Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Novel Therapeutics for the Treatment of Alzheimer’s and Parkinson’s Disease

Author(s): Kavita Singh, Dhananjay Yadav, Pallavi S. Chauhan, Meerambika Mishra and Jun-O Jin*

Volume 26, Issue 7, 2020

Page: [755 - 763] Pages: 9

DOI: 10.2174/1381612826666200107161051

Price: $65

conference banner
Abstract

Neurodegenerative diseases are a leading apprehension in underdeveloped and developed countries. The advancement of medical science is synonymous with an increase in life expectancy, which results in an aging population. Neurodegenerative diseases in the older population are on the rise globally. This review sums up the novel therapeutic approaches for Alzheimer’s and Parkinson's disease. Although most neurodegenerative diseases occur due to the accumulation of misfolded proteins, the mechanisms are not completely understood. Currently, only prophylactic therapies are available and none are preventive in nature. The World Health Organisation estimates that neurodegenerative diseases that cause cognitive and motor impairments will be the second most ubiquitous cause of death in the next decade, hence the need for novel therapeutic targets for these diseases is demanded. In Alzheimer’s, β-amyloid and tau proteinopathies are prevalent, whereas a prion-like increase of α- synuclein is the characteristic trait of Parkinson's disease.

Keywords: Neurodegenerative disease, Alzheimer’s, Parkinson’s, novel therapeutics, proteinopathies, prophylactic.

[1]
Polanco JC, Li C, Bodea LG, Martinez-Marmol R, Meunier FA, Götz J. Amyloid-β and tau complexity - towards improved biomarkers and targeted therapies. Nat Rev Neurol 2018; 14(1): 22-39.
[http://dx.doi.org/10.1038/nrneurol.2017.162] [PMID: 29242522]
[2]
Ballard C, Gauthier S, Corbett A, Brayne C, Aarsland D, Jones E. Alzheimer’s disease. Lancet 2011; 377(9770): 1019-31.
[http://dx.doi.org/10.1016/S0140-6736(10)61349-9] [PMID: 21371747]
[3]
Brookmeyer R, Johnson E, Ziegler-Graham K, Arrighi HM. Forecasting the global burden of Alzheimer’s disease. Alzheimers Dement 2007; 3(3): 186-91.
[http://dx.doi.org/10.1016/j.jalz.2007.04.381]] [PMID: 19595937]
[4]
Amemori T, Jendelova P, Ruzicka J, Urdzikova LM, Sykova E. Alzheimer’s disease: mechanism and approach to cell therapy. Int J Mol Sci 2015; 16(11): 26417-51.
[http://dx.doi.org/10.3390/ijms161125961] [PMID: 26556341]
[5]
Qin L, Wu X, Block ML, et al. Systemic LPS causes chronic neuroinflammation and progressive neurodegeneration. Glia 2007; 55(5): 453-62.
[http://dx.doi.org/10.1002/glia.20467] [PMID: 17203472]
[6]
Jack CR Jr, Wiste HJ, Weigand SD, et al. Age-specific and sex-specific prevalence of cerebral β-amyloidosis, tauopathy, and neurodegeneration in cognitively unimpaired individuals aged 50-95 years: a cross-sectional study. Lancet Neurol 2017; 16(6): 435-44.
[http://dx.doi.org/10.1016/S1474-4422(17)30077-7] [PMID: 28456479]
[7]
Querfurth HW, LaFerla FM. Alzheimer’s disease. N Engl J Med 2010; 362(4): 329-44.
[http://dx.doi.org/10.1056/NEJMra0909142] [PMID: 20107219]
[8]
Yao ZX, Han Z, Drieu K, Papadopoulos V. Ginkgo biloba extract (Egb 761) inhibits beta-amyloid production by lowering free cholesterol levels. J Nutr Biochem 2004; 15(12): 749-56.
[http://dx.doi.org/10.1016/j.jnutbio.2004.06.008] [PMID: 15607648]
[9]
Kanninen K, Heikkinen R, Malm T, et al. Intrahippocampal injection of a lentiviral vector expressing Nrf2 improves spatial learning in a mouse model of Alzheimer’s disease. Proc Natl Acad Sci USA 2009; 106(38): 16505-10.
[http://dx.doi.org/10.1073/pnas.0908397106] [PMID: 19805328]
[10]
Liu Z, Zhou T, Ziegler AC, Dimitrion P, Zuo L. Oxidative stress in neurodegenerative diseases: from molecular mechanisms to clinical applications. Oxid Med Cell Longev 2017; 2017 2525967
[http://dx.doi.org/10.1155/2017/2525967] [PMID: 28785371]
[11]
Mawuenyega KG, Sigurdson W, Ovod V, et al. Decreased clearance of CNS beta-amyloid in Alzheimer’s disease. Science 2010; 330(6012): 1774.
[http://dx.doi.org/10.1126/science.1197623] [PMID: 21148344]
[12]
Frieden C, Garai K. Structural differences between apoE3 and apoE4 may be useful in developing therapeutic agents for Alzheimer’s disease. Proc Natl Acad Sci USA 2012; 109(23): 8913-8.
[http://dx.doi.org/10.1073/pnas.1207022109] [PMID: 22615372]
[13]
Chen J, Li Q, Wang J. Topology of human apolipoprotein E3 uniquely regulates its diverse biological functions. Proc Natl Acad Sci USA 2011; 108(36): 14813-8.
[http://dx.doi.org/10.1073/pnas.1106420108] [PMID: 21873229]
[14]
Zhong N, Weisgraber KH. Understanding the association of apolipoprotein E4 with Alzheimer disease: clues from its structure. J Biol Chem 2009; 284(10): 6027-31.
[http://dx.doi.org/10.1074/jbc.R800009200] [PMID: 18948255]
[15]
Aïd S, Bosetti F. Targeting cyclooxygenases-1 and -2 in neuroinflammation: Therapeutic implications. Biochimie 2011; 93(1): 46-51.
[http://dx.doi.org/10.1016/j.biochi.2010.09.009] [PMID: 20868723]
[16]
Bhaskar K, Maphis N, Xu G, et al. Microglial derived tumor necrosis factor-α drives Alzheimer’s disease-related neuronal cell cycle events. Neurobiol Dis 2014; 62: 273-85.
[http://dx.doi.org/10.1016/j.nbd.2013.10.007] [PMID: 24141019]
[17]
Chen G, Goeddel DV. TNF-R1 signaling: a beautiful pathway. Science 2002; 296(5573): 1634-5.
[http://dx.doi.org/10.1126/science.1071924] [PMID: 12040173]
[18]
Decourt B, Lahiri DK, Sabbagh MN. Targeting tumor necrosis factor alpha for Alzheimer’s disease. Curr Alzheimer Res 2017; 14(4): 412-25.
[PMID: 27697064]
[19]
Beattie EC, Stellwagen D, Morishita W, et al. Control of synaptic strength by glial TNFalpha. Science 2002; 295(5563): 2282-5.
[http://dx.doi.org/10.1126/science.1067859] [PMID: 11910117]
[20]
Rowan M, Klyubin I, Wang Q, Hu N, Anwyl R. Synaptic memory mechanisms: Alzheimer’s disease amyloid beta peptide precursors. Biochem Soc Trans 35(Pt 5): 1219-23.
[21]
Wang Q, Wu J, Rowan MJ, Anwyl R. β-amyloid inhibition of long-term potentiation is mediated via tumor necrosis factor. Eur J Neurosci 2005; 22(11): 2827-32.
[http://dx.doi.org/10.1111/j.1460-9568.2005.04457.x] [PMID: 16324117]
[22]
Mullane K, Williams M. Alzheimer’s therapeutics: continued clinical failures question the validity of the amyloid hypothesis-but what lies beyond? Biochem Pharmacol 2013; 85(3): 289-305.
[http://dx.doi.org/10.1016/j.bcp.2012.11.014] [PMID: 23178653]
[23]
Atwal JK, Chen Y, Chiu C, et al. A therapeutic antibody targeting BACE1 inhibits amyloid-β production in vivo. Sci Transl Med 2011; 3(84) 84ra43
[http://dx.doi.org/10.1126/scitranslmed.3002254]
[24]
Kumar V, Jahan S, Singh S, Khanna VK, Pant AB. Progress toward the development of in vitro model system for chemical-induced developmental neurotoxicity: potential applicability of stem cells. Arch Toxicol 2015; 89(2): 265-7.
[http://dx.doi.org/10.1007/s00204-014-1442-0] [PMID: 25537189]
[25]
Reddy AP, Ravichandran J, Carkaci-Salli N. Neural regeneration therapies for Alzheimer’s and Parkinson’s disease-related disorders. Biochim Biophys Acta Mol Basis of Dis 2019; 1866(4) 165506
[http://dx.doi.org/10.1016/j.bbadis.2019.06.020]
[26]
Binan L, Ajji A, De Crescenzo G, Jolicoeur M. Approaches for neural tissue regeneration. Stem Cell Rev Rep 2014; 10(1): 44-59.
[http://dx.doi.org/10.1007/s12015-013-9474-z] [PMID: 24092527]
[27]
Choong CJ, Baba K, Mochizuki H. Gene therapy for neurological disorders. Expert Opin Biol Ther 2016; 16(2): 143-59.
[http://dx.doi.org/10.1517/14712598.2016.1114096] [PMID: 26642082]
[28]
Carty N, Nash KR, Brownlow M, et al. Intracranial injection of AAV expressing NEP but not IDE reduces amyloid pathology in APP+PS1 transgenic mice. PLoS One 2013; 8(3) e59626
[http://dx.doi.org/10.1371/journal.pone.0059626] [PMID: 23555730]
[29]
Dupont C, Armant DR, Brenner CA. Epigenetics: definition, mechanisms and clinical perspective Seminars in reproductive medicine. Thieme Medical Publishers 2009; pp. 351-7.
[30]
Rogers J, Mastroeni D, Grover A, Delvaux E, Whiteside C, Coleman PD. The epigenetics of Alzheimer’s disease-additional considerations. Neurobiol Aging 2011; 32(7): 1196-7.
[http://dx.doi.org/10.1016/j.neurobiolaging.2011.02.025] [PMID: 21497949]
[31]
Loera-Valencia R, Piras A, Ismail MAM, et al. Targeting Alzheimer’s disease with gene and cell therapies. J Intern Med 2018; 284(1): 2-36.
[http://dx.doi.org/10.1111/joim.12759] [PMID: 29582495]
[32]
Portelius E, Dean RA, Andreasson U, et al. β-site amyloid precursor protein-cleaving enzyme 1(BACE1) inhibitor treatment induces Aβ5-X peptides through alternative amyloid precursor protein cleavage. Alzheimers Res Ther 2014; 6(5-8): 75.
[http://dx.doi.org/10.1186/s13195-014-0075-0] [PMID: 25404952]
[33]
Guo Q, Zheng X, Yang P, et al. Small interfering RNA delivery to the neurons near the amyloid plaques for improved treatment of Alzheimer׳s disease. Acta Pharm Sin B 2019; 9(3): 590-603.
[http://dx.doi.org/10.1016/j.apsb.2018.12.010] [PMID: 31193846]
[34]
Mahley RW. Apolipoprotein E: cholesterol transport protein with expanding role in cell biology. Science 1988; 240(4852): 622-30.
[http://dx.doi.org/10.1126/science.3283935] [PMID: 3283935]
[35]
Lu J, Fu J, Zhong Y, et al. The roles of apolipoprotein E3 and CYP2D6 (rs1065852) gene polymorphisms in the predictability of responses to individualized therapy with donepezil in Han Chinese patients with Alzheimer’s disease. Neurosci Lett 2016; 614: 43-8.
[http://dx.doi.org/10.1016/j.neulet.2015.12.062] [PMID: 26768225]
[36]
Huang Y. Abeta-independent roles of apolipoprotein E4 in the pathogenesis of Alzheimer’s disease. Trends Mol Med 2010; 16(6): 287-94.
[http://dx.doi.org/10.1016/j.molmed.2010.04.004] [PMID: 20537952]
[37]
Aoki K, Uchihara T, Sanjo N, et al. Increased expression of neuronal apolipoprotein E in human brain with cerebral infarction. Stroke 2003; 34(4): 875-80.
[http://dx.doi.org/10.1161/01.STR.0000064320.73388.C6] [PMID: 12649507]
[38]
Zeng XS, Geng WS, Jia JJ, Chen L, Zhang PP. Cellular and molecular basis of neurodegeneration in Parkinson disease. Front Aging Neurosci 2018; 10: 109.
[http://dx.doi.org/10.3389/fnagi.2018.00109] [PMID: 29719505]
[39]
DeMaagd G, Philip A. Parkinson’s disease and its management: part 1: disease entity, risk factors, pathophysiology, clinical presentation, and diagnosis. P T 2015; 40(8): 504-32.
[40]
Tarakad A, Jankovic J. Essential tremor and Parkinson’s disease: exploring the relationship. Tremor Other Hyperkinet Mov (N Y) 2019; 8: 589.
[PMID: 30643667]
[41]
Outeiro TF, Koss DJ, Erskine D, et al. Dementia with Lewy bodies: an update and outlook. Mol Neurodegener 2019; 14(1): 5.
[http://dx.doi.org/10.1186/s13024-019-0306-8] [PMID: 30665447]
[42]
Canário N, Sousa M, Moreira F, et al. Impulsivity across reactive, proactive and cognitive domains in Parkinson’s disease on dopaminergic medication: Evidence for multiple domain impairment. PLoS One 2019; 14(2) e0210880
[http://dx.doi.org/10.1371/journal.pone.0210880] [PMID: 30759108]
[43]
Juárez OH, Calderón GD, Hernández GE, Barragán MG. The role of dopamine and its dysfunction as a consequence of oxidative stress. Oxid Med Cell Longev 2016; 2016 9730467
[44]
Oertel WH. Recent advances in treating Parkinson’s disease. F1000 Res 2017; 6: 260-0.
[http://dx.doi.org/10.12688/f1000research.10100.1] [PMID: 28357055]
[45]
Nagoshi E. Drosophila models of sporadic Parkinson’s disease. Int J Mol Sci 2018; 19(11): 3343.
[http://dx.doi.org/10.3390/ijms19113343] [PMID: 30373150]
[46]
Mishima T, Fujioka S, Fukae J, Yuasa-Kawada J, Tsuboi Y. Modeling Parkinson’s disease and atypical parkinsonian syndromes using induced pluripotent stem cells. Int J Mol Sci 2018; 19(12): 3870.
[http://dx.doi.org/10.3390/ijms19123870] [PMID: 30518093]
[47]
Lv C, Li Q, Liu X, et al. Determination of catecholamines and their metabolites in rat urine by ultra-performance liquid chromatography-tandem mass spectrometry for the study of identifying potential markers for Alzheimer’s disease. J Mass Spectrom 2015; 50(2): 354-63.
[http://dx.doi.org/10.1002/jms.3536] [PMID: 25800017]
[48]
Bridi JC, Hirth F. Mechanisms of α-synuclein induced synaptopathy in Parkinson’s disease. Front Neurosci 2018; 12: 80.
[http://dx.doi.org/10.3389/fnins.2018.00080] [PMID: 29515354]
[49]
Zhang G, Xia Y, Wan F, et al. New perspectives on roles of alpha-synuclein in Parkinson’s disease. Front Aging Neurosci 2018; 10: 370.
[http://dx.doi.org/10.3389/fnagi.2018.00370] [PMID: 30524265]
[50]
Brodie NI, Popov KI, Petrotchenko EV, Dokholyan NV, Borchers CH. Conformational ensemble of native α-synuclein in solution as determined by short-distance crosslinking constraint-guided discrete molecular dynamics simulations. PLOS Comput Biol 2019; 15(3) e1006859
[http://dx.doi.org/10.1371/journal.pcbi.1006859] [PMID: 30917118]
[51]
Ma L, Yang C, Zhang X, et al. C-terminal truncation exacerbates the aggregation and cytotoxicity of α-Synuclein: a vicious cycle in Parkinson’s disease. Biochim Biophys Acta Mol Basis Dis 2018; 1864(12): 3714-25.
[http://dx.doi.org/10.1016/j.bbadis.2018.10.003] [PMID: 30290273]
[52]
Singh SK, Dutta A, Modi G. α-Synuclein aggregation modulation: an emerging approach for the treatment of Parkinson’s disease. Future Med Chem 2017; 9(10): 1039-53.
[http://dx.doi.org/10.4155/fmc-2017-0016] [PMID: 28632413]
[53]
Javed H, Meeran MFN, Azimullah S, Adem A, Sadek B, Ojha SK. Plant extracts and phytochemicals targeting α-synuclein aggregation in parkinson’s disease models. Front Pharmacol 2018; 9: 1555.
[PMID: 30941047]
[54]
Park J-S, Davis RL, Sue CM. Mitochondrial dysfunction in Parkinson’s disease: new mechanistic insights and therapeutic perspectives. Curr Neurol Neurosci Rep 2018; 18(5): 21.
[http://dx.doi.org/10.1007/s11910-018-0829-3] [PMID: 29616350]
[55]
Kausar S, Wang F, Cui H. The role of mitochondria in reactive oxygen species generation and its implications for neurodegenerative diseases. Cells 2018; 7(12): 274.
[http://dx.doi.org/10.3390/cells7120274] [PMID: 30563029]
[56]
Lickteig B, Wimalasena VK, Wimalasena K. N-Methyl-4-phenylpyridinium scaffold-containing lipophilic compounds are potent complex i inhibitors and selective dopaminergic toxins. ACS Chem Neurosci 2019; 10(6): 2977-88.
[http://dx.doi.org/10.1021/acschemneuro.9b00184] [PMID: 30929447]
[57]
Salazar C, Ruiz-Hincapie P, Ruiz LM. The interplay among PINK1/PARKIN/Dj-1 network during mitochondrial quality control in cancer biology: Protein interaction analysis. Cells 2018; 7(10): 154.
[http://dx.doi.org/10.3390/cells7100154] [PMID: 30274236]
[58]
Franco-Iborra S, Cuadros T, Parent A, Romero-Gimenez J, Vila M, Perier C. Defective mitochondrial protein import contributes to complex I-induced mitochondrial dysfunction and neurodegeneration in Parkinson’s disease. Cell Death Dis 2018; 9(11): 1122.
[http://dx.doi.org/10.1038/s41419-018-1154-0] [PMID: 30405116]
[59]
Tekirdag K, Cuervo AM. Chaperone-mediated autophagy and endosomal microautophagy: joint by a chaperone. J Biol Chem 2018; 293(15): 5414-24.
[http://dx.doi.org/10.1074/jbc.R117.818237] [PMID: 29247007]
[60]
Chung CG, Lee H, Lee SB. Mechanisms of protein toxicity in neurodegenerative diseases. Cell Mol Life Sci 2018; 75(17): 3159-80.
[http://dx.doi.org/10.1007/s00018-018-2854-4] [PMID: 29947927]
[61]
Zondler L, Kostka M, Garidel P, et al. Proteasome impairment by α-synuclein. PLoS One 2017; 12(9) e0184040
[http://dx.doi.org/10.1371/journal.pone.0184040] [PMID: 28945746]
[62]
Terron A, Bal-Price A, Paini A, et al. EFSA WG EPI1 Members. An adverse outcome pathway for parkinsonian motor deficits associated with mitochondrial complex I inhibition. Arch Toxicol 2018; 92(1): 41-82.
[http://dx.doi.org/10.1007/s00204-017-2133-4] [PMID: 29209747]
[63]
Pitcairn C, Wani WY, Mazzulli JR. Dysregulation of the autophagic-lysosomal pathway in Gaucher and Parkinson’s disease. Neurobiol Dis 2019; 122: 72-82.
[http://dx.doi.org/10.1016/j.nbd.2018.03.008] [PMID: 29550539]
[64]
Karimi-Moghadam A, Charsouei S, Bell B, Jabalameli MR. Parkinson disease from Mendelian forms to genetic susceptibility: new molecular insights into the neurodegeneration process Cell Mol Neurobiol 2018; 38(6): 1153-78.
[http://dx.doi.org/10.1007/s10571-018-0587-4] [PMID: 29700661]
[65]
Li H, Ham A, Ma TC, et al. Mitochondrial dysfunction and mitophagy defect triggered by heterozygous GBA mutations. Autophagy 2019; 15(1): 113-30.
[http://dx.doi.org/10.1080/15548627.2018.1509818] [PMID: 30160596]
[66]
Hall S, Janelidze S, Surova Y, Widner H, Zetterberg H, Hansson O. Cerebrospinal fluid concentrations of inflammatory markers in Parkinson’s disease and atypical parkinsonian disorders. Sci Rep 2018; 8(1): 13276-6.
[http://dx.doi.org/10.1038/s41598-018-31517-z] [PMID: 30185816]
[67]
Tansey MG, Romero-Ramos M. Immune system responses in Parkinson’s disease: Early and dynamic. Eur J Neurosci 2019; 49(3): 364-83.
[PMID: 30474172]
[68]
Rane MA, Foster JG, Wood SK, Hebert PR, Hennekens CH. Benefits and risks of nonsteroidal anti-inflammatory drugs: methodologic limitations lead to clinical uncertainties. Ther Innov Regul Sci 2019; 53(4): 502-5.
[http://dx.doi.org/10.1177/2168479018794159] [PMID: 30176739]
[69]
Rizek P, Kumar N, Jog MS. An update on the diagnosis and treatment of Parkinson disease. CMAJ 2016; 188(16): 1157-65.
[http://dx.doi.org/10.1503/cmaj.151179] [PMID: 27221269]
[70]
Tibar H, El Bayad K, Bouhouche A, et al. Non-motor symptoms of parkinson’s disease and their impact on quality of life in a cohort of Moroccan patients. Front Neurol 2018; 9: 170-0.
[http://dx.doi.org/10.3389/fneur.2018.00170] [PMID: 29670566]
[71]
Weng M, Xie X, Liu C, Lim K-L, Zhang C-w, Li L. The sources of reactive oxygen species and its possible role in the pathogenesis of Parkinson’s disease. Parkinsons Dis 2018; 2018 9163040
[http://dx.doi.org/10.1155/2018/9163040]
[72]
Tambasco N, Romoli M, Calabresi P. Levodopa in Parkinson’s disease: current status and future developments. Curr Neuropharmacol 2018; 16(8): 1239-52.
[http://dx.doi.org/10.2174/1570159X15666170510143821] [PMID: 28494719]
[73]
Haddad F, Sawalha M, Khawaja Y, Najjar A, Karaman R. Dopamine and levodopa prodrugs for the treatment of Parkinson’s disease. Molecules 2017; 23(1): 40.
[http://dx.doi.org/10.3390/molecules23010040] [PMID: 29295587]
[74]
Nomoto M, Nagai M, Nishikawa N, et al. Pharmacokinetics and safety/efficacy of levodopa pro-drug ONO-2160/carbidopa for Parkinson’s disease. eNeurologicalSci 2018; 13: 8-13.
[http://dx.doi.org/10.1016/j.ensci.2018.09.003] [PMID: 30294682]
[75]
Emamzadeh FN, Surguchov A. Parkinson’s disease: biomarkers, treatment, and risk factors. Front Neurosci 2018; 12: 612.
[http://dx.doi.org/10.3389/fnins.2018.00612] [PMID: 30214392]
[76]
Grall-Bronnec M, Victorri-Vigneau C, Donnio Y, et al. Dopamine agonists and impulse control disorders: a complex association. Drug Saf 2018; 41(1): 19-75.
[http://dx.doi.org/10.1007/s40264-017-0590-6] [PMID: 28861870]
[77]
Dallé E, Mabandla MV. Early life stress, depression and Parkinson’s disease: a new approach. Mol Brain 2018; 11(1): 18.
[http://dx.doi.org/10.1186/s13041-018-0356-9] [PMID: 29551090]
[78]
Pagano G, Niccolini F, Politis M. The serotonergic system in Parkinson’s patients with dyskinesia: evidence from imaging studies. J Neural Transm (Vienna) 2018; 125(8): 1217-23.
[http://dx.doi.org/10.1007/s00702-017-1823-7] [PMID: 29264660]
[79]
Gainetdinov RR, Hoener MC, Berry MD. Trace amines and their receptors. Pharmacol Rev 2018; 70(3): 549-620.
[http://dx.doi.org/10.1124/pr.117.015305] [PMID: 29941461]
[80]
Cascella R, Perni M, Chen SW, et al. Probing the origin of the toxicity of oligomeric aggregates of α-synuclein with antibodies. ACS Chem Biol 2019; 14(6): 1352-62.
[http://dx.doi.org/10.1021/acschembio.9b00312] [PMID: 31050886]
[81]
Chiken S, Nambu A. Mechanism of deep brain stimulation: inhibition, excitation, or disruption? Neuroscientist 2016; 22(3): 313-22.
[http://dx.doi.org/10.1177/1073858415581986] [PMID: 25888630]
[82]
Gallay MN, Moser D, Federau C, Jeanmonod D. Anatomical and technical reappraisal of the pallidothalamic tractotomy with the incisionless transcranial MR-guided focused ultrasound. A technical note. Front Surg 2019; 6: 2.
[http://dx.doi.org/10.3389/fsurg.2019.00002] [PMID: 30733946]
[83]
Doshi BS, Arruda VR. Gene therapy for hemophilia: what does the future hold? Ther Adv Hematol 2018; 9(9): 273-93.
[http://dx.doi.org/10.1177/2040620718791933] [PMID: 30210756]
[84]
Garea-Rodríguez E, Eesmaa A, Lindholm P, et al. Comparative analysis of the effects of neurotrophic factors CDNF and GDNF in a nonhuman primate model of Parkinson’s disease. PLoS One 2016; 11(2) e0149776
[http://dx.doi.org/10.1371/journal.pone.0149776] [PMID: 26901822]
[85]
Seo E-J, Sugimoto Y, Greten HJ, Efferth T. Repurposing of bromocriptine for cancer therapy. Front Pharmacol 2018; 9: 1030-0.
[http://dx.doi.org/10.3389/fphar.2018.01030] [PMID: 30349477]
[86]
Sahney A, Sharma BC, Jindal A, et al. A double-blind randomized controlled trial to assess efficacy of bromocriptine in cirrhotic patients with hepatic parkinsonism. Liver Int 2019; 39(4): 684-93.
[http://dx.doi.org/10.1111/liv.14024] [PMID: 30554466]
[87]
Connolly BS, Lang AE. Pharmacological treatment of Parkinson disease: a review. JAMA 2014; 311(16): 1670-83.
[http://dx.doi.org/10.1001/jama.2014.3654] [PMID: 24756517]
[88]
Massano J, Bhatia KP. Clinical approach to Parkinson’s disease: features, diagnosis, and principles of management. Cold Spring Harb Perspect Med 2012; 2(6) a008870
[http://dx.doi.org/10.1101/cshperspect.a008870] [PMID: 22675666]
[89]
Pessoa RR, Moro A, Munhoz RP, Teive HAG, Lees AJ. Apomorphine in the treatment of Parkinson’s disease: a review. Arq Neuropsiquiatr 2018; 76(12): 840-8.
[http://dx.doi.org/10.1590/0004-282x20180140] [PMID: 30698208]
[90]
Sang Q, Liu X, Wang L, et al. CircSNCA downregulation by pramipexole treatment mediates cell apoptosis and autophagy in Parkinson’s disease by targeting miR-7. Aging (Albany NY) 2018; 10(6): 1281-93.
[http://dx.doi.org/10.18632/aging.101466] [PMID: 29953413]
[91]
Collo G, Cavalleri L, Bono F, et al. Ropinirole and pramipexole promote structural plasticity in human iPSC-derived dopaminergic neurons via BDNF and mTOR signaling. Neural Plast 2018; 2018 4196961
[http://dx.doi.org/10.1155/2018/4196961] [PMID: 29531524]
[92]
Muller T. Real world studies are essential for drug therapy in Parkinson’s disease. Neural Regen Res 2018; 13(9): 1544-5.
[http://dx.doi.org/10.4103/1673-5374.237118] [PMID: 30127111]
[93]
Bette S, Shpiner DS, Singer C, Moore H. Safinamide in the management of patients with Parkinson’s disease not stabilized on levodopa: a review of the current clinical evidence. Ther Clin Risk Manag 2018; 14: 1737-45.
[http://dx.doi.org/10.2147/TCRM.S139545]] [PMID: 30271159]
[94]
Murata M, Odawara T, Hasegawa K, et al. Adjunct zonisamide to levodopa for DLB parkinsonism: A randomized double-blind phase 2 study. Neurology 2018; 90(8): e664-72.
[http://dx.doi.org/10.1212/WNL.0000000000005010] [PMID: 29367449]
[95]
Ferreira JJ, Lees AJ, Poewe W, et al. Effectiveness of opicapone and switching from entacapone in fluctuating Parkinson disease. Neurology 2018; 90(21): e1849-57.
[http://dx.doi.org/10.1212/WNL.0000000000005557] [PMID: 29695590]
[96]
Zhang P-L, Wang Y-X, Chen Y, Zhang C-H, Li C-H. The efficacy of homemade tolcapone in the treatment of patients with Parkinsons disease. Exp Ther Med 2018; 15(1): 127-30.
[PMID: 29375679]
[97]
Fabbri M, Ferreira JJ, Lees A, et al. Opicapone for the treatment of Parkinson’s disease: a review of a new licensed medicine. Mov Disord 2018; 33(10): 1528-39.
[http://dx.doi.org/10.1002/mds.27475] [PMID: 30264443]
[98]
Müller T. Safinamide: an add-on treatment for managing Parkinson’s disease. Clin Pharmacol 2018; 10: 31-41.
[http://dx.doi.org/10.2147/CPAA.S137740] [PMID: 29670409]
[99]
Kim A, Kim YE, Yun JY, et al. Amantadine and the risk of dyskinesia in patients with early Parkinson’s disease: an open-label, pragmatic trial. J Mov Disord 2018; 11(2): 65-71.
[http://dx.doi.org/10.14802/jmd.18005] [PMID: 29860788]
[100]
Torti M, Vacca L, Stocchi F. Istradefylline for the treatment of Parkinson’s disease: is it a promising strategy? Expert Opin Pharmacother 2018; 19(16): 1821-8.
[http://dx.doi.org/10.1080/14656566.2018.1524876] [PMID: 30232916]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy