Next Article in Journal
Decellularized Antler Cancellous Bone Matrix Material Can Serve as Potential Bone Tissue Scaffold
Previous Article in Journal
Lithium Ions as Modulators of Complex Biological Processes: The Conundrum of Multiple Targets, Responsiveness and Non-Responsiveness, and the Potential to Prevent or Correct Dysregulation of Systems during Aging and in Disease
Previous Article in Special Issue
Haloperidol, Olanzapine, and Risperidone Induce Morphological Changes in an In Vitro Model of Human Hippocampal Neurogenesis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Opinion

What Remains to Be Discovered in Schizophrenia Therapeutics: Contributions by Advancing the Molecular Mechanisms of Drugs for Psychosis and Schizophrenia

by
Christoph U. Correll
1,2,3,†,
Massimo Tusconi
4,*,†,
Mauro Giovanni Carta
5 and
Serdar M. Dursun
6
1
Department of Psychiatry, Zucker Hillside Hospital, Northwell Health, Glen Oaks, NY 10128, USA
2
Department of Psychiatry and Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
3
Department of Child and Adolescent Psychiatry, Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
4
University Hospital of Cagliari, 09123 Cagliari, Italy
5
Department of Medical Sciences and Public Health, University of Cagliari, 09124 Cagliari, Italy
6
Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Edmonton, AB T6G 2G5, Canada
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomolecules 2024, 14(8), 906; https://doi.org/10.3390/biom14080906
Submission received: 17 June 2024 / Revised: 14 July 2024 / Accepted: 17 July 2024 / Published: 25 July 2024
(This article belongs to the Special Issue Molecular Insights into the Mechanism of Antipsychotic Drugs)

Abstract

:
Schizophrenia is a frequently debilitating and complex mental disorder affecting approximately 1% of the global population, characterized by symptoms such as hallucinations, delusions, disorganized thoughts and behaviors, cognitive dysfunction, and negative symptoms. Traditional treatment has centered on postsynaptic dopamine antagonists, commonly known as antipsychotic drugs, which aim to alleviate symptoms and improve functioning and the quality of life. Despite the availability of these medications, significant challenges remain in schizophrenia therapeutics, including incomplete symptom relief, treatment resistance, and medication side effects. This opinion article explores advancements in schizophrenia treatment, emphasizing molecular mechanisms, novel drug targets, and innovative delivery methods. One promising approach is novel strategies that target neural networks and circuits rather than single neurotransmitters, acknowledging the complexity of brain region interconnections involved in schizophrenia. Another promising approach is the development of biased agonists, which selectively activate specific signaling pathways downstream of receptors, offering potential for more precise pharmacological interventions with fewer side effects. The concept of molecular polypharmacy, where a single drug targets multiple molecular pathways, is exemplified by KarXT, a novel drug combining xanomeline and trospium to address both psychosis and cognitive dysfunction. This approach represents a comprehensive strategy for schizophrenia treatment, potentially improving outcomes for patients. In conclusion, advancing the molecular understanding of schizophrenia and exploring innovative therapeutic strategies hold promise for addressing the unmet needs in schizophrenia treatment, aiming for more effective and tailored interventions. Future research should focus on these novel approaches to achieve better clinical outcomes and improve the functional level and quality of life for individuals with schizophrenia.

1. Introduction

Schizophrenia is a debilitating and complex mental disorder that affects approximately 1% of the global population [1]. Characterized by a wide range of symptoms, including hallucinations, delusions, cognitive dysfunction, and negative symptoms [2,3,4], schizophrenia poses significant challenges to patients, their families, and the healthcare system [5,6]. For decades, postsynaptic dopamine antagonists, commonly also known as “antipsychotic” drugs, have been the cornerstone of schizophrenia treatment, aiming to alleviate distressing symptoms and improve the quality of life for affected individuals [7]. However, despite the availability of these medications, there are still significant gaps and challenges in schizophrenia therapeutics [8]. This article delves into the evolving landscape of the development of drugs for psychosis and schizophrenia [9], focusing on the molecular mechanisms, novel drug targets, and innovative delivery methods that hold promise in addressing these challenges.

2. Schizophrenia Treatment Gaps and Challenges

Terminology: From “Anti-psychotic” to “Drugs for Psychosis”

The terminology used to describe medications for schizophrenia has evolved over time [10,11]. The term “antipsychotic” has been traditionally used to describe drugs that alleviate psychotic symptoms, such as hallucinations and delusions [12]. While this term accurately reflects the primary action of these drugs, it can be limiting in capturing the broader scope of their effects. To address this limitation and emphasize the underlying mechanisms [13] (Table 1), a more neuroscientific nomenclature has been proposed: “drugs for psychosis” [14,15].
This shift in terminology highlights the fact that these medications [16] primarily target the neural mechanisms underlying psychosis, rather than simply treating the symptoms [17,18,19]. Understanding the molecular pathways and neural circuits involved in psychosis is crucial for the development of more effective treatments [20].

3. “Anti-psychotic” vs. Anti-Schizophrenia Treatments

Schizophrenia is not a monolithic condition solely characterized by psychosis [21]. It encompasses a spectrum of symptoms, including negative symptoms [22] (such as anhedonia and social withdrawal) [23], cognitive dysfunction (such as impaired memory and executive function), and affective symptoms (such as depression and anxiety) [24]. Traditional postsynaptic dopamine antagonist “antipsychotic” drugs primarily target psychosis-related symptoms by modulating neurotransmitter systems, particularly the dopamine system [25,26] (Table 1). However, there is a growing recognition of the need for treatments that address the broader spectrum of schizophrenia symptoms [27].
The term “anti-schizophrenia treatments” is emerging to encompass therapies that target multiple symptom domains, including positive, negative, cognitive, and affective symptoms. These treatments aim to provide comprehensive relief for individuals with schizophrenia and improve their overall functioning and quality of life [28,29,30].

4. Problems and Opportunities of Animal Models and Drug Effects

Animal Models of Schizophrenia

One of the challenges in schizophrenia research lies in the translation of findings from animal models to human patients [31]. Animal models, typically rodents, have been instrumental in understanding the neurobiology of schizophrenia and testing potential drug candidates [32,33]. However, these models have limitations in capturing the complexity of the human condition [34,35]. Schizophrenia is a uniquely human disorder, and animal models can only simulate certain aspects of the disease [36,37]. Thus, while animal models can provide valuable insights into the neurobiology and potential therapeutic interventions [38], they cannot fully replicate the intricate interplay of genetic, environmental, and developmental factors that contribute to schizophrenia in humans [39,40,41] (Table 1).
Animal models are essential for understanding the neurobiological mechanisms of schizophrenia and developing new therapies [37]. Among the most commonly used models are those based on the pharmacologic stimulation of “psychosis-like” disorganized cognitive or behavioral symptoms with dopamine agonists or NMDA antagonists and genetic manipulation, such as mice with mutations in the DISC1, NRG1, and DTNBP1 genes, which replicate some neurophysiological and behavioral characteristics of schizophrenia [41]. Other models include prenatal exposure to infections or stress, mimicking environmental risk factors associated with the disease [42]. Despite their utility, these models have significant limitations. The complex cognitive and symptomatic features of human schizophrenia, such as hallucinations and delusions, cannot be fully replicated in animals [43]. Moreover, differences between the human brain and those of laboratory animals make it challenging to translate findings directly to clinical settings [44]. These limitations highlight the need to develop more sophisticated models and to combine data from animal studies with human research for a more comprehensive understanding of schizophrenia.

5. “Anti-psychotic” Drug Effects in Animal Models

Understanding the effects of drugs for psychosis in animal models is crucial for drug development. These models help researchers assess the efficacy and safety of potential medications before advancing to human clinical trials. However, translating the effects observed in animals to human outcomes can be challenging [45]. Researchers face the dilemma of selecting appropriate animal proxies for schizophrenia symptoms. The choice of behavioral assays and endpoints can significantly impact the interpretation of drug effects. Innovative approaches, such as “smart box screening”, are being explored to address these challenges. Smart box screening involves the use [46] of sophisticated behavioral assays and monitoring systems to assess a drug’s impact on specific receptor systems and neural circuits [47].

6. Established and Emerging Drug Discovery Approaches: Hypothesis-Free Rapid Screening

To address the gaps in schizophrenia therapeutics, researchers are exploring novel drug discovery approaches [48]. The hypothesis-free rapid screening of compounds is one such approach. This method involves systematically testing a wide range of compounds for their effects on specific receptor systems and neural circuitry targets [49]. By using a hypothesis-free rapid screening approach, researchers can identify promising candidates without preconceived notions about their mechanisms of action. This strategy can accelerate drug discovery by identifying compounds that exhibit the desired effects on psychosis-related neural circuits.

7. Reducing Preclinical to Clinical Translation Failures

One of the critical challenges in drug development is the failure to translate promising preclinical findings into successful clinical outcomes [50]. To mitigate these translation failures, researchers are working to establish robust predictive biomarkers and improve the validity of animal models [51,52,53,54,55,56]. The development of reliable biomarkers that correlate with treatment response in schizophrenia patients can aid in patient stratification and the early identification of responders and non-responders [57,58]. This approach can reduce the variability observed in clinical trials and increase the likelihood of success.

8. Lessons Learned from “Anti-psychotic” Drug Development Failures

The history of the development of drugs for psychosis and schizophrenia is marked by both successes and failures. Understanding the reasons behind the failures can guide future research efforts. Some common pitfalls in drug development include the following [9,59]:
  • Lack of specificity: Many drugs currently approved for schizophrenia target multiple receptors, leading to a range of side effects. Identifying more selective drug candidates can improve tolerability [60].
  • Overemphasis on the dopamine system: While dopamine dysregulation plays a role in psychosis, it is not the sole contributor to schizophrenia. Expanding the focus to other neurotransmitter systems is essential [61].
  • Neglect of cognitive and negative symptoms: Traditional “antipsychotic” drugs as studied in people with schizophrenia primarily target positive symptoms (hallucinations and delusions) but often have limited efficacy for cognitive and negative symptoms. Developing treatments for these domains is crucial [62].

9. Beyond Neurotransmitter–Receptor Dyads: Intracellular Signaling as Antipsychotic Targets

Biased Agonism as an Example of Complexity

Schizophrenia’s pathophysiology is not solely reliant on neurotransmitter–receptor interactions. Intracellular signaling pathways within neurons play a crucial role in shaping the disease’s manifestations [63]. One intriguing concept is biased agonism, which refers to the selective activation of specific signaling pathways downstream of a receptor [64,65]. Biased agonists can modulate intracellular signaling in unique ways, offering the potential for more precise and nuanced pharmacological interventions [66,67,68,69,70]. By targeting specific intracellular pathways associated with schizophrenia, researchers may develop drugs with improved efficacy and fewer side effects [71].

10. Novel Approaches to Treat Psychosis and Schizophrenia: Targeting Neural Networks and Circuits

Moving Beyond Single Neurotransmitters and Receptors

Traditional antipsychotic drugs primarily focus on modulating single neurotransmitters (e.g., dopamine) and receptors (e.g., D2 receptors) [72]. However, recent advances in neuroscience have revealed the complexity of neural networks and circuits involved in schizophrenia [73]. Rather than targeting isolated neurotransmitters, researchers are exploring interventions that modulate entire neural networks [74]. This holistic approach recognizes the interconnectedness of brain regions and their contributions to schizophrenia symptomatology [75]. By targeting specific circuits, researchers aim to achieve more precise and effective treatments.

11. Molecular Polypharmacy: One Drug, Multiple Targets—KarXT: A Promising Example

Molecular polypharmacy is an emerging concept in drug development, where a single drug targets multiple molecular pathways implicated in a complex disorder like schizophrenia [76,77]. One promising example is KarXT, a novel drug that simultaneously addresses psychosis and cognitive dysfunction [78]. KarXT combines xanomeline, which targets the muscarinic acetylcholine system, with trospium, an anticholinergic agent [79]. This combination demonstrates the potential of polypharmacy in one pill, providing a comprehensive approach to schizophrenia treatment [80]. By simultaneously addressing different aspects of the disorder, such as positive symptoms and cognitive deficits, KarXT represents a promising advancement in the development of drugs to treat psychosis and schizophrenia (as well as likely symptom domains and disorders beyond that involve presynaptic hyperdopaminergia and/or an imbalance between excitation and inhibition related to GABA and glutamate transmission and interaction [79,81,82].

12. Known and Emerging Molecular Targets for Specific Symptoms

Positive Psychotic Symptoms

Positive psychotic symptoms, such as hallucinations and delusions, have long been associated with dysregulated dopamine transmission in the brain [83]. While this neurotransmitter–receptor dyad remains a critical target, researchers are exploring additional molecular targets to improve treatment efficacy and reduce side effects [84,85].
  • Presynaptic vs. postsynaptic dopamine modulation: Traditional drugs targeting psychosis and schizophrenia primarily act by blocking postsynaptic dopamine D2 receptors. However, the presynaptic modulation of dopamine release is also under investigation as a potential target to fine-tune dopamine transmission [86,87,88,89].
  • GABA and glutamate systems: The dysregulation of the gamma-aminobutyric acid (GABA) and glutamate systems has been implicated in schizophrenia. Novel drugs targeting these systems may offer alternative treatment options [90].
  • Muscarinic acetylcholine system: The muscarinic acetylcholine system plays a role in cognitive function. Drugs that modulate this system, such as xanomeline, are being explored to address cognitive deficits in schizophrenia [91,92].
  • Trace amine-associated receptor 1 (TAAR1): TAAR1 is a receptor involved in modulating dopamine and other neurotransmitters. Targeting TAAR1 may provide a unique approach to regulating neurotransmitter systems in schizophrenia [93].
  • Excitation–inhibition imbalance: Schizophrenia is characterized by an excitation–inhibition imbalance in neural circuits. Modulators that restore this balance are under investigation [94,95,96].

13. Molecular Underpinnings and Targets for Negative Symptoms

Negative symptoms in schizophrenia, including social withdrawal, anhedonia, and apathy, pose significant challenges in treatment. Identifying the molecular underpinnings and targets for these symptoms is crucial for improving the quality of life for affected individuals [97].
  • Dopamine receptors: While positive symptoms are associated with excess dopamine activity, negative symptoms may result from deficits in dopamine transmission. Balancing dopamine receptor activity is a potential approach [98].
  • Glutamate and NMDA receptors: The glutamate system, particularly N-methyl-D-aspartate (NMDA) receptors, has been implicated in negative symptoms. Enhancing NMDA receptor function is under investigation [99] (Table 1).
  • Oxytocin receptors: Oxytocin, a neuropeptide, has shown promise in alleviating negative symptoms [100]. Drugs targeting oxytocin receptors may improve social functioning [101].

14. Molecular Underpinnings and Targets for Cognitive Dysfunction

Cognitive dysfunction is a pervasive and debilitating aspect of schizophrenia. Impaired memory, executive function, and attention significantly impact daily functioning. Identifying the molecular underpinnings and targets for cognitive deficits is crucial for enhancing patients’ cognitive abilities [102].
  • Glutamate and AMPA receptors: Enhancing glutamate signaling, particularly through AMPA receptors, is a potential strategy to improve cognitive function [103] (Table 1).
  • Cholinergic systems: The cholinergic system, including nicotinergic and muscarinic receptors, plays a role in cognitive processes. Drugs targeting these receptors may enhance cognitive abilities [104].
  • Neuroplasticity: Promoting neuroplasticity, the brain’s ability to reorganize and adapt, is another avenue of research for addressing cognitive deficits in schizophrenia [105,106,107].

15. Molecular Underpinnings and Targets for Improved Reward System Functioning

The reward system in the brain is implicated in motivation, pleasure, and goal-directed behavior. The dysregulation of this system can lead to anhedonia and reduced motivation [108], common negative symptoms in schizophrenia [109,110,111]. Identifying molecular targets to restore normal reward system functioning is essential.
  • Dopamine pathways: Modulating dopamine pathways involved in reward processing may help restore motivation and pleasure in individuals with schizophrenia [112,113].
  • Opioid receptors: Opioid receptors are also involved in reward processing. Targeting these receptors may offer therapeutic potential [114].
  • Serotonin receptors: Serotonin receptors, particularly the 5-HT2A subtype, have been implicated in reward dysfunction and may be targeted to restore normal functioning [115] (Table 1).

16. Molecular Underpinnings and Targets for Improved Illness Insight

One of the challenges in schizophrenia treatment is anosognosia, a lack of insight into the illness. Individuals with schizophrenia may not recognize their symptoms or the need for treatment. Identifying molecular targets to improve illness insight is essential for enhancing treatment engagement [116].
  • Dopamine and prefrontal cortex: Dysregulated dopamine signaling in the prefrontal cortex is associated with impaired insight. Targeting this circuitry may help individuals gain a better awareness of their illness [117].
  • Glutamate and cognitive functioning: Improving cognitive function, particularly in areas related to self-awareness, may contribute to increased illness insight [118].
  • Neuroplasticity: Promoting neuroplasticity and cognitive flexibility may enhance individuals’ ability to understand and accept their condition [119].

17. Molecular Underpinnings and Targets of Treatment-Resistant Schizophrenia

Treatment-resistant schizophrenia (TRS) presents a formidable challenge [120,121,122]. Some individuals with schizophrenia do not respond adequately to available treatments [123,124,125,126,127]. Identifying the molecular underpinnings and targets specific to TRS is critical for developing interventions that can break through the treatment resistance barrier.
  • Dopamine dysregulation: While traditional drugs targeting psychosis and schizophrenia primarily target the dopamine system, novel approaches to modulate dopamine pathways are under investigation [9,128].
  • Glutamate dysfunction: Restoring glutamate balance, particularly through NMDA receptor modulation, is a focus in TRS research [129].
  • Cholinergic systems: The cholinergic system, particularly the muscarinic receptors, regulate GABA and glutamate as well as dopamine, and each may be involved in TRS. Drugs targeting these receptors may enhance cognitive abilities [87,130].
  • Inflammatory mechanisms: Inflammation in the brain has been implicated in TRS [131,132,133]. Targeting neuroinflammatory pathways may provide new treatment avenues [134] (Table 1).

18. Molecular Targets for Disease Modification

Beyond symptom management, there is a growing interest in developing treatments that modify the course of schizophrenia. These disease-modifying interventions aim to address the underlying pathophysiological processes and potentially slow down or halt disease progression.
  • Synaptic pruning: Excessive synaptic pruning during adolescence is believed to contribute to schizophrenia development. Interventions that regulate this process are under investigation [135].
  • Neuroinflammation: Chronic neuroinflammation is associated with schizophrenia. Modulating immune responses, microglial activation, and synaptic pruning in the brain may have disease-modifying effects [136] (Table 1).
  • Neuroplasticity: Promoting neuroplasticity and neural repair mechanisms may help mitigate the long-term consequences of schizophrenia [137].

19. Molecular Targets for Improved Safety and Tolerability

Currently, medications approved to treat schizophrenia often come with a range of side effects, including sedation/somnolence and agitation or insomnia, prolactin elevation and sexual dysfunction, neuromotor symptoms, and weight gain and metabolic disturbances [138,139,140]. Identifying molecular targets to avoid adverse effects and enhance the safety and tolerability of agents targeting psychosis and schizophrenia is crucial to improving treatment adherence and overall patient well-being. Balancing efficacy and effectiveness with safety and tolerability is essential. Strategies to reduce weight gain, dyslipidemia, and insulin resistance are actively being explored, as these are among the strongest contributors to the life-shortening features of schizophrenia [141,142] that are partly inherent to the illness, partly related to an unhealthy lifestyle, and partly related to adverse effects of treatments targeting schizophrenia [143].
  • Dopamine receptor subtypes: Selectively targeting specific dopamine receptor subtypes may reduce side effects while preserving efficacy for psychosis [144].
  • Serotonin receptors: Modulating serotonin receptors can influence side effect profiles. Balancing dopamine and serotonin interactions is a focus of research [145].
  • Metabolic pathways: Understanding the metabolic effects of drugs targeting psychosis and schizophrenia and developing interventions to mitigate these adverse effects are crucial. This can involve added molecular activity as part of a drug with efficacy for positive, negative, and/or cognitive symptoms or adding self-standing medications to currently approved antidopaminergic drugs and other treatments targeting psychosis, such as metformin of GLP-1 agonists [146,147].
  • Neuroprotection: Developing neuroprotective agents that shield the brain from the adverse effects of medications targeting psychosis and schizophrenia is a promising avenue. These agents may help prevent structural and functional changes associated with long-term antidopaminergic use or stimulate BDNF or other neurotrophic and antiapoptotic processes [148].

20. Combination Treatments to Minimize Side Effects

Combining multiple medications with complementary mechanisms of action is another strategy to minimize side effects while preserving therapeutic efficacy. This approach can help offset the adverse effects associated with individual drugs, resulting in a more tolerable treatment regimen.
  • Olanzapine plus samidorphan: Olanzapine is an effective medication for schizophrenia, but it is associated with weight gain. Samidorphan, an opioid receptor antagonist, is combined with olanzapine in an attempt to mitigate this side effect, providing a more tolerable treatment option [149,150].
  • Xanomeline plus trospium: A s previously mentioned, KarXT combines xanomeline, which targets the muscarinic acetylcholine system, with trospium, an anticholinergic agent. This combination minimizes the cholinergic side effects associated with muscarinic agonists [151,152].
These combination treatments exemplify the concept of molecular polypharmacy, where multiple drugs with distinct molecular targets work together synergistically to enhance therapeutic outcomes while minimizing side effects.

21. Novel Modes of Delivery for Medications Targeting Psychosis and Schizophrenia

In addition to innovative drug targets, researchers are exploring novel modes of drug delivery to improve the precision, convenience, and adherence of treatments targeting psychosis and schizophrenia. Some of these approaches include the following:
  • Subcutaneous delivery: Subcutaneous injections offer a more controlled and sustained release of medication compared to oral formulations. This mode of delivery can help minimize fluctuations in drug levels and improve adherence [153,154,155].
  • Transdermal patches: Transdermal patches provide a non-invasive and potentially convenient method of drug delivery. They can ensure the steady absorption of medication over an extended period, reducing the need for frequent dosing and improving the pharmacokinetic properties of some drugs [156,157].
  • Intranasal administration: Intranasal sprays and powders allow for rapid drug absorption through the nasal mucosa. This mode of delivery can lead to a faster onset of action, particularly important in managing acute psychotic episodes [158,159].
  • Viral vector-based delivery: Advanced techniques involving viral vectors can be used to target specific brain regions or neural circuits. This precision allows for localized drug delivery, minimizing systemic side effects [160,161,162,163].
These novel delivery methods hold promise in tailoring treatments to individual patient needs, enhancing treatment efficacy, and reducing the burden of daily medication regimens.

22. Pharmacokinetic Mechanisms to Speed Up or Extend Drug Effects

Pharmacokinetic strategies aim to optimize the absorption, distribution, metabolism, and elimination of drugs targeting psychosis and schizophrenia to achieve specific treatment goals. Depending on the clinical scenario, these mechanisms can either speed up drug delivery for rapid release and relief or extend the duration of drug effects to enhance adherence.
  • Rapid-acting drugs for psychosis and schizophrenia: For the management of acute agitation and psychosis, rapid-acting antipsychotic formulations are crucial. Short-acting intramuscular, inhalable, or intranasal administration can provide rapid relief within minutes [159,164,165].
  • Long-acting gastrointestinal delivery: Slowing down the intestinal transit of oral medications can potentially lead to a once-weekly oral depot whereby patients with difficulties with adherence would only require once weekly reminders or supervision [166].
  • Long-acting injectable drugs for psychosis and schizophrenia: To improve treatment adherence, long-acting injectable “antipsychotics” (LAIs) postsynaptic antidopaminergic medications offer extended drug release over weeks or months. LAIs ensure that patients receive consistent treatment without the need for daily dosing, reducing the risk of relapse. Administrations routes include intramuscular and subcutaneous administration [167,168].

23. Conclusions

Schizophrenia remains a complex and challenging mental disorder that affects millions of individuals worldwide. While postsynaptic antidopaminergic drugs have been the cornerstone of treatment for decades, there is still much to discover and improve upon in the field of schizophrenia therapeutics.
Advancements in understanding the molecular mechanisms underlying schizophrenia and the development of novel drug targets offer hope for more effective treatments. Targeting specific symptom domains, such as positive and negative symptoms, cognitive dysfunction, and impaired insight, allows for a more comprehensive approach to schizophrenia management.
Innovative strategies, such as molecular polypharmacy, combination treatments, and novel drug delivery methods, aim to improve treatment efficacy while minimizing side effects. Additionally, pharmacokinetic mechanisms can tailor drug effects to meet the unique needs of individual patients.
The ongoing pursuit of a better understanding of the molecular underpinnings of schizophrenia and the development of innovative treatment approaches provide hope for improved outcomes and a higher quality of life for individuals living with this challenging disorder.

Author Contributions

Conceptualization, C.U.C. and M.T.; methodology, C.U.C. and M.T.; software, M.T.; validation, C.U.C., S.M.D., M.G.C. and M.T.; formal analysis, M.T.; investigation, C.U.C. and M.T.; writing—original draft preparation, C.U.C. and M.T.; writing—review and editing, C.U.C., S.M.D., M.G.C. and M.T.; visualization, C.U.C. and M.T.; supervision, C.U.C. and M.T.; project administration, C.U.C. and M.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

Author Christoph U. Correll has been a consultant and/or advisor to or has received honoraria from: AbbVie, Acadia, Adock Ingram, Alkermes, Allergan, Angelini, Aristo, Biogen, Boehringer-Ingelheim, Bristol-Meyers Squibb, Cardio Diagnostics, Cerevel, CNX Therapeutics, Compass Pathways, Darnitsa, Delpor, Denovo, Eli Lilly, Gedeon Richter, Hikma, Holmusk, IntraCellular Therapies, Jamjoom Pharma, Janssen/J & J, Karuna, LB Pharma, Lundbeck, MedInCell, MedLink, Merck, Mindpax, Mitsubishi Tanabe Pharma, Maplight, Mylan, Neumora Therapeutics, Neurocrine, Neurelis, Newron, Noven, Novo Nordisk, Otsuka, PPD Biotech, Recordati, Relmada, Reviva, Rovi, Sage, Saladax, Sanofi, Seqirus, SK Life Science, Sumitomo Pharma America, Sunovion, Sun Pharma, Supernus, Tabuk, Takeda, Teva, Terran, Tolmar, Vertex, Viatris and Xenon Pharmaceuticals. He provided expert testimony for Janssen, Lundbeck and Otsuka. He served on a Data Safety Monitoring Board for Compass Pathways, Denovo, IntraCellular Therapies, Lundbeck, Relmada, Reviva, Rovi, Supernus, and Teva. He has received grant support from Boehringer-Ingelheim, Janssen and Takeda. He received royalties from UpToDate and is also a stock option holder of Cardio Diagnostics, Kuleon Biosciences, LB Pharma, Medlink, Mindpax, Quantic, Terran. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interNone of the mentioned subjects had any role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

References

  1. Charlson, F.J.; Ferrari, A.J.; Santomauro, D.F.; Diminic, S.; Stockings, E.; Scott, J.G.; McGrath, J.J.; Whiteford, H.A. Global Epidemiology and Burden of Schizophrenia: Findings from the Global Burden of Disease Study 2016. Schizophr. Bull. 2018, 44, 1195–1203. [Google Scholar] [CrossRef]
  2. Correll, C.U.; Schooler, N.R. Negative Symptoms in Schizophrenia: A Review and Clinical Guide for Recognition, Assessment, and Treatment. Neuropsychiatr. Dis. Treat. 2020, 519–534. [Google Scholar] [CrossRef]
  3. Millan, M.J.; Fone, K.; Steckler, T.; Horan, W.P. Negative Symptoms of Schizophrenia: Clinical Characteristics, Pathophysiological Substrates, Experimental Models and Prospects for Improved Treatment. Eur. Neuropsychopharmacol. 2014, 24, 645–692. [Google Scholar] [CrossRef]
  4. Millan, M.J.; Agid, Y.; Brüne, M.; Bullmore, E.T.; Carter, C.S.; Clayton, N.S.; Connor, R.; Davis, S.; Deakin, B.; DeRubeis, R.J.; et al. Cognitive Dysfunction in Psychiatric Disorders: Characteristics, Causes and the Quest for Improved Therapy. Nat. Rev. Drug Discov. 2012, 11, 141–168. [Google Scholar] [CrossRef]
  5. Awad, A.G.; Voruganti, L.N.P. The Burden of Schizophrenia on Caregivers. PharmacoEconomics 2008, 26, 149–162. [Google Scholar] [CrossRef]
  6. Liberman, R.P.; Kopelowicz, A. Recovery from Schizophrenia: A Challenge for the 21st Century. Int. Rev. Psychiatry 2002, 14, 245–255. [Google Scholar] [CrossRef]
  7. Tandon, R.; Targum, S.D.; Nasrallah, H.A.; Ross, R. Consortium, the T.E. in S. Strategies for Maximizing Clinical Effectiveness in the Treatment of Schizophrenia. J. Psychiatr. Pract. 2006, 12, 348. [Google Scholar] [CrossRef]
  8. Wagner, E.; Luykx, J.J.; Strube, W.; Hasan, A. Challenges, Unmet Needs and Future Directions—A Critical Evaluation of the Clinical Trial Landscape in Schizophrenia Research. Expert Rev. Clin. Pharmacol. 2024, 17, 11–18. [Google Scholar] [CrossRef]
  9. Correll, C.U.; Solmi, M.; Cortese, S.; Fava, M.; Højlund, M.; Kraemer, H.C.; McIntyre, R.S.; Pine, D.S.; Schneider, L.S.; Kane, J.M. The Future of Psychopharmacology: A Critical Appraisal of Ongoing Phase 2/3 Trials, and of Some Current Trends Aiming to de-Risk Trial Programmes of Novel Agents. World Psychiatry 2023, 22, 48–74. [Google Scholar] [CrossRef]
  10. Kane, J.M.; Correll, C.U. Past and Present Progress in the Pharmacologic Treatment of Schizophrenia. J. Clin. Psychiatry 2010, 71, 13909. [Google Scholar] [CrossRef]
  11. Cetin, M. Editorial: Treatment of Schizophrenia: Past, Present and Future. Klin. Psikofarmakol. Bül. Bull. Clin. Psychopharmacol. 2015, 25, 95–99. [Google Scholar] [CrossRef]
  12. Bangwal, R.; Bisht, S.; Saklani, S.; Garg, S.; Dhayani, M. Psychotic Disorders, Definition, Sign and Symptoms, Antipsychotic Drugs, Mechanism of Action, Pharmacokinetics & Pharmacodynamics with Side Effects & Adverse Drug Reactions: Updated Systematic Review Article. J. Drug Deliv. Ther. 2020, 10, 163–172. [Google Scholar] [CrossRef]
  13. Khodosevich, K.; Dragicevic, K.; Howes, O. Drug Targeting in Psychiatric Disorders—How to Overcome the Loss in Translation? Nat. Rev. Drug Discov. 2024, 23, 218–231. [Google Scholar] [CrossRef]
  14. Stahl, S.M. Drugs for Psychosis and Mood: Unique Actions at D3, D2, and D1 Dopamine Receptor Subtypes. CNS Spectr. 2017, 22, 375–384. [Google Scholar] [CrossRef]
  15. Zohar, J.; Stahl, S.; Moller, H.-J.; Blier, P.; Kupfer, D.; Yamawaki, S.; Uchida, H.; Spedding, M.; Goodwin, G.M.; Nutt, D. A Review of the Current Nomenclature for Psychotropic Agents and an Introduction to the Neuroscience-Based Nomenclature. Eur. Neuropsychopharmacol. 2015, 25, 2318–2325. [Google Scholar] [CrossRef]
  16. Jung, R.E.; Vartanian, O. (Eds.) The Cambridge Handbook of the Neuroscience of Creativity; Cambridge Handbooks in Psychology; Cambridge University Press: Cambridge, UK, 2018; ISBN 978-1-107-14761-4. [Google Scholar]
  17. Horacek, J.; Bubenikova-Valesova, V.; Kopecek, M.; Palenicek, T.; Dockery, C.; Mohr, P.; Höschl, C. Mechanism of Action of Atypical Antipsychotic Drugs and the Neurobiology of Schizophrenia. CNS Drugs 2006, 20, 389–409. [Google Scholar] [CrossRef]
  18. Davidson, M.; Carpenter, W.T., Jr. Targeted Treatment of Schizophrenia Symptoms as They Manifest, or Continuous Treatment to Reduce the Risk of Psychosis Recurrence. Schizophr. Bull. 2024, 50, 14–21. [Google Scholar] [CrossRef]
  19. Paul, S.M.; Potter, W.Z. Finding New and Better Treatments for Psychiatric Disorders. Neuropsychopharmacology 2024, 49, 3–9. [Google Scholar] [CrossRef]
  20. Howes, O.D.; Bukala, B.R.; Beck, K. Schizophrenia: From Neurochemistry to Circuits, Symptoms and Treatments. Nat. Rev. Neurol. 2024, 20, 22–35. [Google Scholar] [CrossRef]
  21. Jablensky, A. Schizophrenia or Schizophrenias? The Challenge of Genetic Parsing of a Complex Disorder. Am. J. Psychiatry 2015, 172, 105–107. [Google Scholar] [CrossRef]
  22. Mosolov, S.N.; Yaltonskaya, P.A. Primary and Secondary Negative Symptoms in Schizophrenia. Front. Psychiatry 2022, 12, 766692. [Google Scholar] [CrossRef]
  23. Muñoz-Negro, J.E.; Lozano, V.; Ibanez-Casas, I.; de la Cruz, B.G.; Soler, A.; Alcalá, J.A.; Ontiveros, C.; Aznarte, P.; Cervilla, J.A. Negative Symptoms across Psychotic Spectrum Disorders. Eur. J. Psychiatry 2017, 31, 37–41. [Google Scholar] [CrossRef]
  24. Malik, J.A.; Yaseen, Z.; Thotapalli, L.; Ahmed, S.; Shaikh, M.F.; Anwar, S. Understanding Translational Research in Schizophrenia: A Novel Insight into Animal Models. Mol. Biol. Rep. 2023, 50, 3767–3785. [Google Scholar] [CrossRef]
  25. Munawar, N.; Ahsan, K.; Muhammad, K.; Ahmad, A.; Anwar, M.A.; Shah, I.; Al Ameri, A.K.; Al Mughairbi, F. Hidden Role of Gut Microbiome Dysbiosis in Schizophrenia: Antipsychotics or Psychobiotics as Therapeutics? Int. J. Mol. Sci. 2021, 22, 7671. [Google Scholar] [CrossRef] [PubMed]
  26. Chana, G.; Bousman, C.A.; Money, T.T.; Gibbons, A.; Gillett, P.; Dean, B.; Everall, I.P. Biomarker Investigations Related to Pathophysiological Pathways in Schizophrenia and Psychosis. Front. Cell. Neurosci. 2013, 7, 95. [Google Scholar] [CrossRef]
  27. Addington, D.; Anderson, E.; Kelly, M.; Lesage, A.; Summerville, C. Canadian Practice Guidelines for Comprehensive Community Treatment for Schizophrenia and Schizophrenia Spectrum Disorders. Can. J. Psychiatry 2017, 62, 662–672. [Google Scholar] [CrossRef]
  28. Buchanan, R.W.; Kreyenbuhl, J.; Kelly, D.L.; Noel, J.M.; Boggs, D.L.; Fischer, B.A.; Himelhoch, S.; Fang, B.; Peterson, E.; Aquino, P.R.; et al. The 2009 Schizophrenia PORT Psychopharmacological Treatment Recommendations and Summary Statements. Schizophr. Bull. 2010, 36, 71–93. [Google Scholar] [CrossRef]
  29. de Winter, L.; Jelsma, A.; Vermeulen, J.M.; van Weeghel, J.; Hasson-Ohayon, I.; Mulder, C.L.; Boonstra, N.; Veling, W.; de Haan, L. Long-Term Changes in Personal Recovery and Quality of Life Among Patients with Schizophrenia Spectrum Disorders and Different Durations of Illness: A Meta-Analysis. Schizophr. Bull. 2024, sbae045. [Google Scholar] [CrossRef]
  30. Correll, C.U. MD What Are We Looking for in New Antipsychotics? J. Clin. Psychiatry 2011, 72, 1287. [Google Scholar] [CrossRef]
  31. Markou, A.; Chiamulera, C.; Geyer, M.A.; Tricklebank, M.; Steckler, T. Removing Obstacles in Neuroscience Drug Discovery: The Future Path for Animal Models. Neuropsychopharmacology 2009, 34, 74–89. [Google Scholar] [CrossRef]
  32. Powell, S.B.; Geyer, M.A. Overview of Animal Models of Schizophrenia. Curr. Protoc. Neurosci. 2007, 39, 9.24.1–9.24.20. [Google Scholar] [CrossRef] [PubMed]
  33. Lipska, B.K.; Weinberger, D.R. To Model a Psychiatric Disorder in Animals: Schizophrenia As a Reality Test. Neuropsychopharmacology 2000, 23, 223–239. [Google Scholar] [CrossRef] [PubMed]
  34. Lipska, B.K.; Gogos, J.A. Animal Models of Schizophrenia. In Schizophrenia; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2010; pp. 462–481. ISBN 978-1-4443-2729-8. [Google Scholar]
  35. Geyer, M.A.; Olivier, B.; Joëls, M.; Kahn, R.S. From Antipsychotic to Anti-Schizophrenia Drugs: Role of Animal Models. Trends Pharmacol. Sci. 2012, 33, 515–521. [Google Scholar] [CrossRef] [PubMed]
  36. van der Staay, F.J.; Arndt, S.S.; Nordquist, R.E. Evaluation of Animal Models of Neurobehavioral Disorders. Behav. Brain Funct. 2009, 5, 11. [Google Scholar] [CrossRef] [PubMed]
  37. Winship, I.R.; Dursun, S.M.; Baker, G.B.; Balista, P.A.; Kandratavicius, L.; Maia-de-Oliveira, J.P.; Hallak, J.; Howland, J.G. An Overview of Animal Models Related to Schizophrenia. Can. J. Psychiatry 2019, 64, 5–17. [Google Scholar] [CrossRef] [PubMed]
  38. Eyles, D.W. How Do Established Developmental Risk-Factors for Schizophrenia Change the Way the Brain Develops? Transl. Psychiatry 2021, 11, 1–15. [Google Scholar] [CrossRef]
  39. Howes, O.D.; Murray, R.M. Schizophrenia: An Integrated Sociodevelopmental-Cognitive Model. Lancet 2014, 383, 1677–1687. [Google Scholar] [CrossRef] [PubMed]
  40. Fišar, Z. Biological Hypotheses, Risk Factors, and Biomarkers of Schizophrenia. Prog. Neuropsychopharmacol. Biol. Psychiatry 2023, 120, 110626. [Google Scholar] [CrossRef] [PubMed]
  41. Ross, C.A.; Margolis, R.L.; Reading, S.A.J.; Pletnikov, M.; Coyle, J.T. Neurobiology of Schizophrenia. Neuron 2006, 52, 139–153. [Google Scholar] [CrossRef]
  42. Yates, E.F.; Mulkey, S.B. Viral Infections in Pregnancy and Impact on Offspring Neurodevelopment: Mechanisms and Lessons Learned. Pediatr. Res. 2024, 1–9. [Google Scholar] [CrossRef]
  43. Lv, Y.; Wen, L.; Hu, W.-J.; Deng, C.; Ren, H.-W.; Bao, Y.-N.; Su, B.-W.; Gao, P.; Man, Z.-Y.; Luo, Y.-Y.; et al. Schizophrenia in the Genetic Era: A Review from Development History, Clinical Features and Genomic Research Approaches to Insights of Susceptibility Genes. Metab. Brain Dis. 2024, 39, 147–171. [Google Scholar] [CrossRef]
  44. Murai, T.; Bailey, L.; Schultz, L.; Mongeau, L.; DeSana, A.; Silva, A.C.; Roberts, A.C.; Sukoff Rizzo, S.J. Improving Preclinical to Clinical Translation of Cognitive Function for Aging-Related Disorders: The Utility of Comprehensive Touchscreen Testing Batteries in Common Marmosets. Cogn. Affect. Behav. Neurosci. 2024, 24, 325–348. [Google Scholar] [CrossRef]
  45. Denayer, T.; Stöhr, T.; Van Roy, M. Animal Models in Translational Medicine: Validation and Prediction. New Horiz. Transl. Med. 2014, 2, 5–11. [Google Scholar] [CrossRef]
  46. Pratt, J.; Winchester, C.; Dawson, N.; Morris, B. Advancing Schizophrenia Drug Discovery: Optimizing Rodent Models to Bridge the Translational Gap. Nat. Rev. Drug Discov. 2012, 11, 560–579. [Google Scholar] [CrossRef]
  47. Torous, J.; Firth, J.; Mueller, N.; Onnela, J.P.; Baker, J.T. Methodology and Reporting of Mobile Health and Smartphone Application Studies for Schizophrenia. Harv. Rev. Psychiatry 2017, 25, 146. [Google Scholar] [CrossRef]
  48. Bender, A.; Cortés-Ciriano, I. Artificial Intelligence in Drug Discovery: What Is Realistic, What Are Illusions? Part 1: Ways to Make an Impact, and Why We Are Not There Yet. Drug Discov. Today 2021, 26, 511–524. [Google Scholar] [CrossRef]
  49. Meissner, F.; Geddes-McAlister, J.; Mann, M.; Bantscheff, M. The Emerging Role of Mass Spectrometry-Based Proteomics in Drug Discovery. Nat. Rev. Drug Discov. 2022, 21, 637–654. [Google Scholar] [CrossRef]
  50. Becker, R.E.; Greig, N.H. Lost in Translation: Neuropsychiatric Drug Development. Sci. Transl. Med. 2010, 2, 61rv6. [Google Scholar] [CrossRef]
  51. Seyhan, A.A. Lost in Translation: The Valley of Death across Preclinical and Clinical Divide—Identification of Problems and Overcoming Obstacles. Transl. Med. Commun. 2019, 4, 18. [Google Scholar] [CrossRef]
  52. Moore, H. The Role of Rodent Models in The Discovery of New Treatments for Schizophrenia: Updating Our Strategy. Schizophr. Bull. 2010, 36, 1066–1072. [Google Scholar] [CrossRef] [PubMed]
  53. Białoń, M.; Wąsik, A. Advantages and Limitations of Animal Schizophrenia Models. Int. J. Mol. Sci. 2022, 23, 5968. [Google Scholar] [CrossRef]
  54. Gilmour, G.; Arguello, A.; Bari, A.; Brown, V.J.; Carter, C.; Floresco, S.B.; Jentsch, D.J.; Tait, D.S.; Young, J.W.; Robbins, T.W. Measuring the Construct of Executive Control in Schizophrenia: Defining and Validating Translational Animal Paradigms for Discovery Research. Neurosci. Biobehav. Rev. 2013, 37, 2125–2140. [Google Scholar] [CrossRef]
  55. Geyer, M.A. Developing Translational Animal Models for Symptoms of Schizophrenia or Bipolar Mania. Neurotox. Res. 2008, 14, 71–78. [Google Scholar] [CrossRef]
  56. Kraguljac, N.V.; McDonald, W.M.; Widge, A.S.; Rodriguez, C.I.; Tohen, M.; Nemeroff, C.B. Neuroimaging Biomarkers in Schizophrenia. Am. J. Psychiatry 2021, 178, 509–521. [Google Scholar] [CrossRef]
  57. Abi-Dargham, A.; Moeller, S.J.; Ali, F.; DeLorenzo, C.; Domschke, K.; Horga, G.; Jutla, A.; Kotov, R.; Paulus, M.P.; Rubio, J.M.; et al. Candidate Biomarkers in Psychiatric Disorders: State of the Field. World Psychiatry 2023, 22, 236–262. [Google Scholar] [CrossRef]
  58. Goossens, J.; Morrens, M.; Coppens, V. The Potential Use of Peripheral Blood Mononuclear Cells as Biomarkers for Treatment Response and Outcome Prediction in Psychiatry: A Systematic Review. Mol. Diagn. Ther. 2021, 25, 283–299. [Google Scholar] [CrossRef]
  59. Green, M.F.; Harvey, P.D. Cognition in Schizophrenia: Past, Present, and Future. Schizophr. Res. Cogn. 2014, 1, e1–e9. [Google Scholar] [CrossRef]
  60. Wong, E.H.F.; Yocca, F.; Smith, M.A.; Lee, C.-M. Challenges and Opportunities for Drug Discovery in Psychiatric Disorders: The Drug Hunters’ Perspective. Int. J. Neuropsychopharmacol. 2010, 13, 1269–1284. [Google Scholar] [CrossRef]
  61. Miyamoto, S.; Duncan, G.E.; Marx, C.E.; Lieberman, J.A. Treatments for Schizophrenia: A Critical Review of Pharmacology and Mechanisms of Action of Antipsychotic Drugs. Mol. Psychiatry 2005, 10, 79–104. [Google Scholar] [CrossRef]
  62. Keshavan, M.S.; Lawler, A.N.; Nasrallah, H.A.; Tandon, R. New Drug Developments in Psychosis: Challenges, Opportunities and Strategies. Prog. Neurobiol. 2017, 152, 3–20. [Google Scholar] [CrossRef]
  63. Mısır, E.; Akay, G.G. Synaptic Dysfunction in Schizophrenia. Synapse 2023, 77, e22276. [Google Scholar] [CrossRef]
  64. Ward, R.D.; Kellendonk, C.; Kandel, E.R.; Balsam, P.D. Timing as a Window on Cognition in Schizophrenia. Neuropharmacology 2012, 62, 1175–1181. [Google Scholar] [CrossRef]
  65. Kajitani, N.; Okada-Tsuchioka, M.; Inoue, A.; Miyano, K.; Masuda, T.; Boku, S.; Iwamoto, K.; Ohtsuki, S.; Uezono, Y.; Aoki, J.; et al. G Protein-Biased LPAR1 Agonism of Prototypic Antidepressants: Implication in the Identification of Novel Therapeutic Target for Depression. Neuropsychopharmacology 2024, 49, 561–572. [Google Scholar] [CrossRef]
  66. Kellendonk, C.; Simpson, E.H.; Kandel, E.R. Modeling Cognitive Endophenotypes of Schizophrenia in Mice. Trends Neurosci. 2009, 32, 347–358. [Google Scholar] [CrossRef] [PubMed]
  67. Kruyer, A.; Kalivas, P.W.; Scofield, M.D. Astrocyte Regulation of Synaptic Signaling in Psychiatric Disorders. Neuropsychopharmacology 2023, 48, 21–36. [Google Scholar] [CrossRef]
  68. Kaye, A.D.; Edinoff, A.N.; Babin, K.C.; Hebert, C.M.; Hardin, J.L.; Cornett, E.M.; Kaye, A.J.; Kaye, A.M.; Urman, R.D. Pharmacological Advances in Opioid Therapy: A Review of the Role of Oliceridine in Pain Management. Pain Ther. 2021, 10, 1003–1012. [Google Scholar] [CrossRef] [PubMed]
  69. Abi-Dargham, A.; Javitch, J.A.; Slifstein, M.; Anticevic, A.; Calkins, M.E.; Cho, Y.T.; Fonteneau, C.; Gil, R.; Girgis, R.; Gur, R.E.; et al. Dopamine D1R Receptor Stimulation as a Mechanistic Pro-Cognitive Target for Schizophrenia. Schizophr. Bull. 2022, 48, 199–210. [Google Scholar] [CrossRef]
  70. Shen, Y.; McCorvy, J.D.; Martini, M.L.; Rodriguiz, R.M.; Pogorelov, V.M.; Ward, K.M.; Wetsel, W.C.; Liu, J.; Roth, B.L.; Jin, J. D2 Dopamine Receptor G Protein-Biased Partial Agonists Based on Cariprazine. J. Med. Chem. 2019, 62, 4755–4771. [Google Scholar] [CrossRef]
  71. Simpson, E.H.; Kellendonk, C.; Kandel, E. A Possible Role for the Striatum in the Pathogenesis of the Cognitive Symptoms of Schizophrenia. Neuron 2010, 65, 585–596. [Google Scholar] [CrossRef]
  72. Kapur, S.; Mamo, D. Half a Century of Antipsychotics and Still a Central Role for Dopamine D2 Receptors. Prog. Neuropsychopharmacol. Biol. Psychiatry 2003, 27, 1081–1090. [Google Scholar] [CrossRef]
  73. Perez de la Mora, M.; Hernandez-Mondragon, C.; Crespo-Ramirez, M.; Rejon-Orantes, J.; Borroto-Escuela, D.O.; Fuxe, K. Conventional and Novel Pharmacological Approaches to Treat Dopamine-Related Disorders: Focus on Parkinson’s Disease and Schizophrenia. Neuroscience 2020, 439, 301–318. [Google Scholar] [CrossRef] [PubMed]
  74. Stępnicki, P.; Kondej, M.; Kaczor, A.A. Current Concepts and Treatments of Schizophrenia. Molecules 2018, 23, 2087. [Google Scholar] [CrossRef] [PubMed]
  75. van den Heuvel, M.P.; Fornito, A. Brain Networks in Schizophrenia. Neuropsychol. Rev. 2014, 24, 32–48. [Google Scholar] [CrossRef] [PubMed]
  76. Anighoro, A.; Bajorath, J.; Rastelli, G. Polypharmacology: Challenges and Opportunities in Drug Discovery. J. Med. Chem. 2014, 57, 7874–7887. [Google Scholar] [CrossRef] [PubMed]
  77. Kondej, M.; Stępnicki, P.; Kaczor, A.A. Multi-Target Approach for Drug Discovery against Schizophrenia. Int. J. Mol. Sci. 2018, 19, 3105. [Google Scholar] [CrossRef] [PubMed]
  78. Howes, O.D.; Dawkins, E.; Lobo, M.C.; Kaar, S.J.; Beck, K. New Drug Treatments for Schizophrenia: A Review of Approaches to Target Circuit Dysfunction. Biol. Psychiatry, 2024; in press. [Google Scholar] [CrossRef]
  79. Paul, S.M.; Yohn, S.E.; Brannan, S.K.; Neugebauer, N.M.; Breier, A. Muscarinic Receptor Activators as Novel Treatments for Schizophrenia. Biol. Psychiatry, 2024; in press. [Google Scholar] [CrossRef]
  80. Jones, S.E.; Harvey, P.D. Cross-Diagnostic Determinants of Cognitive Functioning: The Muscarinic Cholinergic Receptor as a Model System. Transl. Psychiatry 2023, 13, 1–9. [Google Scholar] [CrossRef] [PubMed]
  81. Kaul, I.; Sawchak, S.; Walling, D.P.; Tamminga, C.A.; Breier, A.; Zhu, H.; Miller, A.C.; Paul, S.M.; Brannan, S.K. Efficacy and Safety of Xanomeline-Trospium Chloride in Schizophrenia: A Randomized Clinical Trial. JAMA Psychiatry 2024. [Google Scholar] [CrossRef] [PubMed]
  82. Kaul, I.; Sawchak, S.; Correll, C.U.; Kakar, R.; Breier, A.; Zhu, H.; Miller, A.C.; Paul, S.M.; Brannan, S.K. Efficacy and Safety of the Muscarinic Receptor Agonist KarXT (Xanomeline–Trospium) in Schizophrenia (EMERGENT-2) in the USA: Results from a Randomised, Double-Blind, Placebo-Controlled, Flexible-Dose Phase 3 Trial. Lancet 2024, 403, 160–170. [Google Scholar] [CrossRef] [PubMed]
  83. Winton-Brown, T.T.; Fusar-Poli, P.; Ungless, M.A.; Howes, O.D. Dopaminergic Basis of Salience Dysregulation in Psychosis. Trends Neurosci. 2014, 37, 85–94. [Google Scholar] [CrossRef]
  84. Kirby, B.P.; Waddington, J.L.; O’Tuathaigh, C.M.P. Advancing a Functional Genomics for Schizophrenia: Psychopathological and Cognitive Phenotypes in Mutants with Gene Disruption. Brain Res. Bull. 2010, 83, 162–176. [Google Scholar] [CrossRef]
  85. Correll, C.U.; Citrome, L. Pharmacologic Treatment of Schizophrenia Beyond Dopamine Receptor Blockade—Has Its Time Come Yet? JAMA Psychiatry 2024, 81, 118–120. [Google Scholar] [CrossRef] [PubMed]
  86. Yun, S.; Yang, B.; Anair, J.D.; Martin, M.M.; Fleps, S.W.; Pamukcu, A.; Yeh, N.-H.; Contractor, A.; Kennedy, A.; Parker, J.G. Antipsychotic Drug Efficacy Correlates with the Modulation of D1 Rather than D2 Receptor-Expressing Striatal Projection Neurons. Nat. Neurosci. 2023, 26, 1417–1428. [Google Scholar] [CrossRef] [PubMed]
  87. Yohn, S.E.; Weiden, P.J.; Felder, C.C.; Stahl, S.M. Muscarinic Acetylcholine Receptors for Psychotic Disorders: Bench-Side to Clinic. Trends Pharmacol. Sci. 2022, 43, 1098–1112. [Google Scholar] [CrossRef] [PubMed]
  88. Correll, C.U.; Abi-Dargham, A.; Howes, O. Emerging Treatments in Schizophrenia. J. Clin. Psychiatry 2022, 83, 39683. [Google Scholar] [CrossRef] [PubMed]
  89. Leo, D.; Mus, L.; Espinoza, S.; Hoener, M.C.; Sotnikova, T.D.; Gainetdinov, R.R. Taar1-Mediated Modulation of Presynaptic Dopaminergic Neurotransmission: Role of D2 Dopamine Autoreceptors. Neuropharmacology 2014, 81, 283–291. [Google Scholar] [CrossRef] [PubMed]
  90. Ľupták, M.; Michaličková, D.; Fišar, Z.; Kitzlerová, E.; Hroudová, J. Novel Approaches in Schizophrenia-from Risk Factors and Hypotheses to Novel Drug Targets. World J. Psychiatry 2021, 11, 277–296. [Google Scholar] [CrossRef]
  91. Melancon, B.J.; Tarr, J.C.; Panarese, J.D.; Wood, M.R.; Lindsley, C.W. Allosteric Modulation of the M1 Muscarinic Acetylcholine Receptor: Improving Cognition and a Potential Treatment for Schizophrenia and Alzheimer’s Disease. Drug Discov. Today 2013, 18, 1185–1199. [Google Scholar] [CrossRef] [PubMed]
  92. Shekhar, A.; Potter, W.Z.; Lightfoot, J.; Lienemann, J.; Dubé, S.; Mallinckrodt, C.; Bymaster, F.P.; McKinzie, D.L.; Felder, C.C. Selective Muscarinic Receptor Agonist Xanomeline as a Novel Treatment Approach for Schizophrenia. Am. J. Psychiatry 2008, 165, 1033–1039. [Google Scholar] [CrossRef] [PubMed]
  93. Dodd, S.; Carvalho, A.F.; Puri, B.K.; Maes, M.; Bortolasci, C.C.; Morris, G.; Berk, M. Trace Amine-Associated Receptor 1 (TAAR1): A New Drug Target for Psychiatry? Neurosci. Biobehav. Rev. 2021, 120, 537–541. [Google Scholar] [CrossRef]
  94. Uliana, D.L.; Lisboa, J.R.F.; Gomes, F.V.; Grace, A.A. The Excitatory-Inhibitory Balance as a Target for the Development of Novel Drugs to Treat Schizophrenia. Biochem. Pharmacol. 2024, 116298. [Google Scholar] [CrossRef]
  95. Howes, O.D.; Shatalina, E. Integrating the Neurodevelopmental and Dopamine Hypotheses of Schizophrenia and the Role of Cortical Excitation-Inhibition Balance. Biol. Psychiatry 2022, 92, 501–513. [Google Scholar] [CrossRef] [PubMed]
  96. Zhu, J.; Zikopoulos, B.; Yazdanbakhsh, A. A Neural Model of Modified Excitation/Inhibition and Feedback Levels in Schizophrenia. Front. Psychiatry 2023, 14, 1199690. [Google Scholar] [CrossRef] [PubMed]
  97. Singh, S.; Khanna, D.; Kalra, S. Role of Neurochemicals in Schizophrenia. Curr. Psychopharmacol. 2020, 9, 144–161. [Google Scholar] [CrossRef]
  98. Kesby, J.P.; Eyles, D.W.; McGrath, J.J.; Scott, J.G. Dopamine, Psychosis and Schizophrenia: The Widening Gap between Basic and Clinical Neuroscience. Transl. Psychiatry 2018, 8, 1–12. [Google Scholar] [CrossRef] [PubMed]
  99. Coyle, J.T.; Tsai, G. The NMDA Receptor Glycine Modulatory Site: A Therapeutic Target for Improving Cognition and Reducing Negative Symptoms in Schizophrenia. Psychopharmacology 2004, 174, 32–38. [Google Scholar] [CrossRef] [PubMed]
  100. Cochran, D.M.; Fallon, D.; Hill, M.; Frazier, J.A. The Role of Oxytocin in Psychiatric Disorders: A Review of Biological and Therapeutic Research Findings. Harv. Rev. Psychiatry 2013, 21, 219. [Google Scholar] [CrossRef] [PubMed]
  101. Jones, C.; Barrera, I.; Brothers, S.; Ring, R.; Wahlestedt, C. Oxytocin and Social Functioning. Dialogues Clin. Neurosci. 2017, 19, 193–201. [Google Scholar] [CrossRef] [PubMed]
  102. Ichinose, M.; Park, S. Chapter 2—Models of Schizophrenia. A Selective Review of Genetic, Neuropharmacological, Cognitive, and Social Approaches. In A Clinical Introduction to Psychosis; Badcock, J.C., Paulik, G., Eds.; Academic Press: New York, NY, USA, 2020; pp. 37–62. ISBN 978-0-12-815012-2. [Google Scholar]
  103. Partin, K.M. AMPA Receptor Potentiators: From Drug Design to Cognitive Enhancement. Curr. Opin. Pharmacol. 2015, 20, 46–53. [Google Scholar] [CrossRef] [PubMed]
  104. Erskine, D.; Taylor, J.-P.; Bakker, G.; Brown, A.J.H.; Tasker, T.; Nathan, P.J. Cholinergic Muscarinic M1 and M4 Receptors as Therapeutic Targets for Cognitive, Behavioural, and Psychological Symptoms in Psychiatric and Neurological Disorders. Drug Discov. Today 2019, 24, 2307–2314. [Google Scholar] [CrossRef]
  105. Kaneko, Y.; Keshavan, M. Cognitive Remediation in Schizophrenia. Clin. Psychopharmacol. Neurosci. 2012, 10, 125–135. [Google Scholar] [CrossRef]
  106. Sehatpour, P.; Iosifescu, D.V.; De Baun, H.M.; Shope, C.; Mayer, M.R.; Gangwisch, J.; Dias, E.; Sobeih, T.; Choo, T.-H.; Wall, M.M.; et al. 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. Psychiatry 2023, 94, 164–173. [Google Scholar] [CrossRef] [PubMed]
  107. Jun, R.; Zhang, W.; Beacher, N.J.; Zhang, Y.; Li, Y.; Lin, D.-T. Dysbindin-1, BDNF, and GABAergic Transmission in Schizophrenia. Front. Psychiatry 2022, 13, 876749. [Google Scholar] [CrossRef]
  108. Zhao, N.O.; Topolski, N.; Tusconi, M.; Salarda, E.M.; Busby, C.W.; Lima, C.N.N.C.; Pillai, A.; Quevedo, J.; Barichello, T.; Fries, G.R. Blood-Brain Barrier Dysfunction in Bipolar Disorder: Molecular Mechanisms and Clinical Implications. Brain Behav. Immun.—Health 2022, 21, 100441. [Google Scholar] [CrossRef] [PubMed]
  109. Liang, S.; Wu, Y.; Hanxiaoran, L.; Greenshaw, A.J.; Li, T. Anhedonia in Depression and Schizophrenia: Brain Reward and Aversion Circuits. Neuropsychiatr. Dis. Treat. 2022, 18, 1385–1396. [Google Scholar] [CrossRef]
  110. Dresp-Langley, B. From Reward to Anhedonia-Dopamine Function in the Global Mental Health Context. Biomedicines 2023, 11, 2469. [Google Scholar] [CrossRef]
  111. Gheiratmand, M.; Rish, I.; Cecchi, G.A.; Brown, M.R.G.; Greiner, R.; Polosecki, P.I.; Bashivan, P.; Greenshaw, A.J.; Ramasubbu, R.; Dursun, S.M. Learning Stable and Predictive Network-Based Patterns of Schizophrenia and Its Clinical Symptoms. npj Schizophr. 2017, 3, 1–12. [Google Scholar] [CrossRef] [PubMed]
  112. Saperia, S.; Da Silva, S.; Siddiqui, I.; Agid, O.; Daskalakis, Z.J.; Ravindran, A.; Voineskos, A.N.; Zakzanis, K.K.; Remington, G.; Foussias, G. Reward-Driven Decision-Making Impairments in Schizophrenia. Schizophr. Res. 2019, 206, 277–283. [Google Scholar] [CrossRef]
  113. Gilleen, J.; Shergill, S.S.; Kapur, S. Impaired Subjective Well-Being in Schizophrenia Is Associated with Reduced Anterior Cingulate Activity during Reward Processing. Psychol. Med. 2015, 45, 589–600. [Google Scholar] [CrossRef]
  114. Rabiner, E.A.; Beaver, J.; Makwana, A.; Searle, G.; Long, C.; Nathan, P.J.; Newbould, R.D.; Howard, J.; Miller, S.R.; Bush, M.A.; et al. Pharmacological Differentiation of Opioid Receptor Antagonists by Molecular and Functional Imaging of Target Occupancy and Food Reward-Related Brain Activation in Humans. Mol. Psychiatry 2011, 16, 826–835. [Google Scholar] [CrossRef]
  115. Aznar, S.; Hervig, M.E.-S. The 5-HT2A Serotonin Receptor in Executive Function: Implications for Neuropsychiatric and Neurodegenerative Diseases. Neurosci. Biobehav. Rev. 2016, 64, 63–82. [Google Scholar] [CrossRef]
  116. Mograbi, D.C.; Hall, S.; Arantes, B.; Huntley, J. The Cognitive Neuroscience of Self-Awareness: Current Framework, Clinical Implications, and Future Research Directions. WIREs Cogn. Sci. 2024, 15, e1670. [Google Scholar] [CrossRef]
  117. Kawahata, I.; Finkelstein, D.I.; Fukunaga, K. Dopamine D1–D5 Receptors in Brain Nuclei: Implications for Health and Disease. Receptors 2024, 3, 155–181. [Google Scholar] [CrossRef]
  118. Meiron, O. Self-Awareness in Schizophrenia: Identifying Common Neural Oscillatory Parameters Underlying Altered Sense of Self-Agency and Reduced Prefrontal Cortex Excitability. Curr. Opin. Behav. Sci. 2024, 58, 101398. [Google Scholar] [CrossRef]
  119. Page, C.E.; Epperson, C.N.; Novick, A.M.; Duffy, K.A.; Thompson, S.M. Beyond the Serotonin Deficit Hypothesis: Communicating a Neuroplasticity Framework of Major Depressive Disorder. Mol. Psychiatry 2024, 1–12. [Google Scholar] [CrossRef]
  120. Reitan, E.C.K.; Iversen, V.C.; Riley, H.; Høye, A. Why Do Patients Want Medication Free Treatment for Psychosis? An Explorative Study on Reasons for Applying to Medication Free Programs. BMC Psychiatry 2024, 24, 127. [Google Scholar] [CrossRef]
  121. Potkin, S.G.; Kane, J.M.; Correll, C.U.; Lindenmayer, J.-P.; Agid, O.; Marder, S.R.; Olfson, M.; Howes, O.D. The Neurobiology of Treatment-Resistant Schizophrenia: Paths to Antipsychotic Resistance and a Roadmap for Future Research. npj Schizophr. 2020, 6, 1. [Google Scholar] [CrossRef]
  122. Howes, O.D.; McCutcheon, R.; Agid, O.; de Bartolomeis, A.; van Beveren, N.J.M.; Birnbaum, M.L.; Bloomfield, M.A.P.; Bressan, R.A.; Buchanan, R.W.; Carpenter, W.T.; et al. Treatment-Resistant Schizophrenia: Treatment Response and Resistance in Psychosis (TRRIP) Working Group Consensus Guidelines on Diagnosis and Terminology. Am. J. Psychiatry 2017, 174, 216–229. [Google Scholar] [CrossRef]
  123. Wagner, E.; Siskind, D.; Falkai, P.; Howes, O.; Correll, C.; Lee, J.; Honer, W.G.; Kane, J.M.; Fernandez-Egea, E.; Barnes, T.R.E.; et al. Clozapine Optimization: A Delphi Consensus Guideline from the Treatment Response and Resistance in Psychosis Working Group. Schizophr. Bull. 2023, 49, 962–972. [Google Scholar] [CrossRef]
  124. Tusconi, M.; Nibbio, G.; Gupta, R.; Carr, E. Editorial: Case Reports in Schizophrenia and Psychotic Disorders. Front. Psychiatry 2023, 14, 1282780. [Google Scholar] [CrossRef]
  125. Tusconi, M.; Sanchez-Gutierrez, T. Editorial: Reviews in Psychiatry 2022: Schizophrenia. Front. Psychiatry 2023, 14, 1237676. [Google Scholar] [CrossRef]
  126. Diniz, E.; Fonseca, L.; Rocha, D.; Trevizol, A.; Cerqueira, R.; Ortiz, B.; Brunoni, A.R.; Bressan, R.; Correll, C.U.; Gadelha, A. Treatment Resistance in Schizophrenia: A Meta-Analysis of Prevalence and Correlates. Braz. J. Psychiatry 2023, 45, 448–458. [Google Scholar] [CrossRef]
  127. Tusconi, M.; Kätzel, D.; Sánchez-Gutiérrez, T. Editorial: Reviews in Psychiatry 2023: Schizophrenia. Front. Psychiatry 2024, 15, 1444818. [Google Scholar] [CrossRef]
  128. Dudzik, P.; Lustyk, K.; Pytka, K. Beyond Dopamine: Novel Strategies for Schizophrenia Treatment. Med. Res. Rev. 2024. [Google Scholar] [CrossRef]
  129. Eisenstein, T.; Furman-Haran, E.; Tal, A. Early Excitatory-Inhibitory Cortical Modifications Following Skill Learning Are Associated with Motor Memory Consolidation and Plasticity Overnight. Nat. Commun. 2024, 15, 906. [Google Scholar] [CrossRef]
  130. Cerri, D.H.; Albaugh, D.L.; Walton, L.R.; Katz, B.; Wang, T.-W.; Chao, T.-H.H.; Zhang, W.; Nonneman, R.J.; Jiang, J.; Lee, S.-H.; et al. Distinct Neurochemical Influences on fMRI Response Polarity in the Striatum. Nat. Commun. 2024, 15, 1916. [Google Scholar] [CrossRef]
  131. Tusconi, M.; Dursun, S.M. Editorial: Further Findings in the Role of Inflammation in the Etiology and Treatment of Schizophrenia. Front. Psychiatry 2024, 15, 1349568. [Google Scholar] [CrossRef]
  132. Chaves, C.; Dursun, S.M.; Tusconi, M.; Hallak, J.E.C. Neuroinflammation and Schizophrenia—Is There a Link? Front. Psychiatry 2024, 15, 1356975. [Google Scholar] [CrossRef]
  133. Tusconi, M.; Fries, G.R. Chapter 11—Neuroprogression in Bipolar Disorder. In Biomarkers in Bipolar Disorders; Machado-Vieira, R., Soares, J.C., Eds.; Academic Press: New York, NY, USA, 2022; pp. 167–189. ISBN 978-0-12-821398-8. [Google Scholar]
  134. Bjørklund, G.; Mkhitaryan, M.; Sahakyan, E.; Fereshetyan, K.; Meguid, N.A.; Hemimi, M.; Nashaat, N.H.; Yenkoyan, K. Linking Environmental Chemicals to Neuroinflammation and Autism Spectrum Disorder: Mechanisms and Implications for Prevention. Mol. Neurobiol. 2024. [Google Scholar] [CrossRef]
  135. Chen, Y.; Li, W.; Lv, L.; Yue, W. Shared Genetic Determinants of Schizophrenia and Autism Spectrum Disorder Implicate Opposite Risk Patterns: A Genome-Wide Analysis of Common Variants. Schizophr. Bull. 2024, sbae044. [Google Scholar] [CrossRef]
  136. Chandra, J. The Potential Role of the P75 Receptor in Schizophrenia: Neuroimmunomodulation and Making Life or Death Decisions. Brain Behav. Immun.—Health 2024, 38, 100796. [Google Scholar] [CrossRef]
  137. Sugden, S.G.; Merlo, G.; Manger, S. Strengthening Neuroplasticity in Substance Use Recovery Through Lifestyle Intervention. Am. J. Lifestyle Med. 2024, 15598276241242016. [Google Scholar] [CrossRef]
  138. Ward, K.; Citrome, L. Tolerability and Safety Outcomes of First-Line Oral Second-Generation Antipsychotics in Patients with Schizophrenia. Expert Opin. Drug Saf. 2024, 23, 399–409. [Google Scholar] [CrossRef] [PubMed]
  139. Pillinger, T.; Howes, O.D.; Correll, C.U.; Leucht, S.; Huhn, M.; Schneider-Thoma, J.; Gaughran, F.; Jauhar, S.; McGuire, P.K.; Taylor, D.M.; et al. Antidepressant and Antipsychotic Side-Effects and Personalised Prescribing: A Systematic Review and Digital Tool Development. Lancet Psychiatry 2023, 10, 860–876. [Google Scholar] [CrossRef] [PubMed]
  140. Solmi, M.; Murru, A.; Pacchiarotti, I.; Undurraga, J.; Veronese, N.; Fornaro, M.; Stubbs, B.; Monaco, F.; Vieta, E.; Seeman, M.V.; et al. Safety, Tolerability, and Risks Associated with First- and Second-Generation Antipsychotics: A State-of-the-Art Clinical Review. Ther. Clin. Risk Manag. 2017, 13, 757–777. [Google Scholar] [CrossRef] [PubMed]
  141. Correll, C.U.; Solmi, M.; Croatto, G.; Schneider, L.K.; Rohani-Montez, S.C.; Fairley, L.; Smith, N.; Bitter, I.; Gorwood, P.; Taipale, H.; et al. Mortality in People with Schizophrenia: A Systematic Review and Meta-Analysis of Relative Risk and Aggravating or Attenuating Factors. World Psychiatry 2022, 21, 248–271. [Google Scholar] [CrossRef] [PubMed]
  142. Chan, J.K.N.; Correll, C.U.; Wong, C.S.M.; Chu, R.S.T.; Fung, V.S.C.; Wong, G.H.S.; Lei, J.H.C.; Chang, W.C. Life Expectancy and Years of Potential Life Lost in People with Mental Disorders: A Systematic Review and Meta-Analysis. eClinicalMedicine 2023, 65. [Google Scholar] [CrossRef] [PubMed]
  143. Firth, J.; Siddiqi, N.; Koyanagi, A.; Siskind, D.; Rosenbaum, S.; Galletly, C.; Allan, S.; Caneo, C.; Carney, R.; Carvalho, A.F.; et al. The Lancet Psychiatry Commission: A Blueprint for Protecting Physical Health in People with Mental Illness. Lancet Psychiatry 2019, 6, 675–712. [Google Scholar] [CrossRef] [PubMed]
  144. Reiner-Link, D.; Madsen, J.S.; Gloriam, D.E.; Bräuner-Osborne, H.; Hauser, A.S. Differential G Protein Activation by the Long and Short Isoforms of the Dopamine D2 Receptor. Br. J. Pharmacol. 2024. [Google Scholar] [CrossRef] [PubMed]
  145. Ballardin, D.; Makrini-Maleville, L.; Seper, A.; Valjent, E.; Rebholz, H. 5-HT4R Agonism Reduces L-DOPA-Induced Dyskinesia via Striatopallidal Neurons in Unilaterally 6-OHDA Lesioned Mice. Neurobiol. Dis. 2024, 106559. [Google Scholar] [CrossRef]
  146. Larsen, J.R.; Vedtofte, L.; Holst, J.J.; Oturai, P.; Kjær, A.; Corell, C.U.; Vilsbøll, T.; Fink-Jensen, A. Does a GLP-1 Receptor Agonist Change Glucose Tolerance in Patients Treated with Antipsychotic Medications? Design of a Randomised, Double-Blinded, Placebo-Controlled Clinical Trial. BMJ Open 2014, 4, e004227. [Google Scholar] [CrossRef]
  147. Siskind, D.; Hahn, M.; Correll, C.U.; Fink-Jensen, A.; Russell, A.W.; Bak, N.; Broberg, B.V.; Larsen, J.; Ishøy, P.L.; Vilsbøll, T.; et al. Glucagon-like Peptide-1 Receptor Agonists for Antipsychotic-Associated Cardio-Metabolic Risk Factors: A Systematic Review and Individual Participant Data Meta-Analysis. Diabetes Obes. Metab. 2019, 21, 293–302. [Google Scholar] [CrossRef] [PubMed]
  148. Kositsyn, Y.M.; de Abreu, M.S.; Kolesnikova, T.O.; Lagunin, A.A.; Poroikov, V.V.; Harutyunyan, H.S.; Yenkoyan, K.B.; Kalueff, A.V. Towards Novel Potential Molecular Targets for Antidepressant and Antipsychotic Pharmacotherapies. Int. J. Mol. Sci. 2023, 24, 9482. [Google Scholar] [CrossRef] [PubMed]
  149. Martin, W.F.; Correll, C.U.; Weiden, P.J.; Jiang, Y.; Pathak, S.; DiPetrillo, L.; Silverman, B.L.; Ehrich, E.W. Mitigation of Olanzapine-Induced Weight Gain with Samidorphan, an Opioid Antagonist: A Randomized Double-Blind Phase 2 Study in Patients with Schizophrenia. Am. J. Psychiatry 2019, 176, 457–467. [Google Scholar] [CrossRef]
  150. Correll, C.U.; Stein, E.; Graham, C.; DiPetrillo, L.; Akerman, S.; Stanford, A.D.; Jiang, Y.; Yagoda, S.; McDonnell, D.; Hopkinson, C. Reduction in Multiple Cardiometabolic Risk Factors with Combined Olanzapine/Samidorphan Compared with Olanzapine: Post Hoc Analyses From a 24-Week Phase 3 Study. Schizophr. Bull. 2023, 49, 454–463. [Google Scholar] [CrossRef] [PubMed]
  151. Johnson, C.R.; Kangas, B.D.; Jutkiewicz, E.M.; Bergman, J.; Coop, A. Drug Design Targeting the Muscarinic Receptors and the Implications in Central Nervous System Disorders. Biomedicines 2022, 10, 398. [Google Scholar] [CrossRef] [PubMed]
  152. Breier, A.; Brannan, S.K.; Paul, S.M.; Miller, A.C. Evidence of Trospium’s Ability to Mitigate Cholinergic Adverse Events Related to Xanomeline: Phase 1 Study Results. Psychopharmacology 2023, 240, 1191–1198. [Google Scholar] [CrossRef]
  153. Baryakova, T.H.; Pogostin, B.H.; Langer, R.; McHugh, K.J. Overcoming Barriers to Patient Adherence: The Case for Developing Innovative Drug Delivery Systems. Nat. Rev. Drug Discov. 2023, 22, 387–409. [Google Scholar] [CrossRef] [PubMed]
  154. Li, W.; Tang, J.; Lee, D.; Tice, T.R.; Schwendeman, S.P.; Prausnitz, M.R. Clinical Translation of Long-Acting Drug Delivery Formulations. Nat. Rev. Mater. 2022, 7, 406–420. [Google Scholar] [CrossRef]
  155. Kane, J.M.; Harary, E.; Eshet, R.; Tohami, O.; Weiser, M.; Leucht, S.; Merenlender-Wagner, A.; Sharon, N.; Davis, G.L.; Suett, M.; et al. Efficacy and Safety of TV-46000, a Long-Acting, Subcutaneous, Injectable Formulation of Risperidone, for Schizophrenia: A Randomised Clinical Trial in the USA and Bulgaria. Lancet Psychiatry 2023, 10, 934–943. [Google Scholar] [CrossRef]
  156. Pastore, M.N.; Kalia, Y.N.; Horstmann, M.; Roberts, M.S. Transdermal Patches: History, Development and Pharmacology. Br. J. Pharmacol. 2015, 172, 2179–2209. [Google Scholar] [CrossRef]
  157. Citrome, L.; Zeni, C.M.; Correll, C.U. Patches: Established and Emerging Transdermal Treatments in Psychiatry. J. Clin. Psychiatry 2019, 80, 21174. [Google Scholar] [CrossRef] [PubMed]
  158. Pandey, V.; Gadeval, A.; Asati, S.; Jain, P.; Jain, N.; Roy, R.K.; Tekade, M.; Soni, V.; Tekade, R.K. Chapter 7—Formulation Strategies for Nose-to-Brain Delivery of Therapeutic Molecules. In Drug Delivery Systems; Tekade, R.K., Ed.; Advances in Pharmaceutical Product Development and Research; Academic Press: New York, NY, USA, 2020; pp. 291–332. ISBN 978-0-12-814487-9. [Google Scholar]
  159. Citrome, L.; Correll, C.U.; San, L.; Zeller, S.L.; Madden, S.; Gutierrez, J.; Lopez-Toledano, M.; Misra, S.N.; Carrazana, E.; Rabinowicz, A.L. Alternative Approaches for Addressing Acute Agitation in Schizophrenia and Bipolar Disorder. Prim. Care Companion CNS Disord. 2024, 26, 23nr03596. [Google Scholar] [CrossRef] [PubMed]
  160. Hermens, W.T.J.M.C.; Verhaagen, J. Viral Vectors, Tools for Gene Transfer in the Nervous System. Prog. Neurobiol. 1998, 55, 399–432. [Google Scholar] [CrossRef] [PubMed]
  161. Cwetsch, A.W.; Pinto, B.; Savardi, A.; Cancedda, L. In Vivo Methods for Acute Modulation of Gene Expression in the Central Nervous System. Prog. Neurobiol. 2018, 168, 69–85. [Google Scholar] [CrossRef] [PubMed]
  162. Moreira, R.; Nóbrega, C.; de Almeida, L.P.; Mendonça, L. Brain-Targeted Drug Delivery—Nanovesicles Directed to Specific Brain Cells by Brain-Targeting Ligands. J. Nanobiotechnol. 2024, 22, 260. [Google Scholar] [CrossRef] [PubMed]
  163. Ogawa, K.; Kato, N.; Kawakami, S. Recent Strategies for Targeted Brain Drug Delivery. Chem. Pharm. Bull. 2020, 68, 567–582. [Google Scholar] [CrossRef] [PubMed]
  164. Ward, K.; Citrome, L. The Treatment of Acute Agitation Associated with Schizophrenia or Bipolar Disorder: Investigational Drugs in Early Stages of Their Clinical Development, and Their Clinical Context and Potential Place in Therapy. Expert Opin. Investig. Drugs 2020, 29, 245–257. [Google Scholar] [CrossRef] [PubMed]
  165. Kim, H.K.; Leonard, J.B.; Corwell, B.N.; Connors, N.J. Safety and Efficacy of Pharmacologic Agents Used for Rapid Tranquilization of Emergency Department Patients with Acute Agitation or Excited Delirium. Expert Opin. Drug Saf. 2021, 20, 123–138. [Google Scholar] [CrossRef]
  166. Lyndra Therapeutics Announces Positive Data from Pivotal Phase 3 Study with Oral Weekly Risperidone (LYN-005) for Schizophrenia. Available online: https://www.biospace.com/article/lyndra-therapeutics-announces-positive-data-from-pivotal-phase-3-study-with-oral-weekly-risperidone-lyn-005-for-schizophrenia/ (accessed on 16 June 2024).
  167. Milz, R.; Benson, C.; Knight, K.; Antunes, J.; Najarian, D.; Lopez Rengel, P.-M.; Wang, S.; Richarz, U.; Gopal, S.; Kane, J.M. The Effect of Longer Dosing Intervals for Long-Acting Injectable Antipsychotics on Outcomes in Schizophrenia. Neuropsychiatr. Dis. Treat. 2023, 19, 531–545. [Google Scholar] [CrossRef]
  168. Haddad, P.M.; Correll, C.U. Long-Acting Antipsychotics in the Treatment of Schizophrenia: Opportunities and Challenges. Expert Opin. Pharmacother. 2023, 24, 473–493. [Google Scholar] [CrossRef]
Table 1. Molecular mechanisms involved in schizophrenia.
Table 1. Molecular mechanisms involved in schizophrenia.
MechanismVariationOutcome
Dopamine DysregulationHyperactivity in Mesolimbic PathwayPositive Symptoms
Hypoactivity in Mesocortical PathwayNegative Symptoms
Glutamate HypofunctionNMDA Receptor Dysfunction and Cortical ExcitabilityCognitive Deficits and Negative Symptoms
Serotonin Imbalance5-HT2A Receptor Overactivity and Dopamine ReleasePositive and Negative Symptoms
Genetic FactorsDISC1, NRG1, and DTNBP1 Genes and Synaptic Function
NeuroinflammationMicroglial Activation and Synaptic Pruning
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Correll, C.U.; Tusconi, M.; Carta, M.G.; Dursun, S.M. What Remains to Be Discovered in Schizophrenia Therapeutics: Contributions by Advancing the Molecular Mechanisms of Drugs for Psychosis and Schizophrenia. Biomolecules 2024, 14, 906. https://doi.org/10.3390/biom14080906

AMA Style

Correll CU, Tusconi M, Carta MG, Dursun SM. What Remains to Be Discovered in Schizophrenia Therapeutics: Contributions by Advancing the Molecular Mechanisms of Drugs for Psychosis and Schizophrenia. Biomolecules. 2024; 14(8):906. https://doi.org/10.3390/biom14080906

Chicago/Turabian Style

Correll, Christoph U., Massimo Tusconi, Mauro Giovanni Carta, and Serdar M. Dursun. 2024. "What Remains to Be Discovered in Schizophrenia Therapeutics: Contributions by Advancing the Molecular Mechanisms of Drugs for Psychosis and Schizophrenia" Biomolecules 14, no. 8: 906. https://doi.org/10.3390/biom14080906

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop