Advancing translational neurotherapeutics: Bridging pharmacy, psychiatry, and neurology for precision brain disorder management

Review
[10.37881/1.1012]
PDF Downloaded : 43 Fulltext Viewed : 97 Views
CROSSMARK LOGO

Advancing translational neurotherapeutics: Bridging pharmacy, psychiatry, and neurology for precision brain disorder management

Review | Volume 10 | Issue 1 | NeuroPharmac 04 2025 | Page 01-08 | Dushad Ram . DOI: 10.37881/1.1012
Authors: Dushad Ram
Department of Clinical Medicine, College of Medicine, Shaqra University, Saudi Arabia
Address of Correspondence:
Dushad Ram
Department of Clinical Medicine, College of Medicine, Shaqra University, Saudi Arabia
Email: dushadram@su.edu.sa
Article Received : 2025-02-20,
Article Accepted : 2025-03-28
Available Online : 2025-04-30
ABSTRACT

Psychiatric and neurological disorders pose a significant global health challenge, marked by rising prevalence, disability, and treatment disparities. This review proposes an integrated translational neurotherapeutic framework bridging pharmacy, psychiatry, and neurology, emphasizing shared pathophysiological mechanisms (neurotransmitter dysregulation, neuroinflammation, and synaptic dysfunction) across conditions such as depression, schizophrenia, Alzheimer's disease, and Parkinson's disease. Emerging strategies leverage pharmacogenomics, AI modeling, and neurotechnologies for personalized interventions, along with novel agents (psychedelics, biologics, and neuromodulators) that target treatment-resistant and progressive disorders. The influence of the gut-brain axis and microbiome necessitates a holistic approach. Pharmacological advancements (rapid-acting antidepressants and targeted antipsychotics) have been enhanced by precision medicine, digital health, and multidisciplinary teams. Addressing translational challenges (preclinical limitations, trial heterogeneity, ethics, and access) requires adaptive trials, biomarkers, and regulatory reforms. Future directions should prioritize early intervention, stratified care, and global equity, supported by interdisciplinary education and policy, aiming to transform brain disorder management through precision, personalization, and innovation.

Keywords: Neurotherapeutics, pharmacy, psychiatry, brain disorders

Introduction

Brain disorders, encompassing psychiatric (depression affecting >300 million, schizophrenia 1%) and neurological (epilepsy 50 million, alzheimer’s >50 million, projected to triple by 2050) disorders, constitute a major global health burden, contributing 13.1% of disability-adjusted life years (DALYs) and incurring trillions in annual costs.[1] Their multifactorial etiology involves genetic, environmental, and neurobiological factors and frequent comorbidities.[2]

Treatment disparities are significant in LMICs, where only 25–31% of patients receive adequate care, compounded by stigma and limited resources.[3] Translational neurotherapeutics integrates psychiatry, neurology, and pharmacy to target shared mechanisms (e.g., synaptic dysfunction and neuroinflammation) using advanced methodologies (e.g., rs-fMRI, pharmacogenomics, and AI) for personalized treatment.[4] Innovations, such as psychedelics, neuromodulation, and nanocarriers, enhance efficacy, while multi-omics and holistic approaches (the gut-brain axis) address underlying causes.

A strategic roadmap emphasizes early detection, cost-effective interventions, interdisciplinary collaboration, adaptive therapies, and global initiatives for equitable and precision-driven care.[5] The integration of molecular insights, drug innovation, and systems biology aims to transform the outcomes of patients with brain disorders.

Shared molecular and physiological mechanisms

Diverse brain disorders, including psychiatric (schizophrenia, depression, and anxiety) and neurological disorders (epilepsy, alzheimer’s disease (AD), parkinson’s disease (PD), and multiple sclerosis (MS), share key molecular and physiological mechanisms that inform translational strategies.

Neurotransmitter Dysregulation: Imbalances in glutamate, GABA, dopamine, and serotonin levels are central. Schizophrenia is characterized by reduced prefrontal GABA levels, elevated glutamate levels, and upregulated metabotropic glutamate receptors.[6] Epilepsy exhibits hyperexcitability via reduced GABAergic inhibition and excess glutamate, whereas anxiety involves low GABA and high glutamate levels.[7] PD involves dopaminergic neuron loss and serotonergic dysfunction, while MDD presents with mesolimbic dopamine deficits and serotonergic disruption.[8]

Neuroinflammation and microglial activation: Proinflammatory cytokines (IL-6 and TNF-α) are implicated in AD, MS, and MDD, promoting amyloid-beta aggregation (AD), demyelination (MS), and impaired serotonin function/neuroplasticity (MDD).[9] Chronically activated microglia amplify damage via the release of cytokines and neurotoxic factors in AD, MS, and PD. Therapeutic modulation (PPAR agonists and curcumin) can shift microglia to a protective phenotype.[10]

Synaptic plasticity and connectivity: Brain-derived neurotrophic factor (BDNF), which is crucial for synaptic plasticity, is disrupted in MDD, AD, autism, and schizophrenia. Reduced BDNF (linked to Val66Met) impairs hippocampal plasticity in MDD/AD, whereas altered BDNF disrupts synaptic pruning in autism and schizophrenia.[11,12] BDNF mimetics and antidepressants aim to restore synaptic health.[13]

Genetic and Epigenetic Overlaps: Genes such as DISC1 and COMT with pleiotropic effects (ZSCAN29/31) are implicated in schizophrenia, MDD, PD, and MS, affecting synaptic function and dopamine metabolism.[14,15] DNA methylation in NRG1/BDNF influences drug responses, with antipsychotic-induced epigenomic shifts and accelerated epigenetic aging offering precision care biomarkers.[16,17]

Translational Strategies: Integrated approaches utilize pharmacogenomics (CYP2D6 and APOE ε4) and innovations (nanocarriers, psychedelics, and CRISPR) to target these mechanisms and enhance CNS delivery/efficacy.[18,19] A strategic roadmap emphasizes interdisciplinary collaboration, multi-omics, and adaptive therapies for personalized and holistic treatment.[5]

 Pharmacological innovations

Next-Generation Antipsychotics: Evolving from the 1970s SGAs, newer agents such as pimavanserin, cariprazine, and brexpiprazole (partial D2 agonists) mitigate metabolic and motor side effects.[20] Emerging drugs (BRD5814, roluperidone, ulotaront, and KarXT) target negative symptoms, cognition, and oxidative stress, with TAAR1 agonists showing promise.[21]

Rapid-Acting Antidepressants: For TRD, ketamine, esketamine (FDA-approved), and psilocybin offer rapid action via NMDA modulation and neuroplasticity[22], enhancing glutamate, synaptogenesis, and BDNF.[23] Alternatives (R-ketamine, dextromethorphan-bupropion, and resolvins) offer efficacy with fewer side effects.[24] Precision medicine utilizes SLC6A4 and CYP2D6 markers.[25,26]

Anti-amyloid and Cannabinoid Therapies: In AD, anti-amyloid monoclonal antibodies (aducanumab and lecanemab) aim to slow the progression.[27] CBD (Epidiolex) reduces seizures (epilepsy) and spasticity (MS) via endocannabinoid modulation, with minimal side effects.[28]

Drug Delivery and Translational Advances: Nanoparticles and intranasal delivery bypass the BBB in AD and schizophrenia.[18,29,30] Sustained-release systems (depot antipsychotics, hydrogels) improve adherence. Preclinical models (organoids and zebrafish) validate targets such as levetiracetam/SV2A in epilepsy.[31]

Interdisciplinary Precision: Pharmacy, psychiatry, and neurology converge using pharmacogenomics and AI-driven fMRI for tailored, real-time, biomarker-guided precision care.[32,33]

 Pharmacogenomics and precision medicine

Genetic polymorphisms in CYP450 enzymes (CYP2D6, CYP2C19, and CYP1A2) significantly affect psychotropic drug metabolism, thereby influencing their efficacy and side effects. Pretreatment genetic testing optimizes antidepressant and antipsychotic dosing.[34] HLA gene variations (e.g., HLA-B1502 and HLA-A3101) predict hypersensitivity reactions to antiepileptic drugs, necessitating screening for specific ethnicities.[35]

Biomarkers, such as neurofilament light chain (NfL), monitor neurodegeneration in Alzheimer’s disease and multiple sclerosis and guide therapeutic adjustments.[36] C-reactive protein (CRP) level predicts psychiatric treatment response in major depressive disorder, informing anti-inflammatory strategies.[37]

Although pharmacogenomics optimizes warfarin dosing via CYP2C9/VKORC1 genotyping, inspiring tailored psychotropic drug administration, implementation challenges include cost, access, and standardization.[38,39] In clozapine treatment for schizophrenia, HLA-DQB10602 and CYP enzyme profiling, along with NfL and CRP monitoring, can mitigate risks and personalize treatment to improve outcomes.[40,41]

 Emerging therapies at the intersection

Psychedelics and Neuroplasticity: Minimal doses of psychedelics (psilocybin, LSD) offer rapid, sustained relief from depression, PTSD, and addiction by targeting 5-HT2A receptors and enhancing neuroplasticity via synaptogenesis, dendritic growth, and BDNF upregulation, which is distinct from the effects.[42]

Neuromodulation Techniques: DBS modulates mood circuits and is effective for Parkinson’s motor symptoms and refractory depression despite cognitive side effects.[43] Noninvasive TMS enhances cortical excitability and synaptic activity, improving antidepressant efficacy and potentially reducing dosage.[44]

Biologics, Gene Therapies, and Cannabinoids: Monoclonal antibodies (aducanumab) targeting amyloid-beta in Alzheimer’s disease represent disease-modifying biologics.[45] AAV-based gene therapies deliver functional genes for rare disorders such as spinal muscular atrophy, demonstrating their safety and efficacy.[46] THC/CBD combinations modulate the endocannabinoid system, effectively managing chronic pain, anxiety, and seizures with analgesic, anxiolytic, and anticonvulsant effects, particularly in pediatric epilepsy.[47,48]

 Translational challenges and opportunities

Preclinical to Clinical Translation: Animal models often poorly replicate the complexity of human brain disorders (depression, PTSD, and neurodegenerative diseases) owing to biological and behavioral differences.[49] Humanized models (stem cells, tissues, and transgenic primates) combined with multi-omics offer more accurate preclinical relevance.[50,51]

Clinical Trial Design: Patient heterogeneity (genetics, severity, and comorbidities) complicates trials and reduces their generalizability.[52] Adaptive designs that adjust parameters based on interim data and biomarker-based stratification enhance precision and efficiency.[53]

Safety, Ethics, and Access Barriers: Emerging therapies (psychedelics and DBS) pose risks (e.g., psychosis and cognitive impairment) and require strict screening.[54,55] Genetic editing (CRISPR) and neuromodulation raise ethical concerns (consent, long-term effects, and misuse), necessitating stringent guidelines.[56] Long approval timelines and access disparities in LMICs highlight equity challenges, necessitating streamlined regulations and global collaboration.[57,58]

Future directions

Gut-Brain Axis and AI in Neuropharmacology: The gut-brain axis and microbiome increasingly modulate drug metabolism and efficacy; specific bacteria influence bioavailability, inflammation, and neurotrophic signaling disorders. Microbiome-based interventions have shown neuroprotective potential. AI/ML enhances precision medicine by analyzing multi-omics data for target identification and response prediction, streamlining drug development, and personalizing treatments.

Clinical Integration and Emerging Therapies: Digital health platforms (wearables) enable real-time biomarker monitoring, adherence improvement, and personalized interventions. Multidisciplinary teams (pharmacists, psychiatrists, and neurologists) are crucial for providing comprehensive care. Neuroprotective compounds that target early pathological mechanisms (oxidative stress and inflammation) have the potential to delay the progression of neurodegenerative diseases. Psychedelic-assisted therapies have shown long-term efficacy for depression, PTSD, and addiction via neuroplasticity and emotional processing, with microbiome composition potentially influencing the response.

Policy, Education, and Funding: Clinicians require training in AI, pharmacogenomics, and digital health for effective implementation of advanced therapies. Robust funding and adaptive regulatory frameworks are vital for neurotherapeutic innovations. Advocacy must address translational barriers, ensure equitable access to treatment, and promote global collaboration.

Conclusion

Advancements in brain disorder treatment are contingent upon the integration of molecular neuroscience, digital health, and precision pharmacology. By elucidating the shared molecular foundations of psychiatric and neurological disorders and incorporating innovations such as pharmacogenomics, neuromodulation, and AI-guided therapies, neurotherapeutics can evolve from merely managing symptoms to modifying and preventing diseases. The clinical translation of these advancements necessitates not only technological progress but also systemic restructuring, including patient stratification, real-time monitoring, and multidisciplinary care.

To effectively address persistent barriers such as regulatory inertia, inequitable access, and inadequate training, it is imperative to engage in sustained investment, foster global cooperation, and develop scalable, context-sensitive interventions. The focus on the gut-brain axis, emerging neuroprotective agents, and long-term outcomes of psychedelic therapy signifies a transition towards a more holistic and personalized approach to mental health care. Ultimately, by dismantling disciplinary silos and centering translational science on patient-specific biology and lived experiences, neurotherapeutics can provide durable and equitable solutions in one of the most complex areas of medicine.

Abbreviations

General medical terms

DALYs – Disability-Adjusted Life Years, LMICs – Low- and Middle-Income Countries, CNS – Central Nervous System, TRD – Treatment-Resistant Depression, PTSD – Post-Traumatic Stress Disorder, BBB – Blood-Brain Barrier.

Disorders

MDD – Major Depressive Disorder, AD – Alzheimer’s Disease, PD – Parkinson’s Disease, MS – Multiple Sclerosis, ASD – Autism Spectrum Disorder.

Genes and molecular biology

BDNF – Brain-Derived Neurotrophic Factor, DISC1 – Disrupted in Schizophrenia 1, COMT – Catechol-O-Methyltransferase, NRG1 – Neuregulin 1, ZSCAN – Zinc Finger and SCAN Domain-Containing Protein, HLA – Human Leukocyte Antigen, AAV – Adeno-Associated Virus, NfL – Neurofilament Light Chain, CRP – C-Reactive Protein, SV2A – Synaptic Vesicle Glycoprotein 2A.

Pharmacogenomics / Enzymes

CYP450 – Cytochrome P450, CYP2D6, CYP2C19, CYP1A2 – Specific isoenzymes of Cytochrome P450 involved in drug metabolism, SLC6A4 – Solute Carrier Family 6 Member 4 (serotonin transporter gene), VKORC1 – Vitamin K Epoxide Reductase Complex Subunit 1.

Medications / Compounds

CBD – Cannabidiol, THC – Tetrahydrocannabinol, SGAs – Second-Generation Antipsychotics, TMS – Transcranial Magnetic Stimulation, DBS – Deep Brain Stimulation, AI/ML – Artificial Intelligence / Machine Learning, CRISPR – Clustered Regularly Interspaced Short Palindromic Repeats.

Genetic risk markers

APOE ε4 – Apolipoprotein E epsilon 4 allele, HLA-B1502, HLA-A3101 – Specific HLA gene variants, HLA-DQB1*0602 – HLA subtype associated with drug hypersensitivity.

Conflict of Interest

The author declares no conflicts of interest relevant to this article.

References

  • 1.
    Ding C, Wu Y, Chen X, et al. Global, regional, and national burden and attributable risk factors of neurological disorders: The Global Burden of Disease study 1990-2019. Front Public Health. 2022;10:952161. [Google Scholar] [PubMed] [CrossRef]
  • 2.
    Paneva J, Leunissen I, Schuhmann T, et al. Using Remotely Supervised At-Home TES for Enhancing Mental Resilience. Front Hum Neurosci. 2022;16:838187. [Google Scholar] [PubMed] [CrossRef]
  • 3.
    Charlson FJ, Ferrari AJ, Santomauro DF, et al. Global Epidemiology and Burden of Schizophrenia: Findings From the Global Burden of Disease Study 2016. Schizophr Bull. 2018;44(6):1195-1203. [Google Scholar] [PubMed] [CrossRef]
  • 4.
    Ngai J. BRAIN 2.0: Transforming neuroscience. Cell. 2022;185(1):4-8. doi:10.1016/j.cell.2021.11.037 [Google Scholar] [PubMed] [CrossRef]
  • 5.
    Stanley MPH, Silbersweig DA, Perez DL. Toward a Unified Classification System for Brain-Mind Disorders: Putting Calls for Integrated Clinical Neuroscience Into Action. Cogn Behav Neurol. 2023;36(4):199-201. [Google Scholar] [PubMed] [CrossRef]
  • 6.
    Marsman A, Mandl RC, Klomp DW, et al. GABA and glutamate in schizophrenia: a 7 T ¹H-MRS study. Neuroimage Clin. 2014;6:398-407. [Google Scholar] [PubMed] [CrossRef]
  • 7.
    Cherubini E, Di Cristo G, Avoli M. Dysregulation of GABAergic Signaling in Neurodevelomental Disorders: Targeting Cation-Chloride Co-transporters to Re-establish a Proper E/I Balance. Front Cell Neurosci. 2022;15:813441. [Google Scholar] [PubMed] [CrossRef]
  • 8.
    Boileau I, Warsh JJ, Guttman M, et al. Elevated serotonin transporter binding in depressed patients with Parkinson's disease: a preliminary PET study with [11C]DASB. Mov Disord. 2008;23(12):1776-1780. [Google Scholar] [PubMed] [CrossRef]
  • 9.
    Kempuraj D, Thangavel R, Selvakumar GP, et al. Brain and Peripheral Atypical Inflammatory Mediators Potentiate Neuroinflammation and Neurodegeneration. Front Cell Neurosci. 2017;11:216. [Google Scholar] [PubMed] [CrossRef]
  • 10.
    Darwish SF, Elbadry AMM, Elbokhomy AS, Salama GA, Salama RM. The dual face of microglia (M1/M2) as a potential target in the protective effect of nutraceuticals against neurodegenerative diseases. Front Aging. 2023;4:1231706. [Google Scholar] [PubMed] [CrossRef]
  • 11.
    Garcia KL, Yu G, Nicolini C, et al. Altered balance of proteolytic isoforms of pro-brain-derived neurotrophic factor in autism. J Neuropathol Exp Neurol. 2012;71(4):289-297. [Google Scholar] [PubMed] [CrossRef]
  • 12.
    Mizui T, Kojima M. Recent Advances in the Biology of BDNF And the Newly Identified Pro-Peptide. J Neurol Neuromedicine (2018) 3(6): 1-4 [Google Scholar] [PubMed] [CrossRef]
  • 13.
    Gholami M, Hozuri F, Abdolkarimi S, et al. Pharmacological and Molecular Evidence of Neuroprotective Curcumin Effects Against Biochemical and Behavioral Sequels Caused by Methamphetamine: Possible Function of CREB-BDNF Signaling Pathway. Basic Clin Neurosci. 2021;12(3):325-338. [Google Scholar] [PubMed] [CrossRef]
  • 14.
    Smeland OB, Kutrolli G, Bahrami S, et al. The shared genetic risk architecture of neurological and psychiatric disorders: a genome-wide analysis. Preprint. medRxiv. 2023;2023.07.21.23292993. [Google Scholar] [PubMed] [CrossRef]
  • 15.
    Martínez-Iglesias O, Naidoo V, Carrera I, Carril JC, Cacabelos N, Cacabelos R. Influence of Metabolic, Transporter, and Pathogenic Genes on Pharmacogenetics and DNA Methylation in Neurological Disorders. Biology (Basel). 2023;12(9):1156. [Google Scholar] [PubMed] [CrossRef]
  • 16.
    Postberg J, Schubert MT, Nin V, Wagner L, Piefke M. A perspective on epigenomic aging processes in the human brain and their plasticity in patients with mental disorders - a systematic review. Neurogenetics. 2024;25(4):351-366. [Google Scholar] [PubMed] [CrossRef]
  • 17.
    Goud Alladi C, Etain B, Bellivier F, Marie-Claire C. DNA Methylation as a Biomarker of Treatment Response Variability in Serious Mental Illnesses: A Systematic Review Focused on Bipolar Disorder, Schizophrenia, and Major Depressive Disorder. Int J Mol Sci. 2018;19(10):3026. [Google Scholar] [PubMed] [CrossRef]
  • 18.
    Rajput A, Pingale P, Dhapte-Pawar V. Nasal delivery of neurotherapeutics via nanocarriers: Facets, aspects, and prospects. Front Pharmacol. 2022;13:979682. [Google Scholar] [PubMed] [CrossRef]
  • 19.
    Ahmad SR, Zeyaullah M, Khan MS, et al. Pharmacogenomics for neurodegenerative disorders - a focused review. Front Pharmacol. 2024;15:1478964. [Google Scholar] [PubMed] [CrossRef]
  • 20.
    Bugarski-Kirola D, Abbs B, Odetalla R, Liu IY, Darwish M, DeKarske D. Adherence to Background Antipsychotic and Pimavanserin in Patients with Schizophrenia: Post Hoc Analyses from the ENHANCE and ADVANCE Studies. Patient Prefer Adherence. 2024;18:207-216. [Google Scholar] [PubMed] [CrossRef]
  • 21.
    Vasiliu O, Mangalagiu AG, Petrescu BM, et al. Next-generation antipsychotics- Trends and perspectives beyond dopaminergic and glutamatergic agents. Eur Psychiatry. 2023;66(Suppl 1):S558-S559. [Google Scholar] [PubMed] [CrossRef]
  • 22.
    Chen MH, Tu PC, Su TP. Next generation antidepressants with novel mechanisms for treatment resistant depression. Prog Brain Res. 2023;278:149-168. [Google Scholar] [PubMed] [CrossRef]
  • 23.
    Duman RS, Aghajanian GK. Synaptic dysfunction in depression: potential therapeutic targets. Science. 2012;338(6103):68-72. [Google Scholar] [PubMed] [CrossRef]
  • 24.
    Jha MK, Mathew SJ. Pharmacotherapies for Treatment-Resistant Depression: How Antipsychotics Fit in the Rapidly Evolving Therapeutic Landscape. Am J Psychiatry. 2023;180(3):190-199. [Google Scholar] [PubMed] [CrossRef]
  • 25.
    Hamilton JM. The role of pharmacogenomics in precision mental health care. J Am Assoc Nurse Pract. 2024;36(3):143-146. [Google Scholar] [PubMed] [CrossRef]
  • 26.
    Alhaddad S, Paulsingh C, Riaz F, Garg G, Umeano L, Iftikhar S, et al. Impact of Pharmacogenomics Testing on Response to Antidepressants and Antipsychotics - A Systematic Review. J Int Med Res. 2024;3(1);1-10. [Google Scholar] [PubMed] [CrossRef]
  • 27.
    Skoczeń A, Rutecka N, Kaczmarek B, Kuśnierz-Gibała A, Kruk A, Kulesza M, et al. Advancements in Alzheimer’s Disease Therapy: The Role of Anti-Amyloid Monoclonal Antibodies - A Literature Review. Journal of Education, Health and Sport. 2025;79:58362. [Google Scholar] [PubMed] [CrossRef]
  • 28.
    Singh K, Bhushan B, Chanchal DK, et al. Emerging Therapeutic Potential of Cannabidiol (CBD) in Neurological Disorders: A Comprehensive Review. Behav Neurol. 2023;2023:8825358. [Google Scholar] [PubMed] [CrossRef]
  • 29.
    Bors LA, Erdő F. Overcoming the Blood–Brain Barrier. Challenges and Tricks for CNS Drug Delivery. Scientia Pharmaceutica. 2019; 87(1):6. [Google Scholar] [PubMed] [CrossRef]
  • 30.
    Mazahir F, Alam MI, Yadav AK. Development of nanomedicines for the treatment of Alzheimer's disease: Raison d'être, strategies, challenges and regulatory aspects. Ageing Res Rev. 2024;98:102318. [Google Scholar] [PubMed] [CrossRef]
  • 31.
    Chakraborty C, Hsu CH, Wen ZH, Lin CS, Agoramoorthy G. Zebrafish: a complete animal model for in vivo drug discovery and development. Curr Drug Metab. 2009;10(2):116-124. [Google Scholar] [PubMed] [CrossRef]
  • 32.
    Siraj S. Precision Medicine Approaches for Psychosis Treatment. Precis. Med. Com. 2023;3(1):01-02. [Google Scholar] [PubMed] [CrossRef]
  • 33.
    Lin E, Lin CH, Lane HY. Precision Psychiatry Applications with Pharmacogenomics: Artificial Intelligence and Machine Learning Approaches. Int J Mol Sci. 2020;21(3):969. [Google Scholar] [PubMed] [CrossRef]
  • 34.
    Alchakee A, Ahmed M, Eldohaji L, Alhaj H, Saber-Ayad M. Pharmacogenomics in Psychiatry Practice: The Value and the Challenges. Int J Mol Sci. 2022;23(21):13485. [Google Scholar] [PubMed] [CrossRef]
  • 35.
    Chen CB, Abe R, Pan RY, et al. An Updated Review of the Molecular Mechanisms in Drug Hypersensitivity [published correction appears in J Immunol Res. 2019 Jan 31;2019:2489429. doi: 10.1155/2019/2489429.]. J Immunol Res. 2018;2018:6431694. [Google Scholar] [PubMed] [CrossRef]
  • 36.
    Srinivasan N, Mehra E, Dommaraju S, Kakavetsis E. Neurogenetics: Precision Medicine-Based Approaches to Neurological Disorders with an Emphasis on Addressing Alzheimer’s Disease and Schizophrenia. bptjm. 2024;4(1):14-33. [Google Scholar] [PubMed] [CrossRef]
  • 37.
    Hornick MG, Potempa LA. Monomeric C-reactive protein as a biomarker for major depressive disorder. Front Psychiatry. 2024;14:1325220. [Google Scholar] [PubMed] [CrossRef]
  • 38.
    Amaro-Álvarez L, Cordero-Ramos J, Calleja-Hernández MÁ. Exploring the impact of pharmacogenetics on personalized medicine: A systematic review. Farm Hosp. 2024;48(6):299-309. [Google Scholar] [PubMed] [CrossRef]
  • 39.
    Hockings JK, Pasternak AL, Erwin AL, Mason NT, Eng C, Hicks JK. Pharmacogenomics: An evolving clinical tool for precision medicine. Cleve Clin J Med. 2020;87(2):91-99. [Google Scholar] [PubMed] [CrossRef]
  • 40.
    Pardiñas AF, Nalmpanti M, Pocklington AJ, et al. Pharmacogenomic Variants and Drug Interactions Identified Through the Genetic Analysis of Clozapine Metabolism. Am J Psychiatry. 2019;176(6):477-486. [Google Scholar] [PubMed] [CrossRef]
  • 41.
    Butcher NJ, Fung WL, Fitzpatrick L, et al. Response to clozapine in a clinically identifiable subtype of schizophrenia. Br J Psychiatry. 2015;206(6):484-491. [Google Scholar] [PubMed] [CrossRef]
  • 42.
    de Vos CMH, Mason NL, Kuypers KPC. Psychedelics and Neuroplasticity: A Systematic Review Unraveling the Biological Underpinnings of Psychedelics. Front Psychiatry. 2021;12:724606. [Google Scholar] [PubMed] [CrossRef]
  • 43.
    van der Steld L de P. Electrical stimulation in special clinical situations. J. Cardiac Arrhythmias. 2024;37:1-14. [Google Scholar] [PubMed] [CrossRef]
  • 44.
    Dell'osso B, Camuri G, Castellano F, et al. Meta-Review of Metanalytic Studies with Repetitive Transcranial Magnetic Stimulation (rTMS) for the Treatment of Major Depression. Clin Pract Epidemiol Ment Health. 2011;7:167-177. [Google Scholar] [PubMed] [CrossRef]
  • 45.
    Ling Q, Herstine JA, Bradbury A, Gray SJ. AAV-based in vivo gene therapy for neurological disorders. Nat Rev Drug Discov. 2023;22(10):789-806. [Google Scholar] [PubMed] [CrossRef]
  • 46.
    Wang S, Xiao L. Progress in AAV-Mediated In Vivo Gene Therapy and Its Applications in Central Nervous System Diseases. Int J Mol Sci. 2025;26(5):2213. [Google Scholar] [PubMed] [CrossRef]
  • 47.
    Legare CA, Raup-Konsavage WM, Vrana KE. Therapeutic Potential of Cannabis, Cannabidiol, and Cannabinoid-Based Pharmaceuticals. Pharmacology. 2022;107(3-4):131-149. [Google Scholar] [PubMed] [CrossRef]
  • 48.
    Nutt DJ, Phillips LD, Barnes MP, et al. A Multicriteria Decision Analysis Comparing Pharmacotherapy for Chronic Neuropathic Pain, Including Cannabinoids and Cannabis-Based Medical Products. Cannabis Cannabinoid Res. 2022;7(4):482-500. [Google Scholar] [PubMed] [CrossRef]
  • 49.
    Peplow PV. Animal models in medical translation: the grand challenge of developing new treatments for human diseases. Front Med Technol. 2024;6:1367521. [Google Scholar] [PubMed] [CrossRef]
  • 50.
    Dionne O, Sabatié S, Laurent B. Deciphering the physiopathology of neurodevelopmental disorders using brain organoids. Brain. 2025;148(1):12-26. [Google Scholar] [PubMed] [CrossRef]
  • 51.
    Jennings CG, Landman R, Zhou Y, et al. Opportunities and challenges in modeling human brain disorders in transgenic primates [published correction appears in Nat Neurosci. 2017 Jun 27;20(7):1033. [Google Scholar] [PubMed] [CrossRef]
  • 52.
    Guha L, Singh N, Kumar H. Challenges in Drug Development for Neurological Disorders. Mishra A, Kulhari H, editors. Drug Delivery Strategies in Neurological Disorders: Challenges and Opportunities. Singapore: Springer Nature. 2023;27-45. [Google Scholar] [PubMed
  • 53.
    Husain M. Smarter adaptive platform clinical trials in neurology. Brain. 2022;145(2):409-410. [Google Scholar] [PubMed] [CrossRef]
  • 54.
    Niazi SK. Safety Concerns in Neurological Clinical Trials: A Challenge That the FDA Must Resolve. Biomedicines. 2024;12(12):2918. [Google Scholar] [PubMed] [CrossRef]
  • 55.
    Schermer M. Ethical issues in deep brain stimulation. Front Integr Neurosci. 2011;5:17. [Google Scholar] [PubMed] [CrossRef]
  • 56.
    Ayanoğlu FB, Elçin AE, Elçin YM. Bioethical issues in genome editing by CRISPR-Cas9 technology. Turk J Biol. 2020;44(2):110-120. [Google Scholar] [PubMed] [CrossRef]
  • 57.
    Lee TL, Sawai T. Navigating equity in global access to genome therapy expanding access to potentially transformative therapies and benefiting those in need requires global policy changes. Front Genet. 2024;15:1381172. [Google Scholar] [PubMed] [CrossRef]
  • 58.
    Martin DE, Van Assche K, Cervantes L, et al. Toward Equity in Global Access to SoHO-based Therapies: Recommendations for Action. Transplantation. 2025;109(1):60-72. [Google Scholar] [PubMed] [CrossRef]
How to Cite This Article: Ram D. Advancing translational neurotherapeutics: Bridging pharmacy, psychiatry, and neurology for precision brain disorder management NeuroPharmac Journal 2025 April; (04): 01-08
©2022 NeuroPharmac J. This is an open-access journal, and articles are distributed under the terms of the Creative Commons AttributionNonCommercial-ShareAlike 4.0 License.
(Visited 97 times, 1 visits today)
RELATED POSTS