Full-text resources of PSJD and other databases are now available in the new Library of Science.
Visit https://bibliotekanauki.pl

PL EN


Preferences help
enabled [disable] Abstract
Number of results
2013 | 13 | 4 | 287–294

Article title

Farmakoterapia dysfunkcji poznawczych w chorobie Alzheimera: obecne strategie i terapie eksperymentalne

Authors

Content

Title variants

EN
Pharmacotherapy of cognitive dysfunctions in Alzheimer’s disease: current strategies and experimental therapies

Languages of publication

PL

Abstracts

PL
Obecnie dostępne metody farmakologicznego leczenia zaburzeń funkcji poznawczych w chorobie Alzheimera (Alzheimer’s disease, AD) opierają się na potencjalizacji przekaźnictwa cholinergicznego (inhibitory cholinesteraz) oraz modulowaniu przekaźnictwa glutaminianergicznego (memantyna). Leki te wywierają zauważalny efekt objawo­wy w zakresie nie tylko funkcji poznawczych, lecz także zachowania i codziennej aktywności. Ich wpływ na naturalny przebieg choroby nie został jak dotąd jednoznacznie potwierdzony i ze względu na mechanizm działania jest uwa­żany za mało prawdopodobny. Wraz z rozwojem badań genetycznych i molekularnych nad patogenezą AD do prób klinicznych wchodzą leki, które mają bezpośrednio oddziaływać na procesy odpowiedzialne za neurodege­nerację i prowadzące do neurotoksycznego działania oligomerów i odkładania się w mózgu złogów białkowych. Niestety, przeprowadzone dotąd badania kliniczne zawiodły oczekiwania – nie potwierdziły skuteczności klinicz­nej i/lub wskazały na problemy z bezpieczeństwem leczenia. Najczęściej wymienianymi przyczynami tych niepo­wodzeń mają być podawanie leków w zbyt późnym stadium choroby i/lub niewystarczająco selektywne strategie doboru pacjentów, skutkujące włączaniem do badań chorych cierpiących na inne niż AD przyczyny zaburzeń po­znawczych. Proponowaną odpowiedzią na niepowodzenia prób klinicznych ma być inkorporowanie biomarkerów procesu chorobowego do nowych badań.
EN
Currently available methods of pharmacological treatment of cognitive dysfunction related to Alzheimer’s dis­ease (AD) are based on augmentation of cholinergic neurotransmission (by inhibiting activity of cholinesterases) or modulation of glutamatergic transmission (by acting on NMDA receptor). Both classes of drug exhibit clinically significant (though modest) symptomatic improvement not only considering cognition but also behaviour and ac­tivities of daily living. Whether or not they modify natural course of the disease, is not clearly confirmed by result of rigorously planned clinical trials and is currently considered unlikely. With the development of genetic and molecu­lar studies on the pathogenesis of AD novel treatment targets emerged recently, importantly these based on amyloid cascade hypothesis and τ protein phosphorylation. Unfortunately, the so far undertaken clinical trials yielded dis­appointing results, either proving not be effective or showing unfavourable side effects profile. The most commonly proposed explanations of these failures include starting trials too late during the disease process and/or insufficiently specific strategies of patients’ selection resulting in inclusion of subjects suffering from other than AD cognitive dys­functions. Biomarkers of disease-specific process are currently leading answer to the experienced trials failures as they are starting to be routinely used in novel studies.

Discipline

Year

Volume

13

Issue

4

Pages

287–294

Physical description

Contributors

author
  • Zakład Psychologii Lekarskiej, Wydział Nauk o Zdrowiu, Uniwersytet Medyczny w Łodzi

References

  • 1. Craig L.A., Hong N.S., McDonald R.J.: Revisiting the cholin­ergic hypothesis in the development of Alzheimer’s disease. Neurosci. Biobehav. Rev. 2011; 35: 1397–1409.
  • 2. Francis P.T.: The interplay of neurotransmitters in Alzheim­er’s disease. CNS Spectr. 2005; 10 supl. 18: 6–9.
  • 3. Whitehouse P.J., Price D.L., Struble R.G. i wsp.: Alzheimer’s disease and senile dementia: loss of neurons in the basal fore­brain. Science 1982; 215: 1237–1239.
  • 4. Farlow M.R., Cummings J.L.: Effective pharmacologic manage­ment of Alzheimer’s disease. Am. J. Med. 2007; 120: 388–397.
  • 5. Li J., Wu H.M., Zhou R.L. i wsp.: Huperzine A for Alzheimer’s disease. Cochrane Database Syst. Rev. 2008; (2): CD005592.
  • 6. Bond M., Rogers G., Peters J. i wsp.: The effectiveness and cost-effectiveness of donepezil, galantamine, rivastigmine and memantine for the treatment of Alzheimer’s disease (review of Technology Appraisal No. 111): a systematic review and eco­nomic model. Health Technol. Assess. 2012; 16: 1–470.
  • 7. Hansen R.A., Gartlehner G., Webb A.P. i wsp.: Efficacy and safety of donepezil, galantamine, and rivastigmine for the treatment of Alzheimer’s disease: a systematic review and meta-analysis. Clin. Interv. Aging 2008; 3: 211–225.
  • 8. Kaduszkiewicz H., Zimmermann T., Beck-Bornholdt H.P., van der Bussche H.: Cholinesterase inhibitors for patients with Alzheimer’s disease: systematic review of randomised clinical trials. BMJ 2005; 331: 321–327.
  • 9. Hansen R.A., Gartlehner G., Lohr K.N., Kaufer D.I.: Functional outcomes of drug treatment in Alzheimer’s disease. Drugs Aging 2007; 24: 155–167.
  • 10. Campbell N., Ayub A., Boustani M.A. i wsp.: Impact of cholinesterase inhibitors on behavioral and psychological symptoms of Alzheimer’s disease: a meta-analysis. Clin. Interv. Aging 2008; 3: 719–728.
  • 11. Rodda J., Morgan S., Walker Z.: Are cholinesterase inhibitors effective in the management of the behavioral and psychological symptoms of dementia in Alzheimer’s disease? A systematic review of randomized, placebo-controlled trials of donepezil, rivastigmine and galantamine. Int. Psychogeriatr. 2009; 21: 813–824.
  • 12. Takeda A., Loveman E., Clegg A. i wsp.: A systematic review of the clinical effectiveness of donepezil, rivastigmine and galantamine on cognition, quality of life and adverse events in Alzheimer’s disease. Int. J. Geriatr. Psychiatry 2006; 21: 17–28.
  • 13. Lockhart I.A., Mitchell S.A., Kelly S.: Safety and tolerability of donepezil, rivastigmine and galantamine for patients with Alzheimer’s disease: systematic review of the ‘real-world’ evidence. Dement. Geriatr. Cogn. Disord. 2009; 28: 389–403.
  • 14. Massoud F., Desmarais J.E., Gauthier S.: Switching cholinesterase inhibitors in older adults with dementia. Int. Psychogeriatr. 2011; 23: 372–378.
  • 15. Sadowsky C., Perez J.A., Bouchard R.W. i wsp.: Switching from oral cholinesterase inhibitors to the rivastigmine transdermal patch. CNS Neurosci. Ther. 2010; 16: 51–60.
  • 16. Sobów T.: Farmakoterapia zaburzeń funkcji poznawczych w chorobie Alzheimera. W: Diagnostyka i leczenie otępień. Rekomendacje zespołu ekspertów Polskiego Towarzystwa Alzheimerowskiego. Medisfera, Otwock 2012.
  • 17. Cummings J., Jones R., Wilkinson D. i wsp.: Effect of donepezil on cognition in severe Alzheimer’s disease: a pooled data analysis. J. Alzheimers Dis. 2010; 21: 843–851.
  • 18. Farlow M.R., Salloway S., Tariot P. i wsp.: Effectiveness and tolerability of high-dose (23 mg/d) versus standard-dose (10 mg/d) donepezil in moderate to severe Alzheimer’s disease: a 24-week, randomized, double-blind study. Clin. Ther. 2010; 32: 1234–1251.
  • 19. Farlow M.R., Grossberg G.T., Sadowsky C.H. i wsp.: A 24-week, randomized, controlled trial of rivastigmine patch 13.3 mg/24 h versus 4.6 mg/24 h in severe Alzheimer’s dementia. CNS Neurosci. Ther. 2013; 19: 745–752.
  • 20. Sobow T.: Combination treatments in Alzheimer’s disease: risks and benefits. Expert Rev. Neurother. 2010; 10: 693–702.
  • 21. Gauthier S., Molinuevo J.L.: Benefits of combined cholinesterase inhibitor and memantine treatment in moderate-severe Alzheimer’s disease. Alzheimers Dement. 2013; 9: 326–331.
  • 22. Danysz W., Parsons C.G., Mobius H.J. i wsp.: Neuroprotective and symptomatological action of memantine relevant for Alzheimer’s disease – a unified glutamatergic hypothesis of mechanism of action. Neurotox. Res. 2000; 2: 85–97.
  • 23. Flirski M., Sobów T.: Memantyna – strategia glutaminergiczna w leczeniu choroby Alzheimera. Farmakoter. Psychiatr. Neurol. 2006; 3–4: 177–186.
  • 24. Herrmann N., Li A., Lanctôt K.: Memantine in dementia: review of the current evidence. Expert Opin. Pharmacother. 2011; 12: 787–800.
  • 25. Schneider L.S., Dagerman K.S., Higgins J.P., McShane R.: Lack of evidence for the efficacy of memantine in mild Alzheimer disease. Arch. Neurol. 2011; 68: 991–998.
  • 26. Muayqil T., Camicioli R.: Systematic review and meta-analysis of combination therapy with cholinesterase inhibitors and memantine in Alzheimer’s disease and other dementias. Dement. Geriatr. Cogn. Dis. Extra 2012; 2: 546–572.
  • 27. Farrimond L.E., Roberts E., McShane R.: Memantine and cholinesterase inhibitor combination therapy for Alzheimer’s disease: a systematic review. BMJ Open 2012; 2: e000917.
  • 28. Atri A., Shaughnessy L.W., Locascio J.J., Growdon J.H.: Long-term course and effectiveness of combination therapyin Alzheimer disease. Alzheimer Dis. Assoc. Disord. 2008; 22: 209–221.
  • 29. Lopez O.L., Becker J.T., Wahed A.S. i wsp.: Long-term effects of the concomitant use of memantine with cholinesterase inhibition in Alzheimer disease. J. Neurol. Neurosurg. Psychiatry 2009; 80: 600–607.
  • 30. Rountree S.D., Atri A., Lopez O.L., Doody R.S.: Effectiveness of antidementia drugs in delaying Alzheimer’s disease progression. Alheimers Dement. 2013; 9: 338–345.
  • 31. Riordan K.C., Hoffman Snyder C.R., Wellik K.E. i wsp.: Effectiveness of adding memantine to an Alzheimer dementia treatment regimen which already includes stable donepezil therapy: a critically appraised topic. Neurologist 2011; 17: 121–123.
  • 32. Sun Y., Lu C.J., Chien K.L. i wsp.: Efficacy of multivitamin supplementation containing vitamins B6 and B12 and folic acid as adjunctive treatment with cholinesterase inhibitor in Alzheimer’s disease: a 26-week, randomized, double-blind, placebo-controlled study in Taiwanese patients. Clin. Ther. 2007; 29: 2204–2214.
  • 33. Aisen P.S., Schneider L.S., Sano M. i wsp.; Alzheimer Disease Cooperative Study: High-dose B vitamin supplementation and cognitive decline in Alzheimer’s disease: a randomized, controlled trial. JAMA 2008; 300: 1774–1783.
  • 34. Kwok T., Lee J., Law C.B. i wsp.: A randomized placebo controlled trial of homocysteine lowering to reduce cognitive decline in older demented people. Clin. Nutr. 2011; 30: 297–302.
  • 35. Smith A.D., Smith S.M., de Jager C.A. i wsp.: Homocysteinelowering by B vitamins slows the rate of accelerated brain atrophy in mild cognitive impairment: a randomized controlled trial. PLoS One 2010; 5: e12244.
  • 36. de Jager C.A., Oulhaj A., Jacoby R. i wsp.: Cognitive and clinical outcomes of homocysteine-lowering B-vitamin treatment in mild cognitive impairment: a randomized controlled trial. Int. J. Geriatr. Psychiatry 2012; 27: 592–600.
  • 37. Sano M., Ernesto C., Thomas R.G. i wsp.: A controlled trial of selegiline, alpha-tocopherol, or both as treatment for Alzheimer’s disease. The Alzheimer’s Disease Cooperative Study. N. Engl. J. Med. 1997; 336: 1216–1222.
  • 38. Sobow T., Kloszewska I.: Donepezil plus vitamin E as a treatment in Alzheimer disease. Alzheimer Dis. Assoc. Disord. 2003; 17: 244.
  • 39. Lloret A., Badía M.C., Mora N.J. i wsp.: Vitamin E paradox in Alzheimer’s disease: it does not prevent loss of cognition and may even be detrimental. J. Alzheimers Dis. 2009; 17: 143–149.
  • 40. Arlt S., Müller-Thomsen T., Beisiegel U., Kontush A.: Effect of one-year vitamin C- and E-supplementation on cerebrospinal fluid oxidation parameters and clinical course in Alzheimer’s disease. Neurochem. Res. 2012; 37: 2706–2714.
  • 41. Galasko D.R., Peskind E., Clark C.M. i wsp.: Antioxidants for Alzheimer disease: a randomized clinical trial with cerebrospinal fluid biomarker measures. Arch. Neurol. 2012; 69: 836–841.
  • 42. Petersen R.C., Thomas R.G., Grundman M. i wsp.; Alzheimer’s Disease Cooperative Study Group: Vitamin E and donepezil for the treatment of mild cognitive impairment. N. Engl. J. Med. 2005; 352: 2379–2388.
  • 43. Freund-Levi Y., Eriksdotter-Jönhagen M., Cederholm T. i wsp.: Omega-3 fatty acid treatment in 174 patients with mild to moderate Alzheimer disease: OmegAD study: a randomized double-blind trial. Arch. Neurol. 2006; 63: 1402–1408.
  • 44. Quinn J.F., Raman R., Thomas R.G. i wsp.: Docosahexaenoic acid supplementation and cognitive decline in Alzheimer disease: a randomized trial. JAMA 2010; 304: 1903–1911.
  • 45. Chiu C.C., Su K.P., Cheng T.C. i wsp.: The effects of omega-3 fatty acids monotherapy in Alzheimer’s disease and mild cognitive impairment: a preliminary randomized double-blind placebo- controlled study. Prog. Neuropsychopharmacol. Biol. Psychiatry 2008; 32: 1538–1544.
  • 46. Lee L.K., Shahar S., Chin A.V., Yusoff N.A.: Docosahexaenoic acid-concentrated fish oil supplementation in subjects with mild cognitive impairment (MCI): a 12-month randomised,double-blind, placebo-controlled trial. Psychopharmacology (Berl.) 2013; 225: 605–612.
  • 47. Schneider L.S., DeKosky S.T., Farlow M.R. i wsp.: A randomized, double-blind, placebo-controlled trial of two doses of Ginkgo biloba extract in dementia of the Alzheimer’s type. Curr. Alzheimer Res. 2005; 2: 541–551.
  • 48. McCarney R., Fisher P., Iliffe S. i wsp.: Ginkgo biloba for mild to moderate dementia in a community setting: a pragmatic, randomised, parallel-group, double-blind, placebo-controlled trial. Int. J. Geriatr. Psychiatry 2008; 23: 1222–1230.
  • 49. Mazza M., Capuano A., Bria P., Mazza S.: Ginkgo biloba and donepezil: a comparison in the treatment of Alzheimer’s dementia in a randomized placebo-controlled double-blind study. Eur. J. Neurol. 2006; 13: 981–985.
  • 50. Le Bars P.L., Katz M.M., Berman N. i wsp.: A placebo-controlled, double-blind, randomized trial of an extract of Ginkgo biloba for dementia. North American EGb Study Group. JAMA 1997; 278: 1327–1332.
  • 51. Ihl R., Tribanek M., Bachinskaya N.; GOTADAY Study Group: Efficacy and tolerability of a once daily formulation of Ginkgo biloba extract EGb 761® in Alzheimer’s disease and vascular dementia: results from a randomised controlled trial. Pharmacopsychiatry 2012; 45: 41–46.
  • 52. DeKosky S.T., Williamson J.D., Fitzpatrick A.L. i wsp.; Ginkgo Evaluation of Memory (GEM) Study Investigators: Ginkgo biloba for prevention of dementia: a randomized controlled trial. JAMA 2008; 300: 2253–2262.
  • 53. Vellas B., Coley N., Ousset P.J. i wsp.: Long-term use of standardised Ginkgo biloba extract for the prevention of Alzheimer’s disease (GuidAge): a randomised placebo-controlled trial. Lancet Neurol. 2012; 11: 851–859.
  • 54. Baum L., Lam C.W., Cheung S.K. i wsp.: Six-month randomized, placebo-controlled, double-blind, pilot clinical trial of curcumin in patients with Alzheimer disease. J. Clin. Psychopharmacol. 2008; 28: 110–113.
  • 55. Ringman J.M., Frautschy S.A., Teng E. i wsp.: Oral curcumin for Alzheimer’s disease: tolerability and efficacy in a 24-week randomized, double blind, placebo-controlled study. Alzheimers Res. Ther. 2012; 4: 43.
  • 56. Bilikiewicz A., Gaus W.: Colostrinin (a naturally occurring, proline-rich, polypeptide mixture) in the treatment of Alzheimer’s disease. J. Alzheimers Dis. 2004; 6: 17–26.
  • 57. Leszek J., Inglot A.D., Janusz M. i wsp.: Colostrinin: a prolinerich polypeptide (PRP) complex isolated from ovine colostrum for treatment of Alzheimer’s disease. A double-blind, placebocontrolled study. Arch. Immunol. Ther. Exp. (Warsz.) 1999; 47: 377–385.
  • 58. Aisen P.S., Gauthier S., Ferris S.H. i wsp.: Tramiprosate in mild-to-moderate Alzheimer’s disease – a randomized, double- blind, placebo-controlled, multi-centre study (the Alphase Study). Arch. Med. Sci. 2011; 7: 102–111.
  • 59. Henderson S.T., Vogel J.L., Barr L.J. i wsp.: Study of the ketogenic agent AC-1202 in mild to moderate Alzheimer’s disease: a randomized, double-blind, placebo-controlled, multicenter trial. Nutr. Metab. (Lond.) 2009; 6: 31.
  • 60. Scheltens P., Twisk J.W., Blesa R. i wsp.: Efficacy of Souvenaid in mild Alzheimer’s disease: results from a randomized, controlled trial. J. Alzheimers Dis. 2012; 31: 225–236.
  • 61. Kamphuis P.J., Verhey F.R., Olde Rikkert M.G. i wsp.: Efficacy of a medical food on cognition in Alzheimer’s disease: results from secondary analyses of a randomized, controlled trial. J. Nutr. Health Aging 2011; 15: 720–724.
  • 62. Shah R.C., Kamphuis P.J., Leurgans S. i wsp.: The S-Connect study: results from a randomized, controlled trial of Souvenaid in mild-to-moderate Alzheimer’s disease. Alzheimers Res. Ther. 2013; 5: 59.
  • 63. Cummings J.L.: Food for thought: Souvenaid in mild Alzheimer’s disease. J. Alzheimers Dis. 2012; 31: 237–238.
  • 64. Chopra K., Misra S., Kuhad A.: Neurobiological aspects of Alzheimer’s disease. Expert Opin. Ther. Targets 2011; 15: 535–555.
  • 65. Swerdlow R.H.: Alzheimer’s disease pathologic cascades: who comes first, what drives what. Neurotox. Res. 2012; 22: 182–194.
  • 66. Green R.C., Schneider L.S., Amato D.A. i wsp.; Tarenflurbil Phase 3 Study Group: Effect of tarenflurbil on cognitive decline and activities of daily living in patients with mild Alzheimer disease: a randomized controlled trial. JAMA 2009; 302: 2557–2564.
  • 67. Siemers E.R., Quinn J.F., Kaye J. i wsp.: Effects of a γ-secretase inhibitor in a randomized study of patients with Alzheimer disease. Neurology 2006; 66: 602–604.
  • 68. Coric V., van Dyck C.H., Salloway S. i wsp.: Safety and tolerability of the γ-secretase inhibitor avagacestat in a phase 2 study of mild to moderate Alzheimer disease. Arch. Neurol. 2012; 69: 1430–1440.
  • 69. Delrieu J., Ousset P.J., Caillaud C., Vellas B.: ‘Clinical trials in Alzheimer’s disease’: immunotherapy approaches. J. Neurochem. 2012; 120 supl. 1: 186–193.
  • 70. Aisen P.S., Cummings J., Schneider L.S.: Symptomatic and nonamyloid/tau based pharmacologic treatment for Alzheimer disease. Cold Spring Harb. Perspect. Med. 2012; 2: a006395.

Document Type

article

Publication order reference

Identifiers

YADDA identifier

bwmeta1.element.psjd-e3bd22d2-90f6-4663-936d-f620a50d362d
JavaScript is turned off in your web browser. Turn it on to take full advantage of this site, then refresh the page.