Effects of Pridopidine on Functional Capacity in Early-Stage Participants from the PRIDE-HD Study

Andrew McGarry, Mika Leinonen, Karl Kieburtz, Michal Geva, C Warren Olanow, Michael Hayden, Andrew McGarry, Mika Leinonen, Karl Kieburtz, Michal Geva, C Warren Olanow, Michael Hayden

Abstract

Background: No pharmacological treatment has been demonstrated to provide a functional benefit for persons with Huntington's disease (HD). Pridopidine is a sigma-1-receptor agonist shown to have beneficial effects in preclinical models of HD.

Objective: To further explore the effect of pridopidine on Total Functional Capacity (TFC) in the recent double-blind, placebo-controlled PRIDE-HD study.

Methods: We performed post-hoc analyses to evaluate the effect of pridopidine on TFC at 26 and 52 weeks. Participants were stratified according to baseline TFC score and analyzed using repeated measures (MMRM) and multiple imputation assuming missing not-at-random (MNAR) and worst-case scenarios.

Results: The pridopidine 45 mg bid dosage demonstrated a beneficial effect on TFC for the entire population at week 52 of 0.87 (nominal p = 0.0032). The effect was more pronounced for early HD participants (HD1/HD2, TFC = 7-13), with a change from placebo of 1.16 (nominal p = 0.0003). This effect remained nominally significant using multiple imputation with missing not at random assumption as a sensitivity analysis. Responder analyses showed pridopidine 45 mg bid reduced the probability of TFC decline in early HD patients at Week 52 (nominal p = 0.02).

Conclusion: Pridopidine 45 mg bid results in a nominally significant reduction in TFC decline at 52 weeks compared to placebo, particularly in patients with early-stage HD.

Keywords: Huntington’s disease; clinical trial; pridopidine; total functional capacity.

Conflict of interest statement

AM and KK have previously received grant support from Teva. MG and MH are previous employees of Teva. AM, KK, CWO are consultants for Prilenia therapeutics.

Figures

Fig. 1
Fig. 1
TFC change from baseline vs. placebo for pridopidine 45 mg bid at Weeks 26 and 52 in All Participants (A,B) and Early HD patients (C,D). Mean±SEM; p-values are nominal and presented for descriptive purposes only. (E) Change in TFC plotted over time in early HD (TFC≥7); *p = 0.036; **p = 0.0003.
Fig. 2
Fig. 2
Mean TFC change from baseline vs. placebo at Week 52 for all participants and early HD cohorts (TFC 7–13): comparison of MMRM to MNAR. Magnitude and p-value for the change in TFC, 45 mg BID vs. placebo, at 52 weeks.

References

    1. McGarry A, Biglan K, Marshall F. Huntington’s disease. Rosenberg’s Molecular and Genetic Basis of Neurological and Psychiatric Disease London; Elsevier, 2015.
    1. Nguyen L, Lucke-Wold BP, Mookerjee SA, Cavendish JZ, Robson MJ, Scandinaro AL, et al. Role of sigma-1 receptors in neurodegenerative diseases. J Pharmacol Sci. 2015;127:17–29.
    1. Hayashi T, Su TP. Sigma-1 receptor chaperones at the ER-mitochondrion interface regulate Ca (2+) signaling and cell survival. Cell. 2007;131(3):596–610.
    1. Kourrich S, Su TP, Fujimoto M, Bonci A. The sigma-1 receptor: Roles in neuronal plasticity and disease. Trends Neurosci. 2012;35(12):762–71.
    1. Ponten H, Kullingsjö J, Lagerkvist S, Martin P, Pettersson F, Sonesson C, et al. In vivo pharmacology of the dopaminergic stabilizer pridopidine. Eur J Pharmacol. 2010;644:88–95.
    1. Sahlholm K, Århem P, Fuxe K, Marcellino D. The dopamine stabilizers ACR16 and (–) OSU6162 display nanomolar affinities at the sigma-1 receptor. Mol Psychiatry. 2013;18:12–14.
    1. Sahlholm K, Sijbesma JW, Maas B, Kwizera C, Marcellino D, Ramakrishnan NK, et al. Pridopidine selectively occupies sigma-1 rather than dopamine D2 receptors at behaviorally active doses. Psychopharmacology (Berl). 2015;232(18):3443–53.
    1. Ryskamp D, Wu J, Geva M, Kusko R, Grossman I, Hayden M, et al. The sigma-1 receptor mediates the beneficial effects of pridopidine in a mouse model of Huntington disease. Neurobiol Dis. 2016;97(Pt A):46–59.
    1. Geva M, Kusko R, Soares H, Fowler KD, Birnberg T, Barash S, et al. Pridopidine activates neuroprotective pathways impaired in Huntington disease. Hum Med Gen. 2016;25(18):3975–87.
    1. Eddings CR, Arbez N, Akimov S. Pridopidine protects neurons from mutant-huntingtin toxicity via the sigma-1 receptor. Neurobiol Dis. 2019;129:118–29.
    1. Wu J, Ryskamp DA, Liang X, Egorova P, Zakharova O, Hung G, et al. Enhanced store-operated calcium entry leads to striatal synaptic loss in a Huntington’s disease mouse model. J Neurosci. 2016;36:125–41.
    1. Squitieri F, Di Pardo A, Favellato M, Amico E, Maglione V, Frati L. Pridopidine, a dopamine stabilizer, improves motor performance and shows neuroprotective effects in Huntington disease R6/2 mouse model. J Cell Mol Med. 2015;19(11):2540–8.
    1. Huntington Study Group HART Investigators. A randomized, double-blind, placebo controlled trial of pridopidine in Huntington’s disease. Mov Disord. 2013;28(10):1407–15.
    1. de Yebenes JG, Landwehrmeyer B, Squitieri F, Reilmann R, Rosser A, Barker RA, et al. Pridopidine for the treatment of motor function in patients with Huntington’s Disease (MermaiHD): A phase 3, randomized, double-blind, placebo-controlled trial. Lancet Neurol. 2011;10(12):1049–57.
    1. Reilmann R, McGarry A, Grachev ID, Savola JM, Borowsky B, Eyal E, et al. Safety and efficacy of pridopidine in patients with Huntington’s disease (PRIDE HD): A phase 2, randomised, placebo-controlled, multicentre, dose-ranging study. Lancet Neurol. 2019;18(2):165–76.
    1. Huntington Study Group. The Unified Huntington’s Disease Rating Scale: Reliability and consistency. Mov Disord. 1996;11:136–42.
    1. Shoulson I, Kurlan R, Rubin AJ Assessment of functional capacity in neurodegenerative movement disorders: Huntington’s disease as a prototype. In: Munsat TL, editors. Quantification of neurological deficit. Boston: Butterworths; 1989. pp. 271-283.
    1. The Huntington Study Group. A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington’s Disease. Neurology. 2001;57:397–404.
    1. Landwehrmeyer GB, Dubois B, de Yébenes JG, Kremer B, Gaus W, Kraus PH, et al. Riluzole in Huntington’s disease: A 3-year, randomized controlled study. Ann Neurol. 2007;62(3):262–72.
    1. McGarry A, McDermott M, Kieburtz K, de Blieck EA, Beal F, Marder K, et al. A randomized, double-blind, placebo-controlled trial of coenzyme Q10 in Huntington disease. Neurology. 2017;88(2):152–9.
    1. Hersch SM, Schifitto G, Oakes D, Bredlau AL, Meyers CM, Nahin R, et al. The CREST-E study of creatine for Huntington disease: A randomized controlled trial. Neurology. 2017;89(6):594–601.
    1. Schobel SA, Palermo G, Auinger P, Long JD, Ma S, Khwaja OS, et al. Motor, cognitive, and functional declines contribute to a single progressive factor in early HD. Neurology. 2017;89(24):2495–502.
    1. Marder K, Zhao H, Myers RH, Cudkowicz M, Kayson E, Kieburtz K, et al. Rate of functional decline in Huntington’s disease. Neurology. 2000;54(2):452–8.
    1. Reilmann R, Olanow CW, McGarry A, Geva M, Leinonen M, Hayden M, et al. Novel PET data and analysis of early HD from PRIDE-HD. International Parkinson and Movement Disorder Society Congress, Nice, France, September 2-26, 2019.
    1. Lucas G, Rymar VV, Sadikot AF, Debonnel G. Further evidence for an antidepressant potential of the selective s1 agonist SAElectrophysiological, morphological and behavioural studies. Int J Neuropsychopharmacol. 2008;11:485–95.
    1. Maurice T, Hiramatsu M, Itoh J, Kameyama T, Hasegawa T, Nabeshima T. Behavioral evidence for a modulating role of sigma ligands in memory processes. I: Attenuation of dizocilpine (MK-801)-induced amnesia. Brain Res. 1994;647:44–56.
    1. Maurice T, Lockhart BP. Neuroprotective and anti-amnesic potentials of sigma (σ) receptor ligands. Prog Neuropsychopharmacol Biol Psychiatry. 1997;21(1):69–102.
    1. Maurice T, Urani A, Phan VL, Romieu P. The interaction between neuroactive steroids and the σ1 receptor function: Behavioral consequences and therapeutic opportunities. Brain Res Rev. 2001;37(1-3):116–32.
    1. Monnet FP, Maurice T. The sigma 1 protein as a target for the non-genomic effects of neuro (active) steroids: Molecular, physiological, and behavioral aspects. J Pharmacol Sci. 2006;100 (2):93–118.
    1. Matsuno K, Senda T, Matsunaga K, Mita S, Kaneto H. Similar ameliorating effects of benzomorphans and 5-HT 2 antagonists on drug-induced impairment of passive avoidance response in mice: Comparison with acetylcholinesterase inhibitors. Psychopharmacology (Berl). 1993;112(1):134–41.
    1. Matsuno K, Senda T, Matsunaga K, Mita S. Ameliorating effects of σ receptor ligands on the impairment of passive avoidance tasks in mice: Involvement in the central acetylcholinergic system. Eur J Pharmacol. 1994;261(1-2):43–51.
    1. Monnet FP, Debonnel G, Junien JL, De Montigny C. N-methyl-D- aspartate-induced neuronal activation is selectively modulated by sigma receptors. Eur J Pharmacol. 1990;179:441–5.
    1. Monnet FP, Blier P, Debonnel G, de Montigny C. Modulation by sigma ligands of N-methyl-D-aspartate-induced [3 H] noradrenaline release in the rat hippocampus: G-protein dependency. Naunyn Schmiedebergs Arch Pharmaco. 1992;346(1):32–9.
    1. Bermack JE, Debonnel G. The role of sigma receptors in depression. J Pharmacol Sci. 2005;97(3):317–36.
    1. Hayashi T, Maurice T, Su TP. Ca2+ signaling via ς1-receptors: Novel regulatory mechanism affecting intracellular Ca2+ concentration. J Pharmacol Exp Ther.. 2000;293(3):788–98.
    1. Hong W, Nuwayhid SJ, Werling LL. Modulation of bradykinin-induced calcium changes in SH-SY5Y cells by neurosteroids and sigma receptor ligands via a shared mechanism. Synapse. 2004;54(2):102–10.
    1. Urani A, Romieu P, Roman FJ, Yamada K, Noda Y, Kamei H, et al. Enhanced antidepressant efficacy of σ1 receptor agonists in rats after chronic intracerebroventricular infusion of β-amyloid-(1– 40) protein. Eur J Pharmacol. 2004;486(2):151–61.
    1. Smith-Dijak AI, Sepers MD, Raymond LA. Alterations in synaptic function and plasticity in Huntington disease. J Neurochem. 2019;150(4):346–65.
    1. Francardo V, Geva M, Bez F, Denis Q, Steiner L, Hayden MR, et al. Pridopidine induces functional neurorestoration via the sigma-1 receptor in a mouse model of Parkinson’s disease. Neurotherapeutics. 2019;16(2):465–79.
    1. Pande AC, Genève J, Scherrer B, Smith F, Leadbetter RA, de Meynard C. A placebo-controlled trial of igmesine in the treatment of major depression. Eur Neuropsychopharmacol. 1999;9(Suppl 5):S138.
    1. Volz HP, Stoll KD. Clinical trials with sigma ligands. Pharmacopsychiatry. 2004;37(S3):214–20.
    1. The Voice of the Patient: Huntington’s Disease.

Source: PubMed

3
Sottoscrivi