Skip to main navigation menu Skip to main content Skip to site footer

Repurposing Pharmaceutical Drugs for the Treatment of Mental Disorders

Abstract

The multifactorial nature of the etiology of mental disorders and the imperfections in diagnostic approaches hinder the development of more effective medications. The repurposing of previously approved therapeutic agents is one of the promising approaches in the search for new treatments for mental disorders. This is related to the fact that, firstly, this approach is less resource- and economically costly compared to other approaches to drug development. Secondly, the methods used to identify drug candidates for repurposing are highly diverse and cover various aspects of the pathogenesis of mental disorders. Research in the field of drug repurposing can improve the understanding of the mechanisms underlying the development of mental pathology while helping to optimize therapeutic and preventive approaches to the treatment of patients with this group of disorders. In this review, the diversity of approaches to drug repurposing is briefly characterized, with a focus on in silico methods. In addition, examples of successful cases of drug repurposing in psychiatry are provided.

Keywords

drug repurposing in psychiatry, main approaches to repurposing, drug targets, genetic studies

PDF (Русский)

References

  1. Vos T., Lim S.S., Abbafati C. et al. Global burden of 369 diseases and injuries in 204 countries and territories, 1990–2019: A systematic analysis for the Global Burden of Disease Study 2019 // The Lancet. – 2020. – Vol. 396 (10258). – Pp. 1204–1222. – https://doi.org/10.1016/S0140-6736(20)30925-9
  2. Riordan H.J., Cutler H.R. The death of CNS drug development: overstatement or Omen? // J Clin Stud. – 2011. – Vol. 3 – Pp. 12–15.
  3. Hughes J.P., Rees S., Kalindjian S.B., Philpott K.L. Principles of early drug discovery. // Br J Pharmacol. – 2011. – Vol. 162 (6). – Pp. 1239–1249. – https://doi.org/10.1111/j.1476-5381.2010.01127.x
  4. Blokhin I.O., Khorkova O., Saveanu R.V., Wahlestedt C. Molecular mechanisms of psychiatric diseases // Neurobiol Dis. – 2020. – Vol. 146. – Art. 105136. – https://doi.org/10.1016/j.nbd.2020.105136
  5. Caban A., Pisarczyk K., Kopacz K. et al. Filling the gap in CNS drug development: evaluation of the role of drug repurposing // J Mark Access Health Policy. – 2017. – Vol. 5 (1). – Art. 1299833. – https://doi.org/10.1080/20016689.2017.1299833
  6. Aminoff M.J., Boller F., Swaab D.F. Handbook of clinical neurology: Neurobiology of psychiatric disorders: foreword. – Vol. 106. – Amsterdam, Netherlands: Elsevier, 2012. – Pp. 2–760.
  7. Shemesh Y., Chen A. A paradigm shift in translational psychiatry through rodent neuroethology // Mol Psychiatry. –2023. – Vol. 28 (3). – Pp. 993–1003. – https://doi.org/10.1038/s41380-022-01913-z
  8. Truong T.T.T., Panizzutti B., Kim J.H., Walder K. Repurposing drugs via network analysis: Opportunities for psychiatric disorders // Pharmaceutics. – 2022. – Vol. 14 (7). – Art. 1464. – https://doi.org/10.3390/pharmaceutics14071464
  9. U.S. Food and Drug Administration. New Molecular Entity (NME) Drug and New Biologic Approvals. – Silver Spring, MD, USA: U.S. Food and Drug Administration, 2019.
  10. U.S. Food and Drug Administration. New Molecular Entity (NME) Drug and New Biologic Approvals. – Silver Spring, MD, USA: U.S. Food and Drug Administration, 2020.
  11. Silva M.C., Haggarty S.J. Human pluripotent stem cell-derived models and drug screening in CNS precision medicine // Ann N Y Acad Sci. – 2020. – Vol. 1471 (1). – Pp. 18–56. – https://doi.org/10.1111/nyas.14012
  12. Murteira S., Ghezaiel Z., Karray S. et al. Drug reformulations and repositioning in pharmaceutical industry and its impact on market access: reassessment of nomenclature // J Mark Access Health Policy. – 2013. – Vol. 1. – https://doi.org/10.3402/jmahp.v1i0.21131
  13. Wang S.M., Lee H.K., Kweon Y.S. et al. Overactive Bladder Successfully Treated with Duloxetine in a Female Adolescent // Clin Psychopharmacol Neurosci. – 2015. – Vol. 13 (2). – Pp. 212–214. – https://doi.org/10.9758/cpn.2015.13.2.212
  14. Deftereos S.N., Dodou E., Andronis C., Persidis A. From depression to neurodegeneration and heart failure: re-examining the potential of MAO inhibitors // Expert Rev Clin Pharmacol. – 2012. – Vol. 5 (4). – Pp. 413–425. – https://doi.org/10.1586/ecp.12.29
  15. Decherchi S., Cavalli A. Thermodynamics and kinetics of drug-target binding by molecular simulation // Chem Rev. – 2020. – Vol. 120 (23). – Pp. 12788–12833. – https://doi.org/10.1021/acs.chemrev.0c00534
  16. Dudley J., Berliocchi L. Drug repositioning: approaches and applications for neurotherapeutics // CRC press: Boca Raton. – 2017. – Vol. 1. – http://dx.doi.org/10.4324/9781315373669
  17. Jumper J., Evans R., Pritzel A. et al. Highly accurate protein structure prediction with AlphaFold // Nature. – 2021. – Vol. 596 (7873). – Pp. 583–589. – https://doi.org/10.1038/s41586-021-03819-2
  18. McNutt A.T., Francoeur P., Aggarwal R. et al. GNINA 1.0: Molecular docking with deep learning // J Cheminformatics. – 2021. – Vol. 13 (1). – Art. 43. – https://doi.org/10.1186/s13321-021-00522-2
  19. Jiang H., Wang J., Cong W. et al. Predicting Protein–Ligand docking structure with graph neural network // J Chem Inf Model. – 2022. – Vol. 62 (12). – Pp. 2923–2932. – https://doi.org/10.1021/acs.jcim.2c00127
  20. Mathai N., Kirchmair J. Similarity-based methods and machine learning approaches for target prediction in early drug discovery: Performance and scope // Int J Mol Sci. – 2020. – Vol. 21 (10). – Art. 3585. – https://doi.org/10.3390/ijms21103585
  21. Daina A., Michielin O., Zoete V. Swiss target prediction: updated data and new features for efficient prediction of protein targets of small molecules // Nucleic Acids Res. –2019. – Vol. 47 (W1). – Pp. W357–W364. – https://doi.org/10.1093/nar/gkz382
  22. Nickel J., Gohlke B.O., Erehman J. et al. SuperPred: update on drug classification and target prediction // Nucleic Acids Res. – 2014. – Vol. 42 (Web Server issue). – Pp. W26– W31. – https://doi.org/10.1093/nar/gku477
  23. Ochoa D., Karim M., Ghoussaini M. et al. Human genetics evidence supports two-thirds of the 2021 FDA-approved drugs // Nat Rev Drug Discov. – 2022. – Vol. 21 (8). – Art. 551. – https://doi.org/10.1038/d41573-022-00120-3
  24. Miyagawa T., Tokunaga K. Genetics of narcolepsy // Hum Genome Var. – 2019. – Vol. 6 (1). – Pp. 1–8. – https://doi.org/10.1038/s41439-018-0033-7
  25. Casamassima F., Hay A.C., Benedetti A. et al. L-type calcium channels and psychiatric disorders: A brief review // Am J Med Genet B Neuropsychiatr Genet. – 2010. – Vol. 153B (8). – Pp. 1373–1390. – https://doi.org/10.1002/ajmg.b.31122
  26. Ostacher M.J., Iosifescu D.V., Hay A. et al. Pilot investigation of Isradipine in the treatment of bipolar depression motivated by genome-wide association // Bipolar Disord. – 2014. – Vol. 16 (2). – Pp. 199–203. – https://doi.org/10.1002/10.1111/bdi.12143
  27. Doggrell S.A., Hancox J.C. Ibutilide-recent molecular insights and accumulating evidence for use in atrial flutter and fibrillation Expert // Opin Investig Drugs. – 2005. – Vol. 14 (5). – Pp. 655–69. – https://doi.org/10.1517/13543784.14.5.655
  28. Pritchard J.L.E., O’Mara T.A., Glubb D.M. Enhancing the Promise of Drug Repositioning through Genetics // Front Pharmacol. – 2017. – Available at: https://www.frontiersin.org/articles/10.3389/fphar.2017.00896 (accessed February 21, 2023).
  29. So H.C., Lau A., Chau C.K.L., Wong S.Y. Translating GWAS findings into therapies for depression and anxiety disorders: Drug repositioning using gene-set analyses reveals enrichment of psychiatric drug classes // Genetics. – 2017. – https://doi.org/10.1101/132563
  30. de Jong S., Vidler L.R., Mokrab Y. et al. Gene-set analysis based on the pharmacological profiles of drugs to identify repurposing opportunities in schizophrenia // J Psychopharmacol Oxf Engl. – 2016. – Vol. 30 (8). – Pp. 826–830. – https://doi.org/10.1177/0269881116653109
  31. Gaspar H., Hübel C., Breen G. Drug Targetor: A web interface to investigate the human druggome for over 500 phenotypes // Bioinforma Oxf Engl. – 2018. – Vol. 35 (14). – Pp. 2515–2517. – https://doi.org/10.1093/bioinformatics/bty982
  32. Bell N., Uffelmann E., van Walree E. et al. Using genome-wide association results to identify drug repurposing candidates // Genetic and Genomic Medicine. – 2022. – https://doi.org/10.1101/2022.09.06.22279660
  33. Yetisgen-Yildiz M., Pratt W. Using statistical and knowledge-based approaches for literature-based discovery // J Biomed Inform. – 2006. – Vol. 39 (6). – Pp. 600–611. – https://doi.org/10.1016/j.jbi.2005.11.010
  34. Yetisgen-Yildiz M. A new evaluation methodology for literature-based discovery systems // J Biomed Inform. – 2009. – Vol. 42 (4). – Pp. 633–643. – https://doi.org/10.1016/j.jbi.2008.12.001
  35. Reay W.R., Atkins J.R., Carr V.J. et al. Pharmacological enrichment of polygenic risk for precision medicine in complex disorders // Sci Rep. – 2020. – Vol. 10 (1). – Art. 879. – https://doi.org/10.1038/s41598-020-57795-0
  36. Smith G.D., Ebrahim S. “Mendelian randomization”: can genetic epidemiology contribute to understanding environmental determinants of disease? // Int J Epidemiol. – 2003. – Vol. 32 (1). – Pp. 1–22. – https://doi.org/10.1093/ije/dyg070
  37. Walker V.M., Davey Smith G., Davies N.M., Martin R.M. Mendelian randomization: a novel approach for the prediction of adverse drug events and drug repurposing opportunities // Int J Epidemiol. – 2017. – Vol. 46 (6). – Pp. 2078–2089. – https://doi.org/10.1093/ije/dyx207
  38. McIntyre R.S., Rosenblat J.D., Nemeroff C.B. et al. Synthesizing the evidence for Ketamine and Esketamine in treatment-resistant depression: An International expert opinion on the available evidence and implementation // Am J Psychiatry. – 2021. – Vol. 178 (5). – Pp. 383–399. – https://doi.org/10.1176/appi.ajp.2020.20081251
  39. Millard L.A.C., Davies N.M., Timpson N.J. et al. MR-PheWAS: Hypothesis prioritization among potential causal effects of body mass index on many outcomes, using Mendelian randomization. // Sci Rep. – 2015. – Vol. 5. – Art. 16645. – https://doi.org/10.1038/srep16645
  40. Burgess S., Thompson S.G. Interpreting findings from Mendelian randomization using the MR-Egger method // Eur J Epidemiol. – 2017. – Vol. 32 (5). – Pp. 377–389. – https://doi.org/10.1007/s10654-017-0255-x
  41. So H.C., Chau C.K.L., Chiu W.T. et al. Analysis of genome-wide association data highlights candidates for drug repositioning in psychiatry // Nat Neurosci. – 2017. – Vol. 20 (10). – Pp. 1342–1349. – https://doi.org/10.1038/nn.4618
  42. Barbeira N.A., Dickinson S.P., Bonazzola R. et al. Exploring the phenotypic consequences of tissue specific gene expression variation inferred from GWAS summary statistics // Nat Commun. – 2018. – Vol. 9 (1). – Art. 1825. – https://doi.org/10.1038/s41467-018-03621-1
  43. Lamb J., Crawford E.D., Peck D. et al. The Connectivity Map: using gene-expression signatures to connect small molecules, genes, and disease // Science. – 2006. – Vol. 313 (5795). – Pp. 1929–1935. – https://doi.org/10.1126/science.1132939
  44. Méndez-Lucio O., Baillif B., Clevert D.A., Rouquié D., Wichard J. De novo generation of hit-like molecules from gene expression signatures using artificial intelligence // Nat Commun. – 2020. – Vol. 11 (1). – Art. 10. – https://doi.org/10.1038/s41467-019-13807-w
  45. Shayakhmetov R., Kuznetsov M., Zhebrak A., et al. Molecular Generation for Desired Transcriptome Changes with Adversarial Autoencoders // Front Pharmacol. – 2020. – Available at: https://www.frontiersin.org/articles/10.3389/fphar.2020.00269 (accessed January 30, 2023).
  46. Lüscher T. D., Schuch V., Beltrão-Braga P.C.B. et al. Drug repositioning for psychiatric and neurological disorders through a network medicine approach // Transl Psychiatry. – 2020. – Vol. 10 (1). – Pp. 1–10. – https://doi.org/10.1038/s41398-020-0827-5
  47. Ban T.A. Fifty years chlorpromazine: A historical perspective // Neuropsychiatric Disease and Treatment. – 2007. – Vol. 3 (4). – Pp. 495–500.
  48. Ferry L., Johnston J.A. Efficacy and safety of bupropion SR for smoking cessation: data from clinical trials and five years of postmarketing experience // Int J Clin Pract. – 2003. – Vol. 57 (3). – Pp. 224–230.
  49. Das J. Repurposing of drugs — the Ketamine story // J Med Chem. – 2020. – Vol. 63 (22). – Pp. 13514–13525. – https://doi.org/10.1021/acs.jmedchem.0c01193
  50. Sofia R.D., Harakal J.J. Evaluation of ketamine HCl for anti-depressant activity // Arch Int Pharmacodyn Ther. – 1975. – Vol. 214 (1). – Pp. 68–74.
  51. Krupitsky E.M., Grinenko A.Y. Ketamine Psychedelic Therapy (KPT): A review of the results of ten years of research // J Psychoactive Drugs. – 1997. – Vol. 29 (2). – Pp. 165– 183. – https://doi.org/10.1080/02791072.1997.10400185
  52. Berman R.M., Cappiello A., Anand A. et al. Antidepressant effects of ketamine in depressed patients // Biol Psychiatry. – 2000. – Vol. 47 (4). – Pp. 351–354. – https://doi.org/10.1016/s0006-3223(99)00230-9
  53. Baldessarini R.J., Wright V. et al. A double-blind placebo controlled study of Desipramine in the treatment of ADD: I. Efficacy // J Am Acad Child Adolesc Psychiatry. – 1989. – Vol. 28. – Pp. 777–784. – https://doi.org/10.1097/00004583-198909000-00022
  54. Angeles A.D.L., Fernando M.B., Hall N.A.L. et al. Induced Pluripotent Stem Cells in Psychiatry: An Overview and Critical Perspective // Biol Psychiatry. – 2021. – Vol. 90 (6). – Pp. 362–372. – https://doi.org/10.1016/j.biopsych.2021.04.008
  55. Sellgren C.M., Gracias J., Watmuff B. et al. Increased synapse elimination by microglia in schizophrenia patient-derived models of synaptic pruning // Nat Neurosci. – 2019. – Vol. 22 (3). – Pp. 374–385 – https://doi.org/10.1038/s41593-018-0334-7
  56. Kishi T., Ikuta T., Matsuda Y. et al. Pharmacological treatment for bipolar mania: A systematic review and network meta-analysis of double-blind randomized controlled trials // Mol Psychiatry. – 2022. – Vol. 27 (2). – Pp. 1136–1144. – https://doi.org/10.1038/s41380-021-01334-4
  57. Chen C.C., Zhou N., Hu N. et al. Acute effects of intravenous sub-anesthetic doses of Ketamine and intranasal inhaled Esketamine on suicidal ideation: A systematic review and meta-analysis // Neuropsychiatr Dis Treat. – 2023. – Vol. 19. – Pp. 587–599. – https://doi.org/10.2147/NDT.S401032
  58. Fountoulakis K.N., Tohen M., Zarate C.A. Lithium treatment of Bipolar disorder in adults: A systematic review of randomized trials and meta-analyses // Eur Neuropsychopharmacol. – 2022. – Vol. 54. – Pp. 100–115. – https://doi.org/10.1016/j.euroneuro.2021.10.003
  59. Masdrakis V.G., Baldwin D.S. Prevention of suicide by clozapine in mental disorders: Systematic review // Eur Neuropsychopharmacol. – 2023. – Vol. 69. – Pp. 4–23. – https://doi.org/10.1016/j.euroneuro.2022.12.0
  60. Mann J.J., Michel C.A., Auerbach R.P. Improving Suicide prevention through evidence-based strategies: A systematic review // Am J Psychiatry. – 2021. – Vol. 178 (7). – Pp. 611– 624. – https://doi.org/10.1176/appi.ajp.2020.20060864

Downloads

Download data is not yet available.