Effects of Cerebrolysin® in Patients With Minimally Conscious State After Stroke: An Observational Retrospective Clinical Study

Jun Yup Kim, Hyun Jung Kim, Hyo Seon Choi, So Young Park, Deog Young Kim, Jun Yup Kim, Hyun Jung Kim, Hyo Seon Choi, So Young Park, Deog Young Kim

Abstract

Introduction: The neurotrophic drug Cerebrolysin is composed of low-molecular-weight peptides and amino acids and has been shown to have neuroprotective and neuroplastic properties. Cerebrolysin has been reported to promote the recovery of motor functions in central nervous system disorders; however, the effects on the consciousness improvements in post-stroke patients have not yet been studied extensively. Therefore, we aimed to examine the effectiveness of Cerebrolysin on improving the consciousness level of stroke patients with minimally conscious state (MCS). Materials and Methods: In this retrospective study we included ischemic and/or hemorrhagic stroke patients with MCS according to the Coma Recovery Scale-Revised (CRS-R), who were admitted to our hospital between 2014 and 2017. All patients received comprehensive rehabilitation therapy including physical and occupational therapy. We compared patients treated with Cerebrolysin against patients who did not receive Cerebrolysin. Patients were included in the verum group if they received 10 mL of Cerebrolysin IV for at least 20 days. CRS-R scores were assessed at admission and discharge. Results: Of 1,531 patients screened, 75 were included in the study (Cerebrolysin, n = 43; control, n = 32). Baseline characteristics were similar between groups. At discharge, ~2 months after onset of stroke, Cerebrolysin-treated patients improved significantly in the CRS-R (p = 0.010) after adjustment for confounders using linear mixed model (LMM), especially in the Oromotor (p = 0.003) and Arousal subscales (p = 0.038). No safety issues were observed. Conclusion: This retrospective study suggests that Cerebrolysin may improve the level of consciousness in stroke patients with MCS, which should be further investigated in a well-designed, double-blind, placebo-controlled, randomized trial.

Keywords: Cerebrolysin; clinical study; consciousness level; minimally conscious state; stroke.

Figures

Figure 1
Figure 1
Flow chart of inclusion and disposition of subjects.
Figure 2
Figure 2
Changes of JFK-CRS total scores in both groups from admission to discharge. *p < 0.05 comparing CRS-R total scores at discharge with scores at admission in each group by paired t-test, and comparing Cerebrolysin group vs. control group over time by linear mixed model (LMM).

References

    1. Institute for Metrics and Health Evaluation . Global Burden of Disease Result Tool. (2016).
    1. Organisation WH. The Top 10 Causes of Death. (2017).
    1. Feigin VL, Forouzanfar MH, Krishnamurthi R, Mensah GA, Connor M, Bennett DA, et al. . Global and regional burden of stroke during 1990–2010: findings from the Global Burden of Disease Study 2010. Lancet. (2014) 383:245–54. 10.1016/S0140-6736(13)61953-4
    1. Flynn RW, MacWalter RS, Doney AS. The cost of cerebral ischaemia. Neuropharmacology. (2008) 55:250–6. 10.1016/j.neuropharm.2008.05.031
    1. Li J, Wang D, Tao W, Dong W, Zhang J, Yang J, et al. . Early consciousness disorder in acute ischemic stroke: incidence, risk factors and outcome. BMC Neurol. (2016) 16:140. 10.1186/s12883-016-0666-4
    1. Balami JS, Chen RL, Grunwald IQ, Buchan AM. Neurological complications of acute ischaemic stroke. Lancet Neurol. (2011) 10:357–71. 10.1016/S1474-4422(10)70313-6
    1. Masliah E, Diez-Tejedor E. The pharmacology of neurotrophic treatment with Cerebrolysin: brain protection and repair to counteract pathologies of acute and chronic neurological disorders. Drugs Today (Barc). (2012) 48(Suppl. A):3–24. 10.1358/dot.2012.48(Suppl.A).1739716
    1. Ziganshina LE, Abakumova T, Vernay L. Cerebrolysin for acute ischaemic stroke. Cochrane Database Syst Rev. (2017) 4:CD007026. 10.1002/14651858.CD007026.pub5
    1. Ladurner G, Kalvach P, Moessler H, Cerebrolysin Study G . Neuroprotective treatment with cerebrolysin in patients with acute stroke: a randomised controlled trial. J Neural Transm (Vienna). (2005) 112:415–28. 10.1007/s00702-004-0248-2
    1. Gharagozli K, Harandi AA, Houshmand S, Akbari N, Muresanu DF, Vester J, et al. . Efficacy and safety of Cerebrolysin treatment in early recovery after acute ischemic stroke: a randomized, placebo-controlled, double-blinded, multicenter clinical trial. J Med Life. (2017) 10:153–60.
    1. Heiss WD, Brainin M, Bornstein NM, Tuomilehto J, Hong Z. Cerebrolysin acute stroke treatment in Asia I. Cerebrolysin in patients with acute ischemic stroke in Asia: results of a double-blind, placebo-controlled randomized trial. Stroke. (2012) 43:630–6. 10.1161/STROKEAHA.111.628537
    1. Bornstein NM, Guekht A, Vester J, Heiss WD, Gusev E, Homberg V, et al. . Safety and efficacy of Cerebrolysin in early post-stroke recovery: a meta-analysis of nine randomized clinical trials. Neurol Sci. (2018) 39:629–40. 10.1007/s10072-017-3214-0
    1. Giacino JT, Kalmar K, Whyte J. The JFK coma recovery scale-revised: measurement characteristics and diagnostic utility. Arch Phys Med Rehabil. (2004) 85:2020–9. 10.1016/j.apmr.2004.02.033
    1. Zhang C, Chopp M, Cui Y, Wang L, Zhang R, Zhang L, et al. . Cerebrolysin enhances neurogenesis in the ischemic brain and improves functional outcome after stroke. J Neurosci Res. (2010) 88:3275–81. 10.1002/jnr.22495
    1. Ruiz de Azua S, Matute C, Stertz L, Mosquera F, Palomino A, de la Rosa I, et al. . Plasma brain-derived neurotrophic factor levels, learning capacity and cognition in patients with first episode psychosis. BMC Psychiatry. (2013) 13:27. 10.1186/1471-244X-13-27
    1. Akai F, Hiruma S, Sato T, Iwamoto N, Fujimoto M, Ioku M, et al. . Neurotrophic factor-like effect of FPF1070 on septal cholinergic neurons after transections of fimbria-fornix in the rat brain. Histol Histopathol. (1992) 7:213–21.
    1. Wronski R, Kronawetter S, Hutter-Paier B, Crailsheim K, Windisch M. A brain derived peptide preparation reduces the translation dependent loss of a cytoskeletal protein in primary cultured chicken neurons. J Neural Transm Suppl. (2000) 59:263–72. 10.1007/978-3-7091-6781-6_28
    1. Bernat JL. Chronic disorders of consciousness. Lancet. (2006) 367:1181–92. 10.1016/S0140-6736(06)68508-5
    1. Clauss R. Disorders of consciousness and pharmaceuticals that act on oxygen based amino acid and monoamine neurotransmitter pathways of the brain. Curr Pharm Des. (2014) 20:4140–53. 10.2174/13816128113196660656
    1. Clauss RP. Neurotransmitters in coma, vegetative and minimally conscious states, pharmacological interventions. Med Hypotheses. (2010). 75:287–90. 10.1016/j.mehy.2010.03.005
    1. Thonnard M, Gosseries O, Demertzi A, Lugo Z, Vanhaudenhuyse A, Bruno MA, et al. . Effect of zolpidem in chronic disorders of consciousness: a prospective open-label study. Funct Neurol. (2013) 28:259–64. 10.11138/FNeur/2013.28.4.259
    1. Worzniak M, Fetters MD, Comfort M. Methylphenidate in the treatment of coma. J Fam Pract. (1997) 44:495–8.
    1. Whyte J, Katz D, Long D, DiPasquale MC, Polansky M, Kalmar K, et al. . Predictors of outcome in prolonged posttraumatic disorders of consciousness and assessment of medication effects: a multicenter study. Arch Phys Med Rehabil. (2005) 86:453–62. 10.1016/j.apmr.2004.05.016
    1. Peeters M, Page G, Maloteaux JM, Hermans E. Hypersensitivity of dopamine transmission in the rat striatum after treatment with the NMDA receptor antagonist amantadine. Brain Res. (2002) 949:32–41. 10.1016/S0006-8993(02)02961-X
    1. Schneider WN, Drew-Cates J, Wong TM, Dombovy ML. Cognitive and behavioural efficacy of amantadine in acute traumatic brain injury: an initial double-blind placebo-controlled study. Brain Inj. (1999) 13:863–72. 10.1080/026990599121061
    1. Meythaler JM, Brunner RC, Johnson A, Novack TA. Amantadine to improve neurorecovery in traumatic brain injury-associated diffuse axonal injury: a pilot double-blind randomized trial. J Head Trauma Rehabil. (2002) 17:300–13. 10.1097/00001199-200208000-00004
    1. Giacino JT, Whyte J, Bagiella E, Kalmar K, Childs N, Khademi A, et al. . Placebo-controlled trial of amantadine for severe traumatic brain injury. N Engl J Med. (2012) 366:819–26. 10.1056/NEJMoa1102609
    1. Sattin D, Leonardi M, Guido D, Consortium on Functioning and Disability in Patients With DOC . Effects on the diagnosis change and on the disability level for individuals with disorder of consciousness: which predictors? Int Clin Psychopharmacol. (2018) 33:163–71. 10.1097/YIC.0000000000000214
    1. Spillantini MG, Aloe L, Alleva E, De Simone R, Goedert M, Levi-Montalcini R. Nerve growth factor mRNA and protein increase in hypothalamus in a mouse model of aggression. Proc Natl Acad Sci USA. (1989) 86:8555–9. 10.1073/pnas.86.21.8555
    1. Aloe L, Bracci-Laudiero L, Bonini S, Manni L. The expanding role of nerve growth factor: from neurotrophic activity to immunologic diseases. Allergy. (1997) 52:883–94. 10.1111/j.1398-9995.1997.tb01247.x
    1. Scaccianoce S, Cigliana G, Nicolai R, Muscolo LA, Porcu A, Navarra D, et al. . Hypothalamic involvement in the activation of the pituitary–adrenocortical axis by nerve growth factor. Neuroendocrinology. (1993) 58:202–9. 10.1159/000126534
    1. Dreyfus CF. Effects of nerve growth factor on cholinergic brain neurons. Trends Pharmacol Sci. (1989) 10:145–9. 10.1016/0165-6147(89)90166-1

Source: PubMed

3
Iratkozz fel