A REVIEW ARTICLE ON TREATING HUNTINGTON’S DISEASE: CURRENT AND EMERGING THERAPEUTIC ASPECTS

Authors

  • NEHA A BARDE Department of Pharmacology, PES Modern College of Pharmacy, Pune, Maharashtra, India
  • PADMAJA S KORE Department of Pharmacology, PES Modern College of Pharmacy, Pune, Maharashtra, India
  • BANDAWANE DD Department of Pharmacology, PES Modern College of Pharmacy, Pune, Maharashtra, India

DOI:

https://doi.org/10.22159/ijms.2025v13i1.53191

Keywords:

Huntington Chorea/Huntington’s disease, Genetics, Pathogenesis, Polyglutamine, ZFP, TALEN

Abstract

A rare genetic neurodegenerative disorder called Huntington’s disease (HD) causes nerve cells to progressively die. When the trinucleotide CAG repeats are <36, it is regarded as “normal.” When the repetitions in the huntingtin gene are >36, polyglutamine (polyQ) tract leads to polyQ poisoning, which in turn causes psychological, genetic, and movement disorders which are hallmarks of HD. One particularly concerning aspect of HD is its controlling inheritance pattern, this indicates that each child born to a parent who exaggerated through the condition has a 50% likelihood of having the lineage of mutated gene that causes the disease. As a result, there is a significant risk that offspring of individuals with HD may also develop the condition. The underlying molecular mechanisms resulting in the visible loss of neurons remain incompletely elucidated, and the current therapeutic approaches primarily aim to alleviate symptoms. Many symptoms can be treated with the HD prescription medications that are currently available. These include prescribed medication for chorea, tranquilizers, calming agents, antidepressants, and non-pharmacological therapy. Additional possible treatments now undergoing clinical research include RNA interference therapies, therapies targeting RNA using tiny molecules, antibody therapies, stem cell therapies, small molecule therapies not targeting RNA, and therapies concentrating on neuroinflammation. Among the potential treatments presently in pre-clinical development stages are zinc-finger protein therapies, transcription activator-like effector nuclease therapies, and rehabilitations involving clustered regularly interspaced short palindromic repeats (CRISPS)/CRISPS-associated system. Therefore, the purpose of this comprehensive review is to discuss the efficacy of current HD medicines and look into the insights of new emerging therapies which are under pre-clinical development stage.

References

Gatto EM, Rojas NG, Persi G, Etcheverry JL, Cesarini ME, Perandones C. Huntington disease: Advances in the understanding of its mechanisms. Clin Park Relat Disord 2020;3:100056.

Shafie A, Ashour AA, Anwar S, Anjum F, Hassan MI. Exploring molecular mechanisms, therapeutic strategies, and clinical manifestations of Huntington’s disease. Arch Pharm Res 2024;47:571-95.

McColgan P, Tabrizi SJ. Huntington’s disease: A clinical review. Eur J Neurol 2018;25:24-34.

Mendez MF. Huntington’s disease: Update and review of neuropsychiatric aspects. Int J Psychiatry Med 1994;24:189-208.

Gil JM, Rego AC. Mechanisms of neurodegeneration in Huntington’s disease. Eur J Neurosci 2008;27:2803-20.

Andhale R, Shrivastava D. Huntington’s disease: A clinical review. Cureus 2022;14:e28484.

Van Duijn E, Kingma EM, Van der Mast RC. Psychopathology in verified Huntington’s disease gene carriers. J Neuropsychiatry Clin Neurosci 2007;19:441-8.

Novak MJ, Tabrizi SJ. Huntington’s disease: Clinical presentation and treatment. Int Rev Neurobiol 2011;98:297-323.

Imarisio S, Carmichael J, Korolchuk V, Chen CW, Saiki S, Rose C, et al. Huntington’s disease: From pathology and genetics to potential therapies. Biochem J 2008;412:191-209.

Dumas EM, Van den Bogaard SJ, Middelkoop HA, Roos RA. A review of cognition in Huntington’s disease. Front Biosci (Schol Ed) 2013;5:1-18.

Montoya A, Price BH, Menear M, Lepage M. Brain imaging and cognitive dysfunctions in Huntington’s disease. J Psychiatry Neurosci 2006;31:21-9.

Kumar A, Ratan RR. Oxidative stress and Huntington’s disease: The good, the bad, and the ugly. J Huntingtons Dis 2016;5:217-37.

Van Eimeren T, Giehl K, Reetz K, Sampaio C, Mestre TA. Neuroimaging biomarkers in Huntington’s disease: Preparing for a new era of therapeutic development. Parkinsonism Relat Disord 2023;114:105488.

Browne SE, Ferrante RJ, Beal MF. Oxidative stress in Huntington’s disease. Brain Pathol 1999;9:147-63.

Morena E, Romano C, Marconi M, Diamant S, Buscarinu MC, Bellucci G, et al. Peripheral biomarkers in manifest and premanifest Huntington’s disease. Int J Mol Sci 2023;24:6051.

Yero T, Rey JA. Tetrabenazine (Xenazine), An FDA-approved treatment option for huntington’s disease-related chorea. P T 2008;33:690-4.

Saft C, Burgunder JM, Dose M, Jung HH, Katzenschlager R, Priller J, et al. Symptomatic treatment options for Huntington’s disease (guidelines of the German Neurological Society). Neurol Res Pract 2023;5:61.

Pérot JB, Brouillet E, Flament J. The contribution of preclinical magnetic resonance imaging and spectroscopy to Huntington’s disease. Front Aging Neurosci 2024;16:1306312.

Paleacu D. Tetrabenazine in the treatment of Huntington’s disease. Neuropsychiatr Dis Treat 2007;3:545-51.

Dash D, Mestre TA. Therapeutic update on Huntington’s disease: Symptomatic treatments and emerging disease-modifying therapies. Neurotherapeutics 2020;17:1645-59.

Quintanilla RA, Johnson GV. Role of mitochondrial dysfunction in the pathogenesis of Huntington’s disease. Brain Res Bull 2009;80:242-7.

Paulsen JS. Cognitive impairment in Huntington disease: Diagnosis and treatment. Curr Neurol Neurosci Rep 2011;11:474-83.

Vinther-Jensen T, Larsen IU, Hjermind LE, Budtz-Jørgensen E, Nielsen TT, Nørremølle A, et al. A clinical classification acknowledging neuropsychiatric and cognitive impairment in Huntington’s disease. Orphanet J Rare Dis 2014;9:114.

Vonsattel JP, Myers RH, Stevens TJ, Ferrante RJ, Bird ED, Richardson EP Jr. Neuropathological classification of Huntington’s disease. J Neuropathol Exp Neurol 1985;44:559-77.

Ogilvie AC, Schultz JL. Memantine Use and cognitive decline in Huntington’s disease: An enroll-HD study. Mov Disord Clin Pract 2023;10:1120-5.

Snowden JS. The neuropsychology of Huntington’s disease. Arch Clin Neuropsychol 2017;32:876-87.

Rosenblatt A, Leroi I. Neuropsychiatry of Huntington’s disease and other basal ganglia disorders. Psychosomatics 2000;41:24-30.

Caron NS, Dorsey ER, Hayden MR. Therapeutic approaches to Huntington disease: From the bench to the clinic. Nat Rev Drug Discov 2018;17:729-50.

Aronin N, DiFiglia M. Huntingtin-lowering strategies in Huntington’s disease: Antisense oligonucleotides, small RNAs, and gene editing. Mov Disord 2014;29:1455-61.

Rook ME, Southwell AL. Antisense oligonucleotide therapy: From design to the huntington disease clinic. BioDrugs 2022;36:105-19.

Leavitt BR, Tabrizi SJ. Antisense oligonucleotides for neurodegeneration. Science 2020;367:1428-9.

Alkanli SS, Alkanli N, Ay A, Albeniz I. CRISPR/Cas9 mediated therapeutic approach in Huntington’s disease. Mol Neurobiol 2023;60:1486-98.

Shin JW, Kim KH, Chao MJ, Atwal RS, Gillis T, MacDonald ME, et al. Permanent inactivation of Huntington’s disease mutation by personalized allele-specific CRISPR/Cas9. Hum Mol Genet 2016;25:4566-76.

Ekman FK, Ojala DS, Adil MM, Lopez PA, Schaffer DV, Gaj T. CRISPR-Cas9-Mediated genome editing increases lifespan and improves motor deficits in a Huntington’s disease mouse model. Mol Ther Nucleic Acids 2019;17:829-39.

Garriga-Canut M, Agustín-Pavón C, Herrmann F, Sánchez A, Dierssen M, Fillat C, et al. Synthetic zinc finger repressors reduce mutant huntingtin expression in the brain of R6/2 mice. Proc Natl Acad Sci U S A 2012;109:E3136-45.

Bashir H. Emerging therapies in Huntington’s disease. Expert Rev Neurother 2019;19:983-95.

Malankhanova TB, Malakhova AA, Medvedev SP, Zakian SM. Modern genome editing technologies in Huntington’s disease research. J Huntingtons Dis 2017;6:19-31.

Fink KD, Deng P, Gutierrez J, Anderson JS, Torrest A, Komarla A, et al. Allele-specific reduction of the mutant Huntingtin allele using transcription activator-like effectors in human Huntington’s Disease fibroblasts. Cell Transplant 2016;25:677-86.

Komatsu H. Innovative therapeutic approaches for Huntington’s disease: From nucleic acids to GPCR-targeting small molecules. Front Cell Neurosci 2021;15:785703.

Song H, Li H, Guo S, Pan Y, Fu Y, Zhou Z, et al. Targeting Gpr52 lowers mutant HTT levels and rescues Huntington’s disease-associated phenotypes. Brain 2018;141:1782-98.

Ali S, Wang P, Murphy RE, Allen JA, Zhou J. Orphan GPR52 as an emerging neurotherapeutic target. Drug Discov Today 2024;29:103922.

Cowan CM, Raymond LA. Selective neuronal degeneration in Huntington’s disease. Curr Top Dev Biol 2006;75:25-71.

Smalley JL, Breda C, Mason RP, Kooner G, Luthi-Carter R, Gant TW, et al. Connectivity mapping uncovers small molecules that modulate neurodegeneration in Huntington’s disease models. J Mol Med (Berl) 2016;94:235-45.

Ferguson MW, Kennedy CJ, Palpagama TH, Waldvogel HJ, Faull RL, Kwakowsky A. Current and possible future therapeutic options for Huntington’s disease. J Cent Nerv Syst Dis 2022;14:11795735221092517.

Rigamonti D, Bauer JH, De-Fraja C, Conti L, Sipione S, Sciorati C, et al. Wild-type huntingtin protects from apoptosis upstream of caspase-3. J Neurosci 2000;20:3705-13.

Rodrigues FB, Wild EJ. Huntington’s disease clinical trials corner: April 2020. J Huntingtons Dis 2020;9:185-97.

Wang X, Zhu S, Drozda M, Zhang W, Stavrovskaya IG, Cattaneo E, et al. Minocycline inhibits caspase-independent and -dependent mitochondrial cell death pathways in models of Huntington’s disease. Proc Natl Acad Sci U S A 2003;100:10483-7.

Ahamad S, Bhat SA. The emerging landscape of small-molecule therapeutics for the treatment of Huntington’s disease. J Med Chem 2022;65:15993-6032.

Wang C, Zhang YF, Guo S, Zhao Q, Zeng Y, Xie Z, et al. GPR52 antagonist reduces huntingtin levels and ameliorates Huntington’s disease-related phenotypes. J Med Chem 2021;64:941-57. Erratum in: J Med Chem 2021;64:1220-1.

Jurcau A, Jurcau MC. Therapeutic strategies in Huntington’s disease: From genetic defect to gene therapy. Biomedicines 2022;10:1895.

Raymond LA, André VM, Cepeda C, Gladding CM, Milnerwood AJ, Levine MS. Pathophysiology of Huntington’s disease: Time-dependent alterations in synaptic and receptor function. Neuroscience 2011;198:252-73.

Denis HL, David LS, Cicchetti F. Antibody-based therapies for Huntington’s disease: Current status and future directions. Neurobiol Dis 2019;132:104569.

Lansita JA, Mease KM, Qiu H, Yednock T, Sankaranarayanan S, Kramer S. Nonclinical development of ANX005: Ahumanized anti-c1q antibody for treatment of autoimmune and neurodegenerative diseases. Int J Toxicol 2017;36:449-62.

Gupta H, Sahi S. High-throughput virtual screening of potential inhibitors of GPR52 using docking and biased sampling method for Huntington’s disease therapy. Mol Divers 2024;28:3331-47.

Feigin A, Evans EE, Fisher TL, Leonard JE, Smith ES, Reader A, et al. Pepinemab antibody blockade of SEMA4D in early Huntington’s disease: A randomized, placebo-controlled, phase 2 trial. Nat Med 2022;28:2183-93. Erratum in: Nat Med 2024;30:606.

Yao Y, Cui X, Al-Ramahi I, Sun X, Li B, Hou J, et al. A striatal-enriched intronic GPCR modulates huntingtin levels and toxicity. Elife 2015;4:05449.

Atwal RS, Xia J, Pinchev D, Taylor J, Epand RM, Truant R. Huntingtin has a membrane association signal that can modulate huntingtin aggregation, nuclear entry and toxicity. Hum Mol Genet 2007;16:2600-15.

Sagredo O, Pazos MR, Valdeolivas S, Fernandez-Ruiz J. Cannabinoids: Novel medicines for the treatment of Huntington’s disease. Recent Pat CNS Drug Discov 2012;7:41-8.

Bhat SA, Ahamad S, Dar NJ, Siddique YH, Nazir A. The emerging landscape of natural small-molecule therapeutics for Huntington’s disease. Curr Neuropharmacol 2023;21:867-89.

Thomas M, Le WD, Jankovic J. Minocycline and other tetracycline derivatives: A neuroprotective strategy in Parkinson’s disease and Huntington’s disease. Clin Neuropharmacol 2003;26:18-23.

Conner LT, Srinageshwar B, Bakke JL, Dunbar GL, Rossignol J. Advances in stem cell and other therapies for Huntington’s disease: An update. Brain Res Bull 2023;199:110673.

Duncan RS, Riordan SM, Gernon MC, Koulen P. Cannabinoids and endocannabinoids as therapeutics for nervous system disorders: Preclinical models and clinical studies. Neural Regen Res 2024; 19:788-99.

Lescaudron L, Naveilhan P, Neveu I. The use of stem cells in regenerative medicine for Parkinson’s and Huntington’s Diseases. Curr Med Chem 2012;19:6018-35.

Van de Roovaart HJ, Nguyen N, Veenstra TD. Huntington’s disease drug development: Aphase 3 pipeline analysis. Pharmaceuticals (Basel) 2023;16:1513.

D’Egidio F, Castelli V, Lombardozzi G, Ammannito F, Cimini A, D’Angelo M. Therapeutic advances in neural regeneration for Huntington’s disease. Neural Regen Res 2024;19:1991-7.

Valadão KM, Luizeti BO, Yamaguchi MU, Issy AC, Bernuci MP. Nanotechnology in improving the treatment of Huntington’s disease: A systematic review. Neurotox Res 2022;40:636-45.

Kim A, Lalonde K, Truesdell A, Gomes Welter P, Brocardo PS, Rosenstock TR, et al. New avenues for the treatment of Huntington’s disease. Int J Mol Sci 2021;22:8363.

Singh S, Hema, Sharma N, Sachdeva M, Behl T, Zahoor I, et al. Focusing the pivotal role of nanotechnology in Huntington’s disease: An insight into the recent advancements. Environ Sci Pollut Res Int 2022;29:73809-27.

Khan A, Özçelik CE, Begli O, Oguz O, Kesici MS, Kasırga TS, et al. Highly potent peptide therapeutics to prevent protein aggregation in Huntington’s disease. ACS Med Chem Lett 2023;14:1821-6.

Published

01-01-2025

How to Cite

NEHA A BARDE, PADMAJA S KORE, & BANDAWANE DD. (2025). A REVIEW ARTICLE ON TREATING HUNTINGTON’S DISEASE: CURRENT AND EMERGING THERAPEUTIC ASPECTS. Innovare Journal of Medical Sciences, 13(1), 6–13. https://doi.org/10.22159/ijms.2025v13i1.53191

Issue

Section

Review Article(s)

Similar Articles

1 2 3 4 5 > >> 

You may also start an advanced similarity search for this article.