1
|
Ye N, Wang Q, Li Y, Zhen X. Current emerging therapeutic targets and clinical investigational agents for schizophrenia: Challenges and opportunities. Med Res Rev 2025; 45:755-787. [PMID: 39300769 DOI: 10.1002/med.22086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/14/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Since the first discovery of antipsychotics in the 1950s, targeting dopaminergic drugs has manifested to well manage the positive symptoms of schizophrenia with limited efficacy for the negative and cognitive symptoms. In past decades, extensive efforts have been undertaken towards the development of innovative agents that can effectively stabilize the dopamine and serotonin systems or target to nondopaminergic pathways, leading to various promising drug candidates entering into clinical trials. Notably, the sigma-2, 5-HT2A, and α1A receptor antagonist roluperidone, as well as a fixed-dose combination of the M1/4 receptor agonist KarXT, have been submitted for NDA applications. The dual agonist ulotaront, which targets TAAR1 and 5-HT1A receptors, and the GlyT1 inhibitor iclepertin have advanced into phase 3 clinical trials. Nevertheless, satisfactory therapeutic strategies for schizophrenia remain elusive. This review highlights current clinical endeavors in developing novel chemical small-molecule entities and fixed-dose combinations for the treatment of schizophrenia since 2017, thus facilitating the efficient development of the next generation of antipsychotics.
Collapse
Affiliation(s)
- Na Ye
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| | - Qi Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Yue Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases & College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, China
| |
Collapse
|
2
|
Lin CH, Lin E, Lane HY. Interpretable machine learning to evaluate relationships between DAO/DAOA (pLG72) protein data and features in clinical assessments, functional outcome, and cognitive function in schizophrenia patients. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2025; 11:27. [PMID: 39987274 PMCID: PMC11846841 DOI: 10.1038/s41537-024-00548-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 12/17/2024] [Indexed: 02/24/2025]
Abstract
Machine learning has been proposed to utilize D-amino acid oxidase (DAO) and DAO activator (DAOA [or pLG72]) protein levels to ascertain disease status in schizophrenia. However, it remains unclear whether machine learning can effectively evaluate clinical features in relation to DAO and DAOA in schizophrenia patients. We employed an interpretable machine learning (IML) framework including linear regression, least absolute shrinkage and selection operator (Lasso) models, and generalized additive models (GAMs) to analyze DAO/DAOA levels using 380 Taiwanese schizophrenia patients. Additionally, we incorporated 27 parameters encompassing demographic variables, clinical assessments, functional outcomes, and cognitive function as features. The IML framework facilitated linear and non-linear relationships between features and DAO/DAOA. DAO levels demonstrated significant associations with the 17-item Hamilton Depression Rating Scale (HAMD17) based on linear regression. The Lasso model identified four features-HAMD17, age, working memory, and overall cognitive function (OCF)-and highlighted HAMD17 as the most significant feature, using DAO from chronically stable patients. Utilizing DAOA from acutely exacerbated patients, the Lasso model also identified four features-OCF, Scale for the Assessment of Negative Symptoms 20-item, quality of life scale (QLS), and category fluency-and emphasized OCF as the most significant feature. Furthermore, GAMs revealed a non-linear relationship between category fluency and DAO in chronically stable patients, as well as between QLS and DAOA in acutely exacerbated patients. The study suggests that an IML framework holds promise for assessing linear and non-linear relationships between DAO/DAOA and various features in clinical assessments, functional outcomes, and cognitive function in patients with schizophrenia.
Collapse
Affiliation(s)
- Chieh-Hsin Lin
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Eugene Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Department of Biostatistics, University of Washington, Seattle, WA, 98195, USA.
| | - Hsien-Yuan Lane
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Department of Psychiatry, China Medical University Hospital, Taichung, Taiwan.
- Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan.
- Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan.
| |
Collapse
|
3
|
Sehatpour P, Kantrowitz JT. Finding the Right Dose: NMDA Receptor-Modulating Treatments for Cognitive and Plasticity Deficits in Schizophrenia and the Role of Pharmacodynamic Target Engagement. Biol Psychiatry 2025; 97:128-138. [PMID: 39218136 PMCID: PMC11634630 DOI: 10.1016/j.biopsych.2024.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Cognitive impairment associated with schizophrenia (CIAS) and related deficits in learning (plasticity) are among the leading causes of disability in schizophrenia. Despite this, there are no Food and Drug Administration-approved treatments for CIAS, and the development of treatments has been limited by numerous phase 2/3 failures of compounds that showed initial promise in small-scale studies. NMDA-type glutamate receptors (NMDARs) have been proposed to play an important role in schizophrenia; moreover, the NMDAR has a well-characterized role in cognition, learning, and neuroplasticity. We review previously published clinical trials in CIAS that focused on NMDAR modulator treatments, focusing on published and recent developments of the use of novel NMDAR-modulating treatments for CIAS both alone and combined with plasticity/learning paradigms to enhance learning. We use this discussion of previous studies to highlight the importance of incorporating pharmacodynamic target engagement biomarkers early in treatment development, which can help predict which compounds will succeed or fail in phase 3. A range of direct and indirect NMDAR modulators are covered, including D-serine, D-cycloserine, memantine, and glycine and first-generation glycine transport inhibitors (e.g., sarcosine and bitopertin), as well as recent positive studies of iclepertin, a novel glycine transport inhibitor, and luvadaxistat, a D-amino acid oxidase inhibitor that increases brain D-serine levels, and indirect noninvasive brain stimulation NMDAR-modulating treatments. Several examples of successful use of pharmacodynamic target engagement biomarkers for dose/drug discovery are emphasized, including the mismatch negativity, auditory steady state, and time-frequency event-related potential approaches.
Collapse
Affiliation(s)
- Pejman Sehatpour
- New York State Psychiatric Institute, New York, New York; College of Physicians and Surgeons, Columbia University, New York, New York; Nathan Kline Institute, Orangeburg, New York
| | - Joshua T Kantrowitz
- New York State Psychiatric Institute, New York, New York; College of Physicians and Surgeons, Columbia University, New York, New York; Nathan Kline Institute, Orangeburg, New York.
| |
Collapse
|
4
|
Biso L, Carli M, Scarselli M, Longoni B. Overview of Novel Antipsychotic Drugs: State of the Art, New Mechanisms, and Clinical Aspects of Promising Compounds. Biomedicines 2025; 13:85. [PMID: 39857669 PMCID: PMC11763187 DOI: 10.3390/biomedicines13010085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
Antipsychotic medications are a vast class of drugs used for the treatment of psychotic disorders such as schizophrenia. Although numerous compounds have been developed since their introduction in the 1950s, several patients do not adequately respond to current treatments, or they develop adverse reactions that cause treatment discontinuation. Moreover, in the past few decades, discoveries in the pathophysiology of psychotic disorders have opened the way for experimenting with novel compounds that have alternative mechanisms of action, with some of them showing promising results in early trials. The scope of this review was to summarize the novel antipsychotics developed, their current experimental status, and their mechanisms of action. In particular, we analyzed the main classes of investigational antipsychotics, such as monoamine, glutamate, acetylcholine, cannabinoid receptor modulators, enzyme inhibitors, ion channel modulators, and mixed receptor modulators. In addition, the safety profiles and adverse effects of these drugs were carefully evaluated, considering the relevance of these aspects for patients' drug adherence and quality of life, especially in the long-term treatment. Lastly, we tried to understand which compounds have greater potential to be approved by the principal drug regulatory agencies in the next years and if they could be used for diseases other than psychotic disorders.
Collapse
Affiliation(s)
| | | | | | - Biancamaria Longoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (L.B.); (M.C.); (M.S.)
| |
Collapse
|
5
|
Yoon J, Mayer MR, Berro T, Brazis S, Kantrowitz JT. Knowing is Half the Battle: The Factors Leading to Efficient Recruitment of Representative Samples in Schizophrenia Research. Pharmaceut Med 2025; 39:29-38. [PMID: 39794624 DOI: 10.1007/s40290-024-00545-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/13/2025]
Abstract
BACKGROUND AND OBJECTIVES Drug development in schizophrenia is limited by the differential scaling of the active treatment and placebo arms of a study, such that, as the number of sites increases, the magnitude of placebo response disproportionately increases. The objective of this article was to identify factors conducive to efficient recruitment as a step towards trial designs allowing recruitment of more participants per site, leading to reduced variability, and potentially a smaller placebo effect. PATIENTS AND METHODS Using the information of 554 individuals, we calculated the percentage of individuals who were screened, consented, and retained in our research, along with rationale for nonconsent. Independent t tests and Chi-squared tests were performed to compare participant characteristics. RESULTS Out of the 273 individuals who were fully screened, 84 did not progress to the consented stage owing to various reasons, leading to a total of 189 individuals who were screened and consented and a total of 365 individuals who were either incompletely screened or not consented into a study. Individuals with an externally validated medical history showed the highest yield in being consented and retained in research as new participants. In particular, chart reviews from clinics were highly efficient (25.8%) in facilitating new participants' enrollment, even when these individuals were not actively/prospectively seeking research. The most common reason for nonconsent was difficulty in telephone or email contact. Consenting and nonconsenting participants were similar in demographics, and recruitment sources only differed in their reported substance use. CONCLUSIONS Referrals and chart reviews from known clinics were the most efficient method in retaining new participants, highlighting the importance of external validation and communication between research and clinical staff within a system. Consenting participants showed no significant differences from the database as a whole, demonstrating that the study samples were representative. Future studies should aim to confirm the present findings at other academic and commercial research centers.
Collapse
Affiliation(s)
- Joohyun Yoon
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA.
| | - Megan R Mayer
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA
| | - Tala Berro
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA
| | - Stephanie Brazis
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Joshua T Kantrowitz
- New York State Psychiatric Institute, 1051 Riverside Drive, New York, NY, 10032, USA
- College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA
| |
Collapse
|
6
|
Terry-Lorenzo RT, Fan RH, Khin NA, Singh JB. Therapeutic potential of D-amino acid oxidase inhibitors for cognitive impairment associated with schizophrenia: learnings from luvadaxistat. Int J Neuropsychopharmacol 2024; 28:pyae066. [PMID: 39756412 PMCID: PMC11712274 DOI: 10.1093/ijnp/pyae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025] Open
Abstract
Hypofunction of the N-methyl-D-aspartate receptor (NMDAR) has been proposed to underlie the pathophysiology of schizophrenia, suggesting that promoting NMDAR activity may alleviate the negative or cognitive symptoms associated with schizophrenia. To circumvent excitotoxicity that may accompany direct agonism of the glutamate binding site on the NMDAR, therapeutic trials have focused on targeting the glycine binding site on the NMDAR. Direct administration of either glycine or D-serine, both of which are endogenous coagonists at the NMDAR glycine site, has yielded mixed outcomes across an array of clinical trials investigating different doses or patient populations. Furthermore, directly administering D-serine and glycine is challenging, and thus attention has turned to alternative, indirect methods that increase endogenous D-serine and glycine levels in the brain, such as D-amino acid oxidase (DAAO) inhibitors and glycine transporter 1 inhibitors, respectively. In this review, we provide an overview of the evidence supporting the potential of NMDAR modulators in general, and DAAO inhibitors in particular, as potential adjunctive treatments for schizophrenia. We also discuss the preclinical and clinical data related to luvadaxistat, an investigational highly selective and potent DAAO inhibitor that was under development for the treatment of the cognitive impairment associated with schizophrenia.
Collapse
Affiliation(s)
| | - Reuben H Fan
- Neurocrine Biosciences, Inc., San Diego, CA, United States
| | - Ni A Khin
- Neurocrine Biosciences, Inc., San Diego, CA, United States
| | | |
Collapse
|
7
|
Bajaj A, Tsukamoto T. Evolution of D-amino acid oxidase inhibitors: From concept to clinic. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 102:301-345. [PMID: 39929584 DOI: 10.1016/bs.apha.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
D-amino acid oxidase (DAAO) is a flavin-dependent peroxisomal monooxygenase with a substrate preference for glycine and certain small hydrophobic D-amino acids. Although the biochemical properties of the enzyme have been extensively studied since 1930s, the therapeutic interest in targeting the enzyme emerged more recently after the physiological significance of endogenous D-serine, a substrate for DAAO, was recognized in 1990s. This triggered a new wave of efforts by many researchers to develop more potent and drug-like DAAO inhibitors with greater translational potential. This chapter recounts the evolution of DAAO inhibitors since then driven by new molecular design strategies guided by structural biology. Some of these inhibitors were investigated in a range of preclinical in vivo studies to assess pharmacokinetics, pharmacodynamics, and behavioral pharmacology. Most importantly, these efforts culminated with the discovery of TAK-831 (luvadaxistat), an orally available brain-penetrant DAAO inhibitor currently under clinical development, representing a true bench-to-bedside success in this field.
Collapse
Affiliation(s)
- Ayush Bajaj
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Takashi Tsukamoto
- Johns Hopkins Drug Discovery, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
8
|
Kambeitz-Ilankovic L, Strube W, Baune BT, Falkai P, Röll L, Leucht S. [Cognitive impairments associated with schizophrenic psychoses : Diagnostics, course and therapy]. DER NERVENARZT 2024:10.1007/s00115-024-01773-8. [PMID: 39589503 DOI: 10.1007/s00115-024-01773-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/04/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Longitudinal outcome studies confirm that the majority of patients with schizophrenic psychoses develop cognitive impairments associated with schizophrenia (CIAS). OBJECTIVE To provide an overview of the epidemiology, diagnostics and evidence for various treatment options for CIAS. MATERIAL AND METHODS Literature review of the current level of evidence regarding the efficacy of different treatment strategies for CIAS. RESULTS Up to 85% of patients with schizophrenic psychoses exhibit CIAS, in some cases even before the development of positive or negative symptoms. The CIAS are associated with extensive individual burden due to impairments in many areas of cognitive and psychosocial functioning relevant to daily life. Various test instruments are available for clinical assessment with the Mental Health's Measurement and Treatment Research to Improve Cognition in Schizophrenia (MATRICS) consensus cognitive battery (MCCB) as an established standard for clinical trials and special clinical issues. The treatment of CIAS warrants a multimodal approach with non-drug strategies (e.g., cognitive remediation, exercise) currently providing the best level of evidence. Noninvasive neurostimulation procedures and dopaminergic antipsychotic drugs of the first and second generations have demonstrated low effectiveness on cognitive function disorders in schizophrenic psychoses. CONCLUSION The CIAS is a frequent disease-immanent symptom in schizophrenic psychoses that should be considered in the clinical routine as it substantially impairs those affected in the functional level and quality of life. Current treatment options are limited but innovative psychosocial interventions show low to moderate effects. In addition, new medications developed based on current neurobiological findings and combinations with psychosocial and neurostimulation procedures could open up new perspectives.
Collapse
Affiliation(s)
- Lana Kambeitz-Ilankovic
- Klinik und Poliklinik für Psychiatrie und Psychotherapie, Universitätsklinikum Köln, Köln, Deutschland
| | - Wolfgang Strube
- Klinik für Psychiatrie, Psychotherapie und Psychosomatik, Medizinische Fakultät, Universität Augsburg, Augsburg, Deutschland
- DZPG (Deutsches Zentrum für Psychische Gesundheit), Standort München/Augsburg, München, Deutschland
| | - Bernhard T Baune
- Klinik für Psychische Gesundheit, Universitätsklinikum Münster, Münster, Deutschland
- Department of Psychiatry, University of Melbourne, Melbourne, Australien
- Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, Australien
| | - Peter Falkai
- DZPG (Deutsches Zentrum für Psychische Gesundheit), Standort München/Augsburg, München, Deutschland
- Klinik für Psychiatrie und Psychotherapie, LMU Klinikum München, München, Deutschland
- Max-Planck-Institut für Psychiatrie, München, Deutschland
| | - Lukas Röll
- Klinik für Psychiatrie und Psychotherapie, LMU Klinikum München, München, Deutschland
- Max-Planck-Institut für Psychiatrie, München, Deutschland
| | - Stefan Leucht
- DZPG (Deutsches Zentrum für Psychische Gesundheit), Standort München/Augsburg, München, Deutschland.
- Klinik und Poliklinik für Psychiatrie und Psychotherapie, Technische Universität München, TUM School of Medicine and Health, Ismaninger Straße 22, 81675, München, Deutschland.
| |
Collapse
|
9
|
Tumdam R, Hussein Y, Garin-Shkolnik T, Stern S. NMDA Receptors in Neurodevelopmental Disorders: Pathophysiology and Disease Models. Int J Mol Sci 2024; 25:12366. [PMID: 39596430 PMCID: PMC11594297 DOI: 10.3390/ijms252212366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are critical components of the mammalian central nervous system, involved in synaptic transmission, plasticity, and neurodevelopment. This review focuses on the structural and functional characteristics of NMDARs, with a particular emphasis on the GRIN2 subunits (GluN2A-D). The diversity of GRIN2 subunits, driven by alternative splicing and genetic variants, significantly impacts receptor function, synaptic localization, and disease manifestation. The temporal and spatial expression of these subunits is essential for typical neural development, with each subunit supporting distinct phases of synaptic formation and plasticity. Disruptions in their developmental regulation are linked to neurodevelopmental disorders, underscoring the importance of understanding these dynamics in NDD pathophysiology. We explore the physiological properties and developmental regulation of these subunits, highlighting their roles in the pathophysiology of various NDDs, including ASD, epilepsy, and schizophrenia. By reviewing current knowledge and experimental models, including mouse models and human-induced pluripotent stem cells (hiPSCs), this article aims to elucidate different approaches through which the intricacies of NMDAR dysfunction in NDDs are currently being explored. The comprehensive understanding of NMDAR subunit composition and their mutations provides a foundation for developing targeted therapeutic strategies to address these complex disorders.
Collapse
Affiliation(s)
- Roshan Tumdam
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| | - Yara Hussein
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| | | | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| |
Collapse
|
10
|
Howes OD, Dawkins E, Lobo MC, Kaar SJ, Beck K. New Drug Treatments for Schizophrenia: A Review of Approaches to Target Circuit Dysfunction. Biol Psychiatry 2024; 96:638-650. [PMID: 38815885 DOI: 10.1016/j.biopsych.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/23/2024] [Accepted: 05/14/2024] [Indexed: 06/01/2024]
Abstract
Schizophrenia is a leading cause of global disease burden. Current drug treatments are associated with significant side effects and have limited efficacy for many patients, highlighting the need to develop new approaches that target other aspects of the neurobiology of schizophrenia. Preclinical, in vivo imaging, postmortem, genetic, and pharmacological studies have highlighted the key role of cortical GABAergic (gamma-aminobutyric acidergic)-glutamatergic microcircuits and their projections to subcortical dopaminergic circuits in the pathoetiology of negative, cognitive, and psychotic symptoms. Antipsychotics primarily act downstream of the dopaminergic component of this circuit. However, multiple drugs are currently in development that could target other elements of this circuit to treat schizophrenia. These include drugs for GABAergic or glutamatergic targets, including glycine transporters, D-amino acid oxidase, sodium channels, or potassium channels. Other drugs in development are likely to primarily act on pathways that regulate the dopaminergic system, such as muscarinic or trace amine receptors or 5-HT2A receptors, while PDE10A inhibitors are being developed to modulate the downstream consequences of dopaminergic dysfunction. Our review considers where new drugs may act on this circuit and their latest clinical trial evidence in terms of indication, efficacy, and side effects. Limitations of the circuit model, including whether there are neurobiologically distinct subgroups of patients, and future directions are also considered. Several drugs based on the mechanisms reviewed have promising clinical data, with the muscarinic agonist KarXT most advanced. If these drugs are approved for clinical use, they have the potential to revolutionize understanding of the pathophysiology and treatment of schizophrenia.
Collapse
Affiliation(s)
- Oliver D Howes
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom; South London and Maudsley NHS Foundation Trust, Maudsley Hospital, London, United Kingdom.
| | - Eleanor Dawkins
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; South London and Maudsley NHS Foundation Trust, Maudsley Hospital, London, United Kingdom
| | - Maria C Lobo
- South London and Maudsley NHS Foundation Trust, Maudsley Hospital, London, United Kingdom
| | - Stephen J Kaar
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; Division of Psychology and Mental Health, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom; Greater Manchester Mental Health National Health Service Foundation Trust, Manchester, United Kingdom
| | - Katherine Beck
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom; South London and Maudsley NHS Foundation Trust, Maudsley Hospital, London, United Kingdom
| |
Collapse
|
11
|
Vita A, Barlati S, Cavallaro R, Mucci A, Riva MA, Rocca P, Rossi A, Galderisi S. Definition, assessment and treatment of cognitive impairment associated with schizophrenia: expert opinion and practical recommendations. Front Psychiatry 2024; 15:1451832. [PMID: 39371908 PMCID: PMC11450451 DOI: 10.3389/fpsyt.2024.1451832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/22/2024] [Indexed: 10/08/2024] Open
Abstract
A considerable proportion of patients with schizophrenia perform below population norms on standardized neuropsychological tests, and the performance of those performing within normal range is lower than predicted based on parental education. Cognitive impairment predates the onset of psychosis, is observed during symptom remission and in non-affected first-degree relatives of patients. At the present time, cognitive deficits are regarded as key features of schizophrenia, important determinants of poor psychosocial outcome and targets for both pharmacological and non-pharmacological treatment strategies. A group of eight key opinion leaders reviewed and discussed latest advances in scientific research and current good clinical practices on assessment, management, and treatment of CIAS. In the present paper they summarize the current evidence, identify main gaps between current knowledge and mental health services clinical practice, and provide practical recommendations to reduce the gap.
Collapse
Affiliation(s)
- Antonio Vita
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Department of Mental Health and Addiction Services, Azienda Socio-Sanitaria Territoriale (ASST) Spedali Civili of, Brescia, Italy
| | - Stefano Barlati
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Department of Mental Health and Addiction Services, Azienda Socio-Sanitaria Territoriale (ASST) Spedali Civili of, Brescia, Italy
| | - Roberto Cavallaro
- Department of Clinical Neurosciences, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) San Raffaele Scientific Institute, Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Armida Mucci
- Department of Mental and Physical Health and Preventive Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Marco A. Riva
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
- Biological Psychiatry Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Paola Rocca
- Department of Neuroscience “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Alessandro Rossi
- Department of Biotechnological and Applied Clinical Sciences, Section of Psychiatry, University of L’Aquila, L’Aquila, Italy
| | - Silvana Galderisi
- Department of Mental and Physical Health and Preventive Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| |
Collapse
|
12
|
Calzavara-Pinton I, Nibbio G, Barlati S, Bertoni L, Necchini N, Zardini D, Baglioni A, Paolini S, Poddighe L, Bulgari V, Lisoni J, Deste G, Vita A. Treatment of Cognitive Impairment Associated with Schizophrenia Spectrum Disorders: New Evidence, Challenges, and Future Perspectives. Brain Sci 2024; 14:791. [PMID: 39199483 PMCID: PMC11352256 DOI: 10.3390/brainsci14080791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 07/29/2024] [Accepted: 08/03/2024] [Indexed: 09/01/2024] Open
Abstract
Cognitive impairment associated with schizophrenia (CIAS) represents one of the core features of the disorder and has a significant impact on functional and rehabilitation outcomes of people living with schizophrenia spectrum disorders (SSD). The aim of this critical review is to highlight the most recent evidence on effective treatments available for CIAS, to discuss the current challenges in this field, and to present future perspectives that may help to overcome them. Concerning psychopharmacological approaches, among the most indicated strategies for the management and prevention of CIAS is to favor second-generation antipsychotic medications and avoid long-term and high-dose treatments with anticholinergic medications and benzodiazepines. Moreover, non-pharmacological approaches such as cognitive remediation and physical exercise-based programs represent evidence-based interventions in the treatment of CIAS that have shown reliable evidence of effectiveness on both cognitive and functional outcomes. These treatments, however, are still delivered to people accessing mental health services with a diagnosis of CIAS in an uneven manner, even in high-income countries. Academic and clinical partnership and collaboration, as well as advocacy from service users, families, carers, and stakeholders' organizations could help to reduce the bench to bedside gap in the treatment of CIAS. Future perspectives include the development of novel pharmacological agents that could be effective in the treatment of CIAS, the implementation of novel technologies such as telemedicine and virtual reality in the delivery of evidence-based interventions to improve accessibility and engagement, and further research in the field of non-invasive brain stimulation.
Collapse
Affiliation(s)
- Irene Calzavara-Pinton
- Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, 25123 Brescia, Italy; (I.C.-P.); (L.P.); (J.L.); (A.V.)
| | - Gabriele Nibbio
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.N.); (L.B.); (N.N.); (D.Z.); (A.B.); (S.P.); (V.B.); (G.D.)
| | - Stefano Barlati
- Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, 25123 Brescia, Italy; (I.C.-P.); (L.P.); (J.L.); (A.V.)
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.N.); (L.B.); (N.N.); (D.Z.); (A.B.); (S.P.); (V.B.); (G.D.)
| | - Lorenzo Bertoni
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.N.); (L.B.); (N.N.); (D.Z.); (A.B.); (S.P.); (V.B.); (G.D.)
| | - Nicola Necchini
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.N.); (L.B.); (N.N.); (D.Z.); (A.B.); (S.P.); (V.B.); (G.D.)
| | - Daniela Zardini
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.N.); (L.B.); (N.N.); (D.Z.); (A.B.); (S.P.); (V.B.); (G.D.)
| | - Antonio Baglioni
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.N.); (L.B.); (N.N.); (D.Z.); (A.B.); (S.P.); (V.B.); (G.D.)
| | - Stefano Paolini
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.N.); (L.B.); (N.N.); (D.Z.); (A.B.); (S.P.); (V.B.); (G.D.)
| | - Laura Poddighe
- Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, 25123 Brescia, Italy; (I.C.-P.); (L.P.); (J.L.); (A.V.)
| | - Viola Bulgari
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.N.); (L.B.); (N.N.); (D.Z.); (A.B.); (S.P.); (V.B.); (G.D.)
| | - Jacopo Lisoni
- Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, 25123 Brescia, Italy; (I.C.-P.); (L.P.); (J.L.); (A.V.)
| | - Giacomo Deste
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.N.); (L.B.); (N.N.); (D.Z.); (A.B.); (S.P.); (V.B.); (G.D.)
- Department of Mental Health, ASST Valcamonica, 25040 Brescia, Italy
| | - Antonio Vita
- Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, 25123 Brescia, Italy; (I.C.-P.); (L.P.); (J.L.); (A.V.)
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.N.); (L.B.); (N.N.); (D.Z.); (A.B.); (S.P.); (V.B.); (G.D.)
| |
Collapse
|
13
|
Murthy V, Hanson E, DeMartinis N, Asgharnejad M, Dong C, Evans R, Ge T, Dunayevich E, Singh JB, Ratti E, Galderisi S. INTERACT: a randomized phase 2 study of the DAAO inhibitor luvadaxistat in adults with schizophrenia. Schizophr Res 2024; 270:249-257. [PMID: 38943928 DOI: 10.1016/j.schres.2024.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/31/2024] [Accepted: 06/15/2024] [Indexed: 07/01/2024]
Abstract
Deficits in N-methyl-d-aspartate receptor (NMDAR) signaling are implicated in the pathogenesis of schizophrenia. Luvadaxistat (TAK-831/NBI-1065844) is an investigational d-amino acid oxidase (DAAO) inhibitor that increases d-serine levels at NMDAR coagonist sites. INTERACT is a phase 2 randomized, placebo-controlled study that evaluated the efficacy and safety of three doses of luvadaxistat, covering a range of DAAO occupancy and d-serine levels, in patients with schizophrenia with persistent negative symptoms. The study included a 14-day, single-blinded placebo run-in period and a 12-week, double-blinded treatment period. The primary efficacy endpoint was the 12-week change from baseline in Positive and Negative Syndrome Scale-Negative Symptom Factor Score (PANSS NSFS). Secondary efficacy endpoints included the 12-week changes from baseline in Brief Assessment of Cognition in Schizophrenia (BACS) score and Schizophrenia Cognition Rating Scale (SCoRS) score. Safety endpoints included adverse event assessments. The full analysis set included all randomized patients (N = 256 [placebo, n = 87; luvadaxistat 50 mg, n = 58; 125 mg, n = 56; 500 mg, n = 55]); 228 patients completed the study. No significant improvements in PANSS NSFS were observed at any dose versus placebo at week 12. Improvements were observed with luvadaxistat 50 mg versus placebo in cognitive endpoints: BACS composite score (nominal one-sided p = 0.031) and SCoRS interviewer total score (nominal one-sided p = 0.011). Luvadaxistat did not significantly improve negative symptoms of schizophrenia. However, luvadaxistat 50 mg met the prespecified secondary endpoints for cognitive performance (BACS) and function (SCoRS), warranting further investigation in patients with cognitive impairment associated with schizophrenia. Luvadaxistat was well-tolerated in INTERACT, with no new safety signals observed. ClinicalTrials.gov: NCT03382639.
Collapse
Affiliation(s)
- Venkatesha Murthy
- Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States.
| | - Elizabeth Hanson
- Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States
| | - Nicholas DeMartinis
- Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States
| | - Mahnaz Asgharnejad
- Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States
| | - Cheng Dong
- Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States
| | - Rebecca Evans
- Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States
| | - Tingting Ge
- Neurocrine Biosciences, Inc., San Diego, California, United States
| | - Eduardo Dunayevich
- Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States; Neurocrine Biosciences, Inc., San Diego, California, United States
| | - Jaskaran B Singh
- Neurocrine Biosciences, Inc., San Diego, California, United States
| | - Emiliangelo Ratti
- Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts, United States
| | | |
Collapse
|
14
|
Garofalo M, De Simone G, Motta Z, Nuzzo T, De Grandis E, Bruno C, Boeri S, Riccio MP, Pastore L, Bravaccio C, Iasevoli F, Salvatore F, Pollegioni L, Errico F, de Bartolomeis A, Usiello A. Decreased free D-aspartate levels in the blood serum of patients with schizophrenia. Front Psychiatry 2024; 15:1408175. [PMID: 39050919 PMCID: PMC11266155 DOI: 10.3389/fpsyt.2024.1408175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction Schizophrenia (SCZ) and autism spectrum disorder (ASD) are neurodevelopmental diseases characterized by different psychopathological manifestations and divergent clinical trajectories. Various alterations at glutamatergic synapses have been reported in both disorders, including abnormal NMDA and metabotropic receptor signaling. Methods We conducted a bicentric study to assess the blood serum levels of NMDA receptors-related glutamatergic amino acids and their precursors, including L-glutamate, L-glutamine, D-aspartate, L-aspartate, L-asparagine, D-serine, L-serine and glycine, in ASD, SCZ patients and their respective control subjects. Specifically, the SCZ patients were subdivided into treatment-resistant and non-treatment-resistant SCZ patients, based on their responsivity to conventional antipsychotics. Results D-serine and D-aspartate serum reductions were found in SCZ patients compared to controls. Conversely, no significant differences between cases and controls were found in amino acid concentrations in the two ASD cohorts analyzed. Discussion This result further encourages future research to evaluate the predictive role of selected D-amino acids as peripheral markers for SCZ pathophysiology and diagnosis.
Collapse
Affiliation(s)
- Martina Garofalo
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Giuseppe De Simone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, Naples, Italy
| | - Zoraide Motta
- ”The Protein Factory 2.0”, Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell’Insubria, Varese, Italy
| | - Tommaso Nuzzo
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
| | - Elisa De Grandis
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal, and Child Health - DINOGMI, University of Genoa, Genoa, Italy
| | - Claudio Bruno
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal, and Child Health - DINOGMI, University of Genoa, Genoa, Italy
- Center of Translational and Experimental Myology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Giannina Gaslini, Genoa, Italy
| | - Silvia Boeri
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal, and Child Health - DINOGMI, University of Genoa, Genoa, Italy
| | - Maria Pia Riccio
- Department of Maternal and Child Health, Unità Operativa semplice di Dipartimento (UOSD) of Child and Adolescent Psychiatry, Azienda Ospedaliera Universitaria (AOU) Federico II, Naples, Italy
| | - Lucio Pastore
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Naples, Italy
| | - Carmela Bravaccio
- Department of Medical and Translational Sciences, Child Neuropsychiatry, Federico II University, Napoli, Italy
| | - Felice Iasevoli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, Naples, Italy
| | - Francesco Salvatore
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- Centro Interuniversitario per Malattie Multigeniche e Multifattoriali e loro Modelli Animali (Federico II, Naples; Tor Vergata, Rome and “G. D’Annunzio”, Chieti-Pescara), Naples, Italy
| | - Loredano Pollegioni
- ”The Protein Factory 2.0”, Dipartimento di Biotecnologie e Scienze della Vita, Università degli Studi dell’Insubria, Varese, Italy
| | - Francesco Errico
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- Dipartimento di Agraria, Università degli Studi di Napoli “Federico II”, Portici, Italy
| | - Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Odontostomatology, University Medical School of Naples “Federico II”, Naples, Italy
| | - Alessandro Usiello
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
| |
Collapse
|
15
|
Faris P, Pischedda D, Palesi F, D’Angelo E. New clues for the role of cerebellum in schizophrenia and the associated cognitive impairment. Front Cell Neurosci 2024; 18:1386583. [PMID: 38799988 PMCID: PMC11116653 DOI: 10.3389/fncel.2024.1386583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Schizophrenia (SZ) is a complex neuropsychiatric disorder associated with severe cognitive dysfunction. Although research has mainly focused on forebrain abnormalities, emerging results support the involvement of the cerebellum in SZ physiopathology, particularly in Cognitive Impairment Associated with SZ (CIAS). Besides its role in motor learning and control, the cerebellum is implicated in cognition and emotion. Recent research suggests that structural and functional changes in the cerebellum are linked to deficits in various cognitive domains including attention, working memory, and decision-making. Moreover, cerebellar dysfunction is related to altered cerebellar circuit activities and connectivity with brain regions associated with cognitive processing. This review delves into the role of the cerebellum in CIAS. We initially consider the major forebrain alterations in CIAS, addressing impairments in neurotransmitter systems, synaptic plasticity, and connectivity. We then focus on recent findings showing that several mechanisms are also altered in the cerebellum and that cerebellar communication with the forebrain is impaired. This evidence implicates the cerebellum as a key component of circuits underpinning CIAS physiopathology. Further studies addressing cerebellar involvement in SZ and CIAS are warranted and might open new perspectives toward understanding the physiopathology and effective treatment of these disorders.
Collapse
Affiliation(s)
- Pawan Faris
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Doris Pischedda
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Fulvia Palesi
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Egidio D’Angelo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
- Digital Neuroscience Center, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
16
|
DeLisi LE. A lifetime of mental health treatments for people with schizophrenia: update and narrative review. Curr Opin Psychiatry 2024; 37:140-146. [PMID: 38512852 DOI: 10.1097/yco.0000000000000926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
PURPOSE OF REVIEW To provide a summary of the most up-to-date thoughts about treatment for schizophrenia at different stages of illness. RECENT FINDINGS The use of Coordinated Specialty Care clinics has arisen as the standard for early on in the treatment of psychosis, providing the notion that recovery is possible. New medications that do not depend on postsynaptic dopamine receptor blockade are soon becoming available. SUMMARY A focus should be made by clinicians to personalize treatment plans for each patient who has the possibility of being diagnosed with a primary psychosis and the plan should be to predict outcomes based on biological markers that include genetic vulnerability, early psychosocial combined with pharmacological treatments as needed and then a plan to determine or maintain treatments going forward into the future. It is important to individualize treatment by stage of illness, as well as characteristics of the individual patient. Research is ongoing to advance knowledge for interventions at each stage from the premorbid period through to chronicity.
Collapse
Affiliation(s)
- Lynn E DeLisi
- Department of Psychiatry, Cambridge Health Alliance, Department of Psychiatry, Harvard Medical School, Cambridge, Massachusetts, USA
| |
Collapse
|
17
|
Okada N, Yahata N, Koshiyama D, Morita K, Sawada K, Kanata S, Fujikawa S, Sugimoto N, Toriyama R, Masaoka M, Koike S, Araki T, Kano Y, Endo K, Yamasaki S, Ando S, Nishida A, Hiraiwa-Hasegawa M, Edden RAE, Sawa A, Kasai K. Longitudinal trajectories of anterior cingulate glutamate and subclinical psychotic experiences in early adolescence: the impact of bullying victimization. Mol Psychiatry 2024; 29:939-950. [PMID: 38182806 PMCID: PMC11176069 DOI: 10.1038/s41380-023-02382-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 12/06/2023] [Accepted: 12/13/2023] [Indexed: 01/07/2024]
Abstract
Previous studies reported decreased glutamate levels in the anterior cingulate cortex (ACC) in non-treatment-resistant schizophrenia and first-episode psychosis. However, ACC glutamatergic changes in subjects at high-risk for psychosis, and the effects of commonly experienced environmental emotional/social stressors on glutamatergic function in adolescents remain unclear. In this study, adolescents recruited from the general population underwent proton magnetic resonance spectroscopy (MRS) of the pregenual ACC using a 3-Tesla scanner. We explored longitudinal data on the association of combined glutamate-glutamine (Glx) levels, measured by MRS, with subclinical psychotic experiences. Moreover, we investigated associations of bullying victimization, a risk factor for subclinical psychotic experiences, and help-seeking intentions, a coping strategy against stressors including bullying victimization, with Glx levels. Finally, path analyses were conducted to explore multivariate associations. For a contrast analysis, gamma-aminobutyric acid plus macromolecule (GABA+) levels were also analyzed. Negative associations were found between Glx levels and subclinical psychotic experiences at both Times 1 (n = 219, mean age 11.5 y) and 2 (n = 211, mean age 13.6 y), as well as for over-time changes (n = 157, mean interval 2.0 y). Moreover, effects of bullying victimization and bullying victimization × help-seeking intention interaction effects on Glx levels were found (n = 156). Specifically, bullying victimization decreased Glx levels, whereas help-seeking intention increased Glx levels only in bullied adolescents. Finally, associations among bullying victimization, help-seeking intention, Glx levels, and subclinical psychotic experiences were revealed. GABA+ analysis revealed no significant results. This is the first adolescent study to reveal longitudinal trajectories of the association between glutamatergic function and subclinical psychotic experiences and to elucidate the effect of commonly experienced environmental emotional/social stressors on glutamatergic function. Our findings may deepen the understanding of how environmental emotional/social stressors induce impaired glutamatergic neurotransmission that could be the underpinning of liability for psychotic experiences in early adolescence.
Collapse
Affiliation(s)
- Naohiro Okada
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan.
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan.
| | - Noriaki Yahata
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
- Institute for Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba, Chiba, 263-8555, Japan
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, National Institutes for Quantum Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba, Chiba, 263-8555, Japan
| | - Daisuke Koshiyama
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kentaro Morita
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kingo Sawada
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
- Center for Research on Counseling and Support Services, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Sho Kanata
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Psychiatry, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-ku, Tokyo, 173-8605, Japan
| | - Shinya Fujikawa
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Noriko Sugimoto
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Rie Toriyama
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Mio Masaoka
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shinsuke Koike
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
- The University of Tokyo Institute for Diversity and Adaptation of Human Mind (UTIDAHM), The University of Tokyo, Komaba 3-8-1, Meguro-ku, Tokyo, 153-8902, Japan
| | - Tsuyoshi Araki
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
- Department of Psychiatry, Teikyo University Mizonokuchi Hospital, Futago 5-1-1, Takatsu-ku, Kawasaki, Kanagawa, 213-8507, Japan
| | - Yukiko Kano
- Department Child Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kaori Endo
- Research Center for Social Science & Medicine, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa 2-1-6, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Syudo Yamasaki
- Research Center for Social Science & Medicine, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa 2-1-6, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Shuntaro Ando
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
- Research Center for Social Science & Medicine, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa 2-1-6, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Atsushi Nishida
- Research Center for Social Science & Medicine, Tokyo Metropolitan Institute of Medical Science, Kamikitazawa 2-1-6, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Mariko Hiraiwa-Hasegawa
- Department of Evolutionary Studies of Biosystems, School of Advanced Sciences, The Graduate University for Advanced Studies (SOKENDAI), Shonan Village, Hayama, Kanagawa, 240-0193, Japan
| | - Richard A E Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, 600 N Wolfe St, Baltimore, MD, 21287, USA
- F. M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, 707 N Broadway Street, Baltimore, MD, 21205, USA
| | - Akira Sawa
- Departments of Psychiatry, Neuroscience, Biomedical Engineering, Genetic Medicine, and Pharmacology, Johns Hopkins University School of Medicine, 600 N Wolfe St, Baltimore, MD, 21287, USA
- Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, 600 N Wolfe St, Baltimore, MD, 21287, USA
| | - Kiyoto Kasai
- Department of Neuropsychiatry, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-8655, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
18
|
Vita A, Nibbio G, Barlati S. Pharmacological Treatment of Cognitive Impairment Associated With Schizophrenia: State of the Art and Future Perspectives. SCHIZOPHRENIA BULLETIN OPEN 2024; 5:sgae013. [PMID: 39144119 PMCID: PMC11207676 DOI: 10.1093/schizbullopen/sgae013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Cognitive Impairment Associated with Schizophrenia (CIAS) represents one of the core dimensions of Schizophrenia Spectrum Disorders (SSD), with an important negative impact on real-world functional outcomes of people living with SSD. Treatment of CIAS represents a therapeutic goal of considerable importance, and while cognition-oriented evidence-based psychosocial interventions are available, effective pharmacological treatment could represent a game-changer in the lives of people with SSD. The present critical review reports and discusses the evidence regarding the effects of several pharmacological agents that are available in clinical practice or are under study, commenting on both current and future perspectives of CIAS treatment. In particular, the effects on CIAS of antipsychotic medications, anticholinergic medications, benzodiazepines, which are currently commonly used in the treatment of SSD, and of iclepertin, d-serine, luvadaxistat, xanomeline-trospium, ulotaront, anti-inflammatory molecules, and oxytocin, which are undergoing regulatory trials or can be considered as experimental agents, will be reported and discussed. Currently, available pharmacological agents do not appear to provide substantial benefits on CIAS, but accurate management of antipsychotic medications and avoiding treatments that can further exacerbate CIAS represent important strategies. Some molecules that are currently being investigated in Phase 2 and Phase 3 trials have provided very promising preliminary results, but more information is currently required to assess their effectiveness in real-world contexts and to provide clear recommendations regarding their use in clinical practice. The results of ongoing and future studies will reveal whether any of these molecules represents the awaited pharmacological game-changer in the treatment of CIAS.
Collapse
Affiliation(s)
- Antonio Vita
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Gabriele Nibbio
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Stefano Barlati
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- Department of Mental Health and Addiction Services, ASST Spedali Civili of Brescia, Brescia, Italy
| |
Collapse
|
19
|
O'Donnell P, Buhl DL, Johannesen J, Lijffijt M. Neural Circuitry-Related Biomarkers for Drug Development in Psychiatry: An Industry Perspective. ADVANCES IN NEUROBIOLOGY 2024; 40:45-65. [PMID: 39562440 DOI: 10.1007/978-3-031-69491-2_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Drug development in psychiatry has been hampered by the lack of reliable ways to determine the neurobiological effects of the assets tested, difficulties in identifying patient subsets more amenable to benefit from a given asset, and issues with executing trials in a manner that would convincingly provide answers. An emerging idea in many companies is to validate tools to address changes in neural circuits by pharmacological tools as a key piece in quantifying the effects of our drugs. Here, we review past, present, and emerging approaches to capture the outcome of the modulation of brain circuits. The field is now ripe for implementing these approaches in drug development.
Collapse
Affiliation(s)
| | - Derek L Buhl
- Precision Medicine, Abbvie, Inc, Cambridge, MA, USA
| | | | | |
Collapse
|
20
|
Kantrowitz JT, Javitt DC. The Less Things Change, the More They Remain the Same: Impaired Neural Plasticity as a Critical Target for Drug Development in Neuropsychiatry. ADVANCES IN NEUROBIOLOGY 2024; 40:801-828. [PMID: 39562464 DOI: 10.1007/978-3-031-69491-2_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Neuropsychiatric disability is related to reduced ability to change in response to clinical interventions, e.g., plasticity. Study of biomarkers and interventional strategies for plasticity, however, are sparse. In this chapter, we focus on the serial frequency discrimination task (SFDT), which is sensitive to impairments in early auditory processing (EAP) and auditory learning and has been most thoroughly studied in dyslexia and schizophrenia. In the SFDT, participants are presented with repeated paired tones ("reference" and "test") and indicate which tone is higher in pitch. Plasticity during the SFDT is critically dependent upon interactions between prefrontal "cognitive control" regions, and lower-level perceptual and motor regions that may be detected using both fMRI and time-frequency event-related potential (TF-ERP) approaches. Additionally, interactions between the cortex and striatum give insights into glutamate/dopamine interaction mechanisms. The SFDT task has been utilized in the development of N-methyl-D-aspartate receptor (NMDAR) targeted medications, which significantly modulate sensory and premotor neurophysiological activity. Deficits in pitch processing play a critical role in impaired neuro- and social cognitive function in schizophrenia and may contribute to similar impairments in dyslexia. Thus, the SFDT may be ideal for development of treatments aimed at amelioration of neuro- and social cognitive deficits across neuropsychiatric disorders.
Collapse
Affiliation(s)
- Joshua T Kantrowitz
- New York State Psychiatric Institute, New York, NY, USA.
- College of Physicians and Surgeons, Columbia University, New York, NY, USA.
- Nathan Kline Institute, Orangeburg, NY, USA.
| | - Daniel C Javitt
- College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Nathan Kline Institute, Orangeburg, NY, USA
| |
Collapse
|
21
|
Javitt DC. Mismatch Negativity (MMN) as a Pharmacodynamic/Response Biomarker for NMDA Receptor and Excitatory/Inhibitory Imbalance-Targeted Treatments in Schizophrenia. ADVANCES IN NEUROBIOLOGY 2024; 40:411-451. [PMID: 39562453 DOI: 10.1007/978-3-031-69491-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
Schizophrenia is a major mental disorder that affects approximately 0.5% of the population worldwide. Persistent negative symptoms and cognitive impairments associated with schizophrenia (CIAS) are key features of the disorder and primary predictors of long-term disability. At the neurochemical level, both CIAS and negative symptoms are potentially attributable to dysfunction or dysregulation of N-methyl-D-aspartate receptor (NMDAR)-mediated neurotransmission within cortical and subcortical brain regions. At present, there are no approved treatments for either CIAS or persistent negative symptoms. Development of novel treatments, moreover, is limited by the lack of biomarkers that can be used translationally across preclinical and early-stage clinical investigation. The present chapter describes the use of mismatch negativity (MMN) as a pharmacodynamic/response (PD/R) biomarker for early-stage clinical investigation of NMDAR targeted therapies for schizophrenia. MMN indexes dysfunction of early auditory processing (EAP) in schizophrenia. In humans, deficits in MMN generation contribute hierarchically to impaired cognition and functional outcome. Across humans, rodents, and primates, MMN has been linked to impaired NMDAR function and resultant disturbances in excitatory/inhibitory (E/I) balance involving interactions between glutamatergic (excitatory) pyramidal and GABAeric (inhibitory) local circuit neurons. In early-stage clinical trials, MMN has shown sensitivity to the acute effects of novel pharmacological treatments. These findings support use of MMN as a pharmacodynamic/response biomarker to support preclinical drug discovery and early-stage proof-of-mechanisms studies in schizophrenia and other related neuropsychiatric disorders.
Collapse
Affiliation(s)
- Daniel C Javitt
- Division of Experimental Therapeutics, College of Physicians and Surgeons, Columbia University, New York, NY, USA.
- Schizophrenia Research Division, Nathan S. Kline Institute for Psychiatric Research, Orangeburg, NY, USA.
| |
Collapse
|
22
|
Li C, Zheng Z, Ha P, Jiang W, Soo C, Ting K. Neural EGFL-like 1, a craniosynostosis-related osteochondrogenic molecule, strikingly associates with neurodevelopmental pathologies. Cell Biosci 2023; 13:227. [PMID: 38102659 PMCID: PMC10725010 DOI: 10.1186/s13578-023-01174-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
Various craniofacial syndromes cause skeletal malformations and are accompanied by neurological abnormalities at different levels, leading to tremendous biomedical, financial, social, and psychological burdens. Accumulating evidence highlights the importance of identifying and characterizing the genetic basis that synchronously modulates musculoskeletal and neurobehavioral development and function. Particularly, previous studies from different groups have suggested that neural EGFL-like-1 (Nell-1), a well-established osteochondrogenic inducer whose biopotency was initially identified in the craniofacial tissues, may also play a vital role in the central nervous system, particularly regarding neurological disorder pathologies. To provide first-hand behavior evidence if Nell-1 also has a role in central nervous system abnormalities, we compared the Nell-1-haploinsufficient (Nell-1+/6R) mice with their wild-type counterparts regarding their repetitive, social communication, anxiety-related, locomotor, sensory processing-related, motor coordination, and Pavlovian learning and memory behaviors, as well as their hippocampus transcriptional profile. Interestingly, Nell-1+/6R mice demonstrated core autism spectrum disorder-like deficits, which could be corrected by Risperidone, an FDA-approved anti-autism, anti-bipolar medicine. Besides, transcriptomic analyses identified 269 differential expressed genes, as well as significantly shifted alternative splicing of ubiquitin B pseudogene Gm1821, in the Nell-1+/6R mouse hippocampus, which confirmed that Nell-1 plays a role in neurodevelopment. Therefore, the current study verifies that Nell-1 regulates neurological development and function for the first time. Moreover, this study opens new avenues for understanding and treating craniofacial patients suffering from skeletal deformities and behavior, memory, and cognition difficulties by uncovering a novel bone-brain-crosstalk network. Furthermore, the transcriptomic analysis provides the first insight into deciphering the mechanism of Nell-1 in neurodevelopment.
Collapse
Affiliation(s)
- Chenshuang Li
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Zhong Zheng
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Pin Ha
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Wenlu Jiang
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Chia Soo
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Orthopedic Hospital Research Center and David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- MacDonald Research Laboratories (MRL), 675 Charles E. Young Dr. South Room 2641A, Box 951759, Los Angeles, CA, 90095-1759, USA.
| | - Kang Ting
- American Dental Association Forsyth Institute, 245 First Street, Cambridge, MA, 02142, USA.
| |
Collapse
|
23
|
Souza INDO, Roychaudhuri R, de Belleroche J, Mothet JP. d-Amino acids: new clinical pathways for brain diseases. Trends Mol Med 2023; 29:1014-1028. [PMID: 37770379 DOI: 10.1016/j.molmed.2023.09.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 09/30/2023]
Abstract
Free d-amino acids (d-AAs) are emerging as a novel and important class of signaling molecules in many organs, including the brain and endocrine systems. There has been considerable progress in our understanding of the fundamental roles of these atypical messengers, with increasingly recognized implications in a wide range of neuropathologies, including schizophrenia (SCZ), epilepsy, Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), substance abuse, and chronic pain, among others. Research has enabled the discovery that d-serine, d-aspartate and more recently d-cysteine are essential for the healthy development and function of the central nervous system (CNS). We discuss recent progress that has profoundly transformed our vision of numerous physiological processes but has also shown how d-AAs are now offering therapeutic promise in clinical settings for several human diseases.
Collapse
Affiliation(s)
- Isis Nem de Oliveira Souza
- Biophotonics and Synapse Physiopathology Team, Laboratoire LuMIn UMR9024 Université Paris-Saclay, ENS Paris-Saclay, CNRS, CentraleSupelec, 91190 Gif-sur-Yvette, France; Molecular Pharmacology Laboratory, Biomedical Sciences Institute, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robin Roychaudhuri
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Center for Birth Defects, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jacqueline de Belleroche
- Neurogenetics Group, Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Jean-Pierre Mothet
- Biophotonics and Synapse Physiopathology Team, Laboratoire LuMIn UMR9024 Université Paris-Saclay, ENS Paris-Saclay, CNRS, CentraleSupelec, 91190 Gif-sur-Yvette, France.
| |
Collapse
|
24
|
Fradley R, Goetghebeur P, Miller D, Burley R, Almond S, Gruart I Massó A, Delgado García JM, Zhu B, Howley E, Neill JC, Grayson B, Gaskin P, Carlton M, Gray I, Serrats J, Davies CH. Luvadaxistat: A Novel Potent and Selective D-Amino Acid Oxidase Inhibitor Improves Cognitive and Social Deficits in Rodent Models for Schizophrenia. Neurochem Res 2023; 48:3027-3041. [PMID: 37289348 PMCID: PMC10471729 DOI: 10.1007/s11064-023-03956-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/17/2023] [Accepted: 05/21/2023] [Indexed: 06/09/2023]
Abstract
N-methyl-D-aspartate (NMDA) receptor hypofunctionality is a well-studied hypothesis for schizophrenia pathophysiology, and daily dosing of the NMDA receptor co-agonist, D-serine, in clinical trials has shown positive effects in patients. Therefore, inhibition of D-amino acid oxidase (DAAO) has the potential to be a new therapeutic approach for the treatment of schizophrenia. TAK-831 (luvadaxistat), a novel, highly potent inhibitor of DAAO, significantly increases D-serine levels in the rodent brain, plasma, and cerebrospinal fluid. This study shows luvadaxistat to be efficacious in animal tests of cognition and in a translational animal model for cognitive impairment in schizophrenia. This is demonstrated when luvadaxistat is dosed alone and in conjunction with a typical antipsychotic. When dosed chronically, there is a suggestion of change in synaptic plasticity as seen by a leftward shift in the maximum efficacious dose in several studies. This is suggestive of enhanced activation of NMDA receptors in the brain and confirmed by modulation of long-term potentiation after chronic dosing. DAAO is highly expressed in the cerebellum, an area of increasing interest for schizophrenia, and luvadaxistat was shown to be efficacious in a cerebellar-dependent associative learning task. While luvadaxistat ameliorated the deficit seen in sociability in two different negative symptom tests of social interaction, it failed to show an effect in endpoints of negative symptoms in clinical trials. These results suggest that luvadaxistat potentially could be used to improve cognitive impairment in patients with schizophrenia, which is not well addressed with current antipsychotic medications.
Collapse
Affiliation(s)
- Rosa Fradley
- Neuroscience Drug Discovery Unit, Takeda, Cambridge, UK
| | | | - David Miller
- Neuroscience Drug Discovery Unit, Takeda, Cambridge, UK
| | | | - Sarah Almond
- Neuroscience Drug Discovery Unit, Takeda, Cambridge, UK
| | | | | | - Bin Zhu
- Neuroscience Drug Discovery Unit, Takeda, Cambridge, UK
| | - Eimear Howley
- Neuroscience Drug Discovery Unit, Takeda, Cambridge, UK
| | - Jo C Neill
- Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
| | - Ben Grayson
- Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
| | - Philip Gaskin
- Neuroscience Drug Discovery Unit, Takeda, Cambridge, UK
| | - Mark Carlton
- Neuroscience Drug Discovery Unit, Takeda, Cambridge, UK
| | - Ian Gray
- Neuroscience Drug Discovery Unit, Takeda, Cambridge, UK
| | - Jordi Serrats
- Neuroscience Drug Discovery Unit, Takeda California, 9625 Towne Centre Dr, San Diego, CA, 92121, USA.
| | - Ceri H Davies
- Takeda Pharmaceuticals Company Limited, Fujisawa, Kanagawa, Japan
| |
Collapse
|
25
|
Tsapakis EM, Diakaki K, Miliaras A, Fountoulakis KN. Novel Compounds in the Treatment of Schizophrenia-A Selective Review. Brain Sci 2023; 13:1193. [PMID: 37626549 PMCID: PMC10452918 DOI: 10.3390/brainsci13081193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Schizophrenia is a chronic neuropsychiatric syndrome that significantly impacts daily function and quality of life. All of the available guidelines suggest a combined treatment approach with pharmacologic agents and psychological interventions. However, one in three patients is a non-responder, the effect on negative and cognitive symptoms is limited, and many drug-related adverse effects complicate clinical management. As a result, discovering novel drugs for schizophrenia presents a significant challenge for psychopharmacology. This selective review of the literature aims to outline the current knowledge on the aetiopathogenesis of schizophrenia and to present the recently approved and newly discovered pharmacological substances in treating schizophrenia. We discuss ten novel drugs, three of which have been approved by the FDA (Olanzapine/Samidorphan, Lumateperone, and Pimavanserin). The rest are under clinical trial investigation (Brilaroxazine, Xanomeline/Trospium, Emraclidine, Ulotaront, Sodium Benzoate, Luvadaxistat, and Iclepertin). However, additional basic and clinical research is required not only to improve our understanding of the neurobiology and the potential novel targets in the treatment of schizophrenia, but also to establish more effective therapeutical interventions for the syndrome, including the attenuation of negative and cognitive symptoms and avoiding dopamine blockade-related adverse effects.
Collapse
Affiliation(s)
| | - Kalliopi Diakaki
- Department of Psychiatry, Academic General Hospital, 711 10 Heraklion, Greece
| | - Apostolos Miliaras
- Department of Psychiatry, Academic General Hospital, 711 10 Heraklion, Greece
| | | |
Collapse
|
26
|
Meyer JM, Correll CU. Increased Metabolic Potential, Efficacy, and Safety of Emerging Treatments in Schizophrenia. CNS Drugs 2023; 37:545-570. [PMID: 37470979 PMCID: PMC10374807 DOI: 10.1007/s40263-023-01022-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/21/2023]
Abstract
Patients with schizophrenia experience a broad range of detrimental health outcomes resulting from illness severity, heterogeneity of disease, lifestyle behaviors, and adverse effects of antipsychotics. Because of these various factors, patients with schizophrenia have a much higher risk of cardiometabolic abnormalities than people without psychiatric illness. Although exposure to many antipsychotics increases cardiometabolic risk factors, mortality is higher in patients who are not treated versus those who are treated with antipsychotics. This indicates both direct and indirect benefits of adequately treated illness, as well as the need for beneficial medications that result in fewer cardiometabolic risk factors and comorbidities. The aim of the current narrative review was to outline the association between cardiometabolic dysfunction and schizophrenia, as well as discuss the confluence of factors that increase cardiometabolic risk in this patient population. An increased understanding of the pathophysiology of schizophrenia has guided discovery of novel treatments that do not directly target dopamine and that not only do not add, but may potentially minimize relevant cardiometabolic burden for these patients. Key discoveries that have advanced the understanding of the neural circuitry and pathophysiology of schizophrenia now provide possible pathways toward the development of new and effective treatments that may mitigate the risk of metabolic dysfunction in these patients. Novel targets and preclinical and clinical data on emerging treatments, such as glycine transport inhibitors, nicotinic and muscarinic receptor agonists, and trace amine-associated receptor-1 agonists, offer promise toward relevant therapeutic advancements. Numerous areas of investigation currently exist with the potential to considerably progress our knowledge and treatment of schizophrenia.
Collapse
Affiliation(s)
- Jonathan M Meyer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA.
| | - Christoph U Correll
- Department of Psychiatry, The Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY, USA
- Department of Psychiatry and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany
| |
Collapse
|
27
|
Matrisciano F. Epigenetic regulation of metabotropic glutamate 2/3 receptors: Potential role for ultra-resistant schizophrenia? Pharmacol Biochem Behav 2023:173589. [PMID: 37348609 DOI: 10.1016/j.pbb.2023.173589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/05/2023] [Accepted: 06/15/2023] [Indexed: 06/24/2023]
Abstract
Schizophrenia is a severe and debilitating psychiatric disorder characterized by early cognitive deficits, emotional and behavioral abnormalities resulted by a dysfunctional gene x environment interaction. Genetic and epigenetic abnormalities in cortical parvalbumin-positive GABAergic interneurons lead to alterations in glutamate-mediated excitatory neurotransmission, synaptic plasticity, and neuronal development. Epigenetic alterations during pregnancy or early phases of postnatal life are associated with schizophrenia vulnerability as well as inflammatory processes which are at the basis of brain pathology. An epigenetic animal model of schizophrenia showed specific changes in promoter DNA methylation activity of genes related to schizophrenia such as reelin, BDNF and GAD67, and altered expression and function of mGlu2/3 receptors in the frontal cortex. Although antipsychotic medications represent the main treatment for schizophrenia and generally show an optimal efficacy profile for positive symptoms and relatively poor efficacy for negative or cognitive symptoms, a considerable percentage of individuals show poor response, do not achieve a complete remission, and approximately 30 % of patients show treatment-resistance. Here, we explore the potential role of epigenetic abnormalities linked to metabotropic glutamate 2/3 receptors changes in expression and function as key molecular factors underlying the difference in response to antipsychotics.
Collapse
Affiliation(s)
- Francesco Matrisciano
- The Psychiatric Institute, Department of Psychiatry, College of Medicine, University of Illinois Chicago (UIC), Chicago, IL, USA.
| |
Collapse
|