOI] [PubMed] [Google Scholar]
- Belujon P, Grace AA (2015) Regulation of dopamine system responsivity and its adaptive and pathological response to stress. Proceedings of the Royal Society of London B: Biological Sciences 282:20142516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benes FM (2000) Emerging principles of altered neural circuitry in schizophrenia. Brain Research Reviews 31:251–269. [DOI] [PubMed] [Google Scholar]
- Benes FM, Berretta S (2001) GABAergic interneurons: implications for understanding schizophrenia and bipolar disorder. Neuropsychopharmacology 25:1–27. [DOI] [PubMed] [Google Scholar]
- Berman RM, Marcus RN, Swanink R, McQuade RD, Carson WH, Corey-Lisle PK, Khan A (2007) The efficacy and safety of aripiprazole as adjunctive therapy in major depressive disorder: a multicenter, randomized, double-blind, placebo-controlled study. The Journal of clinical psychiatry 68:843–853. [DOI] [PubMed] [Google Scholar]
- Blaha CD, Lane RF (1987) Chronic treatment with classical and atypical antipsychotic drugs differentially decreases dopamine release in striatum and nucleus accumbens in vivo. Neuroscience letters 78:199–204. [DOI] [PubMed] [Google Scholar]
- Borgwardt SJ, Riecher-Rössler A, Dazzan P, Chitnis X, Aston J, Drewe M, Gschwandtner U, Haller S, Pflüger M, Rechsteiner E (2007) Regional gray matter volume abnormalities in the at risk mental state. Biological psychiatry 61:1148–1156. [DOI] [PubMed] [Google Scholar]
- Bortolozzi A, Diaz-Mataix L, Toth M, Celada P, Artigas F (2007) In vivo actions of aripiprazole on serotonergic and dopaminergic systems in rodent brain. Psychopharmacology 191:745–758. [DOI] [PubMed] [Google Scholar]
- Brown AS (2006) Prenatal infection as a risk factor for schizophrenia. Schizophrenia bulletin 32:200–202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buchanan RW, Javitt DC, Marder SR, Schooler NR, Gold JM, McMahon RP, Heresco-Levy U, Carpenter WT (2007) The Cognitive and Negative Symptoms in Schizophrenia Trial (CONSIST): the efficacy of glutamatergic agents for negative symptoms and cognitive impairments. American Journal of Psychiatry 164:1593–1602. [DOI] [PubMed] [Google Scholar]
- Bugarski-Kirola D, Wang A, Abi-Saab D, Blättler T (2014) A phase II/III trial of bitopertin monotherapy compared with placebo in patients with an acute exacerbation of schizophrenia–results from the CandleLyte study. European Neuropsychopharmacology 24:1024–1036. [DOI] [PubMed] [Google Scholar]
- Bugarski-Kirola D, Iwata N, Sameljak S, Reid C, Blaettler T, Millar L, Marques TR, Garibaldi G, Kapur S (2016) Efficacy and safety of adjunctive bitopertin versus placebo in patients with suboptimally controlled symptoms of schizophrenia treated with antipsychotics: results from three phase 3, randomised, double-blind, parallel-group, placebo-controlled, multicentre studies in the SearchLyte clinical trial programme. The Lancet Psychiatry 3:1115–1128. [DOI] [PubMed] [Google Scholar]
- Bugarski-Kirola D, Blaettler T, Arango C, Fleischhacker WW, Garibaldi G, Wang A, Dixon M, Bressan RA, Nasrallah H, Lawrie S (2017) Bitopertin in negative symptoms of schizophrenia—Results from the phase III FlashLyte and DayLyte studies. Biological psychiatry 82:8–16. [DOI] [PubMed] [Google Scholar]
- Bunney B, Grace A (1978) Acute and chronic haloperidol treatment: comparison of effects on nigral dopaminergic cell activity. Life sciences 23:1715–1727. [DOI] [PubMed] [Google Scholar]
- Bunney B, Walters J, Roth R, Aghajanian G (1973) Dopaminergic neurons: effect of antipsychotic drugs and amphetamine on single cell activity. The Journal of pharmacology and experimental therapeutics 185:560. [PubMed] [Google Scholar]
- Burris K, Molski T, Xu C, Ryan E, Tottori K, Kikuchi T, Yocca F, Molinoff P (2002) Aripiprazole, a novel antipsychotic, is a high-affinity partial agonist at human dopamine D2 receptors. The Journal of pharmacology and experimental therapeutics 302:381–389. [DOI] [PubMed] [Google Scholar]
- Canetta SE, Brown AS (2012) Prenatal infection, maternal immune activation, and risk for schizophrenia. Translational neuroscience 3:320–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carlsson A, Lindqvist M (1963) Effect of chlorpromazine or haloperidol on formation of 3-methoxytyramine and normetanephrine in mouse brain. Acta pharmacologica et toxicologica 20:140–144. [DOI] [PubMed] [Google Scholar]
- Carpenter WT, Koenig JI (2008) The evolution of drug development in schizophrenia: past issues and future opportunities. Neuropsychopharmacology 33:2061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cartmell J, Monn JA, Schoepp DD (1999) The metabotropic glutamate 2/3 receptor agonists LY354740 and LY379268 selectively attenuate phencyclidine versus d-amphetamine motor behaviors in rats. Journal of Pharmacology and Experimental Therapeutics 291:161–170. [PubMed] [Google Scholar]
- Cartmell J, Monn JA, Schoepp DD (2000) The mGlu2/3 receptor agonist LY379268 selectively blocks amphetamine ambulations and rearing. European journal of pharmacology 400:221–224. [DOI] [PubMed] [Google Scholar]
- Castner SA, Williams GV (2007) From vice to virtue: insights from sensitization in the nonhuman primate. Progress in Neuro-Psychopharmacology and Biological Psychiatry 31:1572–1592. [DOI] [PubMed] [Google Scholar]
- Chiodo LA, Bunney BS (1983) Typical and atypical neuroleptics: differential effects of chronic administration on the activity of A9 and A10 midbrain dopaminergic neurons. Journal of Neuroscience 3:1607–1619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chiodo LA, Bunney BS (1985) Possible mechanisms by which repeated clozapine administration differentially affects the activity of two subpopulations of midbrain dopamine neurons. Journal of Neuroscience 5:2539–2544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chouinard G, Jones BD (1980) Neuroleptic-induced supersensitivity psychosis: clinical and pharmacologic characteristics. Am J Psychiatry 137:16–21. [DOI] [PubMed] [Google Scholar]
- Chouinard G, Samaha A-N, Chouinard V-A, Peretti C-S, Kanahara N, Takase M, Iyo M (2017) Antipsychotic-Induced Dopamine Supersensitivity Psychosis: Pharmacology, Criteria, and Therapy. Psychotherapy and Psychosomatics 86:189–219. [DOI] [PubMed] [Google Scholar]
- Cohen MM, Jing D, Yang RR, Tottenham N, Lee FS, Casey B (2013) Early-life stress has persistent effects on amygdala function and development in mice and humans. Proceedings of the National Academy of Sciences 110:18274–18278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Conley RR, Buchanan RW (1997) Evaluation of treatment-resistant schizophrenia. Schizophrenia bulletin 23:663–674. [DOI] [PubMed] [Google Scholar]
- Conn PJ, Christopoulos A, Lindsley CW (2009) Allosteric modulators of GPCRs: a novel approach for the treatment of CNS disorders. Nature reviews Drug discovery 8:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corcoran C, Walker E, Huot R, Mittal V, Tessner K, Kestler L, Malaspina D (2003) The stress cascade and schizophrenia: etiology and onset. Schizophrenia bulletin 29:671–692. [DOI] [PubMed] [Google Scholar]
- Correll CU, Schenk EM (2008) Tardive dyskinesia and new antipsychotics. Current opinion in psychiatry 21:151–156. [DOI] [PubMed] [Google Scholar]
- Correll CU, Rubio JM, Kane JM (2018) What is the risk‐benefit ratio of long‐term antipsychotic treatment in people with schizophrenia? World Psychiatry 17:149–160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Correll CU, Frederickson AM, Kane JM, Manu P (2007) Does Antipsychotic Polypharmacy Increase the Risk for Metabolic Syndrome? Schizophr Res 89:91–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coyle JT, Tsai G, Goff D (2003) Converging evidence of NMDA receptor hypofunction in the pathophysiology of schizophrenia. Annals of the New York Academy of Sciences 1003:318–327. [DOI] [PubMed] [Google Scholar]
- Creese I, Iversen SD (1975) The pharmacological and anatomical substrates of the amphetamine response in the rat. Brain research 83:419–436. [DOI] [PubMed] [Google Scholar]
- Creese I, Burt DR, Snyder SH (1976) Dopamine receptor binding predicts clinical and pharmacological potencies of antischizophrenic drugs. Science 192:481–483. [DOI] [PubMed] [Google Scholar]
- Dahan L, Husum H, Mnie-Filali O, Arnt J, Hertel P, Haddjeri N (2009) Effects of bifeprunox and aripiprazole on rat serotonin and dopamine neuronal activity and anxiolytic behaviour. Journal of Psychopharmacology 23:177–189. [DOI] [PubMed] [Google Scholar]
- De Hert M, Detraux J, Van Winkel R, Yu W, Correll CU (2012) Metabolic and cardiovascular adverse effects associated with antipsychotic drugs. Nature Reviews Endocrinology 8:114. [DOI] [PubMed] [Google Scholar]
- Demjaha A, Murray RM, McGuire PK, Kapur S, Howes OD (2012) Dopamine synthesis capacity in patients with treatment-resistant schizophrenia. American Journal of Psychiatry 169:1203–1210. [DOI] [PubMed] [Google Scholar]
- Demjaha A, Egerton A, Murray RM, Kapur S, Howes OD, Stone JM, McGuire PK (2014) Antipsychotic treatment resistance in schizophrenia associated with elevated glutamate levels but normal dopamine function. Biological psychiatry 75:e11–e13. [DOI] [PubMed] [Google Scholar]
- Deutch AY, Tam S-Y, Freeman AS, Bowers MB Jr, Roth RH (1987) Mesolimbic and mesocortical dopamine activation induced by phencyclidine: contrasting pattern to striatal response. European journal of pharmacology 134:257–264. [DOI] [PubMed] [Google Scholar]
- Deutch AY, Moghaddam B, Innis RB, Krystal JH, Aghajanian GK, Bunney BS, Charney DS (1991) Mechanisms of action of atypical antipsychotic drugs: implications for novel therapeutic strategies for schizophrenia. Schizophrenia research 4:121–156. [DOI] [PubMed] [Google Scholar]
- Do KQ, Cuenod M, Hensch TK (2015) Targeting oxidative stress and aberrant critical period plasticity in the developmental trajectory to schizophrenia. Schizophrenia bulletin 41:835–846. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Downing AM, Kinon BJ, Millen BA, Zhang L, Liu L, Morozova MA, Brenner R, Rayle TJ, Nisenbaum L, Zhao F (2014) A double-blind, placebo-controlled comparator study of LY2140023 monohydrate in patients with schizophrenia. BMC psychiatry 14:351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Du Y, Grace AA (2013) Peripubertal diazepam administration prevents the emergence of dopamine system hyperresponsivity in the MAM developmental disruption model of schizophrenia. Neuropsychopharmacology 38:1881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Du Y, Grace AA (2016a) Amygdala hyperactivity in MAM model of schizophrenia is normalized by peripubertal diazepam administration. Neuropsychopharmacology 41:2455–2462. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Du Y, Grace AA (2016b) Loss of parvalbumin in the hippocampus of MAM schizophrenia model rats is attenuated by peripubertal diazepam. International Journal of Neuropsychopharmacology 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dunlop J, Brandon NJ (2015) Schizophrenia drug discovery and development in an evolving era: are new drug targets fulfilling expectations? Journal of Psychopharmacology 29:230–238. [DOI] [PubMed] [Google Scholar]
- Emsley R, Chiliza B, Asmal L, Harvey BH (2013) The nature of relapse in schizophrenia. BMC psychiatry 13:50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Evins AE, Fitzgerald SM, Wine L, Rosselli R, Goff DC (2000) Placebo-controlled trial of glycine added to clozapine in schizophrenia. American Journal of Psychiatry 157:826–828. [DOI] [PubMed] [Google Scholar]
- Evins AE, Amico E, Posever TA, Toker R, Goff DC (2002) D-Cycloserine added to risperidone in patients with primary negative symptoms of schizophrenia. Schizophrenia Research 56:19–23. [DOI] [PubMed] [Google Scholar]
- Farde L, Nordström A-L, Wiesel F-A, Pauli S, Halldin C, Sedvall G (1992) Positron emission tomographic analysis of central D1 and D2 dopamine receptor occupancy in patients treated with classical neuroleptics and clozapine: relation to extrapyramidal side effects. Archives of general psychiatry 49:538–544. [DOI] [PubMed] [Google Scholar]
- Faries D, Ascher-Svanum H, Zhu B, Correll C, Kane J (2005) Antipsychotic monotherapy and polypharmacy in the naturalistic treatment of schizophrenia with atypical antipsychotics. Bmc Psychiatry 5:26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fell MJ, McKinzie DL, Monn JA, Svensson KA (2012) Group II metabotropic glutamate receptor agonists and positive allosteric modulators as novel treatments for schizophrenia. Neuropharmacology 62:1473–1483. [DOI] [PubMed] [Google Scholar]
- Flagstad P, Mørk A, Glenthøj BY, Van Beek J, Michael-Titus AT, Didriksen M (2004) Disruption of neurogenesis on gestational day 17 in the rat causes behavioral changes relevant to positive and negative schizophrenia symptoms and alters amphetamine-induced dopamine release in nucleus accumbens. Neuropsychopharmacology 29:2052. [DOI] [PubMed] [Google Scholar]
- Fleischhacker WW, Uchida H (2014) Critical review of antipsychotic polypharmacy in the treatment of schizophrenia. International Journal of Neuropsychopharmacology 17:1083–1093. [DOI] [PubMed] [Google Scholar]
- Floresco SB, Todd CL, Grace AA (2001) Glutamatergic afferents from the hippocampus to the nucleus accumbens regulate activity of ventral tegmental area dopamine neurons. Journal of Neuroscience 21:4915–4922. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Floresco SB, West AR, Ash B, Moore H, Grace AA (2003) Afferent modulation of dopamine neuron firing differentially regulates tonic and phasic dopamine transmission. Nature neuroscience 6:968. [DOI] [PubMed] [Google Scholar]
- Fraley ME (2009) Positive allosteric modulators of the metabotropic glutamate receptor 2 for the treatment of schizophrenia. Expert opinion on therapeutic patents 19:1259–1275. [DOI] [PubMed] [Google Scholar]
- Gastambide F, Cotel M-C, Gilmour G, O’neill MJ, Robbins TW, Tricklebank MD (2012) Selective remediation of reversal learning deficits in the neurodevelopmental MAM model of schizophrenia by a novel mGlu5 positive allosteric modulator. Neuropsychopharmacology 37:1057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gill KM, Grace AA (2014) Corresponding decrease in neuronal markers signals progressive parvalbumin neuron loss in MAM schizophrenia model. International Journal of Neuropsychopharmacology 17:1609–1619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gill KM, Cook JM, Poe MM, Grace AA (2014) Prior antipsychotic drug treatment prevents response to novel antipsychotic agent in the methylazoxymethanol acetate model of schizophrenia. Schizophrenia bulletin 40:341–350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gill KM, Lodge DJ, Cook JM, Aras S, Grace AA (2011) A novel α5GABAAR-positive allosteric modulator reverses hyperactivation of the dopamine system in the MAM model of schizophrenia. Neuropsychopharmacology 36:1903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goff DC (2014) Bitopertin: the good news and bad news. JAMA psychiatry 71:621–622. [DOI] [PubMed] [Google Scholar]
- Goff DC, Herz L, Posever T, Shih V, Tsai G, Henderson DC, Freudenreich O, Evins AE, Yovel I, Zhang H (2005) A six-month, placebo-controlled trial of D-cycloserine co-administered with conventional antipsychotics in schizophrenia patients. Psychopharmacology 179:144–150. [DOI] [PubMed] [Google Scholar]
- Goldstein JM, Litwin LC, Sutton EB, Malick JB (1993) Seroquel: electrophysiological profile of a potential atypical antipsychotic. Psychopharmacology 112:293–298. [DOI] [PubMed] [Google Scholar]
- Gomes FV, Grace AA (2016) Prefrontal cortex dysfunction increases susceptibility to schizophrenia-like changes induced by adolescent stress exposure. Schizophrenia bulletin 43:592–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gomes FV, Grace AA (2017) Adolescent stress as a driving factor for schizophrenia development—a basic science perspective. Schizophrenia bulletin 43:486–489. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gomes FV, Rincón-Cortés M, Grace AA (2016) Adolescence as a period of vulnerability and intervention in schizophrenia: insights from the MAM model. Neuroscience & Biobehavioral Reviews 70:260–270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grace A (1991) Phasic versus tonic dopamine release and the modulation of dopamine system responsivity: a hypothesis for the etiology of schizophrenia. Neuroscience 41:1–24. [DOI] [PubMed] [Google Scholar]
- Grace A, Bunney B (1986) Induction of depolarization block in midbrain dopamine neurons by repeated administration of haloperidol: analysis using in vivo intracellular recording. Journal of Pharmacology and Experimental Therapeutics 238:1092–1100. [PubMed] [Google Scholar]
- Grace AA (2016) Dysregulation of the dopamine system in the pathophysiology of schizophrenia and depression. Nature reviews Neuroscience 17:524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grace AA, Bunney BS (1984) The control of firing pattern in nigral dopamine neurons: burst firing. Journal of neuroscience 4:2877–2890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grace AA, Gomes FV (2018) The circuitry of dopamine system regulation and its disruption in schizophrenia: insights into treatment and prevention. Schizophrenia bulletin. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grace AA, Bunney BS, Moore H, Todd CL (1997) Dopamine-cell depolarization block as a model for the therapeutic actions of antipsychotic drugs. Trends in neurosciences 20:31–37. [DOI] [PubMed] [Google Scholar]
- Grace AA, Floresco SB, Goto Y, Lodge DJ (2007) Regulation of firing of dopaminergic neurons and control of goal-directed behaviors. Trends in neurosciences 30:220–227. [DOI] [PubMed] [Google Scholar]
- Green MF, Kern RS, Braff DL, Mintz J (2000) Neurocognitive deficits and functional outcome in schizophrenia: are we measuring the “right stuff”? Schizophrenia bulletin 26:119–136. [DOI] [PubMed] [Google Scholar]
- Greenslade RG, Mitchell SN (2004) Selective action of (–)-2-oxa-4-aminobicyclo [3.1. 0] hexane-4, 6-dicarboxylate (LY379268), a group II metabotropic glutamate receptor agonist, on basal and phencyclidine-induced dopamine release in the nucleus accumbens shell. Neuropharmacology 47:1–8. [DOI] [PubMed] [Google Scholar]
- Harlow HF, Dodsworth RO, Harlow MK (1965) Total social isolation in monkeys. Proceedings of the National Academy of Sciences 54:90–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haro JM, Salvador-Carulla L (2006) The SOHO (Schizophrenia Outpatient Health Outcome) Study. CNS drugs 20:293–301. [DOI] [PubMed] [Google Scholar]
- Harrison PJ (2004) The hippocampus in schizophrenia: a review of the neuropathological evidence and its pathophysiological implications. Psychopharmacology 174:151–162. [DOI] [PubMed] [Google Scholar]
- Harvey PD, Koren D, Reichenberg A, Bowie CR (2005) Negative symptoms and cognitive deficits: what is the nature of their relationship? Schizophrenia bulletin 32:250–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heckers S, Rauch S, Goff D, Savage C, Schacter D, Fischman A, Alpert N (1998) Impaired recruitment of the hippocampus during conscious recollection in schizophrenia. Nature neuroscience 1:318. [DOI] [PubMed] [Google Scholar]
- Heilbronner SR, Rodriguez-Romaguera J, Quirk GJ, Groenewegen HJ, Haber SN (2016) Circuit-based corticostriatal homologies between rat and primate. Biological psychiatry 80:509–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heim C, Nemeroff CB (2001) The role of childhood trauma in the neurobiology of mood and anxiety disorders: preclinical and clinical studies. Biological psychiatry 49:1023–1039. [DOI] [PubMed] [Google Scholar]
- Heresco-Levy U, Ermilov M, Lichtenberg P, Bar G, Javitt DC (2004) High-dose glycine added to olanzapine and risperidone for the treatment of schizophrenia. Biological psychiatry 55:165–171. [DOI] [PubMed] [Google Scholar]
- Heresco-Levy U, Javitt DC, Ermilov M, Mordel C, Horowitz A, Kelly D (1996) Double-blind, placebo-controlled, crossover trial of glycine adjuvant therapy for treatment-resistant schizophrenia. The British Journal of Psychiatry 169:610–617. [DOI] [PubMed] [Google Scholar]
- Heresco-Levy U, Javitt DC, Ermilov M, Mordel C, Silipo G, Lichtenstein M (1999) Efficacy of high-dose glycine in the treatment of enduring negative symptoms of schizophrenia. Archives of general psychiatry 56:29–36. [DOI] [PubMed] [Google Scholar]
- Heresco-Levy U, Ermilov M, Shimoni J, Shapira B, Silipo G, Javitt DC (2002) Placebo-controlled trial of D-cycloserine added to conventional neuroleptics, olanzapine, or risperidone in schizophrenia. American Journal of Psychiatry 159:480–482. [DOI] [PubMed] [Google Scholar]
- Heresco-Levy U, Javitt DC, Ebstein R, Vass A, Lichtenberg P, Bar G, Catinari S, Ermilov M (2005) D-serine efficacy as add-on pharmacotherapy to risperidone and olanzapine for treatment-refractory schizophrenia. Biological psychiatry 57:577–585. [DOI] [PubMed] [Google Scholar]
- Ho B-C, Andreasen NC, Ziebell S, Pierson R, Magnotta V (2011) Long-term antipsychotic treatment and brain volumes: a longitudinal study of first-episode schizophrenia. Archives of general psychiatry 68:128–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holt DJ, Kunkel L, Weiss AP, Goff DC, Wright CI, Shin LM, Rauch SL, Hootnick J, Heckers S (2006) Increased medial temporal lobe activation during the passive viewing of emotional and neutral facial expressions in schizophrenia. Schizophrenia research 82:153–162. [DOI] [PubMed] [Google Scholar]
- Holtzman C, Trotman H, Goulding S, Ryan A, Macdonald A, Shapiro D, Brasfield J, Walker E (2013) Stress and neurodevelopmental processes in the emergence of psychosis. Neuroscience 249:172–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Homayoun H, Moghaddam B (2007) NMDA receptor hypofunction produces opposite effects on prefrontal cortex interneurons and pyramidal neurons. Journal of Neuroscience 27:11496–11500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Homayoun H, Jackson ME, Moghaddam B (2005) Activation of metabotropic glutamate 2/3 receptors reverses the effects of NMDA receptor hypofunction on prefrontal cortex unit activity in awake rats. Journal of neurophysiology 93:1989–2001. [DOI] [PubMed] [Google Scholar]
- Howes O, Bose S, Turkheimer F, Valli I, Egerton A, Stahl D, Valmaggia L, Allen P, Murray R, McGuire P (2011a) Progressive increase in striatal dopamine synthesis capacity as patients develop psychosis: a PET study. Molecular psychiatry 16:885. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howes OD, Kapur S (2014) A neurobiological hypothesis for the classification of schizophrenia: type A (hyperdopaminergic) and type B (normodopaminergic). The British Journal of Psychiatry 205:1–3. [DOI] [PubMed] [Google Scholar]
- Howes OD, McCutcheon R, Owen MJ, Murray RM (2017) The role of genes, stress, and dopamine in the development of schizophrenia. Biological psychiatry 81:9–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howes OD, Kambeitz J, Kim E, Stahl D, Slifstein M, Abi-Dargham A, Kapur S (2012) The nature of dopamine dysfunction in schizophrenia and what this means for treatment: meta-analysis of imaging studies. Archives of general psychiatry 69:776–786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howes OD, Bose SK, Turkheimer F, Valli I, Egerton A, Valmaggia LR, Murray RM, McGuire P (2011b) Dopamine synthesis capacity before onset of psychosis: a prospective [18F]-DOPA PET imaging study. American Journal of Psychiatry 168:1311–1317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howes OD, Montgomery AJ, Asselin M-C, Murray RM, Valli I, Tabraham P, Bramon-Bosch E, Valmaggia L, Johns L, Broome M (2009) Elevated striatal dopamine function linked to prodromal signs of schizophrenia. Archives of general psychiatry 66:13–20. [DOI] [PubMed] [Google Scholar]
- Ichikawa J, Meltzer HY (1991) Differential effects of repeated treatment with haloperidol and clozapine on dopamine release and metabolism in the striatum and the nucleus accumbens. Journal of Pharmacology and Experimental Therapeutics 256:348–357. [PubMed] [Google Scholar]
- Inoue A, Miki S, Seto M, Kikuchi T, Morita S, Ueda H, Misu Y, Nakata Y (1997) Aripiprazole, a novel antipsychotic drug, inhibits quinpriole-evoked GTPase activity but does not up-regulate dopamine D 2 receptor following repeated treatment in the rat striatum. European journal of pharmacology 321:105–111. [DOI] [PubMed] [Google Scholar]
- J Marek G (2015) When is a Proof-of-Concept (POC) not a POC? Pomaglumetad (LY2140023) as a Case Study for Antipsychotic Efficacy. Current pharmaceutical design 21:3788–3796. [DOI] [PubMed] [Google Scholar]
- Javitt DC (2012) Glycine transport inhibitors in the treatment of schizophrenia In: Novel Antischizophrenia Treatments, pp 367–399: Springer. [DOI] [PubMed] [Google Scholar]
- Javitt DC, Silipo G, Cienfuegos A, Shelley A-M, Bark N, Park M, Lindenmayer J-P, Suckow R, Zukin SR (2001) Adjunctive high-dose glycine in the treatment of schizophrenia. International Journal of Neuropsychopharmacology 4:385–391. [DOI] [PubMed] [Google Scholar]
- Johnston M, Carman A, Coyle J (1981) Effects of fetal treatment with methylazoxymethanol acetate at various gestational dates on the neurochemistry of the adult neocortex of the rat. Journal of neurochemistry 36:124–128. [DOI] [PubMed] [Google Scholar]
- Jordan S, Koprivica V, Dunn R, Tottori K, Kikuchi T, Altar CA (2004) In vivo effects of aripiprazole on cortical and striatal dopaminergic and serotonergic function. European journal of pharmacology 483:45–53. [DOI] [PubMed] [Google Scholar]
- Kane J, Honigfeld G, Singer J, Meltzer H (1988) Clozapine for the treatment-resistant schizophrenic: a double-blind comparison with chlorpromazine. Archives of general psychiatry 45:789–796. [DOI] [PubMed] [Google Scholar]
- Kane JM (1989) The current status of neuroleptic therapy. The Journal of clinical psychiatry. [PubMed] [Google Scholar]
- Kane JM, Fleischhacker WW, Hansen L, Perlis R, Pikalov A III, Assunção-Talbott S (2009) Akathisia: an updated review focusing on second-generation antipsychotics. Journal of Clinical Psychiatry 70:627. [DOI] [PubMed] [Google Scholar]
- Kapur S (2003) Psychosis as a state of aberrant salience: a framework linking biology, phenomenology, and pharmacology in schizophrenia. American journal of Psychiatry 160:13–23. [DOI] [PubMed] [Google Scholar]
- Kapur S, Seeman P (2001) Does fast dissociation from the dopamine D2 receptor explain the action of atypical antipsychotics?: A new hypothesis. American Journal of Psychiatry 158:360–369. [DOI] [PubMed] [Google Scholar]
- Kapur S, Remington G (2001) Dopamine D 2 receptors and their role in atypical antipsychotic action: still necessary and may even be sufficient. Biological psychiatry 50:873–883. [DOI] [PubMed] [Google Scholar]
- Kapur S, VanderSpek SC, Brownlee BA, Nobrega JN (2003) Antipsychotic dosing in preclinical models is often unrepresentative of the clinical condition: a suggested solution based on in vivo occupancy. Journal of Pharmacology and Experimental Therapeutics 305:625–631. [DOI] [PubMed] [Google Scholar]
- Kapur S, Zipursky R, Jones C, Remington G, Houle S (2000) Relationship between dopamine D2 occupancy, clinical response, and side effects: a double-blind PET study of first-episode schizophrenia. American Journal of Psychiatry 157:514–520. [DOI] [PubMed] [Google Scholar]
- Kapur S, Arenovich T, Agid O, Zipursky R, Lindborg S, Jones B (2005) Evidence for onset of antipsychotic effects within the first 24 hours of treatment. American Journal of Psychiatry 162:939–946. [DOI] [PubMed] [Google Scholar]
- Kegeles LS, Abi-Dargham A, Zea-Ponce Y, Rodenhiser-Hill J, Mann JJ, Van Heertum RL, Cooper TB, Carlsson A, Laruelle M (2000) Modulation of amphetamine-induced striatal dopamine release by ketamine in humans: implications for schizophrenia. Biological psychiatry 48:627–640. [DOI] [PubMed] [Google Scholar]
- Kegeles LS, Abi-Dargham A, Frankle WG, Gil R, Cooper TB, Slifstein M, Hwang D-R, Huang Y, Haber SN, Laruelle M (2010) Increased synaptic dopamine function in associative regions of the striatum in schizophrenia. Archives of general psychiatry 67:231–239. [DOI] [PubMed] [Google Scholar]
- Kesby J, Eyles D, McGrath J, Scott J (2018) Dopamine, psychosis and schizophrenia: the widening gap between basic and clinical neuroscience. Translational psychiatry 8:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kikuchi T, Tottori K, Uwahodo Y, Hirose T, Miwa T, Oshiro Y, Morita S (1995) 7-(4-[4-(2, 3-Dichlorophenyl)-1-piperazinyl] butyloxy)-3, 4-dihydro-2 (1H)-quinolinone (OPC-14597), a new putative antipsychotic drug with both presynaptic dopamine autoreceptor agonistic activity and postsynaptic D2 receptor antagonistic activity. Journal of Pharmacology and Experimental Therapeutics 274:329–336. [PubMed] [Google Scholar]
- Kinon BJ, Millen BA, Zhang L, McKinzie DL (2015) Exploratory Analysis for a Targeted Patient Population Responsive to the Metabotropic Glutamate 2/3 Receptor Agonist Pomaglumetad Methionil in Schizophrenia. Biological Psychiatry 78:754–762. [DOI] [PubMed] [Google Scholar]
- Kishimoto T, Robenzadeh A, Leucht C, Leucht S, Watanabe K, Mimura M, Borenstein M, Kane JM, Correll CU (2012) Long-acting injectable vs oral antipsychotics for relapse prevention in schizophrenia: a meta-analysis of randomized trials. Schizophrenia bulletin 40:192–213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kraguljac NV, White DM, Reid MA, Lahti AC (2013) Increased hippocampal glutamate and volumetric deficits in unmedicated patients with schizophrenia. JAMA psychiatry 70:1294–1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krystal JH, Anticevic A (2015) Toward illness phase–specific pharmacotherapy for schizophrenia. Biological psychiatry 78:738–740. [DOI] [PubMed] [Google Scholar]
- Krystal JH, Karper LP, Seibyl JP, Freeman GK, Delaney R, Bremner JD, Heninger GR, Bowers MB, Charney DS (1994) Subanesthetic effects of the noncompetitive NMDA antagonist, ketamine, in humans: psychotomimetic, perceptual, cognitive, and neuroendocrine responses. Archives of general psychiatry 51:199–214. [DOI] [PubMed] [Google Scholar]
- Kurita M, Holloway T, García-Bea A, Kozlenkov A, Friedman AK, Moreno JL, Heshmati M, Golden SA, Kennedy PJ, Takahashi N (2012) HDAC2 regulates atypical antipsychotic responses through the modulation of mGlu2 promoter activity. Nature neuroscience 15:1245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lahti AC, Koffel B, LaPorte D, Tamminga CA (1995) Subanesthetic doses of ketamine stimulate psychosis in schizophrenia. Neuropsychopharmacology 13:9. [DOI] [PubMed] [Google Scholar]
- Lane RF, Blaha CD (1987) Chronic haloperidol decreases dopamine release in striatum and nucleus accumbens in vivo: depolarization block as a possible mechanism of action. Brain research bulletin 18:135–138. [DOI] [PubMed] [Google Scholar]
- Lannes B, Micheletti G, Warter J-M, Kempf E, Di Scala G (1991) Behavioural, pharmacological and biochemical effects of acute and chronic administration of ketamine in the rat. Neuroscience letters 128:177–181. [DOI] [PubMed] [Google Scholar]
- Laruelle M, Abi-Dargham A (1999) Dopamine as the wind of the psychotic fire: new evidence from brain imaging studies. Journal of Psychopharmacology 13:358–371. [DOI] [PubMed] [Google Scholar]
- Laruelle M, Abi-Dargham A, Gil R, Kegeles L, Innis R (1999) Increased dopamine transmission in schizophrenia: relationship to illness phases. Biological psychiatry 46:56–72. [DOI] [PubMed] [Google Scholar]
- Laruelle M, Abi-Dargham A, Van Dyck CH, Gil R, D’Souza CD, Erdos J, McCance E, Rosenblatt W, Fingado C, Zoghbi SS (1996) Single photon emission computerized tomography imaging of amphetamine-induced dopamine release in drug-free schizophrenic subjects. Proceedings of the National Academy of Sciences 93:9235–9240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lavreysen H, Langlois X, Ahnaou A, Drinkenburg W, te Riele P, Biesmans I, Van der Linden I, Peeters L, Megens A, Wintmolders C (2013) Pharmacological characterization of JNJ-40068782, a new potent, selective, and systemically active positive allosteric modulator of the mGlu2 receptor and its radioligand [3H] JNJ-40068782. Journal of Pharmacology and Experimental Therapeutics 346:514–527. [DOI] [PubMed] [Google Scholar]
- Lawler CP, Prioleau C, Lewis MM, Mak C, Jiang D, Schetz JA, Gonzalez AM, Sibley DR, Mailman RB (1999) Interactions of the novel antipsychotic aripiprazole (OPC-14597) with dopamine and serotonin receptor subtypes. Neuropsychopharmacology 20:612–627. [DOI] [PubMed] [Google Scholar]
- Legault M, Wise RA (1999) Injections of N-methyl-D-aspartate into the ventral hippocampus increase extracellular dopamine in the ventral tegmental area and nucleus accumbens. Synapse 31:241–249. [DOI] [PubMed] [Google Scholar]
- Leucht C, Heres S, Kane JM, Kissling W, Davis JM, Leucht S (2011) Oral versus depot antipsychotic drugs for schizophrenia—a critical systematic review and meta-analysis of randomised long-term trials. Schizophrenia research 127:83–92. [DOI] [PubMed] [Google Scholar]
- Leucht S, Busch R, Hamann J, Kissling W, Kane JM (2005) Early-onset hypothesis of antipsychotic drug action: a hypothesis tested, confirmed and extended. Biological psychiatry 57:1543–1549. [DOI] [PubMed] [Google Scholar]
- Leucht S, Corves C, Arbter D, Engel RR, Li C, Davis JM (2009) Second-generation versus first-generation antipsychotic drugs for schizophrenia: a meta-analysis. The Lancet 373:31–41. [DOI] [PubMed] [Google Scholar]
- Leucht S, Tardy M, Komossa K, Heres S, Kissling W, Salanti G, Davis JM (2012) Antipsychotic drugs versus placebo for relapse prevention in schizophrenia: a systematic review and meta-analysis. The Lancet 379:2063–2071. [DOI] [PubMed] [Google Scholar]
- Leucht S, Cipriani A, Spineli L, Mavridis D, Örey D, Richter F, Samara M, Barbui C, Engel RR, Geddes JR (2013) Comparative efficacy and tolerability of 15 antipsychotic drugs in schizophrenia: a multiple-treatments meta-analysis. The Lancet 382:951–962. [DOI] [PubMed] [Google Scholar]
- Lewis DA, Levitt P (2002) Schizophrenia as a disorder of neurodevelopment. Annual review of neuroscience 25:409–432. [DOI] [PubMed] [Google Scholar]
- Li Z, Ichikawa J, Dai J, Meltzer HY (2004) Aripiprazole, a novel antipsychotic drug, preferentially increases dopamine release in the prefrontal cortex and hippocampus in rat brain. European journal of pharmacology 493:75–83. [DOI] [PubMed] [Google Scholar]
- Lieberman J, Chakos M, Wu H, Alvir J, Hoffman E, Robinson D, Bilder R (2001) Longitudinal study of brain morphology in first episode schizophrenia. Biological psychiatry 49:487–499. [DOI] [PubMed] [Google Scholar]
- Lieberman JA, Safferman AZ, Pollack S, Szymanski S, Johns C, Howard A, Kronig M, Bookstein P, Kane JM (1994) Clinical effects of clozapine in chronic schizophrenia: response to treatment and predictors of outcome. American Journal of Psychiatry 151:1744–1752. [DOI] [PubMed] [Google Scholar]
- Lieberman JA, Stroup TS, McEvoy JP, Swartz MS, Rosenheck RA, Perkins DO, Keefe RS, Davis SM, Davis CE, Lebowitz BD (2005) Effectiveness of antipsychotic drugs in patients with chronic schizophrenia. N Engl j Med 2005:1209–1223. [DOI] [PubMed] [Google Scholar]
- Lipska BK, Weinberger DR (2000) To model a psychiatric disorder in animals: schizophrenia as a reality test. Neuropsychopharmacology 23:223–239. [DOI] [PubMed] [Google Scholar]
- Lipska BK, Jaskiw GE, Weinberger DR (1993) Postpubertal emergence of hyperresponsiveness to stress and to amphetamine after neonatal excitotoxic hippocampal damage: a potential animal model of schizophrenia. Neuropsychopharmacology 9:67. [DOI] [PubMed] [Google Scholar]
- Lipska BK, Jaskiw GE, Chrapusta S, Karoum F, Weinberger DR (1992) Ibotenic acid lesion of the ventral hippocampus differentially affects dopamine and its metabolites in the nucleus accumbens and prefrontal cortex in the rat. Brain research 585:1–6. [DOI] [PubMed] [Google Scholar]
- Lisman JE, Coyle JT, Green RW, Javitt DC, Benes FM, Heckers S, Grace AA (2008) Circuit-based framework for understanding neurotransmitter and risk gene interactions in schizophrenia. Trends in neurosciences 31:234–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Litman RE, Smith MA, Doherty JJ, Cross A, Raines S, Gertsik L, Zukin SR (2016) AZD8529, a positive allosteric modulator at the mGluR2 receptor, does not improve symptoms in schizophrenia: A proof of principle study. Schizophrenia research 172:152–157. [DOI] [PubMed] [Google Scholar]
- Lodge D, Grace A (2006a) The laterodorsal tegmentum is essential for burst firing of ventral tegmental area dopamine neurons. Proceedings of the National Academy of Sciences of the United States of America 103:5167–5172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lodge DJ, Grace AA (2006b) The hippocampus modulates dopamine neuron responsivity by regulating the intensity of phasic neuron activation. Neuropsychopharmacology 31:1356. [DOI] [PubMed] [Google Scholar]
- Lodge DJ, Grace AA (2007) Aberrant hippocampal activity underlies the dopamine dysregulation in an animal model of schizophrenia. Journal of Neuroscience 27:11424–11430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lodge DJ, Grace AA (2011) Hippocampal dysregulation of dopamine system function and the pathophysiology of schizophrenia. Trends in pharmacological sciences 32:507–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- López-Muñoz F, Alamo C, Cuenca E, Shen WW, Clervoy P, Rubio G (2005) History of the discovery and clinical introduction of chlorpromazine. Annals of Clinical Psychiatry 17:113–135. [DOI] [PubMed] [Google Scholar]
- Lorrain D, Baccei C, Bristow L, Anderson J, Varney M (2003) Effects of ketamine and n-methyl-d-aspartate on glutamate and dopamine release in the rat prefrontal cortex: modulation by a group II selective metabotropic glutamate receptor agonist LY379268. Neuroscience 3:697–706. [DOI] [PubMed] [Google Scholar]
- Lupien SJ, McEwen BS, Gunnar MR, Heim C (2009) Effects of stress throughout the lifespan on the brain, behaviour and cognition. Nature reviews neuroscience 10:434. [DOI] [PubMed] [Google Scholar]
- Maeda K, Sugino H, Akazawa H, Amada N, Shimada J, Futamura T, Yamashita H, Ito N, McQuade RD, Mørk A (2014) Brexpiprazole I: in vitro and in vivo characterization of a novel serotonin-dopamine activity modulator. Journal of Pharmacology and Experimental Therapeutics 350:589–604. [DOI] [PubMed] [Google Scholar]
- Mailman RB, Gay EA (2004) Novel mechanisms of drug action: functional selectivity at D2 dopamine receptors. Medicinal chemistry research 13:115–126. [Google Scholar]
- Mamo D, Graff A, Mizrahi R, Shammi CM, Romeyer F, Kapur S (2007) Differential effects of aripiprazole on D 2, 5-HT 2, and 5-HT 1A receptor occupancy in patients with schizophrenia: a triple tracer PET study. American Journal of Psychiatry 164:1411–1417. [DOI] [PubMed] [Google Scholar]
- Meaney M, Diorio J, Francis D, Widdowson J, LaPlante P, Caldji C, Sharma S, Seckl J, Plotsky P (1996) Early environmental regulation of forebrain glucocorticoid receptor gene expression: implications for adrenocortical responses to stress. Developmental neuroscience 18:49–72. [DOI] [PubMed] [Google Scholar]
- Medoff DR, Holcomb HH, Lahti AC, Tamminga CA (2001) Probing the human hippocampus using rCBF: contrasts in schizophrenia. Hippocampus 11:543–550. [DOI] [PubMed] [Google Scholar]
- Melis M, Mereu G, Lilliu V, Quartu M, Diana M, Gessa GL (1998) Haloperidol does not produce dopamine cell depolarization-block in paralyzed, unanesthetized rats. Brain research 783:127–132. [DOI] [PubMed] [Google Scholar]
- Meltzer HY, Bastani B, Kwon KY, Ramirez L, Burnett S, Sharpe J (1989) A prospective study of clozapine in treatment-resistant schizophrenic patients. Psychopharmacology 99:S68–S72. [DOI] [PubMed] [Google Scholar]
- Mereu G, Lilliu V, Vargiu P, Muntoni AL, Diana M, Gessa GL (1995) Depolarization inactivation of dopamine neurons: an artifact? Journal of Neuroscience 15:1144–1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meyer U, Feldon J (2010) Epidemiology-driven neurodevelopmental animal models of schizophrenia. Progress in neurobiology 90:285–326. [DOI] [PubMed] [Google Scholar]
- Mezler M, Geneste H, Gault L, Marek GJ (2010) LY-2140023, a prodrug of the group II metabotropic glutamate receptor agonist LY-404039 for the potential treatment of schizophrenia. Curr Opin Investig Drugs 11:833–845. [PubMed] [Google Scholar]
- Milev P, Ho B-C, Arndt S, Andreasen NC (2005) Predictive values of neurocognition and negative symptoms on functional outcome in schizophrenia: a longitudinal first-episode study with 7-year follow-up. American Journal of Psychiatry 162:495–506. [DOI] [PubMed] [Google Scholar]
- Miyamoto S, Duncan G, Marx C, Lieberman J (2005) Treatments for schizophrenia: a critical review of pharmacology and mechanisms of action of antipsychotic drugs. Molecular psychiatry 10:79. [DOI] [PubMed] [Google Scholar]
- Mizrahi R, Addington J, Rusjan PM, Suridjan I, Ng A, Boileau I, Pruessner JC, Remington G, Houle S, Wilson AA (2012) Increased stress-induced dopamine release in psychosis. Biological psychiatry 71:561–567. [DOI] [PubMed] [Google Scholar]
- Modinos G, Allen P, Grace AA, McGuire P (2015) Translating the MAM model of psychosis to humans. Trends in neurosciences 38:129–138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moghaddam B, Bunney BS (1993) Depolarization inactivation of dopamine neurons: terminal release characteristics. Synapse 14:195–200. [DOI] [PubMed] [Google Scholar]
- Moghaddam B, Adams BW (1998) Reversal of phencyclidine effects by a group II metabotropic glutamate receptor agonist in rats. Science 281:1349–1352. [DOI] [PubMed] [Google Scholar]
- Moghaddam B, Adams B, Verma A, Daly D (1997) Activation of glutamatergic neurotransmission by ketamine: a novel step in the pathway from NMDA receptor blockade to dopaminergic and cognitive disruptions associated with the prefrontal cortex. Journal of Neuroscience 17:2921–2927. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moore H, Todd CL, Grace AA (1998) Striatal extracellular dopamine levels in rats with haloperidol-induced depolarization block of substantia nigra dopamine neurons. Journal of Neuroscience 18:5068–5077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moore H, Jentsch JD, Ghajarnia M, Geyer MA, Grace AA (2006) A neurobehavioral systems analysis of adult rats exposed to methylazoxymethanol acetate on E17: implications for the neuropathology of schizophrenia. Biological psychiatry 60:253–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mothet J-P, Parent AT, Wolosker H, Brady RO, Linden DJ, Ferris CD, Rogawski MA, Snyder SH (2000) D-serine is an endogenous ligand for the glycine site of the N-methyl-D-aspartate receptor. Proceedings of the National Academy of Sciences 97:4926–4931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murray RM, Quattrone D, Natesan S, van Os J, Nordentoft M, Howes O, Di Forti M, Taylor D (2016) Should psychiatrists be more cautious about the long-term prophylactic use of antipsychotics? The British Journal of Psychiatry 209:361–365. [DOI] [PubMed] [Google Scholar]
- Nestler EJ, Hyman SE (2010) Animal models of neuropsychiatric disorders. Nature neuroscience 13:1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nicoletti F, Bockaert J, Collingridge G, Conn P, Ferraguti F, Schoepp D, Wroblewski J, Pin J (2011) Metabotropic glutamate receptors: from the workbench to the bedside. Neuropharmacology 60:1017–1041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nutt DJ, Lingford-Hughes A, Erritzoe D, Stokes PR (2015) The dopamine theory of addiction: 40 years of highs and lows. Nature Reviews Neuroscience 16:305. [DOI] [PubMed] [Google Scholar]
- O’Donnell P (2013) How can animal models be better utilized. Schizophrenia: evolution and synthesis MIT Press, Cambridge:205–216. [Google Scholar]
- Pantelis C, Velakoulis D, McGorry PD, Wood SJ, Suckling J, Phillips LJ, Yung AR, Bullmore ET, Brewer W, Soulsby B (2003) Neuroanatomical abnormalities before and after onset of psychosis: a cross-sectional and longitudinal MRI comparison. The Lancet 361:281–288. [DOI] [PubMed] [Google Scholar]
- Patil ST, Zhang L, Martenyi F, Lowe SL, Jackson KA, Andreev BV, Avedisova AS, Bardenstein LM, Gurovich IY, Morozova MA (2007) Activation of mGlu2/3 receptors as a new approach to treat schizophrenia: a randomized Phase 2 clinical trial. Nature medicine 13:1102–1107. [DOI] [PubMed] [Google Scholar]
- Pehrson AL, Moghaddam B (2010) Impact of metabotropic glutamate 2/3 receptor stimulation on activated dopamine release and locomotion. Psychopharmacology 211:443–455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perez SM, Lodge DJ (2013) Hippocampal interneuron transplants reverse aberrant dopamine system function and behavior in a rodent model of schizophrenia. Molecular psychiatry 18:1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perez SM, Shah A, Asher A, Lodge DJ (2013) Hippocampal deep brain stimulation reverses physiological and behavioural deficits in a rodent model of schizophrenia. International Journal of Neuropsychopharmacology 16:1331–1339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perkins DO, Gu H, Boteva K, Lieberman JA (2005) Relationship between duration of untreated psychosis and outcome in first-episode schizophrenia: a critical review and meta-analysis. American Journal of Psychiatry 162:1785–1804. [DOI] [PubMed] [Google Scholar]
- Poels EM, Kegeles LS, Kantrowitz JT, Javitt DC, Lieberman JA, Abi-Dargham A, Girgis RR (2014) Glutamatergic abnormalities in schizophrenia: a review of proton MRS findings. Schizophrenia research 152:325–332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Powell CM, Miyakawa T (2006) Schizophrenia-relevant behavioral testing in rodent models: a uniquely human disorder? Biological psychiatry 59:1198–1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pruessner JC, Champagne F, Meaney MJ, Dagher A (2004) Dopamine release in response to a psychological stress in humans and its relationship to early life maternal care: a positron emission tomography study using [11C] raclopride. Journal of Neuroscience 24:2825–2831. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pucak ML, Grace AA (1994) Evidence that systemically administered dopamine antagonists activate dopamine neuron firing primarily by blockade of somatodendritic autoreceptors. Journal of Pharmacology and Experimental Therapeutics 271:1181–1192. [PubMed] [Google Scholar]
- Remington G, Kapur S (2010) Antipsychotic dosing: how much but also how often? Schizophrenia bulletin 36:900–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Remington G, Saha A, Chong S-A, Shammi C (2005) Augmentation strategies in clozapine-resistant schizophrenia. CNS drugs 19:843–872. [DOI] [PubMed] [Google Scholar]
- Robinson DG, Woerner MG, Alvir JMJ, Bilder RM, Hinrichsen GA, Lieberman JA (2002) Predictors of medication discontinuation by patients with first-episode schizophrenia and schizoaffective disorder. Schizophrenia Research 57:209–219. [DOI] [PubMed] [Google Scholar]
- Robinson DG, Woerner MG, Alvir JMJ, Geisler S, Koreen A, Sheitman B, Chakos M, Mayerhoff D, Bilder R, Goldman R (1999) Predictors of treatment response from a first episode of schizophrenia or schizoaffective disorder. American Journal of Psychiatry 156:544–549. [DOI] [PubMed] [Google Scholar]
- Robinson TE, Becker JB (1986) Enduring changes in brain and behavior produced by chronic amphetamine administration: a review and evaluation of animal models of amphetamine psychosis. Brain research reviews 11:157–198. [DOI] [PubMed] [Google Scholar]
- Rorick-Kehn LM, Johnson BG, Knitowski KM, Salhoff CR, Witkin JM, Perry KW, Griffey KI, Tizzano JP, Monn JA, McKinzie DL (2007) In vivo pharmacological characterization of the structurally novel, potent, selective mGlu2/3 receptor agonist LY404039 in animal models of psychiatric disorders. Psychopharmacology 193:121–136. [DOI] [PubMed] [Google Scholar]
- Ruby E, Rothman K, Corcoran C, Goetz RR, Malaspina D (2017) Influence of early trauma on features of schizophrenia. Early intervention in psychiatry 11:322–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samaha A-N, Seeman P, Stewart J, Rajabi H, Kapur S (2007) “Breakthrough” dopamine supersensitivity during ongoing antipsychotic treatment leads to treatment failure over time. Journal of Neuroscience 27:2979–2986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samaha A-N, Reckless GE, Seeman P, Diwan M, Nobrega JN, Kapur S (2008) Less is more: antipsychotic drug effects are greater with transient rather than continuous delivery. Biological psychiatry 64:145–152. [DOI] [PubMed] [Google Scholar]
- Schobel SA, Lewandowski NM, Corcoran CM, Moore H, Brown T, Malaspina D, Small SA (2009) Differential targeting of the CA1 subfield of the hippocampal formation by schizophrenia and related psychotic disorders. Archives of general psychiatry 66:938–946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schobel SA, Chaudhury NH, Khan UA, Paniagua B, Styner MA, Asllani I, Inbar BP, Corcoran CM, Lieberman JA, Moore H (2013) Imaging patients with psychosis and a mouse model establishes a spreading pattern of hippocampal dysfunction and implicates glutamate as a driver. Neuron 78:81–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Seeman P (1987) Dopamine receptors and the dopamine hypothesis of schizophrenia. Synapse 1:133–152. [DOI] [PubMed] [Google Scholar]
- Seeman P, Lee T (1975) Antipsychotic drugs: direct correlation between clinical potency and presynaptic action on dopamine neurons. Science 188:1217–1219. [DOI] [PubMed] [Google Scholar]
- Shapiro DA, Renock S, Arrington E, Chiodo LA, Li-Xin L, Sibley DR, Roth BL, Mailman R (2003) Aripiprazole, a novel atypical antipsychotic drug with a unique and robust pharmacology. Neuropsychopharmacology 28:1400. [DOI] [PubMed] [Google Scholar]
- Silvestri S, Seeman MV, Negrete J-C, Houle S, Shammi C, Remington GJ, Kapur S, Zipursky RB, Wilson AA, Christensen BK (2000) Increased dopamine D2 receptor binding after long-term treatment with antipsychotics in humans: a clinical PET study. Psychopharmacology 152:174–180. [DOI] [PubMed] [Google Scholar]
- Slifstein M, van de Giessen E, Van Snellenberg J, Thompson JL, Narendran R, Gil R, Hackett E, Girgis R, Ojeil N, Moore H (2015) Deficits in prefrontal cortical and extrastriatal dopamine release in schizophrenia: a positron emission tomographic functional magnetic resonance imaging study. JAMA psychiatry 72:316–324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Snyder SH (1973) Amphetamine psychosis: a” model” schizophrenia mediated by catecholamines. American Journal of Psychiatry 130:61–67. [DOI] [PubMed] [Google Scholar]
- Sonnenschein SF, Gill KM, Grace AA (2019) State-dependent effects of the D 2 partial agonist aripiprazole on dopamine neuron activity in the MAM neurodevelopmental model of schizophrenia. Neuropsychopharmacology 44:572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stahl S, Grady M (2004) A critical review of atypical antipsychotic utilization: comparing monotherapy with polypharmacy and augmentation. Current medicinal chemistry 11:313–327. [DOI] [PubMed] [Google Scholar]
- Stauffer VL, Millen BA, Andersen S, Kinon BJ, LaGrandeur L, Lindenmayer J, Gomez JC (2013) Pomaglumetad methionil: no significant difference as an adjunctive treatment for patients with prominent negative symptoms of schizophrenia compared to placebo. Schizophrenia research 150:434–441. [DOI] [PubMed] [Google Scholar]
- Stockton ME, Rasmussen K (1996) Electrophysiological effects of olanzapine, a novel atypical antipsychotic, on A9 and A10 dopamine neurons. Neuropsychopharmacology 14:97. [DOI] [PubMed] [Google Scholar]
- Tadokoro S, Nonomura N, Kanahara N, Hashimoto K, Iyo M (2017) Reduction of severity of recurrent psychotic episode by sustained treatment with aripiprazole in a schizophrenic patient with dopamine supersensitivity: a case report. Clinical Psychopharmacology and Neuroscience 15:79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tadokoro S, Okamura N, Sekine Y, Kanahara N, Hashimoto K, Iyo M (2011) Chronic treatment with aripiprazole prevents development of dopamine supersensitivity and potentially supersensitivity psychosis. Schizophrenia bulletin 38:1012–1020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Takase M, Kanahara N, Oda Y, Kimura H, Watanabe H, Iyo M (2015) Dopamine supersensitivity psychosis and dopamine partial agonist: a retrospective survey of failure of switching to aripiprazole in schizophrenia. Journal of Psychopharmacology 29:383–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tamminga CA, Stan AD, Wagner AD (2010) The hippocampal formation in schizophrenia. American Journal of Psychiatry 167:1178–1193. [DOI] [PubMed] [Google Scholar]
- Thomson AM, Walker V, Flynn DM (1989) Glycine enhances NMDA-receptor mediated synaptic potentials in neocortical slices. Nature 338:422. [DOI] [PubMed] [Google Scholar]
- Tregellas JR, Smucny J, Harris JG, Olincy A, Maharajh K, Kronberg E, Eichman LC, Lyons E, Freedman R (2014) Intrinsic hippocampal activity as a biomarker for cognition and symptoms in schizophrenia. American Journal of Psychiatry 171:549–556. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai G, Yang P, Chung L-C, Lange N, Coyle JT (1998) D-serine added to antipsychotics for the treatment of schizophrenia. Biological psychiatry 44:1081–1089. [DOI] [PubMed] [Google Scholar]
- Tsai G, Lane H-Y, Yang P, Chong M-Y, Lange N (2004) Glycine transporter I inhibitor, N-methylglycine (sarcosine), added to antipsychotics for the treatment of schizophrenia. Biological psychiatry 55:452–456. [DOI] [PubMed] [Google Scholar]
- Umbricht D, Alberati D, Martin-Facklam M, Borroni E, Youssef EA, Ostland M, Wallace TL, Knoflach F, Dorflinger E, Wettstein JG (2014) Effect of bitopertin, a glycine reuptake inhibitor, on negative symptoms of schizophrenia: a randomized, double-blind, proof-of-concept study. JAMA psychiatry 71:637–646. [DOI] [PubMed] [Google Scholar]
- Urban JD, Vargas GA, Von Zastrow M, Mailman RB (2007) Aripiprazole has functionally selective actions at dopamine D2 receptor-mediated signaling pathways. Neuropsychopharmacology 32:67–77. [DOI] [PubMed] [Google Scholar]
- Urs NM, Peterson SM, Caron MG (2017) New concepts in dopamine D2 receptor biased signaling and implications for schizophrenia therapy. Biological psychiatry 81:78–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Valenti O, Cifelli P, Gill KM, Grace AA (2011) Antipsychotic drugs rapidly induce dopamine neuron depolarization block in a developmental rat model of schizophrenia. Journal of Neuroscience 31:12330–12338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Os J, Kenis G, Rutten BP (2010) The environment and schizophrenia. Nature 468:203. [DOI] [PubMed] [Google Scholar]
- Van Rossum J (1967) The significance of dopamine-receptor blockade for the action of neuroleptic drugs. Neuropsychopharmacology. Proceedings 5th Collegium Internationale Neruropsychopharmacologicum, 1967. [Google Scholar]
- Velakoulis D, Wood SJ, Wong MT, McGorry PD, Yung A, Phillips L, Smith D, Brewer W, Proffitt T, Desmond P (2006) Hippocampal and amygdala volumes according to psychosis stage and diagnosis: A magnetic resonance imaging study of chronic schizophrenia, first-episode psychosis, and ultra–high-risk individuals. Archives of general psychiatry 63:139–149. [DOI] [PubMed] [Google Scholar]
- Viguera AC, Baldessarini RJ, Hegarty JD, van Kammen DP, Tohen M (1997) Clinical risk following abrupt and gradual withdrawal of maintenance neuroleptic treatment. Archives of general psychiatry 54:49–55. [DOI] [PubMed] [Google Scholar]
- Vollenweider FX, Vontobel P, Øye I, Hell D, Leenders KL (2000) Effects of (S)-ketamine on striatal dopamine: a [11C] raclopride PET study of a model psychosis in humans. Journal of psychiatric research 34:35–43. [DOI] [PubMed] [Google Scholar]
- Weiser M, Heresco-Levy U, Davidson M, Javitt DC, Werbeloff N, Gershon AA, Abramovich Y, Amital D, Doron A, Konas S (2012) A multicenter, add-on randomized controlled trial of low-dose d-serine for negative and cognitive symptoms of schizophrenia. The Journal of clinical psychiatry 73:e728–734. [DOI] [PubMed] [Google Scholar]
- Weiss AP, Schacter DL, Goff DC, Rauch SL, Alpert NM, Fischman AJ, Heckers S (2003) Impaired hippocampal recruitment during normal modulation of memory performance in schizophrenia. Biological psychiatry 53:48–55. [DOI] [PubMed] [Google Scholar]
- White FJ, Wang RY (1983) Differential effects of classical and atypical antipsychotic drugs on A9 and A10 dopamine neurons. Science 221:1054–1057. [DOI] [PubMed] [Google Scholar]
- Wood SJ, Pantelis C, Velakoulis D, Yücel M, Fornito A, McGorry PD (2008) Progressive changes in the development toward schizophrenia: studies in subjects at increased symptomatic risk. Schizophrenia bulletin 34:322–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yokoi F, Gründer G, Biziere K, Stephane M, Dogan AS, Dannals RF, Ravert H, Suri A, Bramer S, Wong DF (2002) Dopamine D 2 and D 3 receptor occupancy in normal humans treated with the antipsychotic drug aripiprazole (OPC 14597): a study using positron emission tomography and [11 C] raclopride. Neuropsychopharmacology 27:248. [DOI] [PubMed] [Google Scholar]
- Zimmerman EC, Bellaire M, Ewing SG, Grace AA (2013) Abnormal stress responsivity in a rodent developmental disruption model of schizophrenia. Neuropsychopharmacology 38:2131–2139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zipursky RB, Reilly TJ, Murray RM (2012) The myth of schizophrenia as a progressive brain disease. Schizophrenia Bulletin 39:1363–1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zohar J, Stahl S, Moller H-J, Blier P, Kupfer D, Yamawaki S, Uchida H, Spedding M, Goodwin GM, Nutt D (2015) A review of the current nomenclature for psychotropic agents and an introduction to the Neuroscience-based Nomenclature. European Neuropsychopharmacology 25:2318–2325. [DOI] [PubMed] [Google Scholar]
- Zuckerman L, Rehavi M, Nachman R, Weiner I (2003) Immune activation during pregnancy in rats leads to a postpubertal emergence of disrupted latent inhibition, dopaminergic hyperfunction, and altered limbic morphology in the offspring: a novel neurodevelopmental model of schizophrenia. Neuropsychopharmacology 28:1778. [DOI] [PubMed] [Google Scholar]
