This site is for US healthcare professionals only

Primary Endpoint - “Off” Time

Primary Endpoint

NOURIANZ® is the first and only adjunct therapy that reduces “off” time
by lifting the brake of adenosine1,2

Study description: Randomized, 12-week, multicenter, double-blind, placebo-controlled studies of NOURIANZ 20 mg and 40 mg as adjunctive treatment to levodopa treatment in patients with PD (mean age=65 years) experiencing “off” time (mean approximately 6 hours/day).

In Studies 1 & 2, patients were randomized to once-daily treatment with NOURIANZ 20 mg, 40 mg, or placebo with the primary endpoint of change from baseline in % of awake time spent in the “off” state:

  • Study 1 (N=195) was conducted in the U.S. and Canada
  • Study 2 (N=225) was conducted in the U.S.

In Studies 3 & 4, patients were randomized equally to treatment with NOURIANZ 20 mg, 40 mg, or placebo with the primary endpoint of change from baseline spent in “off” time/day

  • Study 3 (N=357) was conducted in Japan
  • Study 4 (N=366) was conducted in Japan

Secondary endpoint in all 4 studies was change from baseline in “on” time without troublesome dyskinesia. Endpoints measured using 24-hour patient diaries.1,2

All enrolled patients were on stable doses of baseline therapy for the duration of the studies. Baseline therapy was levodopa/carbidopa (Studies 1, 2, 3, and 4) or levodopa/benserazide (Studies 3 and 4) for all patients with or without other concomitant baseline medications for PD, including dopamine agonists (85%), COMT inhibitors (38%), MAO-B inhibitors (40%), anticholinergics (13%), and/or amantadine (33%).1,2

COMT, catechol-o-methyl transferase; MAO, monoamine oxidase.

Based on 24-hour patient diaries in Studies 3 & 4: NOURIANZ reduced “off” time1,3

Bar chart shows NOURIANZ® (istradefylline) reduced “off” time hours in studies 3 and 4 Bar chart shows NOURIANZ® (istradefylline) reduced “off” time hours in studies 3 and 4

aLeast squares (LS) mean change from baseline (ANCOVA).
ANCOVA, analysis of covariance.

  • At Week 12, patients taking NOURIANZ in clinical trials saw a significant decrease in “off” time from baseline compared to placebo in all 4 studies (N=1,143 PD patients)1

Studies 1 and 2: Change in “off” time from baseline to endpoint (12 weeks),
daily awake “off” time (% of awake hours)1,3

Table shows NOURIANZ® (istradefylline) change in “off” time from baseline to endpoint (12 weeks), daily awake “off” time (% of awake hours) in studies 1 and 2 Table shows NOURIANZ® (istradefylline) reduced the percentage of awake time “off” in studies 1 and 2
  • Pre-specified analysis for Study 1 was ANOVA, and for Study 2 was ANCOVA*

*Least squares mean change from baseline (ANOVA in Study 1, ANCOVA in Study 2).
ANOVA, analysis of variance; ANCOVA, analysis of covariance.

Secondary Endpoint - “On” Time

Secondary Endpoint

NOURIANZ lifts the brake of adenosine to help increase good “on” time1,2*

Based on 24-hour patient diaries: NOURIANZ reduced “off” time and was shown to increase good “on” time without troublesome dyskinesia at Week 121,3*

 
Bar chart shows NOURIANZ® (istradefylline) adjunctive therapy reduced “off” time and increased good “on” time in all 4 studies Bar chart shows NOURIANZ® (istradefylline) reduced “off” time and increased good “on” time in all 4 studies

*Good “on” time=“on” time without troublesome dyskinesia plus “on” time with non-troublesome dyskinesia.

aLeast squares mean change from baseline (ANOVA in Study 1, ANCOVA in Studies 2, 3, and 4).
ANOVA, analysis of variance; ANCOVA, analysis of covariance.
Baseline therapy was levodopa/carbidopa and/or levodopa/benserazide for all patients with or without
other concomitant medications for PD.

  • At Week 12, patients treated with NOURIANZ experienced an increase in good “on” time.1,3*
    • A change from baseline in good “on” time without troublesome dyskinesia (i.e., “on” time without dyskinesia plus “on” time with non-troublesome dyskinesia) was a secondary efficacy endpoint
  • Increases from baseline with NOURIANZ 20 mg ranged from 0.90 to 1.35 hours. Nominal  P-values: P=0.135 in Study 2; P=0.085 in Study 3; P=0.008 in Study 4
  • Increases from baseline with NOURIANZ 40 mg ranged from 0.85 to 1.45 hours. Nominal  P-values: P=0.026 in Study 1; P=0.048 in Study 3; P=0.008 in Study 4
  • Increases from baseline with placebo ranged from 0.28 to 0.80 hours

NOURIANZ in combination with levodopa may cause dyskinesia or exacerbate pre-existing dyskinesia.

NOURIANZ was evaluated in patients taking levodopa treatment with or without
other concomitant baseline medications for PD1,3

Other concomitant medications for PD taken in NOURIANZ clinical trials.

Table shows NOURIANZ® (istradefylline) was evaluated in patients on anticholinergics, amantadine, MAO-B inhibitors, COMT inhibitors and dopamine agonists for PD Table shows percentage of patients who were on other PD medications in NOURIANZ® (istradefylline) clinical trials


COMT, catechol-o-methyltransferase; MAO-B, monoamine oxidase B.

Actor Portrayals.

Actor Portrayals.

arrow

References: 1. NOURIANZ. Prescribing Information. Kyowa Kirin, Inc; 2023. Accessed November 1, 2023. https://www.nourianzhcp.com/assets/pdf/nourianz-full-prescribing-information.pdf. 2. Kalia LV, Brotchie JM, Fox SH. Novel nondopaminergic targets for motor features of Parkinson’s disease: review of recent trials. Mov Disord. 2013;28(2):131-144.

References: 1. Kalia LV, Brotchie JM, Fox SH. Novel nondopaminergic targets for motor features of Parkinson's disease: review of recent trials. Mov Disord. 2013;28(2):131-144. 2. Mori A. Mode of action of adenosine A2A receptor antagonists as symptomatic treatment for Parkinson’s disease. Int Rev Neurobiol. 2014;119:87-116. 3. Varani K, Vincenzi F, Tosi A, et al. A2A adenosine receptor overexpression and functionality, as well as TNF-α levels, correlate with motor symptoms in Parkinson’s disease. FASEB J. 2010;24(2):587-598. doi:10.1096/fj.09-141044. 4. Fuxe K, Marcellino D, Genedani S, Agnati L. Adenosine A2A receptors, dopamine D2 receptors and their interactions in Parkinson's disease. Mov Disord. 2007;22(14):1990-2017. doi: 10.1002/mds.21440. 5. Morelli M, Di Paolo T, Wardas J, Calon F, Xiao D, Schwarzschild MA. Role of adenosine A2A receptors in parkinsonian motor impairment and L-DOPA-induced motor complications. Prog Neurobiol. 2007;83(5):293-309. 6. Morelli M, Blandini F, Simola N, Hauser RA. A2A receptor antagonism and dyskinesia in Parkinson's disease. Parkinsons Dis. 2012;2012:489853. doi: 10.1155/2012/489853. 7. Mishina M, Ishiwata K. Adenosine receptor PET imaging in human brain. Int Rev Neurobiol. 2014;119:51-69. doi:10.1016/B978-0-12-801022-8.00002-7. 8. The voice of the patient: Parkinson’s disease. Silver Spring, MD: US Food and Drug Administration; April 2016. https://www.fda.gov/media/124392/download. Accessed June 11, 2019. 9. Hickey P, Stacy M. Available and emerging treatments for Parkinson’s disease: a review. Drug Des Devel Ther. 2011;5:241-254. 10. Stocchi F, Antonini A, Barone P, et al. Early DEtection of wEaring off in Parkinson disease: the DEEP study. Parkinsonism Relat Disord. 2014;20(2):204-211.

References: 1. NOURIANZ. Prescribing Information. Kyowa Kirin, Inc; 2020. Accessed April 1, 2021. https://www.nourianzhcp.com/assets/pdf/nourianz-full-prescribing-information.pdf   2. Kalia LV, Brotchie JM, Fox SH. Novel nondopaminergic targets for motor features of Parkinson’s disease: review of recent trials. Mov Disord. 2013;28(2):131-144. 3. Jenner P. Istradefylline, a novel adenosine A2A receptor antagonist, for the treatment of Parkinson’s disease. Expert Opin Investig Drugs. 2005;14(6):729-738. 4. Brichta L, Greengard P, Flajolet M. Advances in the pharmacological treatment of Parkinson’s disease: targeting neurotransmitter systems. Trends Neurosci. 2013;36(9):543-554. 5. Kaakkola S, Wurtman RJ. Effects of COMT inhibitors on striatal dopamine metabolism: a microdialysis study. Brain Res. 1992;587(2):241-249. 6. Kong P, Zhang B, Lei P, et al. Neuroprotection of MAO-B inhibitor and dopamine agonist in Parkinson disease. Int J Clin Exp Med. 2015;8(1):431-439. 7. Ossola B, Schendzielorz N, Chen SH, et al. Amantadine protects dopamine neurons by a dual action: reducing activation of microglia and inducing expression of GDNF in astroglia. Neuropharmacology. 2011;61(4):574-582. 8. Rubí B, Maechler P. Minireview: new roles for peripheral dopamine on metabolic control and tumor growth: let’s seek the balance. Endocrinology. 2010;151(12):5570-5581. doi:10.1210/en.2010-0745. 9. Gerlach M, Double K, Arzberger T, Leblhuber F, Tatschner T, Riederer P. Dopamine receptor agonists in current clinical use: comparative dopamine receptor binding profiles defined in the human striatum. J Neural Transm (Vienna). 2003;110(10):1119-1127. 10. Ishibashi K, Miura Y, Wagatsuma K, Toyohara J, Ishiwata K, Ishii K. Adenosine A2A receptor occupancy by long-term istradefylline administration in Parkinson’s disease. Mov Disord. 2021;36(1):268-269. doi:10.1002/mds.28378.

References: 1. NOURIANZ. Prescribing Information. Kyowa Kirin, Inc; 2020. Accessed April 1, 2021. https://www.nourianzhcp.com/assets/pdf/nourianz-full-prescribing-information.pdf   2. Kalia LV, Brotchie JM, Fox SH. Novel nondopaminergic targets for motor features of Parkinson’s disease: review of recent trials. Mov Disord. 2013;28(2):131-144. 3. Data on file. Kyowa Kirin Pharmaceutical Development, Inc., Princeton, NJ.

References: 1. NOURIANZ. Prescribing Information. Kyowa Kirin, Inc; 2020. Accessed April 1, 2021. https://www.nourianzhcp.com/assets/pdf/nourianz-full-prescribing-information.pdf  2. Data on file. Kyowa Kirin Pharmaceutical Development, Inc., Princeton, NJ.

Reference: 1. NOURIANZ. Prescribing Information. Kyowa Kirin, Inc; 2020. Accessed April 1, 2021. https://www.nourianzhcp.com/assets/pdf/nourianz-full-prescribing-information.pdf

Reference: 1. NOURIANZ. Prescribing Information. Kyowa Kirin, Inc; 2020. Accessed April 1, 2021. https://www.nourianzhcp.com/assets/pdf/nourianz-full-prescribing-information.pdf