touchNEUROLOGY touchNEUROLOGY
Alzheimer's Disease & Dementia
Read Time: 2 mins

Combination Drug Therapy for the Treatment of Alzheimer’s Disease

Copy Link
Published Online: May 15th 2012 European Neurological Review, 2012;7(2):92-102 DOI: http://doi.org/10.17925/ENR.2012.07.02.92
Authors: X Antón Álvarez, Carlos Linares, Eliezer Masliah
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

Alzheimer’s disease (AD) is a complex and progressive neurodegenerative disorder resulting in continuous deterioration of cognition, daily living abilities and motor functions and consequently has a huge social and familial burden. To date, the drugs approved for AD treatment provide only modest symptomatic effects. At present, the combined therapy with memantine plus one cholinesterase inhibitor (ChEI) is the best option for the treatment of moderate-to-severe AD. This combination has demonstrated higher clinical efficacy than monotherapy with ChEIs, with similar safety and tolerability in several randomised-controlled clinical trials (RCTs). Recent long-term observational studies have shown that combination therapy slows the rate of cognitive and functional deterioration, delays the placement of patients in nursing homes and also provides evidence that it is more effective when initiated early. None of the drugs for AD tested in Phase III trials show evidence of disease modification. A few studies have shown that the newer drugs, particularly anti-amyloid and neurotrophic agents, may provide improved disease-modifying treatments of AD in the near future. Meanwhile, combination therapy with available drugs is the most effective AD treatment.

Keywords

Alzheimer’s disease, combination therapy, memantine, disease-modifying treatments, cholinesterase inhibitor

Article:

Alzheimer’s Disease – A Complex Neurodegenerative Disorder with a Multifactorial Pathogenesis
Alzheimer’s disease (AD), the main cause of dementia, is a neurodegenerative disorder characterised by a progressive decline of cognitive functions (memory, language, praxis, judgement and thinking, orientation and executive functions), increasing disabilities in daily living function and the presence of behavioural and psychological symptoms.1–3 In the late to end-stages of AD, motor functions also deteriorate and thus AD causes a continuous loss of mental and physical autonomy in patients and produces a parallel increase in dependency and burden on carer-givers as the disease progresses.

The pathogenic process of AD is complex and may start decades before the condition is diagnosed (see Figure 1). Early molecular alterations give rise to structural changes, which precede the onset of the first symptoms and the later development of the full clinical picture, usually required for diagnosis and treatment. AD aetiopathogenesis is multifactorial and may include genetic mutations and/or risk factors,4,5 abnormal processing and deposition of beta-amyloid (Abeta) and tau proteins,6,7 inflammation mechanisms,8–10 deficits of neurotrophic factors,8,11,12 metabolic dysfunctions,13–15 oxidative stress,15,16 excitotoxicity17,18 and alterations in neurotransmitters such as acetylcholine, noradrenaline, serotonine and glutamate.18–20 These pathogenic factors influence the development of the typical AD neuropathology (senile plaques, neurofibrillary tangles, synaptic loss and neuronal apoptosis-degeneration) underlying cognitive impairment and other dementia symptoms (see Figure 1).21,22 Progressive impairment of cognition reduces the abilities of AD patients to recall events, to deal with complex mental activities, to stay orientated, to plan and execute tasks and to communicate and interact with others.

The Need for Multimodal Intervention in Alzheimer’s Disease
Drugs currently approved for the treatment of AD, i.e. cholinesterase inhibitors (ChEIs) and memantine, are intended tocounteract the pathological consequences of neurotransmitter alterations associated with the disease. Donepezil, galantamine and rivastigmine (ChEIs) enhance cholinergic neurotransmission by inhibiting cholinesterase activity and constitute the first-line standard therapy for mild-to-moderate AD.23,24

To view the full article in PDF or eBook formats, please click on the icons above.

Article Information:
Disclosure

The authors have no conflicts of interest to declare.

Correspondence

X Antón Álvarez, Fundación Antidemencia Al Andalus, PO Box 7010, 15002-A Coruña, Spain. E: xantonal@yahoo.es

Support

The publication of this article was funded by Merz Pharmaceuticals GmbH. The views and opinions expressed are those of the authors and not necessarily those of Merz Pharmaceuticals GmbH.

Received

2012-04-04T00:00:00

References

  1. Ballard C, Gauthier S, Corbett A, et al., Alzheimer’s disease, Lancet, 2011;377:1019–31.
  2. Blennow K, de Leon MJ, Zetterberg H, Alzheimer’s disease, Lancet, 2006;368:387–403.
  3. Prince M, Jackson J, World Alzheimer Report 2009. Available at: www.alz.co.uk/research/files/WorldAlzheimerReport.pdf (accessed 16 May 2012).
  4. Bertram L, Tanzi RE, Thirty years of Alzheimer’s disease genetics: the implications of systematic meta-analyses, Nat Rev Neurosci, 2008;9:768–78.
  5. Lambert JC, Amouyel P, Genetics of Alzheimer’s disease: new evidences for an old hypothesis?, Curr Opin Genet Dev, 2011;21:295–301.
  6. Ittner LM, Gotz J, Amyloid-beta and tau—a toxic pas de deux in Alzheimer’s disease, Nat Rev Neurosci, 2011;12:65–72.
  7. Palop JJ, Mucke L, Amyloid-beta-induced neuronal dysfunction in Alzheimer’s disease: from synapses toward neural networks, Nat Neurosci, 2010;13:812–8.
  8. Alvarez A, Cacabelos R, Sanpedro C, et al., Serum TNF-alpha levels are increased and correlate negatively with free IGF-I in Alzheimer disease, Neurobiol Aging, 2007;28:533–6.
  9. McGeer EG, McGeer PL, Neuroinflammation in Alzheimer’s disease and mild cognitive impairment: a field in its infancy, J Alzheimers Dis, 2010;19:355–61.
  10. Wyss-Coray T, Inflammation in Alzheimer disease: driving force, bystander or beneficial response?, Nat Med, 2006;12:1005–15.
  11. Mufson EJ, Counts SE, Fahnestock M, et al., Cholinotrophic molecular substrates of mild cognitive impairment in the elderly, Curr Alzheimer Res, 2007;4:340–50.
  12. Schindowski K, Belarbi K, Buee L, Neurotrophic factors in Alzheimer’s disease: role of axonal transport, Genes Brain Behav, 2008;7(Suppl. 1):43–56.
  13. Kapogiannis D, Mattson MP, Disrupted energy metabolism and neuronal circuit dysfunction in cognitive impairment and Alzheimer’s disease, Lancet Neurol, 2011;10:187–98.
  14. Mosconi L, Berti V, Glodzik L, et al., Pre-clinical detection of Alzheimer’s disease using FDG-PET, with or without amyloid imaging, J Alzheimers Dis, 2010;20:843–54.
  15. Mosconi L, Pupi A, De Leon MJ, Brain glucose hypometabolism and oxidative stress in preclinical Alzheimer’s disease, Ann N Y Acad Sci, 2008;1147:180–95.
  16. Smith MA, Zhu X, Tabaton M, et al., Increased iron and free radical generation in preclinical Alzheimer disease and mild cognitive impairment, J Alzheimers Dis, 2010;19:363–72.
  17. Bezprozvanny I, Mattson MP, Neuronal calcium mishandling and the pathogenesis of Alzheimer’s disease, Trends Neurosci, 2008;31:454–63.
  18. Bordji K, Becerril-Ortega J, Buisson A, Synapses, NMDA receptor activity and neuronal Abeta production in Alzheimer’s disease, Rev Neurosci, 2011;22:285–94.
  19. Mufson EJ, Counts SE, Perez SE, et al., Cholinergic system during the progression of Alzheimer’s disease: therapeutic implications, Expert Rev Neurother, 2008;8:1703–18.
  20. Szot P, White SS, Greenup JL, et al., Compensatory changes in the noradrenergic nervous system in the locus ceruleus and hippocampus of postmortem subjects with Alzheimer’s disease and dementia with Lewy bodies, J Neurosci, 2006;26:467–78.
  21. Crews L, Masliah E, Molecular mechanisms of neurodegeneration in Alzheimer’s disease, Hum Mol Genet, 2010;19:R12–20.
  22. Nelson PT, Braak H, Markesbery WR, Neuropathology and cognitive impairment in Alzheimer disease: a complex but coherent relationship, J Neuropathol Exp Neurol, 2009;68:1–14.
  23. Burns A, O’Brien J, Auriacombe S, et al., Clinical practice with anti-dementia drugs: a consensus statement from British Association for Psychopharmacology, J Psychopharmacol, 2006;20:732–55.
  24. Farlow MR, Cummings JL, Effective pharmacologic management of Alzheimer’s disease, Am J Med, 2007;120:388–97.
  25. Herrmann N, Li A, Lanctot K, Memantine in dementia: a review of the current evidence, Expert Opin Pharmacother, 2011;12:787–800.
  26. Martinez-Coria H, Green KN, Billings LM, et al., Memantine improves cognition and reduces Alzheimer’s-like neuropathology in transgenic mice, Am J Pathol, 2010;176:870–80.
  27. Parsons CG, Stoffler A, Danysz W, Memantine: a NMDA receptor antagonist that improves memory by restoration of homeostasis in the glutamatergic system—too little activation is bad, too much is even worse, Neuropharmacology, 2007;53:699–723.
  28. Cummings JL, Treatment of Alzheimer’s disease: the role of symptomatic agents in an era of disease-modifying therapies, Rev Neurol Dis, 2007;4:57–62.
  29. Farlow MR, Miller ML, Pejovic V, Treatment options in Alzheimer’s disease: maximizing benefit, managing expectations, Dement Geriatr Cogn Disord, 2008;25:408–22.
  30. McShane R, Areosa Sastre A, Minakaran N, Memantine for dementia, Cochrane Database Syst Rev, 2006;CD003154.
  31. Raina P, Santaguida P, Ismaila A, et al., Effectiveness of cholinesterase inhibitors and memantine for treating dementia: evidence review for a clinical practice guideline, Ann Intern Med, 2008;148:379–97.
  32. Seow D, Gauthier S, Pharmacotherapy of Alzheimer disease, Can J Psychiatry, 2007;52:620–9.
  33. Cortes F, Nourhashemi F, Guerin O, et al., Prognosis of Alzheimer’s disease today: a two-year prospective study in 686 patients from the REAL-FR Study, Alzheimers Dement, 2008;4:22–9.
  34. Gillette-Guyonnet S, Andrieu S, Nourhashemi F, et al., Long-term progression of Alzheimer’s disease in patients under antidementia drugs, Alzheimers Dement, 2011;7:579–92.
  35. Salloway S, Mintzer J, Weiner MF, et al., Disease-modifying therapies in Alzheimer’s disease, Alzheimers Dement, 2008;4:65–79.
  36. Amit T, Avramovich-Tirosh Y, Youdim MB, et al., Targeting multiple Alzheimer’s disease etiologies with multimodal neuroprotective and neurorestorative iron chelators, FASEB J, 2008;22:1296–305.
  37. Chopra K, Misra S, Kuhad A, Current perspectives on pharmacotherapy of Alzheimer’s disease, Expert Opin Pharmacother, 2011;12:335–50.
  38. Frautschy SA, Cole GM, Why pleiotropic interventions are needed for Alzheimer’s disease, Mol Neurobiol, 2010;41:392–409.
  39. Chow VW, Savonenko AV, Melnikova T, et al., Modeling an anti-amyloid combination therapy for Alzheimer’s disease, Sci Transl Med, 2010;2:13ra1.
  40. Ihl R, Frolich L, Winblad B, et al., World Federation of Societies of Biological Psychiatry (WFSBP) guidelines for the biological treatment of Alzheimer’s disease and other dementias, World J Biol Psychiatry, 2011;12:2–32.
  41. Patel L, Grossberg GT, Combination therapy for Alzheimer’s disease, Drugs Aging, 2011;28:539–46.
  42. Schmitt B, Bernhardt T, Moeller HJ, et al., Combination therapy in Alzheimer’s disease: a review of current evidence, CNS Drugs, 2004;18:827–44.
  43. Deiana S, Harrington CR, Wischik CM, et al., Methylthioninium chloride reverses cognitive deficits induced by scopolamine: comparison with rivastigmine, Psychopharmacology (Berl), 2009;202:53–65.
  44. Atri A, Shaughnessy LW, Locascio JJ, et al., Long-term course and effectiveness of combination therapy in Alzheimer disease, Alzheimer Dis Assoc Disord, 2008;22:209–21.
  45. Choi SH, Park KW, Na DL, et al., Tolerability and efficacy of memantine add-on therapy to rivastigmine transdermal patches in mild to moderate Alzheimer’s disease: a multicenter, randomized, open-label, parallel-group study, Curr Med Res Opin, 2011;27:1375–83.
  46. Cummings JL, Schneider E, Tariot PN, et al., Behavioral effects of memantine in Alzheimer disease patients receiving donepezil treatment, Neurology, 2006;67:57–63.
  47. Dantoine T, Auriacombe S, Sarazin M, et al., Rivastigmine monotherapy and combination therapy with memantine in patients with moderately severe Alzheimer’s disease who failed to benefit from previous cholinesterase inhibitor treatment, Int J Clin Pract, 2006;60:110–8.
  48. Farlow MR, Alva G, Meng X, et al., A 25-week, open-label trial investigating rivastigmine transdermal patches with concomitant memantine in mild-to-moderate Alzheimer’s disease: a post hoc analysis, Curr Med Res Opin, 2010;26:263–9.
  49. Feldman HH, Schmitt FA, Olin JT, Activities of daily living in moderate-to-severe Alzheimer disease: an analysis of the treatment effects of memantine in patients receiving stable donepezil treatment, Alzheimer Dis Assoc Disord, 2006;20:263–8.
  50. Lopez OL, Becker JT, Wahed AS, et al., Long-term effects of the concomitant use of memantine with cholinesterase inhibition in Alzheimer disease, J Neurol Neurosurg Psychiatry, 2009;80:600–7.

Further Resources

Share this Article
Related Content In Alzheimer's Disease & Dementia
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72