Abstract
N-methyl-D-aspartate glutamate receptor (NMDAR) hypofunction is implicated in the impaired neuroplasticity and cognitive impairments associated with schizophrenia (CIAS). We hypothesized that enhancing NMDAR function by inhibiting the glycine transporter-1 (GLYT1) would improve neuroplasticity and thereby augment benefits of non-pharmacological cognitive training (CT) strategies. This study examined whether co-administration of a GLYT1 inhibitor and computerized CT would have synergistic effects on CIAS. Stable outpatients with schizophrenia participated in this double-blind, placebo-controlled, within-subject, crossover augmentation study. Participants received placebo or GLYT1 inhibitor (PF-03463275), for two 5-week periods separated by 2 weeks of washout. PF-03463275 doses (40 or 60 mg twice daily) were selected to produce high GLYT1 occupancy. To limit pharmacodynamic variability, only cytochrome P450 2D6 extensive metabolizers were included. Medication adherence was confirmed daily. Participants received 4 weeks of CT in each treatment period. Cognitive performance (MATRICS Consensus Cognitive Battery) and psychotic symptoms (Positive and Negative Syndrome Scale) were assessed in each period. 71 participants were randomized. PF-03463275 in combination with CT was feasible, safe, and well-tolerated at the doses prescribed but did not produce greater improvement in CIAS compared to CT alone. PF-03463275 was not associated with improved CT learning parameters. Participation in CT was associated with improvement in MCCB scores.
Keywords: Cognition, schizophrenia, psychosis, NMDA, glycine transporter, remediation
1. Introduction:
Cognitive impairments associated with schizophrenia (CIAS) encompass deficits in memory, attention, executive functioning, vocabulary, visuospatial skills, and learning (Heinrichs and Zakzanis, 1998), represent a significant decrement from premorbid ability (Keefe et al., 2005) and affect functional status more than schizophrenia’s core disease-defining symptoms (Bowie et al., 2006; Green et al., 2000; Green et al., 2004; Tsang et al., 2010). Antipsychotic drugs that block dopamine (DA) D2 receptors help manage psychosis (Kapur and Mamo, 2003) but CIAS persist (Buchanan et al., 2007a; Buchanan et al., 2007b; Goldberg et al., 1993; Harvey et al., 2001; Keefe et al., 2007). Non-DA pharmacological targets have been explored for the treatment of CIAS including histaminic (H3 antagonism), cholinergic (Alpha7 nicotinic receptor agonism), and N-methyl-D-aspartate receptor (NMDAR agonism via glycine modulatory site) neurotransmitter systems. However, these endeavors have yet to elucidate effective treatments (Girgis et al., 2019), and CIAS remain an important area of unmet medical need for drug development (Yang et al., 2017).
Arguably, the only existing evidence-based treatment for CIAS is cognitive training (CT), or targeted exercises of memory, information processing, attention, and executive functioning, but trials of CT have found modest efficacy (Lejeune et al., 2021; Vita et al., 2021). Well-established deficits in learning and neuroplasticity (Keshavan et al., 2015) could impede the acquisition, or generalization, of trained skills to broader cognitive and functional domains. Indeed, individual response to CT has been associated with baseline differences in behavioral indices of learning potential (Davidson et al., 2016) and in a neurophysiologic metric associated with information processing capacity (Castelluccio et al., 2020).
Pharmacologic and neurostimulation (Jahshan et al., 2017; Jahshan et al., 2020; Krystal et al., 2003) approaches to enhance neuroplasticity have been examined as ways to augment response to CT. Work in this area has focused on increasing NMDAR activity via its high-affinity glycine co-agonist site (glycineB). While enhancing NMDA signaling is a promising strategy, efforts to do so directly by glycine or D-serine intake may be limited by poor brain bioavailability of these drugs. For example, whereas levels of D-serine have been associated with CT response, adjunctive D-serine does not appear to enhance response to CT (D'Souza et al., 2013; Panizzutti et al., 2019).
An alternative strategy is to boost endogenous glycine levels by administering a glycine transporter-1 inhibitor (GLYT1-I). GLYT1-I’s raise glycine levels in hippocampal slice preparations (Harsing and Matyus, 2013) and in human cerebrospinal fluid (CSF) (Hofmann et al., 2016), and, in turn, increase glycine occupancy at the glycineB co-agonist site of NMDAR (Bergeron et al., 1998). In a preliminary signal-finding study, our group evaluated dose-related effects of the GLYT1-I PF-03463275 following 1 week of daily administration in healthy adults and patients with schizophrenia (D'Souza et al., 2018). Endpoints included Positron Emission Tomography (PET) measures of receptor occupancy, working memory task-based fMRI, and an electroencephalographic (EEG) biomarker of neuroplasticity based on long-term potentiation (LTP) of visual evoked potentials (VEP). No appreciable effects were observed in healthy participants; patients, showed a dose-related enhancement of LTP with peak effects at 40 mg BID. More recent evidence suggest GLYT1-I may also have therapeutic effects on CIAS (Fleischhacker et al., 2021).
Collectively, these studies suggest that GLYT1 is a potential pharmacologic mechanism for intervention in CIAS. To test this proposal, the current study paired GLYT1 administration with a computer-based CT intervention, providing a standardized curriculum over which task-based learning and generalization to untrained cognitive endpoints could be evaluated. Based on our previous dose-finding study, we chose the most efficacious and highest doses of PF-03463275 (40mg BID and 60mg BID respectively) to achieve optimal GLYT1-I occupancy and augmentation of CT.
2. Methods and Materials:
2.1. Study Design:
(Figure 1) In a double-blind, placebo-controlled, crossover study, stable antipsychotic-treated outpatients with schizophrenia were randomized to either placebo or active PF-03463275 twice daily in addition to their antipsychotic for 2 dosing periods, each lasting approximately 5 weeks. The first week lead-in of each treatment period was to evaluate the neurophysiologic effects of medication alone (i.e., without CT) and to confirm medication adherence and tolerability. After the week-long lead-in period, subjects received CT in addition to placebo/active PF-03463275 for 4 weeks. After completing the first treatment period, there was a 2-week washout period to ensure clearance of the study drug which has a half-life of 3-6 hours (Buchy et al., 2015)(Buchy et al., 2015)(Buchy et al., 2015), before beginning the second treatment period, which was identical to the first, except that subjects who received active drug received placebo and vice-versa (Table S1).
Figure 1:
Study Design Overview. Participants were randomized to one of 3 sequences in a 1:1:1 ratio: A) PF-03463275 60 mg BID in the first treatment period followed by placebo in the second treatment period, B) placebo in the first treatment period followed by either (50%) PF-03463275 40 or (50%) 60 mg in the second treatment period, or C) PF-03463275 40 mg BID first followed by placebo.
All participants received placebo and only 1 of 2 active conditions in the study; thus, each active drug condition had about half the number of participants as the placebo condition.
In the first week of each study period, participants received study drug only and no cognitive training.
Cognitive training began after 1 week of study drug exposure in each treatment period.
MCCB was collected before (pre-drug) and after (post-drug) 5 weeks of treatment with study drug in each period. PANSS was administered before and twice during each 5-week treatment period.
2.2. Regulatory:
This study was conducted with approvals from the Institutional Review Boards of the VA Connecticut Healthcare System and Yale University School of Medicine, under an approved IND, (IND #118,880), and registered on clinicaltrials.gov (NCT01911676).
2.3. Screening / Inclusion/Exclusion criteria (See Supplemental Material):
Adults 20 to 65 years old: 1) with a DSM-IV schizophrenia or schizoaffective disorder; 2) on maintenance antipsychotic (except clozapine); 3) clinically stable (no antipsychotic changes or psychiatric hospitalizations in the preceding 3 months); 4) English speaking; 5) with capacity to provide written consent; and 6) were CYP2D6 extensive metabolizers, were included in the study. PF-03463275 is primarily metabolized by CYP2D6, so, to prevent large differences in CYP2D6 plasma levels, only participants who were extensive metabolizers were included, whereas those with CYP2D6 genotypes for ultra-rapid, intermediate, and poor metabolism were excluded.
2.4. Recruitment:
Participants were recruited by advertisements, flyers, referral from providers, and word of mouth. Research staff explained study details, risks, and procedures before obtaining written informed consent.
2.5. Randomization:
Stratified block randomization, with block size of 6 and stratification by Intelligence Quotient (WTAR estimated FSIQ 71-90 and >91), was used to assign subjects to one of 4 treatment sequences across two 5-week treatment phases that were separated by a 2-week washout phase (Figure 1 and Table S1): PF-03463275 60 mg BID followed by placebo, PF-03463275 40 mg BID followed by placebo, and placebo followed by either PF-03463275 40 or 60 mg BID. The study pharmacist generated the randomization sequence and prepared the study medications; all other study staff and participants were blinded to the condition.
2.6. PF-04363275:
PF-04363275 was provided by Pfizer Inc. (Groton, CT). Study medication adherence was visually confirmed by videoconferencing twice daily on weekdays using cellphone assisted remote observation of medication adherence (CAROMA) (DeWorsop et al., 2016). The dose of PF-03463275 for the clinical trial was selected based on the results of convergent approaches conducted in the earlier phase of the study: 1) receptor occupancy (PET); 2) reversal of ketamine-induced impairments in working memory (fMRI); and 3) augmentation of visually-evoked LTP. We found 60 mg BID produced the highest receptor occupancy, but that target engagement signal was stronger at 40 mg BID (D'Souza et al., 2018).
2.7. Cognitive Training:
All participants attended computer-based cognitive training appointments in a supervised clinic over weeks 2-5 of each treatment period. A training curriculum was developed using the HAPPYneuron program (detailed in Table S2 and Supplemental Methods). HAPPYneuron has been effective in prior studies of schizophrenia (Franck et al., 2013; Vianin et al., 2014). Participants were encouraged to complete 5 sessions of weekly training over a total of 8 weeks and received incentive payments in increasing intervals at the completion of each 5th session.
2.8. Outcome Measures:
2.8.1. Cognitive Outcome
The primary outcome measure was change in the MATRICS Consensus Cognitive Battery (MCCB) composite score. The MCCB was administered before the start and at the end of each of the two periods (Figure 1). The MCCB composite score and a calculated cognitive composite, which omitted the social cognition test, were calculated to assess the overall effect of treatment, with changes in specific cognitive domains including verbal and visual learning and working memory examined as exploratory analyses.
2.8.2. PANSS:
The Positive and Negative Syndrome Scale (PANSS) (Kay et al., 1989) is a 30-item clinician-rated assessment of symptoms of schizophrenia based on a semi-structured interview with subscales for positive, negative and general symptoms. The same group of raters administered PANSS for all participants thrice in each period: before randomization and during the second and fourth weeks of each period.
2.8.3. Participant Self-Assessments:
Subjective assessment of cognitive and perceptual experience was measured using the Patient’s Assessment of Own Functioning (PAOFI) (Chelune et al., 1986) while self-assessment of attentional processes and perceptual processes was measured using the Sensory Gating Inventory (SGI) (Hetrick et al., 2012).
2.9. Statistical Analyses:
This report is restricted to the cognitive and symptom measures. EEG biomarkers were assessed at Days 1 and 7 of each period, replicating our prior study design (D'Souza et al., 2018), and will be reported elsewhere. Target sample size was 30 per sequence for a total of 90, based on power calculations (Supplementary Methods). All dependent measures were assessed for normality using normal probability plots. Descriptive statistics were calculated prior to analyses. Each dependent variable (MCCB overall and cognitive composite scores, PANSS total and 3 PANSS subscales), was analyzed using a mixed effects model with dose (placebo, 40 mg BID, 60 mg BID), visit, their interaction, and period (first or second) as main effect. The time points were pre- versus post-drug for MCCB, and pre-drug and two post-drug assessments at two and five weeks for PANSS. Interactions involving period were also assessed but dropped from the models for parsimony. Subject was the clustering factor. The best-fitting variance-covariance structure for the repeated measures in each model was selected based on Schwartz’ Bayesian Criterion. Least square means and standard errors were calculated to describe the patterns of means for each outcome. Post-hoc mean comparisons were tested to explain significant effects in the models. For each outcome, we also fit models with drug as a two-level factor (i.e., placebo vs. 40mg and 60mg doses combined). Exploratory analyses (see Supplement for details) included repeated measures ANOVA (rmANOVA) to examine: 1) effects of time, period, and dose on PAOFI and SGI; 2) effects of dose on performance in CT exercises.
3. Results:
71 participants were randomized as shown in Figure 2. Demographics of participants in each of the four randomization assignments are shown in Table 1, and each of the four randomized groups were well-matched for age, education, and IQ.
Figure 2:
Consort Diagram. Of the 71 participants randomized, 6 were terminated by the PI (4 for substance misuse, 1 for abnormal ECG and 1 for wrist drop), 10 withdrew (5 no longer wanted to take study medication, 2 wanted to look for job, 1 had conflict with study activities and other responsibilities, 1 had housing problems) and 1 withdrew and was lost to follow-up. There were 55 completers who did all study activities except follow up.
Table 1:
Participant Demographics:
Abbreviations: SD = standard deviation; WTAR = Wechsler Test of Adult Reading.
| Randomization Assignment | Period 1: 40 mg Period 2: Placebo |
Period 1: Placebo Period 2: 40 mg |
Period 1: Placebo Period 2: 60 mg |
Period 1: 60 mg Period 2: Placebo |
|---|---|---|---|---|
| n | 22 | 12 | 13 | 24 |
| Average age in years (SD) | 46.1 (11.4) | 50.9 (12.4) | 40.9 (14.1) | 48.8 (11.7) |
| Number male (%) | 20 (90.9%) | 7 (58.3%) | 9 (69.2%) | 21 (87.5%) |
| Education, years (SD) | 12.1 (1.9) | 12.3 (2.7) | 12.5 (2.8) | 12.4 (2.4) |
| IQ from WTAR (SD) | 92.2 (10.5) | 96.9 (10.6) | 96.0 (10.8) | 94.4 (11.0) |
| Parent education, years (SD) | 12.5 (3.1) | 11.4 (2.4) | 13.4 (3.2) | 12.1 (3.6) |
| African American (%) | 16 (72.7%) | 5 (41.7%) | 6 (46.1%) | 15 (62.5%) |
| Caucasian (%) | 6 (27.3%) | 7 (58.3%) | 5 (38.5%) | 7 (29.2%) |
| Hispanic Ethnicity (%) | 2 (9.1%) | 3 (25%) | 2 (15.4%) | 2 (8.3%) |
3.1. Safety:
There were no serious adverse events. The rates of non-serious adverse events were low, and there were no differences in adverse event rates between placebo and PF-03463275 (Table S3).
3.2. Cognition:
MCCB MATRICS overall composite and cognitive composite scores, tested either as three independent conditions (PF-04363275 40 mg, PF-04363275 60 mg, and placebo) or with active conditions collapsed in comparison to placebo, increased with time; there were significant main effects of pre- versus post-drug visit (Figures 3 and S1), and of period 1 (before cross over) versus period 2 (after cross over), but no significant study drug condition * visit interaction (Tables S4 and S5). Exploratory analyses of all 10 MCCB subtests revealed no significant visit by study drug condition interactions.
Figure 3:
MCCB overall composite score for all participants collected before (pre-drug) and after (post-drug) 5 weeks of treatment with study drug in each period. All participants were randomized to receive placebo and only 1 of 2 active conditions (PF-03463275 40 or 60 mg) in the study; thus, the active drug conditions each have about half the participants as the placebo condition. X-axis represents dose, and y-axis represents MCCB overall composite score. Error bars represent SEM.
3.3. PANSS
PANSS assessments were performed at three visits in each period, pre-drug, after 2 weeks of study drug, and after 5 weeks of study drug (Figure 1). PANSS total scores and positive, negative, and general subscales decreased over time, with significant main effects of both visit and period, however the visit * study drug condition interactions were not significant for any of the four measures (Figures S2-S5), regardless of whether active doses were treated independently (Tablet S6) or combined (Table S7). A main effect of condition was detected for PANSS total in comparison of combined active conditions to placebo, and for PANSS positive scores tested using independent or combined active conditions (Tables S6 and S7). Condition effects appeared driven by the pre-drug PANSS total and positive scores being higher in active drug than placebo (Figures S2 and S3).
3.4. Exploratory Analyses:
RmANOVA’s revealed no significant main effect of pre- versus post-drug visit, period, or study drug condition on PAOFI or SGI (Supplementary Figures S6, S7), in contrast to other cognitive and clinical measures that improved with time. There was no effect of dose on two different measures of CT performance (Supplement and Figures S8-S9).
4. Discussion:
We tested the hypothesis that pharmacological enhancement of neuroplasticity with the GLYT1-I (PF-03463275) combined with CT would have synergistic effects on CIAS. PF-03463275 treatment, at doses demonstrated to engage the target, was safe and well-tolerated but did not produce a greater improvement in CIAS compared to CT alone. MCCB scores did, however, improve over time regardless of drug condition (active or placebo), as demonstrated by an improvement in period 2 over period 1 and pre- versus post-drug dosing. This improvement in MCCB performance was of effect size (Hedge’s g=0.31) comparable to prior studies of CT in patients with schizophrenia without a combined pharmacologic intervention; Hedge’s g ranging between 0.19-0.33 and Cohen’s d=0.45 (Lejeune et al., 2021; Wykes et al., 2011).
Clinical trials of NMDAR co-agonists (glycine, D-serine, D-cycloserine and D-alanine) and GLYT1-I’s to improve CIAS have produced mixed results. Glycine, low-dose D-serine, and D-cycloserine did not improve cognition in large studies (Buchanan et al., 2007b; Weiser et al., 2012). GLYT1-I’s bitopertin, sarcosine and ORG-25935 did not improve CIAS (Kantrowitz et al., 2017; Lin et al., 2017; Schoemaker et al., 2014). In contrast, in a recent placebo-controlled randomized phase 2 trial, BI 425809 without cognitive remediation improved cognition after 12 weeks of treatment in patients with schizophrenia (Fleischhacker et al., 2021). Two studies of NMDA receptor co-agonists as augmentation to CT have also had conflicting results: D-serine did not augment the effects of CT (D'Souza et al., 2013), and D-cycloserine was associated with improved performance on the trained auditory discrimination task but not with generalized improvements in cognitive outcomes (Cain et al., 2014). Evaluated in the present study, GLYT1-I did not appear to improve learning or peak performance of CT exercises.
PF-03463275 also failed to treat positive or negative symptoms, despite promising improvements in negative symptoms with GLYT1-I’s sarcosine in both chronic and antipsychotic-naïve acutely symptomatic patients (Lane et al., 2005; Lane et al., 2010; Tsai et al., 2004), and bitopertin (Umbricht et al., 2014), the results were not replicated in later studies of sarcosine (Lin et al., 2017; Strzelecki et al., 2015), phase 3 studies of bitopertin (Bugarski-Kirola et al., 2017), or in large studies of GLYT1-I’s AMG 747 and ORG-25935 (Dunayevich et al., 2017; Schoemaker et al., 2014). Though small earlier studies with NMDAR co-agonists suggested improvements in positive and negative symptoms (Heresco-Levy et al., 2004; Heresco-Levy et al., 1996; Heresco-Levy et al., 1999; Javitt et al., 1994; Tsai et al., 2006), large studies of NMDAR co-agonists glycine, D-cycloserine, and low-dose D-serine failed to treat negative symptoms (Buchanan et al., 2007b; Weiser et al., 2012). NMDAR co-agonists might be helpful in combination with behavioral therapy or in specific patients. D-cycloserine enhanced the effects of cognitive behavioral therapy for delusions (Gottlieb et al., 2011). D-serine at higher doses (60 mg/day) improved negative symptoms in those at clinical high risk for psychosis (Kantrowitz et al., 2015) and in schizophrenia in manner that correlated with improvements in the electrophysiological biomarker auditory mismatch negativity (MMN) (Kantrowitz et al., 2018). Glycine treatment in a small study improved MMN and negative symptoms, and there was suggestion of correlation between the two (Greenwood et al., 2018). These studies support target engagement of NMDAR measured with amplified evoked potentials and symptoms.
Reliable biomarkers of NMDAR-mediated neuroplasticity have been difficult to identify. While in our prior study LTP improved with the 40 mg dose of PF-03463275 (D'Souza et al., 2018), D-cycloserine failed to increase LTP (Forsyth et al., 2017). Interpretation of these results is complicated by inconsistent detection of the visual LTP deficit in schizophrenia (Wynn et al., 2019), and recent studies that challenge whether high frequency visual stimulation reliably results in potentiated VEP in healthy people (Dias et al., 2022). Auditory MMN is another proposed EEG measure of NMDAR-mediated synaptic plasticity and has been used to detect target engagement of NMDAR modulating compounds in other studies. However, although the MMN deficit in schizophrenia is robust and found repeatedly (Erickson et al., 2016), NMDAR modulators have had inconsistent effects on MMN (Kantrowitz et al., 2016; Kantrowitz et al., 2017), possibly due to inverted-U dose response curves (Sehatpour et al., 2023); improvements in cognition with BI 425809 were not correlated with MMN changes (Schultheis et al., 2022); and more recent studies in healthy people suggest MMN may not be dependent upon NMDAR functioning (Harms et al., 2021).
There are several possible reasons why we failed to find a synergistic effect of PF-03463275 and CT on cognitive test performance.
First, dose–response relationships for GlyT1-I’s are complicated, with reduced effects observed at higher doses (Javitt, 2009; Umbricht, 2018). This study took several steps to maximize target engagement with PF-03463275. First, the doses of 40 and 60 mg were selected based on in vivo PET imaging data showing ideal occupancy and effects on an electrophysiological index of neuroplasticity (D'Souza et al., 2018). Second, to limit variability in PF-03463275 levels, only CYP2D6 extensive metabolizers were included. Third, daily medication adherence was visually confirmed with CAROMA. Despite these controls, neither dose of the study drug improved MCCB scores in patients receiving CT. Perhaps a wider dose range should have been tested.
Second, the pathophysiology of schizophrenia is heterogenous - perhaps only those individuals with impaired NMDAR functioning benefit from facilitating NMDAR activity. There are no biomarkers at present to identify patients who might benefit from glycineB modulators. Variations in glycine/NMDAR function may predict different outcomes to treatment targeting NMDAR function. For instance, variations in the gene encoding glycine decarboxylase, the enzyme that catabolizes glycine, may influence response to glycineB modulators. Triplication of GLDC would be expected to result in low levels of brain glycine and D-serine. In a small proof-of-concept study, glycine improved psychotic and mood symptoms in individuals with psychosis who showed triplication of the gene encoding GLDC (Bodkin et al., 2019).
Third, improvements in cognitive test performance may be more likely to be detected in patients who are earlier in their illness relative to our sample. Participants in this study were, on average, in their mid-40s, so we cannot exclude the possibility that younger participants, earlier in their course of illness, would have had greater benefit of CT with PF-03463275, as older participants seem to benefit less overall from CT (Wykes et al., 2009). In contrast, participants in the BI 425809 study that showed improvements in MCCB scores were younger (mean age ~36 years) and earlier in their illness (Fleischhacker et al., 2021).
Fourth, GLYT1-I’s may have subtly different effects on glycine levels in different individuals. The GLYT1 also works in reverse to increase extracellular glycine (Huang et al., 2004), in addition to the uptake of glycine, to tightly regulate synaptic and extrasynaptic glycine concentrations so that GLYT1 inhibition could decrease extrasynaptic glycine.
Our study design permits specific examination of the impact of GLYT1 inhibition on distinct cognitive and CT outcomes, including learning potential, achievement in CT, cognitive functioning, and self-assessed community functioning. We hypothesized that treatment with PF-03463275 would improve the neurobiological substrate of learning, thus augmenting the impact of CT on cognitive performance. Our results demonstrated no dose-related effect on the CT-related change in MCCB, suggesting that treatment with PF-03463275 did not augment the effects of CT. Further, lack of drug by time interaction on cognitive testing was consistent with a similar lack in improvement on CT. One possible interpretation of the present findings is that PF-03463275 failed to restore/enhance plasticity adequately, and therefore failed to augment the impact of CT. Another possibility is that detecting effects of an augmenting intervention in context of another, especially in a cross-over design, is difficult. A parallel group study comparing drug alone, CT alone, and the combined intervention may have produced clearer results, however, this would not have been feasible given our aim to evaluate PF-03463275 at more than one dose.
Finally, despite a significant improvement in MCCB performance over time in our study, there was no accompanying improvement in the PAOFI or SGI. This is consistent with the recognition that, in contrast to objective functional measures, PAOFI scores are not strongly predicted by neuropsychological test results (Chelune et al., 1986). It is possible that the lack of improvement in participants’ perceived functioning is related to the trial’s modest duration. Nevertheless, it is important to recognize the divergence between this improvement on cognitive testing and the lack of improvement reported by patients, particularly as patient-reported measures are increasingly identified as primary outcomes for clinical trials.
This study’s design has several strengths and limitations. Dosing choices were based on carefully conducted pharmacokinetic/pharmacodynamic studies that employed central target engagement along with functionally relevant outcomes: PET occupancy of GLYT1, LTP, and attenuation of BOLD response associated with ketamine-induced WM deficits. The PET study results were that 10 mg, 20 mg, 40 mg, and 60 mg of PF-03463275 twice daily for 1 week occupied ~44%, 61%, 76%, and 83% of GLYT1 respectively (D'Souza et al., 2018). This led to two doses tested in this study - it is possible that the range of doses tested in this paradigm was too narrow. The optimal occupancy for GLYT1-I’s is unknown, but a large study of bitopertin indicated that low to medium range occupancy was optimal (Umbricht et al., 2014). There may be several targets that a drug may engage, starting at the receptor and many levels downstream (e.g., LTP). For example, in this case, the most proximal target would be engagement of the glycine transporter. That was definitively demonstrated in vivo using PET (D'Souza et al., 2018). In the same study, 40 mg PF-03463275 enhanced LTP. However, further research is necessary to directly and conclusively link GLYT1 occupancy and/or LTP with the primary outcome of interest – improved cognitive test performance, and whether this measure of LTP is a reliable as a biomarker for neuroplasticity. The study was designed to specifically examine whether PF-03463275 could augment the effects of CT on CIAS. All participants in this study received CT, and the expected CT associated improvements occurred. The lack of a PF-03463275 alone arm prevented examination of drug alone on CIAS, as observed by Fleischhacker and colleagues with BI 425809 (Fleischhacker et al., 2021), but this was not the focus of this study. The within-subject cross over design is a more powerful design but one that might be prone to carryover effects; however, analyzing just the first treatment period alone did not reveal any significant effects that could have carried over to the next treatment period beyond the washout period. Likewise, in a within-subject design, practice effects from repeatedly taking the MCCB may have made it harder to detect drug effects. The duration of treatment with PF-03463275 on the MCCB outcome measure was 5 weeks, and perhaps this was too short, given that the Phase 2 12-week trial of BI 425809 where MCCB improvement continued between 6 and 12 weeks. Use of cognitive training to test the study hypothesis necessitated frequent appointments, which challenged retention. Stringent inclusion criteria to limit pharmacokinetic variability in PF-03463275 prolonged screening and restricted recruitment. While it is possible that the study could not detect a statistically significant improvement in MCCB with PF-03463275 due to insufficient sample size or baseline group differences (despite randomization) in race or sex (randomization was stratified by IQ only), there was no robust improvement in cognition with PF-03463275 above the effects of CT in this sample of people with chronic schizophrenia.
While our study failed to reveal positive findings, Phase 3 studies with GLYT1-Is remain in progress, and the positive results of the phase 2 trial of GLYT1-I BI 425809 are encouraging.
Many parameters must be optimized for a medication to augment the small to moderate effects of CT, including the drug itself, drug dose, duration of drug, timing of drug in relation to CT, the duration and intensity of the CT, the choice of CT program, and the study outcomes for people in varying stages of a heterogeneous illness. Nonetheless, improving deficient neuroplasticity in schizophrenia to enhance CT remains an attractive strategy. Robust, in vivo measures of neuroplasticity and their relationship to learning would help optimize the many variables in clinical trials of cognitive enhancers in CIAS.
Supplementary Material
Acknowledgements:
We thank the staff of the Schizophrenia Neuropharmacology Research Group at Yale (SNRGY) and the Clinical Neuroscience Research Unit (CNRU) at the Connecticut Mental Health Center (CMHC) of the Connecticut Department of Mental Health and Addiction Services (DMHAS) and VA Connecticut Healthcare System.
Funding:
This work was supported by National Center for Advancing Translational Science (NCATS) Grant No. 1UH2TR000960-01, National Institute on Alcohol Abuse and Alcoholism Grant No. P50AA012870, Yale Center for Clinical Investigation Grant No. UL1 RR024139, and the Department of Veterans Affairs through its support for the Veterans Affairs National Center for Posttraumatic Stress Disorder (JHK). TS was supported by CTSA Grant Number UL1 TR001863 from the National Center for Advancing Translational Science (NCATS), components of the National Institutes of Health (NIH), and NIH Roadmap for Medical Research. MR was supported by a Veterans Affairs VISN1 Career Development Award. The contents of this publication are solely the responsibility of the authors and do not necessarily represent the official view of NIH. Pfizer Pharmaceuticals contributed PF-03463275 and matched placebo. This work was funded in part by the State of Connecticut, Department of Mental Health and Addiction Services, but this publication does not express the views of the Department of Mental Health and Addiction Services or the State of Connecticut. The views and opinions expressed are those of the authors.
Abbreviations
- BOLD
blood oxygen level dependent imaging
- CAROMA
cellphone assisted remote observation of medication adherence
- CIAS
cognitive impairments associated with schizophrenia
- CSF
cerebrospinal fluid
- CT
cognitive training
- DA
dopamine
- EEG
electroencephalography
- fMRI
functional magnetic resonance imaging
- FSIQ
full scale IQ
- GLYT1
glycine transporter-1
- GLYT1-I
glycine transporter-1 inhibitor
- LTP
long term potentiation
- MCCB
MATRICS Consensus Cognitive Battery
- MMN
Mismatch negativity
- NMDAR
N-methyl-D-aspartate glutamate receptor
- PAOFI
Patient’s Assessment of Own Functioning
- PET
positron emission tomography
- PANSS
Positive and Negative Syndrome Scale
- rmANOVA
repeated measures analysis of variance
- SD
standard deviation
- SGI
Sensory Gating Inventory
- VEP
Visual Evoked Potential
- WTAR
Wechsler Test of Adult Reading
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 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.
Disclosures: DCD receives research funding administered through Yale University from the US National Institute of Health, US Dept. of Veteran Affairs, Takeda, Biogen, Boehringer Ingelheim, Ceruvia, Heffter Institute, and Wallace Foundation. DCD has served as a paid consultant to Jazz Pharmaceuticals, Biohaven, and Abide. JHK has consulting agreements (less than US $10,000 per year) with the following: AstraZeneca Pharmaceuticals, Biogen, Idec, MA, Biomedisyn Corporation, Bionomics, Limited (Australia), Boehringer Ingelheim International, COMPASS Pathways, Limited, United Kingdom, Concert Pharmaceuticals, Inc, Epiodyne, Inc, EpiVario, Inc, Heptares Therapeutics, Limited (UK), Janssen Research \& Development, Otsuka America, Pharmaceutical, Inc, Perception Neuroscience Holdings, Inc, Spring Care, Inc, Sunovion Pharmaceuticals, Inc, Takeda Industries and Taisho Pharmaceutical Co., Ltd.; serves on the scientific advisory boards of Bioasis Technologies, Inc, Biohaven Pharmaceuticals, BioXcel Therapeutics, Inc (Clinical Advisory Board), BlackThorn Therapeutics, Inc, Cadent Therapeutics (Clinical Advisory Board), Cerevel Therapeutics, LLC., EpiVario, Inc, Lohocla Research Corporation, PsychoGenics, Inc; is on the board of directors of Inheris Biopharma, Inc; has stock options with Biohaven Pharmaceuticals Medical Sciences, BlackThorn Therapeutics, Inc, EpiVario, Inc and Terran Life Sciences; is a cosponsor of a patent for the intranasal administration of ketamine for the treatment of depression that was licensed by Janssen Pharmaceuticals, the maker of S-ketamine; and has a patent related to the use of riluzole to treat anxiety disorders that was licensed by Biohaven Medical Sciences. JHK is editor of Biological Psychiatry with income greater than $10,000. He has fiduciary responsibility for the International College of Neuropsychopharmacology as president of this organization. MR receives research grant support administered through the Yale University School of Medicine from Insys Therapeutics, Roche and Bioxcel and has been a consultant for Bioexcel. JKJ is currently employed full-time at Sage Therapeutics. Sage Therapeutics was not involved in any aspect of the study, including study design; data collection, analyses or interpretation; or writing this manuscript. The other authors report no biomedical financial interests of potential conflicts of interest.
ClinicalTrials.gov: Translational Neuroscience Optimization of GLYT1 Inhibitor (NCATS); https://clinicaltrials.gov/ct2/show/NCT01911676; NCT01911676.
References
- Bergeron R, Meyer TM, Coyle JT, Greene RW, 1998. Modulation of N-methyl-D-aspartate receptor function by glycine transport. Proceedings of the National Academy of Sciences of the United States of America. 95(26), 15730–15734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bodkin JA, Coleman MJ, Godfrey LJ, Carvalho CM, Morgan CJ, Suckow RF, Anderson T, Öngür D, Kaufman MJ, Lewandowski KE, 2019. Targeted treatment of individuals with psychosis carrying a copy number variant containing a genomic triplication of the glycine decarboxylase gene. Biological psychiatry 86(7), 523–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bowie CR, Reichenberg A, Patterson TL, Heaton RK, Harvey PD, 2006. Determinants of real-world functional performance in schizophrenia subjects: correlations with cognition, functional capacity, and symptoms. Am J Psychiatry 163(3), 418–425. [DOI] [PubMed] [Google Scholar]
- Buchanan RW, Freedman R, Javitt DC, Abi-Dargham A, Lieberman JA, 2007a. Recent advances in the development of novel pharmacological agents for the treatment of cognitive impairments in schizophrenia. Schizophrenia bulletin 33(5), 1120–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buchanan RW, Javitt DC, Marder SR, Schooler NR, Gold JM, McMahon RP, Heresco-Levy U, Carpenter WT, 2007b. The Cognitive and Negative Symptoms in Schizophrenia Trial (CONSIST): the efficacy of glutamatergic agents for negative symptoms and cognitive impairments. Am J Psychiatry 164(10), 1593–1602. [DOI] [PubMed] [Google Scholar]
- Buchy L, Cadenhead K, Cannon T, Cornblatt B, McGlashan T, Perkins D, Seidman L, Tsuang M, Walker E, Woods S, 2015. Substance use in individuals at clinical high risk of psychosis. Psychological Medicine 45(11), 2275–2284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bugarski-Kirola D, Blaettler T, Arango C, Fleischhacker WW, Garibaldi G, Wang A, Dixon M, Bressan RA, Nasrallah H, Lawrie S, Napieralski J, Ochi-Lohmann T, Reid C, Marder SR, 2017. Bitopertin in Negative Symptoms of Schizophrenia-Results From the Phase III FlashLyte and DayLyte Studies. Biol Psychiat 82(1), 8–16. [DOI] [PubMed] [Google Scholar]
- Cain CK, McCue M, Bello I, Creedon T, Tang DI, Laska E, Goff DC, 2014. d-Cycloserine augmentation of cognitive remediation in schizophrenia. Schizophr Res 153(1-3), 177–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castelluccio BC, Kenney JG, Johannesen JK, 2020. Individual Alpha Peak Frequency Moderates Transfer of Learning in Cognitive Remediation of Schizophrenia. J Int Neuropsychol Soc 26(1), 19–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chelune GJ, Heaton RK, Lehman RAW, 1986. Neuropsychological and personalitycorrelates of patients' complaints of disability., in: Goldstein G, Tarter RE (Eds.), Advances in Clinical Neuropsychology. Plenum Press, New York, NY, pp. 95–126. [Google Scholar]
- D'Souza DC, Carson RE, Driesen N, Johannesen J, Ranganathan M, Krystal JH, Yale Gly TSG, 2018. Dose-Related Target Occupancy and Effects on Circuitry, Behavior, and Neuroplasticity of the Glycine Transporter-1 Inhibitor PF-03463275 in Healthy and Schizophrenia Subjects. Biol Psychiatry 84(6), 413–421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- D'Souza DC, Radhakrishnan R, Perry E, Bhakta S, Singh NM, Yadav R, Abi-Saab D, Pittman B, Chaturvedi SK, Sharma MP, Bell M, Andrade C, 2013. Feasibility, safety, and efficacy of the combination of D-serine and computerized cognitive retraining in schizophrenia: an international collaborative pilot study. Neuropsychopharmacology 38(3), 492–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davidson CA, Johannesen JK, Fiszdon JM, 2016. Role of learning potential in cognitive remediation: Construct and predictive validity. Schizophr Res 171(1-3), 117–124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeWorsop D, Creatura G, Bluez G, Thurnauer H, Forselius-Bielen K, Ranganathan M, Deaso E, Bhat JA, D'Souza DC, 2016. Feasibility and success of cell-phone assisted remote observation of medication adherence (CAROMA) in clinical trials. Drug Alcohol Depend 163, 24–30. [DOI] [PubMed] [Google Scholar]
- Dias JW, McClaskey CM, Rumschlag JA, Harris KC, 2022. Sensory tetanisation to induce long-term-potentiation-like plasticity: A review and reassessment of the approach. Eur J Neurosci 56(12), 6115–6140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dunayevich E, Buchanan RW, Chen CY, Yang J, Nilsen J, Dietrich JM, Sun H, Marder S, 2017. Efficacy and safety of the glycine transporter type-1 inhibitor AMG 747 for the treatment of negative symptoms associated with schizophrenia. Schizophr Res 182, 90–97. [DOI] [PubMed] [Google Scholar]
- Erickson MA, Ruffle A, Gold JM, 2016. A Meta-Analysis of Mismatch Negativity in Schizophrenia: From Clinical Risk to Disease Specificity and Progression. Biol Psychiatry 79(12), 980–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fleischhacker WW, Podhorna J, Groschl M, Hake S, Zhao Y, Huang S, Keefe RSE, Desch M, Brenner R, Walling DP, Mantero-Atienza E, Nakagome K, Pollentier S, 2021. Efficacy and safety of the novel glycine transporter inhibitor BI 425809 once daily in patients with schizophrenia: a double-blind, randomised, placebo-controlled phase 2 study. Lancet Psychiatry 8(3), 191–201. [DOI] [PubMed] [Google Scholar]
- Forsyth JK, Bachman P, Mathalon DH, Roach BJ, Ye E, Asarnow RF, 2017. Effects of Augmenting N-Methyl-D-Aspartate Receptor Signaling on Working Memory and Experience-Dependent Plasticity in Schizophrenia: An Exploratory Study Using Acute d-cycloserine. Schizophr Bull 43(5), 1123–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Franck N, Duboc C, Sundby C, Amado I, Wykes T, Demily C, Launay C, Le Roy V, Bloch P, Willard D, Todd A, Petitjean F, Foullu S, Briant P, Grillon ML, Deppen P, Verdoux H, Bralet MC, Januel D, Riche B, Roy P, Other members of the Cognitive Remediation, N., Vianin P, 2013. Specific vs general cognitive remediation for executive functioning in schizophrenia: a multicenter randomized trial. Schizophr Res 147(1), 68–74. [DOI] [PubMed] [Google Scholar]
- Girgis RR, Zoghbi AW, Javitt DC, Lieberman JA, 2019. The past and future of novel, non-dopamine-2 receptor therapeutics for schizophrenia: A critical and comprehensive review. J Psychiatr Res 108, 57–83. [DOI] [PubMed] [Google Scholar]
- Goldberg TE, Greenberg RD, Griffin SJ, Gold JM, Kleinman JE, Pickar D, Schulz SC, Weinberger DR, 1993. The effect of clozapine on cognition and psychiatric symptoms in patients with schizophrenia. The British journal of psychiatry : the journal of mental science 162, 43–48. [DOI] [PubMed] [Google Scholar]
- Gottlieb JD, Cather C, Shanahan M, Creedon T, Macklin EA, Goff DC, 2011. D-cycloserine facilitation of cognitive behavioral therapy for delusions in schizophrenia. Schizophr Res 131(1-3), 69–74. [DOI] [PMC free article] [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"? Schizophr Bull 26(1), 119–136. [DOI] [PubMed] [Google Scholar]
- Green MF, Kern RS, Heaton RK, 2004. Longitudinal studies of cognition and functional outcome in schizophrenia: implications for MATRICS. Schizophr Res 72(1), 41–51. [DOI] [PubMed] [Google Scholar]
- Greenwood LM, Leung S, Michie PT, Green A, Nathan PJ, Fitzgerald P, Johnston P, Solowij N, Kulkarni J, Croft RJ, 2018. The effects of glycine on auditory mismatch negativity in schizophrenia. Schizophr Res 191, 61–69. [DOI] [PubMed] [Google Scholar]
- Harms L, Parras GG, Michie PT, Malmierca MS, 2021. The Role of Glutamate Neurotransmission in Mismatch Negativity (MMN), A Measure of Auditory Synaptic Plasticity and Change-detection. Neuroscience 456, 106–113. [DOI] [PubMed] [Google Scholar]
- Harsing LG Jr., Matyus P, 2013. Mechanisms of glycine release, which build up synaptic and extrasynaptic glycine levels: the role of synaptic and non-synaptic glycine transporters. Brain Res Bull 93, 110–119. [DOI] [PubMed] [Google Scholar]
- Harvey PD, Bowie CR, Friedman JI, 2001. Cognition in schizophrenia. Current psychiatry reports 3(5), 423–428. [DOI] [PubMed] [Google Scholar]
- Heinrichs RW, Zakzanis KK, 1998. Neurocognitive deficit in schizophrenia: a quantitative review of the evidence. Neuropsychology 12(3), 426–445. [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. Biol Psychiatry 55(2), 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. Br J Psychiatry 169(5), 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. Arch Gen Psychiatry 56(1), 29–36. [DOI] [PubMed] [Google Scholar]
- Hetrick WP, Erickson MA, Smith DA, 2012. Phenomenological dimensions of sensory gating. Schizophr Bull 38(1), 178–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hofmann C, Pizzagalli F, Boetsch C, Alberati D, Ereshefsky L, Jhee S, Patat A, Boutouyrie-Dumont B, Martin-Facklam M, 2016. Effects of the glycine reuptake inhibitors bitopertin and RG7118 on glycine in cerebrospinal fluid: results of two proofs of mechanism studies in healthy volunteers. Psychopharmacology (Berl) 233(13), 2429–2439. [DOI] [PubMed] [Google Scholar]
- Huang H, Barakat L, Wang D, Bordey A, 2004. Bergmann glial GlyT1 mediates glycine uptake and release in mouse cerebellar slices. J Physiol 560(Pt 3), 721–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jahshan C, Rassovsky Y, Green MF, 2017. Enhancing Neuroplasticity to Augment Cognitive Remediation in Schizophrenia. Front Psychiatry 8, 191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jahshan C, Wynn JK, Roach BJ, Mathalon DH, Green MF, 2020. Effects of Transcranial Direct Current Stimulation on Visual Neuroplasticity in Schizophrenia. Clin EEG Neurosci 51(6), 382–389. [DOI] [PubMed] [Google Scholar]
- Javitt DC, 2009. Glycine transport inhibitors for the treatment of schizophrenia: symptom and disease modification. Current opinion in drug discovery & development 12(4), 468–478. [PubMed] [Google Scholar]
- Javitt DC, Zylberman I, Zukin SR, Heresco-Levy U, Lindenmayer JP, 1994. Amelioration of negative symptoms in schizophrenia by glycine. Am J Psychiatry 151(8), 1234–1236. [DOI] [PubMed] [Google Scholar]
- Kantrowitz JT, Epstein ML, Beggel O, Rohrig S, Lehrfeld JM, Revheim N, Lehrfeld NP, Reep J, Parker E, Silipo G, Ahissar M, Javitt DC, 2016. Neurophysiological mechanisms of cortical plasticity impairments in schizophrenia and modulation by the NMDA receptor agonist D-serine. Brain 139(Pt 12), 3281–3295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kantrowitz JT, Epstein ML, Lee M, Lehrfeld N, Nolan KA, Shope C, Petkova E, Silipo G, Javitt DC, 2018. Improvement in mismatch negativity generation during d-serine treatment in schizophrenia: Correlation with symptoms. Schizophr Res 191, 70–79. [DOI] [PubMed] [Google Scholar]
- Kantrowitz JT, Nolan KA, Epstein ML, Lehrfeld N, Shope C, Petkova E, Javitt DC, 2017. Neurophysiological Effects of Bitopertin in Schizophrenia. J Clin Psychopharmacol 37(4), 447–451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kantrowitz JT, Woods SW, Petkova E, Cornblatt B, Corcoran CM, Chen H, Silipo G, Javitt DC, 2015. D-serine for the treatment of negative symptoms in individuals at clinical high risk of schizophrenia: a pilot, double-blind, placebo-controlled, randomised parallel group mechanistic proof-of-concept trial. Lancet Psychiatry 2(5), 403–412. [DOI] [PubMed] [Google Scholar]
- Kapur S, Mamo D, 2003. Half a century of antipsychotics and still a central role for dopamine D2 receptors. Progress in neuro-psychopharmacology & biological psychiatry 27(7), 1081–1090. [DOI] [PubMed] [Google Scholar]
- Kay SR, Opler LA, Lindenmayer JP, 1989. The Positive and Negative Syndrome Scale (PANSS): rationale and standardisation. Br J Psychiatry Suppl(7), 59–67. [PubMed] [Google Scholar]
- Keefe RS, Bilder RM, Davis SM, Harvey PD, Palmer BW, Gold JM, Meltzer HY, Green MF, Capuano G, Stroup TS, McEvoy JP, Swartz MS, Rosenheck RA, Perkins DO, Davis CE, Hsiao JK, Lieberman JA, Investigators, C., Neurocognitive Working, G., 2007. Neurocognitive effects of antipsychotic medications in patients with chronic schizophrenia in the CATIE Trial. Arch Gen Psychiatry 64(6), 633–647. [DOI] [PubMed] [Google Scholar]
- Keefe RS, Eesley CE, Poe MP, 2005. Defining a cognitive function decrement in schizophrenia. Biol Psychiatry 57(6), 688–691. [DOI] [PubMed] [Google Scholar]
- Keshavan MS, Mehta UM, Padmanabhan JL, Shah JL, 2015. Dysplasticity, metaplasticity, and schizophrenia: Implications for risk, illness, and novel interventions. Dev Psychopathol 27(2), 615–635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Krystal JH, D'Souza DC, Mathalon D, Perry E, Belger A, Hoffman R, 2003. NMDA receptor antagonist effects, cortical glutamatergic function, and schizophrenia: toward a paradigm shift in medication development. Psychopharmacology (Berl) 169(3-4), 215–233. [DOI] [PubMed] [Google Scholar]
- Lane HY, Chang YC, Liu YC, Chiu CC, Tsai GE, 2005. Sarcosine or D-serine add-on treatment for acute exacerbation of schizophrenia: a randomized, double-blind, placebo-controlled study. Arch Gen Psychiatry 62(11), 1196–1204. [DOI] [PubMed] [Google Scholar]
- Lane HY, Lin CH, Huang YJ, Liao CH, Chang YC, Tsai GE, 2010. A randomized, double-blind, placebo-controlled comparison study of sarcosine (N-methylglycine) and D-serine add-on treatment for schizophrenia. Int J Neuropsychopharmacol 13(4), 451–460. [DOI] [PubMed] [Google Scholar]
- Lejeune JA, Northrop A, Kurtz MM, 2021. A Meta-analysis of Cognitive Remediation for Schizophrenia: Efficacy and the Role of Participant and Treatment Factors. Schizophr Bull. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin CY, Liang SY, Chang YC, Ting SY, Kao CL, Wu YH, Tsai GE, Lane HY, 2017. Adjunctive sarcosine plus benzoate improved cognitive function in chronic schizophrenia patients with constant clinical symptoms: A randomised, double-blind, placebo-controlled trial. World J Biol Psychiatry 18(5), 357–368. [DOI] [PubMed] [Google Scholar]
- Panizzutti R, Fisher M, Garrett C, Man WH, Sena W, Madeira C, Vinogradov S, 2019. Association between increased serum d-serine and cognitive gains induced by intensive cognitive training in schizophrenia. Schizophr Res 207, 63–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schoemaker JH, Jansen WT, Schipper J, Szegedi A, 2014. The selective glycine uptake inhibitor org 25935 as an adjunctive treatment to atypical antipsychotics in predominant persistent negative symptoms of schizophrenia: results from the GIANT trial. J Clin Psychopharmacol 34(2), 190–198. [DOI] [PubMed] [Google Scholar]
- Schultheis C, Rosenbrock H, Mack SR, Vinisko R, Schuelert N, Plano A, Sussmuth SD, 2022. Quantitative electroencephalography parameters as neurophysiological biomarkers of schizophrenia-related deficits: A Phase II substudy of patients treated with iclepertin (BI 425809). Transl Psychiat 12(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sehatpour P, Iosifescu DV, De Baun HM, Shope C, Mayer MR, Gangwisch J, Dias E, Sobeih T, Choo T, Wall MM, Medalia A, Saperstein AM, Kegeles LS, Girgis RR, Carlson M, Kantrowitz JT, 2023. Dose-Dependent Augmentation of Neuroplasticity-Based Auditory Learning in Schizophrenia: A Double-Blind, Placebo-Controlled, Randomized, Target Engagement Clinical Trial of the NMDA Glutamate Receptor Agonist D-serine. Biol Psychiat. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strzelecki D, Podgorski M, Kaluzynska O, Gawlik-Kotelnicka O, Stefanczyk L, Kotlicka-Antczak M, Gmitrowicz A, Grzelak P, 2015. Supplementation of Antipsychotic Treatment with the Amino Acid Sarcosine Influences Proton Magnetic Resonance Spectroscopy Parameters in Left Frontal White Matter in Patients with Schizophrenia. Nutrients 7(10), 8767–8782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tsai G, Lane HY, Yang P, Chong MY, Lange N, 2004. Glycine transporter I inhibitor, N-methylglycine (sarcosine), added to antipsychotics for the treatment of schizophrenia. Biol Psychiatry 55(5), 452–456. [DOI] [PubMed] [Google Scholar]
- Tsai GE, Yang P, Chang YC, Chong MY, 2006. D-alanine added to antipsychotics for the treatment of schizophrenia. Biol Psychiatry 59(3), 230–234. [DOI] [PubMed] [Google Scholar]
- Tsang HW, Leung AY, Chung RC, Bell M, Cheung WM, 2010. Review on vocational predictors: a systematic review of predictors of vocational outcomes among individuals with schizophrenia: an update since 1998. Aust N Z J Psychiatry 44(6), 495–504. [DOI] [PubMed] [Google Scholar]
- Umbricht D, 2018. Nailing the Right Dose! The Key for Developing Glycine Transporter Inhibitors for Schizophrenia. Biological psychiatry 84(6), 394–395. [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, Bausch A, Garibaldi G, Santarelli L, 2014. Effect of bitopertin, a glycine reuptake inhibitor, on negative symptoms of schizophrenia: a randomized, double-blind, proof-of-concept study. JAMA Psychiatry 71(6), 637–646. [DOI] [PubMed] [Google Scholar]
- Vianin P, Urben S, Magistretti P, Marquet P, Fornari E, Jaugey L, 2014. Increased activation in Broca's area after cognitive remediation in schizophrenia. Psychiatry Res 221(3), 204–209. [DOI] [PubMed] [Google Scholar]
- Vita A, Barlati S, Ceraso A, Nibbio G, Ariu C, Deste G, Wykes T, 2021. Effectiveness, Core Elements, and Moderators of Response of Cognitive Remediation for Schizophrenia: A Systematic Review and Meta-analysis of Randomized Clinical Trials. JAMA Psychiatry. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weiser M, Heresco-Levy U, Davidson M, Javitt DC, Werbeloff N, Gershon AA, Abramovich Y, Amital D, Doron A, Konas S, Levkovitz Y, Liba D, Teitelbaum A, Mashiach M, Zimmerman Y, 2012. A multicenter, add-on randomized controlled trial of low-dose d-serine for negative and cognitive symptoms of schizophrenia. J Clin Psychiatry 73(6), e728–734. [DOI] [PubMed] [Google Scholar]
- Wykes T, Huddy V, Cellard C, McGurk SR, Czobor P, 2011. A meta-analysis of cognitive remediation for schizophrenia: methodology and effect sizes. Am J Psychiatry 168(5), 472–485. [DOI] [PubMed] [Google Scholar]
- Wykes T, Reeder C, Landau S, Matthiasson P, Haworth E, Hutchinson C, 2009. Does age matter? Effects of cognitive rehabilitation across the age span. Schizophr Res 113(2-3), 252–258. [DOI] [PubMed] [Google Scholar]
- Wynn JK, Roach BJ, McCleery A, Marder SR, Mathalon DH, Green MF, 2019. Evaluating visual neuroplasticity with EEG in schizophrenia outpatients. Schizophr Res 212, 40–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang YS, Marder SR, Green MF, 2017. Repurposing Drugs for Cognition in Schizophrenia. Clin Pharmacol Ther 101(2), 191–193. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.



