Association of Glycolysis-Enhancing α-1 Blockers With Risk of Developing Parkinson Disease

Jacob E Simmering, Michael J Welsh, Lei Liu, Nandakumar S Narayanan, Anton Pottegård, Jacob E Simmering, Michael J Welsh, Lei Liu, Nandakumar S Narayanan, Anton Pottegård

Abstract

Importance: Parkinson disease (PD) is a common neurodegenerative disease. A treatment that prevents or delays development of PD is a critical unmet need. Terazosin and closely related drugs were recently discovered to enhance glycolysis and reduce PD progression in animal models and human clinical databases.

Objective: To determine whether use of terazosin, doxazosin, and alfuzosin is associated with a decreased risk of developing PD.

Design, setting, and participants: This cohort study used active comparator control and propensity score-matched data from Danish nationwide health registries, including the Danish National Prescription Registry, the Danish National Patient Registry, and the Danish Civil Registration System, from January 1996 to December 2017 and data from the Truven Health Analytics MarketScan database from January 2001 to December 2017. Men without PD who newly initiated terazosin/doxazosin/alfuzosin therapy or tamsulosin therapy, which is used for a similar indication (benign prostatic hyperplasia or unspecified urinary problems) but does not enhance glycolysis, and had at least 1 year of follow-up after medication start were included. In Denmark, the database included all residents, while the Truven database is a compilation of insurance claims across the US. Data were analyzed from February 2019 to July 2020.

Exposures: Patients who used terazosin/doxazosin/alfuzosin vs tamsulosin. Additional dose-response analyses were carried out.

Main outcomes and measures: Differences in the hazard of developing PD identified by diagnoses or use of PD-specific medications between patients who ever used terazosin/doxazosin/alfuzosin or tamsulosin.

Results: A cohort of 52 365 propensity score-matched pairs of terazosin/doxazosin/alfuzosin and tamsulosin users were identified in the Danish registries, of which all were male and the mean (SD) age was 67.9 (10.4) years, and 94 883 propensity score-matched pairs were identified in the Truven database, of which all were male and the mean (SD) age was 63.8 (11.1) years. Patients in the Danish cohort who used terazosin/doxazosin/alfuzosin had a hazard ratio (HR) for developing PD of 0.88 (95% CI, 0.81-0.98), and patients in the Truven cohort had an HR of 0.63 (95% CI, 0.58-0.69). There was a dose-response association with short-duration, medium-duration, and long-duration use of terazosin/doxazosin/alfuzosin users having a decreasing HR in both the Danish cohort (short: HR, 0.95; 95% CI, 0.84-1.07; medium: HR, 0.88; 95% CI, 0.77-1.01; long: HR, 0.79; 95% CI, 0.66-0.95) and Truven cohort (short: HR, 0.70; 95% CI, 0.64-0.76; medium: HR, 0.58; 95% CI, 0.52-0.64; long: HR, 0.46; 95% CI, 0.36-0.57).

Conclusions and relevance: These data suggest that users of terazosin/doxazosin/alfuzosin are at lower hazard of developing PD compared with users of tamsulosin. Future work is needed to further assess this association.

Conflict of interest statement

Conflict of Interest Disclosures: None reported.

Figures

Figure 1.. Cumulative Incidence of Parkinson Disease…
Figure 1.. Cumulative Incidence of Parkinson Disease (PD) in the Danish and Truven Cohorts
A, Kaplan-Meier plot of Danish nationwide health registries, including the Danish National Prescription Registry, the Danish National Patient Registry, and the Danish Civil Registration System. B, Kaplan-Meier plot of Truven Health Analytics MarketScan database. The lines denote the Kaplan-Meier estimated cumulative incidence of PD by cohort while the shaded regions denote the 95% CIs. The Truven cohort terminates at 15 years, as that is the longest possible follow-up in that database, while the Danish cohort has 5 additional years of follow-up possible. In both samples, use of terazosin (TZ), doxazosin (DZ), or alfuzosin (AZ) was associated with lower cumulative incidence of PD compared with use of tamsulosin.
Figure 2.. Estimated Hazard Ratio by Cumulative…
Figure 2.. Estimated Hazard Ratio by Cumulative Number of Defined Daily Doses
A, Estimated hazard of developing Parkinson disease among those in the Danish nationwide health registries, including the Danish National Prescription Registry, the Danish National Patient Registry, and the Danish Civil Registration System. B, Estimated hazard of developing Parkinson disease among those in the Truven Health Analytics MarketScan database. Values are hazard ratios, and error bars indicate 95% CIs. Increasing intensity and duration of treatment as measured by defined daily doses was associated with a decreasing estimated hazard ratio.

References

    1. Ascherio A, Schwarzschild MA. The epidemiology of Parkinson’s disease: risk factors and prevention. Lancet Neurol. 2016;15(12):1257-1272. doi:10.1016/S1474-4422(16)30230-7
    1. de Lau LM, Breteler MM. Epidemiology of Parkinson’s disease. Lancet Neurol. 2006;5(6):525-535. doi:10.1016/S1474-4422(06)70471-9
    1. Hirsch L, Jette N, Frolkis A, Steeves T, Pringsheim T. The incidence of Parkinson’s disease: a systematic review and meta-analysis. Neuroepidemiology. 2016;46(4):292-300. doi:10.1159/000445751
    1. Burghaus L, Hilker R, Thiel A, et al. . Deep brain stimulation of the subthalamic nucleus reversibly deteriorates stuttering in advanced Parkinson’s disease. J Neural Transm (Vienna). 2006;113(5):625-631. doi:10.1007/s00702-005-0341-1
    1. Hilker R, Portman AT, Voges J, et al. . Disease progression continues in patients with advanced Parkinson’s disease and effective subthalamic nucleus stimulation. J Neurol Neurosurg Psychiatry. 2005;76(9):1217-1221. doi:10.1136/jnnp.2004.057893
    1. Verschuur CVM, Suwijn SR, Boel JA, et al. ; LEAP Study Group . Randomized delayed-start trial of levodopa in Parkinson’s disease. N Engl J Med. 2019;380(4):315-324. doi:10.1056/NEJMoa1809983
    1. Biglan KM, Oakes D, Lang AE, et al. ; Parkinson Study Group STEADY‐PD III Investigators . A novel design of a phase III trial of isradipine in early Parkinson disease (STEADY-PD III). Ann Clin Transl Neurol. 2017;4(6):360-368. doi:10.1002/acn3.412
    1. Kalia LV, Kalia SK, Lang AE. Disease-modifying strategies for Parkinson’s disease. Mov Disord. 2015;30(11):1442-1450. doi:10.1002/mds.26354
    1. LeWitt PA. Levodopa therapy for Parkinson’s disease: pharmacokinetics and pharmacodynamics. Mov Disord. 2015;30(1):64-72. doi:10.1002/mds.26082
    1. Saxena U. Bioenergetics failure in neurodegenerative diseases: back to the future. Expert Opin Ther Targets. 2012;16(4):351-354. doi:10.1517/14728222.2012.664135
    1. Hoyer S. Brain glucose and energy metabolism during normal aging. Aging (Milano). 1990;2(3):245-258. doi:10.1007/BF03323925
    1. Cunnane S, Nugent S, Roy M, et al. . Brain fuel metabolism, aging, and Alzheimer’s disease. Nutrition. 2011;27(1):3-20. doi:10.1016/j.nut.2010.07.021
    1. Firbank MJ, Yarnall AJ, Lawson RA, et al. . Cerebral glucose metabolism and cognition in newly diagnosed Parkinson’s disease: ICICLE-PD study. J Neurol Neurosurg Psychiatry. 2017;88(4):310-316. doi:10.1136/jnnp-2016-313918
    1. Hattingen E, Magerkurth J, Pilatus U, et al. . Phosphorus and proton magnetic resonance spectroscopy demonstrates mitochondrial dysfunction in early and advanced Parkinson’s disease. Brain. 2009;132(pt 12):3285-3297. doi:10.1093/brain/awp293
    1. Schapira AH, Mann VM, Cooper JM, et al. . Anatomic and disease specificity of NADH CoQ1 reductase (complex I) deficiency in Parkinson’s disease. J Neurochem. 1990;55(6):2142-2145. doi:10.1111/j.1471-4159.1990.tb05809.x
    1. Schapira AH, Cooper JM, Dexter D, Clark JB, Jenner P, Marsden CD. Mitochondrial complex I deficiency in Parkinson’s disease. J Neurochem. 1990;54(3):823-827. doi:10.1111/j.1471-4159.1990.tb02325.x
    1. Pyle A, Anugrha H, Kurzawa-Akanbi M, Yarnall A, Burn D, Hudson G. Reduced mitochondrial DNA copy number is a biomarker of Parkinson’s disease. Neurobiol Aging. 2016;38:216.e7-216.e10. doi:10.1016/j.neurobiolaging.2015.10.033
    1. Blesa J, Phani S, Jackson-Lewis V, Przedborski S. Classic and new animal models of Parkinson’s disease. J Biomed Biotechnol. 2012;2012:845618. doi:10.1155/2012/845618
    1. Schapira AH. Mitochondria in the aetiology and pathogenesis of Parkinson’s disease. Lancet Neurol. 2008;7(1):97-109. doi:10.1016/S1474-4422(07)70327-7
    1. Schapira AH. Mitochondrial dysfunction in neurodegenerative diseases. Neurochem Res. 2008;33(12):2502-2509. doi:10.1007/s11064-008-9855-x
    1. Chen X, Zhao C, Li X, et al. . Terazosin activates Pgk1 and Hsp90 to promote stress resistance. Nat Chem Biol. 2015;11(1):19-25. doi:10.1038/nchembio.1657
    1. Cai R, Zhang Y, Simmering JE, et al. . Enhancing glycolysis attenuates Parkinson’s disease progression in models and clinical databases. J Clin Invest. 2019;129(10):4539-4549. doi:10.1172/JCI129987
    1. Achari R, Laddu A. Terazosin: a new alpha adrenoceptor blocking drug. J Clin Pharmacol. 1992;32(6):520-523. doi:10.1177/009127009203200605
    1. de Groat WC, Yoshiyama M, Ramage AG, Yamamoto T, Somogyi GT. Modulation of voiding and storage reflexes by activation of α1-adrenoceptors. Eur Urol. 1999;36(suppl 1):68-73. doi:10.1159/000052324
    1. Pottegård A, Schmidt SAJ, Wallach-Kildemoes H, Sørensen HT, Hallas J, Schmidt M. Data resource profile: the Danish National Prescription registry. Int J Epidemiol. 2017;46(3):798-798f.
    1. Schmidt M, Schmidt SA, Sandegaard JL, Ehrenstein V, Pedersen L, Sørensen HT. The Danish National Patient registry: a review of content, data quality, and research potential. Clin Epidemiol. 2015;7:449-490. doi:10.2147/CLEP.S91125
    1. Schmidt M, Pedersen L, Sørensen HT. The Danish Civil Registration System as a tool in epidemiology. Eur J Epidemiol. 2014;29(8):541-549. doi:10.1007/s10654-014-9930-3
    1. World Health Organization . Guidelines for ATC classification and DDD assignment. Accessed February 21, 2019.
    1. St Germaine-Smith C, Metcalfe A, Pringsheim T, et al. . Recommendations for optimal ICD codes to study neurologic conditions: a systematic review. Neurology. 2012;79(10):1049-1055. doi:10.1212/WNL.0b013e3182684707
    1. Szumski NR, Cheng EM. Optimizing algorithms to identify Parkinson’s disease cases within an administrative database. Mov Disord. 2009;24(1):51-56. doi:10.1002/mds.22283
    1. Elixhauser A, Steiner C, Harris DR, Coffey RM. Comorbidity measures for use with administrative data. Med Care. 1998;36(1):8-27. doi:10.1097/00005650-199801000-00004
    1. Stürmer T, Wyss R, Glynn RJ, Brookhart MA. Propensity scores for confounder adjustment when assessing the effects of medical interventions using nonexperimental study designs. J Intern Med. 2014;275(6):570-580. doi:10.1111/joim.12197
    1. Surmeier DJ. Determinants of dopaminergic neuron loss in Parkinson’s disease. FEBS J. 2018;285(19):3657-3668. doi:10.1111/febs.14607
    1. Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goedert M. alpha-Synuclein in filamentous inclusions of Lewy bodies from Parkinson’s disease and dementia with lewy bodies. Proc Natl Acad Sci U S A. 1998;95(11):6469-6473. doi:10.1073/pnas.95.11.6469
    1. Singleton AB, Farrer M, Johnson J, et al. . alpha-Synuclein locus triplication causes Parkinson’s disease. Science. 2003;302(5646):841. doi:10.1126/science.1090278
    1. Patel A, Malinovska L, Saha S, et al. . ATP as a biological hydrotrope. Science. 2017;356(6339):753-756. doi:10.1126/science.aaf6846
    1. Hayes MH, Peuchen EH, Dovichi NJ, Weeks DL. Dual roles for ATP in the regulation of phase separated protein aggregates in Xenopus oocyte nucleoli. Elife. 2018;7:7. doi:10.7554/eLife.35224

Source: PubMed

3
Předplatit