Hostname: page-component-8448b6f56d-c47g7 Total loading time: 0 Render date: 2024-04-18T01:53:42.160Z Has data issue: false hasContentIssue false

Placebo-controlled cross-over study of the monoaminergic stabiliser (−)-OSU6162 in mental fatigue following stroke or traumatic brain injury

Published online by Cambridge University Press:  24 June 2014

Birgitta Johansson*
Affiliation:
Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
Arvid Carlsson
Affiliation:
Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
Maria L. Carlsson
Affiliation:
Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
Magdalena Karlsson
Affiliation:
Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
Marie K.L. Nilsson
Affiliation:
Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
Elisabeth Nordquist-Brandt
Affiliation:
Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
Lars Rönnbäck
Affiliation:
Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
*
Birgitta Johansson, Department of Clinical Neuroscience and Rehabilitation, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Per Dubbsgatan 14, 1tr, SE 413 45 Gothenburg, Sweden. Tel: +46-31-3421000; Fax: +031-3422467; E-mail: birgitta.johansson@neuro.gu.se

Extract

Objective: Mental fatigue occurring after a stroke or traumatic brain injury (TBI) often results in difficulties returning to work and pursuing social activities. No effective treatment of this condition is available today. In this study, we have tested a novel pharmacological strategy using the monoaminergic stabiliser (−)-OSU6162.

Methods: (−)-OSU6162 was given orally for 4 weeks in doses increasing from 15 to 45 mg b.i.d. to 12 patients suffering from mental fatigue, following upon stroke (n=6) or TBI (n=6). (−)-OSU6162 was compared with placebo using a double-blind, randomised cross-over design. Patients included were well rehabilitated physically with no gross impairment in cognitive functions other than those related to the mental fatigue.

Results: (−)-OSU6162 caused a remarkable improvement in mental stamina, as evaluated by a self-assessment scale on mental fatigue. Statistical significance was reached on the primary endpoint (Mental Fatigue Scale). There was a trend towards improvement in the secondary endpoints processing speed and attention. Principal component analysis showed an overall positive treatment effect in 7 of 12 patients. Beneficial responses were seen already during the first few days of active drug treatment. Increasing dosage caused no further improvement. Adverse reactions consisted of short-lasting mild nausea and attenuated appetite. These side effects disappeared upon dose reduction.

Conclusion: The monoaminergic stabiliser (−)-OSU6162 offers promise as a candidate for treatment of mental fatigue after a stroke or TBI.

Type
Research Article
Copyright
Copyright © Cambridge University Press 2012

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Chaudhuri, A, Behan, PO. Fatigue in neurological disorders. Lancet 2004;363:978988.CrossRefGoogle ScholarPubMed
Deluca, J. Fatigue as a window to the brain. Cambridge, Massachusetts, London, England: A Bradford Book, The MIT Press, 2005.CrossRefGoogle Scholar
Rödholm, M, Starmark, J-E, Svensson, E, Von Essen, C. Asteno-emotional disorder after aneurysmal SAH: reliability, symptomatology and relation to outcome. Acta Neurol Scand 2001;103:379385.CrossRefGoogle ScholarPubMed
Rönnbäck, L, Hansson, E. On the potential role of glutamate transport in mental fatigue. J Neuroinflammation 2004;1:22.CrossRefGoogle ScholarPubMed
Lindqvist, G, Malmgren, H. Organic mental disorders as hypothetical pathogenetic processes. Acta Psychiatr Scand 1993;88(Suppl. 373):517.CrossRefGoogle Scholar
Staub, F, Bogousslavsky, J. Fatigue after stroke: a major but neglected issue. Cerebrovasc Dis 2001;12:7581.CrossRefGoogle Scholar
Belmont, A, Agar, N, Hugeron, C, Gallais, B, Azouvi, P. Fatigue and traumatic brain injury. Ann Readapt Med Phys 2006;49:283288.CrossRefGoogle ScholarPubMed
Kohl, AD, Wylie, GR, Genova, HM, Hillary, F, Deluca, J. The neural correlates of cognitive fatigue in traumatic brain injury using functional MRI. Brain Inj 2009;23:420432.CrossRefGoogle ScholarPubMed
Ziino, C, Ponsford, J. Vigilance and fatigue following traumatic brain injury. J Int Neuropsychol Soc 2006;12:100110.CrossRefGoogle ScholarPubMed
Ashman, TA, Cantor, JB, Gordon, WA et al. Objective measurement of fatigue following traumatic brain injury. J Head Trauma Rehabil 2008;23:3340.CrossRefGoogle ScholarPubMed
Johansson, B, Berglund, P, Rönnbäck, L. Mental fatigue and impaired information processing after mild and moderate traumatic brain injury. Brain Inj 2009;23(13-14):10271040.CrossRefGoogle ScholarPubMed
Johansson, B, Rönnbäck, L. Mental fatigue: a common long term consequence after a brain injury. In: Agrawal, A, ed. Brain injury - functional aspects, rehabilitation and prevention. Rijeka, Croatia: InTech, 2012: 316.Google Scholar
Brennan, AR, Arnsten, FT. Neuronal mechanisms underlying attention deficit hyperactivity disorder. Ann N Y Acad Sci 2008;1129:236245.CrossRefGoogle ScholarPubMed
Posner, IM, Rothbart, MK. Research on attention networks as a model for the integration of psychological science. Annu Rev Psychol 2007;58:123.CrossRefGoogle Scholar
Azmitia, EC. Serotonin neurons, neuroplasticity, and homeostasis of neural tissue. Neuropsychopharmacology 1999;21(Suppl. 2):33S45S.CrossRefGoogle ScholarPubMed
Rung, JP, Rung, E, Helgeson, L et al. Effects of (−)-OSU6162 and ACR16 on motor activity in rats, indicating a unique mechanism of dopaminergic stabilization. J Neural Transm 2008;115:899908.CrossRefGoogle ScholarPubMed
Lahti, R, Tamminga, CA, Carlsson, A. Stimulating and inhibitory effects of the dopamine stabilizer (−)-OSU6162 on dopamine D(2) receptor function in vitro. J Neural Transm 2007;114:11431146.CrossRefGoogle Scholar
Ekesbo, A, Andrén, PE, Gunne, LM, Tedroff, J. (−)-OSU 6162 inhibits levodopa-induced dyskinesias in a monkey model of Parkinson's disease. Neuroreport 1997;8:25672570.CrossRefGoogle Scholar
Tedroff, J, Ekesbo, A, Rydin, E, Sonesson, S, Waters, AN, Carlsson, A. Clinical effects of (-)-OSU6162 in very advanced Parkinson's disease. A study report to the Swedish Medical Products Agency, March 9, 2000.Google Scholar
Tedroff, J, Ekesbo, A, Sonesson, S, Waters, N, Carlsson, A. Long-lasting improvement following (−)-OSU6162 in patients with Huntington's disease. Neurology 1999;53:16051606.CrossRefGoogle ScholarPubMed
Gefvert, O, Lindström, LH, Dahlbäck, O et al. (−)-OSU6162 induces a rapid onset of antipsychotic effect after a single dose. A double-blind placebo-controlled pilot study. Scandinavian Society for Psychopharmacology 41st annual meeting, Copenhagen, Denmark. Nord J Psychiatry 2000;54:9394.Google Scholar
Lundberg, T, Tedroff, J, Waters, N et al. Safety of early clinical experience with (-)-OSU6162, a dopaminergic stabilizer with antipsychotic properties. In: SCNP 43rd Annual and 2nd Mediterranean Meeting, Juan-les-Pins, France. Nord J Psychiatry 2002;56:24.Google Scholar
Burstein, ES, Carlsson, ML, Owens, M et al. II. In vitro evidence that (−)-OSU6162 and (+)-OSU6162 produce their behavioral effects through 5-HT2A serotonin and D2 dopamine receptors. J Neural Transm 2011;118:15231533.CrossRefGoogle Scholar
Carlsson, ML, Burstein, ES, Kloberg, A et al. I. In vivo evidence for partial agonist effects of (−)-OSU6162 and (+)-OSU6162 on 5-HT2A serotonin receptors. J Neural Transm 2011;118:15111522.CrossRefGoogle ScholarPubMed
Rodríguez, CA, Azie, NE, Adams, G et al. Single oral dose safety, tolerability, and pharmacokinetics of PNU-96391 (=OSU6162) in healthy volunteers. J Clin Pharmacol 2004;44:276283.CrossRefGoogle Scholar
Johansson, B, Starmark, A, Berglund, P, Rödholm, M, Rönnbäck, L. A self-assessment questionnaire for mental fatigue and related symptoms after neurological disorders and injuries. Brain Inj 2010;24:212.CrossRefGoogle ScholarPubMed
Rönnbäck, L, Johansson, B. Long-lasting mental fatigue after traumatic brain injury or stroke – a new perspective. Saarbrucken: LAP Lambert Academic Publishing, 2012.Google Scholar
Svanborg, P, asberg, M. A new self-rating scale for depression and anxiety states based on the Comprehensive Psychopathological Rating Scale. Acta Psychiatr Scand 1994;89:2128.CrossRefGoogle ScholarPubMed
asberg, M, Montgomery, SA, Perris, C, Schalling, D, Sedvall, G. A comprehensive psychopathological rating scale. Acta Psychiatr Scand 1978;271(Suppl):527.CrossRefGoogle Scholar
Holbrook, M, Skilbeck, CE. An activity index for use with stroke patients. Age Aging 1983;12:166170.CrossRefGoogle ScholarPubMed
Wechsler, D. Wechsler Adult Intelligence Scale (WAIS-III). 3rd ednin Swedish Stockholm: Pearson Assessment, 2003.Google Scholar
Ellis, DC, Kaplan, E, Kramer, JH. Delis-Kaplan Executive Function System – D-KEFS. San Antonio, TX: The Psychological Corporation, 2001.Google Scholar
Reitan, RM, Wolfson, D. The Halstead-Reitan neuropsychological test battery. Theory and clinical interpretation. Tucson, AZ: Neuropsychology Press, 1985.Google Scholar
Madison, S. Läsdiagnos. Lund: Läs och skrivcentrum, 2003.Google Scholar
Johansson, B, Rönnbäck, L. Mental fatigue and cognitive impairment after an almost neurological recovered stroke. ISRN Psychiatry 2012. [E-pub ahead of print; DOI: 10.5402/2012/686425].CrossRefGoogle ScholarPubMed
Senn, S. Cross-over trials in clinical research. 2nd edn. Chichester: Wiley, 2002.CrossRefGoogle Scholar