359
Views
28
CrossRef citations to date
0
Altmetric
Review

Advances in understanding genomic markers and pharmacogenetics of Parkinson’s disease

, , &
Pages 433-448 | Received 10 Nov 2015, Accepted 22 Feb 2016, Published online: 09 Mar 2016

References

  • Alonso-Navarro H, Jiménez-Jiménez FJ, García-Martín E, et al. Genomic and pharmacogenomic biomarkers of Parkinson’s disease. Curr Drug Metab. 2014;15:129–181.
  • Agúndez JAG, García-Martín E, Alonso-Navarro H, et al. Anti-Parkinson’s disease drugs and pharmacogenetic considerations. Expert Opin Drug Metab Toxicol. 2013;9:859–874.
  • Othman AA, Syed SA, Newman AH, et al. Transport, metabolism, and in vivo population pharmacokinetics of the chloro benztropine analogs, a class of compounds extensively evaluated in animal models of drug abuse. J Pharmacol Exp Ther. 2007;320:344–353.
  • Zanger UM, Turpeinen M, Klein K, et al. Functional pharmacogenetics/genomics of human cytochromes P450 involved in drug biotransformation. Anal Bioanal Chem. 2008;392:1093–1108.
  • Maurer G, Schreier E, Delaborde S, et al. Fate and disposition of bromocriptine in animals and man. II: absorption, elimination and metabolism. Eur J Drug Metab Pharmacokinet. 1983;8:51–62.
  • Matsubayashi K, Matsumoto H, Fukui Y. Contribution of cytochrome P450 3A pathway to bromocriptine metabolism and effects of ferrous iron and hypoxia-re-oxygenation on its elimination in the perfused rat liver. J Pharm Pharmacol. 1997;49:551–557.
  • Nakatsuka A, Nagai M, Yabe H, et al. Effect of clarithromycin on the pharmacokinetics of cabergoline in healthy controls and in patients with Parkinson’s disease. J Pharmacol Sci. 2006;100:59–64.
  • Christensen J, Dupont E, ØStergaard K. Cabergoline plasma concentration is increased during concomitant treatment with itraconazole. Mov Disord. 2002;17:1360–1362.
  • Lautala P, Ethell BT, Taskinen J, et al. The specificity of glucuronidation of entacapone and tolcapone by recombinant human UDP-glucuronosyltransferases. Drug Metab Dispos. 2000;28:1385–1389.
  • Bialecka M, Kurzawski M, Klodowska-Duda G, et al. The association of functional catechol-O-methyltransferase haplotypes with risk of Parkinson’s disease, levodopa treatment response, and complications. Pharmacogenet Genomics. 2008;18:815–821.
  • Bychkov E, Ahmed MR, Dalby KN, et al. Dopamine depletion and subsequent treatment with L-DOPA, but not the long-lived dopamine agonist pergolide, enhances activity of the Akt pathway in the rat striatum. J Neurochem. 2007;102:699–711.
  • Wynalda MA, Wienkers LC. Assessment of potential interactions between dopamine receptor agonists and various human cytochrome P450 enzymes using a simple in vitro inhibition screen. Drug Metab Dispos. 1997;25:1211–1214.
  • Arbouw ME, Movig KL, Egberts TC, et al. Clinical and pharmacogenetic determinants for the discontinuation of non-ergoline dopamine agonists in Parkinson’s disease. Eur J Clin Pharmacol. 2009;65:1245–1251.
  • Liu YZ, Tang BS, Yan XX, et al. Association of the DRD2 and DRD3 polymorphisms with response to pramipexole in Parkinson’s disease patients. Eur J Clin Pharmacol. 2009;65:679–683.
  • PharmGkb. The pharmacogenomics knowledgebase. 2016 [cited 2016 Jan 31]. Available from: http://www.pharmgkb.org
  • DrugBank. Open data drug & drug target database. 2016 [cited 2016 Jan 31]. Available from: http://www.drugbank.ca/
  • Kaye CM, Nicholls B. Clinical pharmacokinetics of ropinirole. Clin Pharmacokinet. 2000;39:243–254.
  • Kamada T, Chow T, Hiroi T, et al. Metabolism of selegiline hydrochloride, a selective monoamine b-type inhibitor, in human liver microsomes. Drug Metab Pharmacokinet. 2002;17:199–206.
  • Benetton SA, Fang C, Yang YO, et al. P450 phenotyping of the metabolism of selegiline to desmethylselegiline and methamphetamine. Drug Metab Pharmacokinet. 2007;22:78–87.
  • Watanabe T, Sakuyama K, Sasaki T, et al. Functional characterization of 26 CYP2B6 allelic variants (CYP2B6.2-CYP2B6.28, except CYP2B6.22). Pharmacogenet Genomics. 2010;20:459–462.
  • Lu Y, Mo C, Zeng Z, et al. CYP2D6*4 allele polymorphism increases the risk of Parkinson’s disease: evidence from meta-analysis. PLoS One. 2013;8:e84413.
  • Wang T, Wang B. Association between Glutathione S-transferase M1/Glutathione S-transferase T1 polymorphisms and Parkinson’s disease: a meta-analysis. J Neurol Sci. 2014;338:65–70.
  • Wang D, Zhai JX, Zhang LM, et al. Null genotype of GSTT1 contributes to increased Parkinson’s disease risk in Caucasians: evidence from a meta-analysis. Mol Biol Rep. 2014;41:7423–7430.
  • Dai D, Wang Y, Wang L, et al. Polymorphisms of DRD2 and DRD3 genes and Parkinson’s disease: a meta-analysis. Biomed Rep. 2014;2:275–281.
  • Zhai D, Li S, Zhao Y, et al. SLC6A3 is a risk factor for Parkinson’s disease: a meta-analysis of sixteen years’ studies. Neurosci Lett. 2014;564:99–104.
  • Gao L, Gao H. Association between 5-HTTLPR polymorphism and Parkinson’s disease: a meta analysis. Mol Biol Rep. 2014;41:6071–6082.
  • Liu Y, Wang Z, Zhang B. The relationship between monoamine oxidase B (MAOB) A644G polymorphism and Parkinson disease risk: a meta-analysis. Ann Saudi Med. 2014;34:12–17.
  • Sun YX, Wang XH, Xu AH, et al. Functional polymorphisms of the MAO gene with Parkinson disease susceptibility: a meta-analysis. J Neurol Sci. 2014;345:97–105.
  • Zhang Y, Piao X, Wu J, et al. A meta-analysis on relationship of MAOB intron 13 polymorphisms, interactions with smoking/COMT H158L polymorphisms with the risk of PD. Int J Neurosci. 2016;126:400–407.
  • Jiménez-Jiménez FJ, Alonso-Navarro H, García-Martín E, et al. COMT gene and risk for Parkinson’s disease: a systematic review and meta-analysis. Pharmacogenet Genomics. 2014;24:331–339.
  • Wu YL, Ding XX, Sun YH, et al. Methylenetetrahydrofolate reductase (MTHFR) C677T/A1298C polymorphisms and susceptibility to Parkinson’s disease: a meta-analysis. J Neurol Sci. 2013;335:14–21.
  • Zhu ZG, Ai QL, Wang WM, et al. Meta-analysis supports association of a functional SNP (rs1801133) in the MTHFR gene with Parkinson’s disease. Gene. 2013;531:78–83.
  • Zhu Y, Zhu RX, He ZY, et al. Association of MTHFR C677T with total homocysteine plasma levels and susceptibility to Parkinson’s disease: a meta-analysis. Neurol Sci. 2015;36:945–951.
  • Dai D, Lin P, Wang Y, et al. Association of NQO1 and TNF polymorphisms with Parkinson’s disease: A meta-analysis of 15 genetic association studies. Biomed Rep. 2014;2:713–718.
  • Xia J, Xu H, Jiang H, et al. The association between the C282Y and H63D polymorphisms of HFE gene and the risk of Parkinson’s disease: a meta-analysis. Neurosci Lett. 2015;595:99–103.
  • Jin J, Wu P, Li W, et al. Interleukin-10-1082A/G and −592C/A polymorphisms with risk of Parkinson’s disease: a meta-analysis. Int J Neurosci. 2014;124:852–858.
  • Guo X, Tang P, Li X, et al. Association between two α-2-macroglobulin gene polymorphisms and Parkinson’s disease: a meta-analysis. Int J Neurosci. 2016;126:193–198.
  • Dai L, Wang D, Meng H, et al. Association between the BDNF G196A and C270T polymorphisms and Parkinson’s disease: a meta-analysis. Int J Neurosci. 2013;123:675–683.
  • Lee YH, Song GG. BDNF 196 G/A and 270 C/T polymorphisms and susceptibility to Parkinson’s disease: a meta-analysis. J Mot Behav. 2014;46:59–66.
  • Mariani S, Ventriglia M, Simonelli I, et al. Meta-analysis study on the role of bone-derived neurotrophic factor Val66Met polymorphism in Parkinson’s disease. Rejuvenation Res. 2015;18:40–47.
  • Zhu R, Zhu Y, Liu X, et al. Fibroblast growth factor 20 (FGF20) gene polymorphism and risk of Parkinson’s disease: a meta-analysis. Neurol Sci. 2014;35:1889–1894.
  • Lu SS, Gong FF, Feng F, et al. Association of microtubule associated protein tau/Saitohin (MAPT/STH) MAPT_238bp/STH Q7R polymorphisms and Parkinson’s disease: A meta-analysis. Biochem Biophys Res Commun. 2014;453:653–661.
  • Gao Z, Fu HJ, Xue JJ, et al. Genetic polymorphisms in VDR, ESR1 and ESR2 genes may contribute to susceptibility to Parkinson’s disease: a meta-analysis. Mol Biol Rep. 2014;41:4463–4474.
  • Jiménez-Jiménez FJ, García-Martín E, Alonso-Navarro H, et al. PITX3 and risk for Parkinson’s disease: a systematic review and meta-analysis. Eur Neurol. 2014;71:49–56.
  • Qiu G, Fu C, Liang GH. Association between PITX3 promoter polymorphism and risk of Parkinson’s disease: the impact of ethnicity and onset age. Neurosci Lett. 2014;561:128–133.
  • Zhang ZT, He YC, Ma XJ, et al. Association between vitamin D receptor gene polymorphisms and susceptibility to Parkinson’s disease: a meta-analysis. Neurosci Lett. 2014;578:122–127.
  • Lee YH, Kim JH, Song GG. Vitamin D receptor polymorphisms and susceptibility to Parkinson’s disease and Alzheimer’s disease: a meta-analysis. Neurol Sci. 2014;35:1947–1953.
  • Li C, Qi H, Wei S, et al. Vitamin D receptor gene polymorphisms and the risk of Parkinson’s disease. Neurol Sci. 2015;36:247–255.
  • Niu MY, Wang L, Xie AM. ApaI, BsmI, FokI, and TaqI polymorphisms in the vitamin D receptor gene and Parkinson’s disease. Chin Med J (Engl). 2015;128:1809–1814.
  • Su G, Dou H, Zhao L, et al. The angiotensin-converting enzyme (ACE) I/D polymorphism in Parkinson’s disease. J Renin Angiotensin Aldosterone Syst. 2015;16:428–433.
  • Song GG, Lee YH. The insertion/deletion polymorphism in the angiotensin-converting enzyme and susceptibility to schizophrenia or Parkinson’s disease: a meta-analysis. J Renin Angiotensin Aldosterone Syst. 2015;16:434–442.
  • Yu X, Wang F, Zhang JP. Meta analysis of the association of rs7702187 SNP in SEMA5A gene with risk of Parkinson’s disease. Eur Rev Med Pharmacol Sci. 2014;18:900–904.
  • Chen Y, Li S, Su L, et al. Association of progranulin polymorphism rs5848 with neurodegenerative diseases: a meta-analysis. J Neurol. 2015;262:814–822.
  • Ma ZG, Liu TW, Bo YL. HLA-DRA rs3129882 A/G polymorphism was not a risk factor for Parkinson’s disease in Chinese-based populations: a meta-analysis. Int J Neurosci. 2015;125:241–246.
  • Zhu RL, Lu XC, Tang LJ, et al. Association between HLA rs3129882 polymorphism and Parkinson’s disease: a meta-analysis. Eur Rev Med Pharmacol Sci. 2015;19:423–432.
  • Yuan Y, Tong Q, Zhou X, et al. The association between glycogen synthase kinase 3 beta polymorphisms and Parkinson’s disease susceptibility: a meta-analysis. Gene. 2013;524:133–138.
  • Zhang Y, Wang ZZ, Sun HM. A meta-analysis of the relationship of the Parkin p.Val380Leu polymorphism to Parkinson’s disease. Am J Med Genet B Neuropsychiatr Genet. 2013;162B:35–44.
  • Sun S, Zhao Y, Jin G, et al. Lack of association between UCHL1 S18Y gene polymorphism and Parkinson’s disease in the Asian population: a meta-analysis. Neurol Sci. 2014;35:1867–1876.
  • Zhu R, Zhu Y, Liu X, et al. UCH-L1 S18Y variant and risk of Parkinson’s disease in Asian populations: an updated meta-analysis. Neurodegener Dis. 2014;14:194–203.
  • Liu Y, Chen YY, Liu H, et al. Association between ubiquitin carboxy-terminal hydrolase-L1 S18Y variant and risk of Parkinson’s disease: the impact of ethnicity and onset age. Neurol Sci. 2015;36:179–188.
  • Dai D, Wang Y, Zhou X, et al. Meta-analyses of seven GIGYF2 polymorphisms with Parkinson’s disease. Biomed Rep. 2014;2:886–892.
  • Chang KH, Chen CM, Chen YC, et al. Association between PARK16 and Parkinson’s disease in the Han Chinese population: a meta-analysis. Neurobiol Aging. 2013;34:2442.e5–2442.e9.
  • Liu J, Zhang HX. Significant study of population stratification, sensitivity analysis and trim and fill analyses on GBA mutation and Parkinson’s disease. Am J Med Genet B Neuropsychiatr Genet. 2014;165B:96–102.
  • Ma ZG, He F, Xu J. Quantitative assessment of the association between GAK rs1564282 C/T polymorphism and the risk of Parkinson’s disease. J Clin Neurosci. 2015;22:1077–1080.
  • Zhang X, Niu M, Li H, et al. RIT2 rs12456492 polymorphism and the risk of Parkinson’s disease: a meta-analysis. Neurosci Lett. 2015;602:167–171.
  • Huo Q, Li T, Zhao P, et al. Association between rs6812193 polymorphism and sporadic Parkinson’s disease susceptibility. Neurol Sci. 2015;36:1479–1481.
  • Zhang X, Ye YL, Wang YN, et al. Aldehyde dehydrogenase 2 genetic variations may increase susceptibility to Parkinson’s disease in Han Chinese population. Neurobiol Aging. 2015;36:2660.e9–2660.e13.
  • Kumudini N, Uma A, Naushad SM, et al. Sexual dimorphism in xenobiotic genetic variants-mediated risk for Parkinson’s disease. Neurol Sci. 2014;35:897–903.
  • Kiyohara C, Miyake Y, Koyanagi M, et al. MDR1 C3435T polymorphism and interaction with environmental factors in risk of Parkinson’s disease: a case-control study in Japan. Drug Metab Pharmacokinet. 2013;28:138–143.
  • Yang X, Xu P, Zhao Q, et al. Polymorphism in the vesicular monoamine transporter 2 gene decreases the risk of Parkinson’s disease in Han Chinese Men. Parkinsons Dis. 2015;2015:903164. [Epub 2015 Jul 12]. doi:10.1155/2015/903164.
  • Rhodes SL, Buchanan DD, Ahmed I, et al. Pooled analysis of iron-related genes in Parkinson’s disease: association with transferrin. Neurobiol Dis. 2014;62:172–178.
  • Ayuso P, Martínez C, Pastor P, et al. An association study between Heme oxygenase-1 genetic variants and Parkinson’s disease. Front Cell Neurosci. 2014;8:298.
  • von Otter M, Bergström P, Quattrone A, et al. Genetic associations of Nrf2-encoding NFE2L2 variants with Parkinson’s disease a multicenter study. BMC Med Genet. 2014;15:131.
  • Simon KC, Eberly S, Gao X, et al. Mendelian randomization of serum urate and Parkinson disease progression. Ann Neurol. 2014;76:862–868.
  • Gao J, Teng J, Liu H, et al. Association of RAGE gene polymorphisms with sporadic Parkinson’s disease in Chinese Han population. Neurosci Lett. 2014;559:158–162.
  • He X, Zhang L, Yao X, et al. Association studies of MMP-9 in Parkinson’s disease and amyotrophic lateral sclerosis. PLoS One. 2013;8:e73777.
  • Zhao J, Han X, Xue L, et al. Association of TLR4 gene polymorphisms with sporadic Parkinson’s disease in a Han Chinese population. Neurol Sci. 2015;36:1659–1665.
  • Zhu K, Teng J, Zhao J, et al. Association of TLR9 polymorphisms with sporadic Parkinson’s disease in Chinese Han population. Int J Neurosci. 2015:1–5. [Epub ahead of print].
  • Yang X, Liu C, Zhang J, et al. Association of histamine N-methyltransferase Thr105Ile polymorphism with Parkinson’s disease and schizophrenia in Han Chinese: a case-control study. PLoS One. 2015;10:e0119692.
  • Rayaprolu S, Mullen B, Baker M, et al. TREM2 in neurodegeneration: evidence for association of the p.R47H variant with frontotemporal dementia and Parkinson’s disease. Mol Neurodegener. 2013;8:19.
  • Haghnejad L, Emamalizadeh B, Jamshidi J, et al. Variation in the miRNA-433 binding site of FGF20 is a risk factor for Parkinson’s disease in Iranian population. J Neurol Sci. 2015;355:72–74.
  • Wang V, Chuang TC, Kao MC, et al. Polymorphic Ala-allele carriers at residue 1170 of HER2 associated with Parkinson’s disease. J Neurol Sci. 2013;325:115–119.
  • Gusdon AM, Fang F, Chen J, et al. Association of the mt-ND2 5178A/C polymorphism with Parkinson’s disease. Neurosci Lett. 2015;587:98–101.
  • Mateo I, González-Aramburu I, Pozueta A, et al. Reduced serum progranulin level might be associated with Parkinson’s disease risk. Eur J Neurol. 2013;20:1571–1573.
  • Jesús S, Gómez-Garre P, Carrillo F, et al. Genetic association of sirtuin genes and Parkinson’s disease. J Neurol. 2013;260:2237–2241.
  • Liu H, Han X, Li Y, et al. Association of P2X7 receptor gene polymorphisms with sporadic Parkinson’s disease in a Han Chinese population. Neurosci Lett. 2013;546:42–45.
  • Greenbaum L, Rigbi A, Lipshtat N, et al. Association of nicotine dependence susceptibility gene, CHRNA5, with Parkinson’s disease age at onset: gene and smoking status interaction. Parkinsonism Relat Disord. 2013;19:72–76.
  • Zhang LM, Zhang XP, Chen YQ, et al. Association of CHRNA4 gene rs1044396 and rs1044397 polymorphisms with Parkinson’s disease symptoms and smoking. Genet Mol Res. 2015;14:5112–5122.
  • Goldman SM, Kamel F, Ross GW, et al. Peptidoglycan recognition protein genes and risk of Parkinson’s disease. Mov Disord. 2014;29:1171–1180.
  • Yamada-Fowler N, Fredrikson M, Söderkvist P. Caffeine interaction with glutamate receptor gene GRIN2A: Parkinson’s disease in Swedish population. PLoS One. 2014;9:e99294.
  • Zhang P, Liu L, Huang J, et al. Non-SMC condensin I complex, subunit D2 gene polymorphisms are associated with Parkinson’s disease: a Han Chinese study. Genome. 2014;57:253–257.
  • Cervera-Carles L, Pagonabarraga J, Pascual-Sedano B, et al. Copy number variation analysis of the 17q21.31 region and its role in neurodegenerative diseases. Am J Med Genet B Neuropsychiatr Genet. 2015. [Epub ahead of print]. doi:10.1002/ajmg.b.32390.
  • Sonmez E, Ozel MD, Islek EE, et al. Association of rs62063857 variant of the saitohin gene with Parkinson’s disease. Cell Mol Neurobiol. 2015;35:115–121.
  • Yang X, Zhao Q, An R, et al. Association of the functional SNP rs2275294 in ZNF512B with risk of amyotrophic lateral sclerosis and Parkinson’s disease in Han Chinese. Amyotroph Lateral Scler Frontotemporal Degener. 2015;17:142–147.
  • Jian CD, Huang JM, Meng LQ, et al. SNCA rs3822086 C>T polymorphism increases the susceptibility to Parkinson’s disease in a Chinese Han Population. Genet Test Mol Biomarkers. 2015;19:481–487.
  • de Mena L, Samaranch LL, Coto E, et al. Mutational screening of PARKIN identified a 3ʹ UTR variant (rs62637702) associated with Parkinson’s disease. J Mol Neurosci. 2013;50:264–269.
  • Trabzuni D, Ryten M, Emmett W, et al. Fine-mapping, gene expression and splicing analysis of the disease associated LRRK2 locus. PLoS One. 2013;8:e70724.
  • Beecham GW, Dickson DW, Scott WK, et al. PARK10 is a major locus for sporadic neuropathologically confirmed Parkinson disease. Neurology. 2015;84:972–980.
  • Wan JY, Edwards KL, Hutter CM, et al. Association mapping of the PARK10 region for Parkinson’s disease susceptibility genes. Parkinsonism Relat Disord. 2014;20:93–98.
  • Guo XY, Chen YP, Song W, et al. An association analysis of the rs1572931 polymorphism of the RAB7L1 gene in Parkinson’s disease, amyotrophic lateral sclerosis and multiple system atrophy in China. Eur J Neurol. 2014;21:1337–1343.
  • Goudarzian M, Khaligh A, Fourozan R, et al. The rs1572931 polymorphism of the RAB7L1 gene promoter is associated with reduced risk of Parkinson’s disease. Neurol Res. 2015;37:1029–1031.
  • Xia H, Luo Q, Li XX, et al. Association between PARK16 gene polymorphisms and susceptibility of Parkinson’s disease in a Chinese population. Genet Mol Res. 2015;14:2978–2985.
  • Wang L, Cheng L, Li NN, et al. Genetic analysis of SLC41A1 in Chinese Parkinson’s disease patients. Am J Med Genet B Neuropsychiatr Genet. 2015;168:706–711.
  • Yu RL, Guo JF, Wang YQ, et al. The single nucleotide polymorphism Rs12817488 is associated with Parkinson’s disease in the Chinese population. J Clin Neurosci. 2015;22:1002–1004.
  • Li NN, Tan EK, Chang XL, et al. Genetic association study between STK39 and CCDC62/HIP1R and Parkinson’s disease. PLoS One. 2013;8:e79211.
  • Wang JY, Gong MY, Ye YL, et al. The RIT2 and STX1B polymorphisms are associated with Parkinson’s disease. Parkinsonism Relat Disord. 2015;21:300–302.
  • Pihlstrøm L, Axelsson G, Bjørnarå KA, et al. Supportive evidence for 11 loci from genome-wide association studies in Parkinson’s disease. Neurobiol Aging. 2013;34:1708.e7–1708.e13.
  • Chung SJ, Jung Y, Hong M, et al. Alzheimer’s disease and Parkinson’s disease genome-wide association study top hits and risk of Parkinson’s disease in Korean population. Neurobiol Aging. 2013;34:2695.e1–2695.e7.
  • Wissemann WT, Hill-Burns EM, Zabetian CP, et al. Association of Parkinson disease with structural and regulatory variants in the HLA region. Am J Hum Genet. 2013;93:984–993.
  • Hill-Burns EM, Wissemann WT, Hamza TH, et al. Identification of a novel Parkinson’s disease locus via stratified genome-wide association study. BMC Genomics. 2014;15:118.
  • Nalls MA, Pankratz N, Lill CM, et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat Genet. 2014;46:989–993.
  • Guo JF, Li K, Yu RL, et al. Polygenic determinants of Parkinson’s disease in a Chinese population. Neurobiol Aging. 2015;36:1765.
  • Escott-Price V, International Parkinson’s Disease Genomics Consortium, Nalls MA, et al. Polygenic risk of Parkinson disease is correlated with disease age at onset. Ann Neurol. 2015;77:582–591.
  • Quadri M, Yang X, Cossu G, et al. An exome study of Parkinson’s disease in Sardinia, a Mediterranean genetic isolate. Neurogenetics. 2015;16:55–64.
  • Fallon SJ, Smulders K, Esselink RA, et al. Differential optimal dopamine levels for set-shifting and working memory in Parkinson’s disease. Neuropsychologia. 2015;77:42–51.
  • Białecka M, Kurzawski M, Roszmann A, et al. BDNF G196A (Val66Met) polymorphism associated with cognitive impairment in Parkinson’s disease. Neurosci Lett. 2014;561:86–90.
  • Mata IF, Leverenz JB, Weintraub D, et al. GBA Variants are associated with a distinct pattern of cognitive deficits in Parkinson’s disease. Mov Disord. 2016;31:95–102.
  • Winder-Rhodes SE, Evans JR, Ban M, et al. Glucocerebrosidase mutations influence the natural history of Parkinson’s disease in a community-based incident cohort. Brain. 2013;136:392–399.
  • Devos D, Lejeune S, Cormier-Dequaire F, et al. Dopa-decarboxylase gene polymorphisms affect the motor response to L-dopa in Parkinson’s disease. Parkinsonism Relat Disord. 2014;20:170–175.
  • Moreau C, Meguig S, Corvol JC, et al. Polymorphism of the dopamine transporter type 1 gene modifies the treatment response in Parkinson’s disease. Brain. 2015;138:1271–1283.
  • Kwak Y, Bohnen NI, Müller ML, et al. Task-dependent interactions between dopamine D2 receptor polymorphisms and L-DOPA in patients with Parkinson’s disease. Behav Brain Res. 2013;245:128–136.
  • Cheshire P, Bertram K, Ling H, et al. Influence of single nucleotide polymorphisms in COMT, MAO-A and BDNF genes on dyskinesias and levodopa use in Parkinson’s disease. Neurodegener Dis. 2014;13:24–28.
  • Hao H, Shao M, An J, et al. Association of Catechol-O-Methyltransferase and monoamine oxidase B gene polymorphisms with motor complications in parkinson’s disease in a Chinese population. Parkinsonism Relat Disord. 2014;20:1041–1045.
  • Lee JY, Cho J, Lee EK, et al. Differential genetic susceptibility in diphasic and peak-dose dyskinesias in Parkinson’s disease. Mov Disord. 2011;26:73–79.
  • Rieck M, Schumacher-Schuh AF, Altmann V, et al. DRD2 haplotype is associated with dyskinesia induced by levodopa therapy in Parkinson’s disease patients. Pharmacogenomics. 2012;13:1701–1710.
  • Rieck M, Schumacher-Schuh AF, Callegari-Jacques SM, et al. Is there a role for ADORA2A polymorphisms in levodopa-induced dyskinesia in Parkinson’s disease patients? Pharmacogenomics. 2015;16:573–582.
  • Kaplan N, Vituri A, Korczyn AD, et al. Sequence variants in SLC6A3, DRD2, and BDNF genes and time to levodopa-induced dyskinesias in Parkinson’s disease. J Mol Neurosci. 2014;53:183–188.
  • Gao C, Pang H, Luo XG, et al. LRRK2 G2385R variant carriers of female Parkinson’s disease are more susceptible to motor fluctuation. J Neurol. 2013;260:2884–2889.
  • Clark LN, Ross BM, Wang Y, et al. Mutations in the glucocerebrosidase gene are associated with early-onset Parkinson disease. Neurology. 2007;69:1270–1277.
  • Lesage S, Anheim M, Condroyer C, et al. Large-scale screening of the Gaucher’s disease-related glucocerebrosidase gene in Europeans with Parkinson’s disease. Hum Mol Genet. 2011;20:202–210.
  • Pascale E, Purcaro C, Passarelli E, et al. Genetic polymorphism of Angiotensin-Converting Enzyme is not associated with the development of Parkinson’s disease and of L-dopa-induced adverse effects. J Neurol Sci. 2009;276:18–21.
  • Lin JJ, Yueh KC, Lin SZ, et al. Genetic polymorphism of the angiotensin converting enzyme and L-dopa-induced adverse effects in Parkinson’s disease. J Neurol Sci. 2007;252:130–134.
  • Molchadski I, Korczyn AD, Cohen OS, et al. The role of apolipoprotein E polymorphisms in levodopa-induced dyskinesia. Acta Neurol Scand. 2011;123:117–121.
  • Greenbaum L, Goldwurm S, Zozulinsky P, et al. Do tardive dyskinesia and L-dopa induced dyskinesia share common genetic risk factors? An exploratory study. J Mol Neurosci. 2013;51:380–388.
  • Wu H, Dong F, Wang Y, et al. Catechol-O-methyltransferase Val158Met polymorphism: modulation of wearing-off susceptibility in a Chinese cohort of Parkinson’s disease. Parkinsonism Relat Disord. 2014;20:1094–1096.
  • Liu J, Chen P, Guo M, et al. Association of COMT Val158Met polymorphism with wearing-off susceptibility in Parkinson’s disease. Neurol Sci. 2015;36:621–623.
  • Li W, Chen Y, Yin B, et al. Pain in Parkinson’s disease associated with COMT gene polymorphisms. Behav Neurol. 2014;2014:304203.
  • Hoenicka J, García-Ruiz PJ, Ponce G, et al. The addiction-related gene ANKK1 in Parkinsonian patients with impulse control disorder. Neurotox Res. 2015;27:205–208.
  • Zainal Abidin S, Tan EL, Chan SC, et al. DRD and GRIN2B polymorphisms and their association with the development of impulse control behaviour among Malaysian Parkinson’s disease patients. BMC Neurol. 2015;15:59.
  • Kurzawski M, Bialecka M, Drozdzik M. Pharmacogenetic considerations in the treatment of Parkinson’s disease. Neurodegener Dis Manag. 2015;5:27–35.
  • LeWitt PA. Levodopa therapy for Parkinson’s disease: pharmacokinetics and pharmacodynamics. Mov Disord. 2015;30:64–72.
  • Stoessl AJ. Central pharmacokinetics of levodopa: lessons from imaging studies. Mov Disord. 2015;30:73–79.
  • Dezsi L, Vecsei L. Clinical implications of irregular ADMET properties with levodopa and other antiparkinson’s drugs. Expert Opin Drug Metab Toxicol. 2014;10:409–424.
  • Kumagai T, Nagayama H, Ota T, et al. Sex differences in the pharmacokinetics of levodopa in elderly patients with Parkinson disease. Clin Neuropharmacol. 2014;37:173–176.
  • Umeh CC, Perez A, Augustine EF, et al. No sex differences in use of dopaminergic medication in early Parkinson disease in the US and Canada – baseline findings of a multicenter trial. PloS One. 2014;9:e112287.
  • Nagayama H, Kajimoto Y, Kumagai T, et al. Pharmacokinetics of levodopa before and after gastrointestinal resection in Parkinson’s disease. Case Rep Neurol. 2015;7:181–185.
  • Nagashima Y, Kondo T, Sakata M, et al. Effects of soybean ingestion on pharmacokinetics of levodopa and motor symptoms of Parkinson’s disease – in relation to the effects of Mucuna pruriens. J Neurol Sci. 2016;361:229–234.
  • Kyaw WT, Nagai M, Kaneta M, et al. Effect of nicotine on the pharmacokinetics of levodopa. Clin Neuropharmacol. 2013;36:46–51.
  • Yin B, Chen Y, Zhang L. Association between Catechol-O-Methyltransferase (COMT) gene polymorphisms, Parkinson’s disease, and levodopa efficacy. Mol Diag Ther. 2013. [Epub ahead of print].
  • Torkaman-Boutorabi A, Shahidi GA, Choopani S, et al. The catechol-O-methyltransferase and monoamine oxidase B polymorphisms and levodopa therapy in the Iranian patients with sporadic Parkinson’s disease. Acta Neurobiol Exp (Wars). 2012;72:272–282.
  • CPIC. Clinical pharmacogenetics implementation consortium. 2016 [cited 2016 Jan 31]. Available from: http://www.pharmgkb.org/page/cpic
  • Caudle KE, Klein TE, Hoffman JM, et al. Incorporation of pharmacogenomics into routine clinical practice: the Clinical Pharmacogenetics Implementati on Consortium (CPIC) guideline development process. Curr Drug Metab. 2014;15:209–217.
  • Agundez JA, Abad-Santos F, Aldea A, et al. Toward a clinical practice guide in pharmacogenomics testing for functional polymorphisms of drug-metabolizing enzymes. Gene/drug pairs and barriers perceived in Spain. Front Genet. 2012;3:273.
  • Quinones LA, Lavanderos MA, Cayun JP, et al. Perception of the usefulness of drug/gene pairs and barriers for pharmacogenomics in Latin America. Curr Drug Metab. 2014;15:202–208.
  • Relling MV, Klein TE. CPIC: clinical pharmacogenetics implementation consortium of the pharmacogenomics research network. Clin Pharmacol Ther. 2011;89:464–467.
  • Rocha JF, Falcao A, Santos A, et al. Effect of opicapone and entacapone upon levodopa pharmacokinetics during three daily levodopa administrations. Eur J Clin Pharmacol. 2014;70:1059–1071.
  • Siddiqi SH, Abraham NK, Geiger CL, et al. The human experience with intravenous levodopa. Front Pharmacol. 2015;6:307.
  • Othman AA, Chatamra K, Mohamed ME, et al. Jejunal Infusion of levodopa-carbidopa intestinal gel versus oral administration of levodopa-carbidopa tablets in japanese subjects with advanced Parkinson’s disease: pharmacokinetics and pilot efficacy and safety. Clin Pharmacokinet. 2015;54:975–984.
  • Poewe W, Antonini A. Novel formulations and modes of delivery of levodopa. Mov Disord. 2015;30:114–120.
  • Othman AA, Dutta S. Population pharmacokinetics of levodopa in subjects with advanced Parkinson’s disease: levodopa-carbidopa intestinal gel infusion vs. oral tablets. Br J Clin Pharmacol. 2014;78:94–105.
  • Pilleri M, Antonini A. Novel levodopa formulations in the treatment of Parkinson’s disease. Expert Rev Neurother. 2014;14:143–149.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.