An open-label multicenter study to assess the safety of dextromethorphan/quinidine in patients with pseudobulbar affect associated with a range of underlying neurological conditions

Gary L Pattee, James P Wymer, Catherine Lomen-Hoerth, Stanley H Appel, Andrea E Formella, Laura E Pope, Gary L Pattee, James P Wymer, Catherine Lomen-Hoerth, Stanley H Appel, Andrea E Formella, Laura E Pope

Abstract

Background: Pseudobulbar affect (PBA) is associated with neurological disorders or injury affecting the brain, and characterized by frequent, uncontrollable episodes of crying and/or laughing that are exaggerated or unrelated to the patient's emotional state. Clinical trials establishing dextromethorphan and quinidine (DM/Q) as PBA treatment were conducted in patients with amyotrophic lateral sclerosis (ALS) or multiple sclerosis (MS). This trial evaluated DM/Q safety in patients with PBA secondary to any neurological condition affecting the brain.

Objective: To evaluate the safety and tolerability of DM/Q during long-term administration to patients with PBA associated with multiple neurological conditions.

Methods: Fifty-two-week open-label study of DM/Q 30/30 mg twice daily. Safety measures included adverse events (AEs), laboratory tests, electrocardiograms (ECGs), vital signs, and physical examinations.

Clinical trial registration: #NCT00056524.

Results: A total of 553 PBA patients with >30 different neurological conditions enrolled; 296 (53.5%) completed. The most frequently reported treatment-related AEs (TRAEs) were nausea (11.8%), dizziness (10.5%), headache (9.9%), somnolence (7.2%), fatigue (7.1%), diarrhea (6.5%), and dry mouth (5.1%). TRAEs were mostly mild/moderate, generally transient, and consistent with previous controlled trials. Serious AEs (SAEs) were reported in 126 patients (22.8%), including 47 deaths, mostly due to ALS progression and respiratory failure. No SAEs were deemed related to DM/Q treatment by investigators. ECG results suggested no clinically meaningful effect of DM/Q on myocardial repolarization. Differences in AEs across neurological disease groups appeared consistent with the known morbidity of the primary neurological conditions. Study interpretation is limited by the small size of some disease groups, the lack of a specific efficacy measure and the use of a DM/Q dose higher than the eventually approved dose.

Conclusions: DM/Q was generally well tolerated over this 52 week trial in patients with PBA associated with a wide range of neurological conditions.

Keywords: Dextromethorphan/quinidine; Pseudobulbar affect; Safety; Tolerability.

Figures

Figure 1.
Figure 1.
Proportion of patients remaining in study, by study week.

References

    1. Colamonico J, Formella A, Bradley W. Pseudobulbar affect: burden of illness in the USA. Adv Ther. 2012;29:775–98.
    1. Wortzel HS, Oster TJ, Anderson CA, Arciniegas DB. Pathological laughing and crying: epidemiology, pathophysiology and treatment. CNS Drugs. 2008;22:531–45.
    1. Parvizi J, Arciniegas DB, Bernardini GL. Diagnosis and management of pathological laughter and crying. Mayo Clin Proc. 2006;81:1482–6.
    1. Work S, Colamonico JA, Bradley WG, Kaye RE. Pseudobulbar affect: an under-recognized and undertreated neurological disorder. Adv Ther. 2011;28:586–601.
    1. Brooks BR, Crumpacker D, Fellus J. PRISM: a novel research tool to assess the prevalence of pseudobulbar affect symptoms across neurological conditions. PLoS One. 2013;8:1–8.
    1. Lopez OL, Gonzalez MP, Becker JT. Symptoms of depression and psychosis in Alzheimer’s disease and frontotemporal dementia: exploration of underlying mechanisms. Neuropsychiatry Neuropsychol Behav Neurol. 1996;9:154–61.
    1. Feinstein A, Feinstein K, Gray T, O’Connor P. Prevalence and neurobehavioral correlates of pathological laughing and crying in multiple sclerosis. Arch Neurol. 1997;54:1116–21.
    1. Siddiqui MS, Fernandez HH, Garvan CW. Inappropriate crying and laughing in Parkinson disease and movement disorders. World J Biol Psychiatry. 2009;10:234–40.
    1. House A, Dennis M, Molyneux A. Emotionalism after stroke. BMJ. 1989;298:991–4.
    1. Tateno A, Jorge RE, Robinson RG. Pathological laughing and crying following traumatic brain injury. J Neuropsychiatry Clin Neurosci. 2004;16:426–34.
    1. Gallagher JP. Pathologic laughter and crying in ALS: a search for their origin. Acta Neurol Scand. 1989;80:114–17.
    1. Parvizi J, Anderson SW, Martin CO. Pathological laughter and crying: a link to the cerebellum. Brain. 2001;124:1708–19.
    1. Parvizi J, Joseph J, Press DZ, Schmahmann JD. Pathological laughing and crying in patients with multiple system atrophy-cerebellar type. Mov Disord. 2007;22:798–803.
    1. Parvizi J, Coburn KL, Shillcutt SD. Neuroanatomy of pathological laughing and crying: a report of the American Neuropsychiatric Association Committee on Research. J Neuropsychiatry Clin Neurosci. 2009;21:75–87.
    1. Nuedexta (dextromethorphan hydrobromide and quinidine sulfate) capsules. Full prescribing information. Available at: [Last accessed 14 March 2014]
    1. European Medicines Agency. Nuedexta. Summary of product characteristics. Available at: [Last accessed 14 March 2014]
    1. Choi DW, Peters S, Viseskul V. Dextrorphan and levorphanol selectively block N-Methyl-D-aspartate receptor-mediated neurotoxicity on cortical neurons. J Pharmacol Exp Ther. 1987;242:713–20.
    1. Musacchio JM, Klein M, Canoll PD. Dextromethorphan and sigma ligands: common sites but diverse effects. Life Sci. 1989;45:1721–32.
    1. Werling LL, Keller A, Frank JG, Nuwayhid SJ. A comparison of the binding profiles of dextromethorphan, memantine, fluoxetine and amitriptyline: treatment of involuntary emotional expression disorder. Exp Neurol. 2007;207:248–57.
    1. Zhang Y, Britto MR, Valderhaug KL. Dextromethorphan: enhancing its systemic availability by way of low-dose quinidine-mediated inhibition of cytochrome P4502D6. Clin Pharmacol Ther. 1992;51:647–55.
    1. Pope LE, Khalil MH, Berg JE. Pharmacokinetics of dextromethorphan after single or multiple dosing in combination with quinidine in extensive and poor metabolizers. J Clin Pharmacol. 2004;44:1132–42.
    1. Brooks BR, Thisted RA, Appel SH, AVP-923 ALS Study Group Treatment of pseudobulbar affect in ALS with dextromethorphan/quinidine: a randomized trial. Neurology. 2004;63:1364–70.
    1. Panitch HS, Thisted RA, Smith RA, Pseudobulbar Affect in Multiple Sclerosis Study Group Randomized, controlled trial of dextromethorphan/quinidine for pseudobulbar affect in multiple sclerosis. Ann Neurol. 2006;59:780–7.
    1. Pioro EP, Brooks BR, Cummings J, Safety, Tolerability, and Efficacy Results Trial of AVP-923 in PBA Investigators Dextromethorphan plus ultra low-dose quinidine reduces pseudobulbar affect. Ann Neurol. 2010;68:693–702.
    1. Gil J, Funalot B, Verschueren A. Causes of death amongst French patients with amyotrophic lateral sclerosis: a prospective study. Eur J Neurol. 2008;15:1245–51.
    1. Spataro R, Lo Re M, Piccoli T. Causes and place of death in Italian patients with amyotrophic lateral sclerosis. Acta Neurol Scand. 2010;122:217–23.
    1. Bensimon G, Lacomblez L, Meininger V. A controlled trial of riluzole in amyotrophic lateral sclerosis. ALS/Riluzole Study Group. N Engl J Med. 1994;330:585–91.
    1. Louwerse ES, Weverling GJ, Bossuyt PM. Randomized, double-blind, controlled trial of acetylcysteine in amyotrophic lateral sclerosis. Arch Neurol. 1995;52:559–64.
    1. Rooney J, Byrne S, Heverin M. Survival analysis of Irish amyotrophic lateral sclerosis patients diagnosed from 1995–2010. PLoS One. 2013;8:e74733.
    1. Zoccolella S, Beghi E, Palagano G, SLAP Registry Analysis of survival and prognostic factors in amyotrophic lateral sclerosis: a population based study. J Neurol Neurosurg Psychiatry. 2008;79:33–7.
    1. Murphy M, Quinn S, Young J. Increasing incidence of ALS in Canterbury, New Zealand: a 22-year study. Neurology. 2008;71:1889–95.
    1. del Aquila MA, Longstreth WT, Jr, McGuire V. Prognosis in amyotrophic lateral sclerosis: a population-based study. Neurology. 2003;60:813–19.
    1. Chiò A, Mora G, Leone M, Piemonte and Valle d’Aosta Register for ALS (PARALS) Early symptom progression rate is related to ALS outcome: a prospective population-based study. Neurology. 2002;59:99–103.
    1. Logroscino G, Traynor BJ, Hardiman O, EURALS Descriptive epidemiology of amyotrophic lateral sclerosis: new evidence and unsolved issues. J Neurol Neurosurg Psychiatry. 2008;79:6–11.
    1. Millul A, Beghi E, Logroscino G. Survival of patients with amyotrophic lateral sclerosis in a population-based registry. Neuroepidemiology. 2005;25:114–19.
    1. Quinidine Sulfate Tablets USP Revised: June 2005 Rx only. Available at: [Last accessed 14 March 2014]
    1. Holford NH, Coates PE, Guentert TW. The effect of quinidine and its metabolites on the electrocardiogram and systolic time intervals: concentration–effect relationships. Br J Clin Pharmacol. 1981;11:187–95.
    1. Coplen SE, Antman EM, Berlin JA. Efficacy and safety of quinidine therapy for maintenance of sinus rhythm after cardioversion. A meta-analysis of randomized control trials. Circulation. 1990;82:1106–16.
    1. Morganroth J, Goin JE. Quinidine-related mortality in the short-to-medium-term treatment of ventricular arrhythmias. A meta-analysis. Circulation. 1991;84:1977–83.
    1. Grace AA, Camm AJ. Quinidine. N Engl J Med. 1998;338:35–45.

Source: PubMed

3
Prenumerera