Nilotinib Effects on Safety, Tolerability, and Potential Biomarkers in Parkinson Disease: A Phase 2 Randomized Clinical Trial

Fernando L Pagan, Michaeline L Hebron, Barbara Wilmarth, Yasar Torres-Yaghi, Abigail Lawler, Elizabeth E Mundel, Nadia Yusuf, Nathan J Starr, Muhammad Anjum, Joy Arellano, Helen H Howard, Wangke Shi, Sanjana Mulki, Tarick Kurd-Misto, Sara Matar, Xiaoguang Liu, Jaeil Ahn, Charbel Moussa, Fernando L Pagan, Michaeline L Hebron, Barbara Wilmarth, Yasar Torres-Yaghi, Abigail Lawler, Elizabeth E Mundel, Nadia Yusuf, Nathan J Starr, Muhammad Anjum, Joy Arellano, Helen H Howard, Wangke Shi, Sanjana Mulki, Tarick Kurd-Misto, Sara Matar, Xiaoguang Liu, Jaeil Ahn, Charbel Moussa

Abstract

Importance: This study evaluated nilotinib safety and its effects on biomarkers as a potential disease-modifying drug in Parkinson disease.

Objectives: To assess nilotinib effects on safety and pharmacokinetics and measure the change in exploratory biomarkers in patients with moderately severe Parkinson disease.

Design, setting, and participants: This was a single-center, phase 2, randomized, double-blind, placebo-controlled trial with 300 patients approached in clinic; of these, 200 declined to participate, 100 were screened, 25 were excluded, and 75 were randomized 1:1:1 into placebo; nilotinib, 150-mg; or nilotinib, 300-mg groups. Recruitment started on May 17, 2017, and ended April 28, 2018, and follow-up ended August 10, 2019. Parkinson disease was confirmed according to the UK Brain Bank diagnostic criteria and symptoms were stabilized with use of optimal levodopa and/or dopamine agonists and other medications used in Parkinson disease.

Interventions: Nilotinib vs placebo, administered orally once daily for 12 months followed by a 3-month washout period.

Main outcomes and measures: It was hypothesized that nilotinib is safe and can be detected in the cerebrospinal fluid, where it alters exploratory biomarkers via inhibition of Abelson tyrosine kinase and potentially improves clinical outcomes.

Results: Of the 75 patients included in the study, 55 were men (73.3%); mean (SD) age was 68.4 (8.2) years. Doses of 150 or 300 mg of nilotinib were reasonably safe, although more serious adverse events were detected in the nilotinib (150 mg: 6 [24%]; 300 mg: 12 [48%]) vs placebo (4 [16%]) groups. The 150-mg nilotinib group showed an increase in cerebrospinal fluid levels of the dopamine metabolites homovanillic acid (159.80nM; 90% CI, 7.04-312.60nM; P = .04) and 3,4-dihydroxyphenylacetic acid (4.87nM; 90% CI, 1.51-8.23nM; P = .01), and the 300-mg nilotinib group showed an increase in 3,4-dihydroxyphenylacetic acid (7.52nM; 90% CI, 2.35-12.69nM; P = .01). The nilotinib 150-mg but not the nilotinib 300-mg group demonstrated a reduction of α-synuclein oligomers (-0.04 pg/mL; 90% CI, -0.08 to 0.01 pg/mL; P = .03). A significant reduction of hyperphosphorylated tau levels was seen in the nilotinib 150-mg (-10.04 pg/mL; 90% CI, -17.41 to -2.67 pg/mL; P = .01) and nilotinib 300-mg (-12.05 pg/mL; 90% CI, -19.21 to -4.90 pg/mL; P = .01) groups.

Conclusions and relevance: In this study, nilotinib appeared to be reasonably safe and detectable in the cerebrospinal fluid. Exploratory biomarkers were altered in response to nilotinib. Taken together, these data will guide the development of a phase 3 study to investigate the effects of nilotinib therapy in patients with Parkinson disease.

Trial registration: ClinicalTrials.gov identifier: NCT02954978.

Conflict of interest statement

Conflict of Interest Disclosures: Dr Pagan reported receiving personal fees and other support from Acadia, AbbVie, Accorda, and Adamas; personal fees from Teva; grants and other from Medtronic; grants, personal fees, and other from US World Meds; grants from the National Institutes of Health National Institute on Aging; grants from Alzheimer's Association and National Institutes of Health as co–principal investigator, and Parkinson Foundation, and personal fees and other from Sunovion and Merz outside the submitted work. Dr Moussa reported having a patent to US 9,474,753 B2 issued and being listed as an inventor on several patent publications to use nilotinib as a potential treatment for neurodegenerative diseases. Dr Moussa and Georgetown University own the intellectual property on the use of nilotinib for neurodegenerative disorders. No other disclosures were reported.

Figures

Figure 1.. CONSORT Flow Diagram
Figure 1.. CONSORT Flow Diagram
Phase 2, randomized, double-blind, placebo-controlled trial to evaluate nilotinib hydrochloride effects on safety, tolerability, biomarkers, and clinical outcomes in Parkinson disease. For values given as 20-21 in the bottom boxes, biomarkers were analyzed and only those samples who gave a reading were reported. A few samples did not produce a value when analyzed. PI indicates principal investigator; SAE, serious adverse event.
Figure 2.. Data Representing the Pharmacokinetics and…
Figure 2.. Data Representing the Pharmacokinetics and Pharmacodynamics Effects of 1-Year Nilotinib Treatment Compared With Placebo in Parkinson Disease
Cerebrospinal fluid (CSF) levels of homovanillic acid HVA (A), 3,4-dihydroxyphenylacetic acid (DOPAC) (B), total α-synuclein (C), oligomeric α-synuclein (D), ratio of oligomeric/total α-synuclein (E), total tau (F), hyperphosphorylated tau (P-tau181) (G), and ratio of p-tau181/total tau (H). Total of 20 to 21 patients per group. aP < .05.

References

    1. Spillantini MG, Schmidt ML, Lee VM, Trojanowski JQ, Jakes R, Goedert M. Alpha-synuclein in Lewy bodies. Nature. 1997;388(6645):839-840. doi:10.1038/42166
    1. Spillantini MG, Crowther RA, Jakes R, Hasegawa M, Goedert M. α-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. Goedert M. Alpha-synuclein and neurodegenerative diseases. Nat Rev Neurosci. 2001;2(7):492-501. doi:10.1038/35081564
    1. Deremer DL, Ustun C, Natarajan K. Nilotinib: a second-generation tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia. Clin Ther. 2008;30(11):1956-1975. doi:10.1016/j.clinthera.2008.11.014
    1. Skorski T. BCR-ABL1 kinase: hunting an elusive target with new weapons. Chem Biol. 2011;18(11):1352-1353. doi:10.1016/j.chembiol.2011.11.001
    1. Mahon FX, Hayette S, Lagarde V, et al. . Evidence that resistance to nilotinib may be due to BCR-ABL, Pgp, or Src kinase overexpression. Cancer Res. 2008;68(23):9809-9816. doi:10.1158/0008-5472.CAN-08-1008
    1. Fowler AJ, Hebron M, Missner AA, et al. . Multikinase Abl/DDR/Src inhibition produces optimal effects for tyrosine kinase inhibition in neurodegeneration. Drugs R D. 2019;19(2):149-166. doi:10.1007/s40268-019-0266-z
    1. Hebron ML, Lonskaya I, Sharpe K, et al. . Parkin ubiquitinates Tar-DNA binding protein-43 (TDP-43) and promotes its cytosolic accumulation via interaction with histone deacetylase 6 (HDAC6). J Biol Chem. 2013;288(6):4103-4115. doi:10.1074/jbc.M112.419945
    1. Hebron ML, Lonskaya I, Moussa CE. Nilotinib reverses loss of dopamine neurons and improves motor behavior via autophagic degradation of α-synuclein in Parkinson’s disease models. Hum Mol Genet. 2013;22(16):3315-3328. doi:10.1093/hmg/ddt192
    1. Hebron ML, Lonskaya I, Olopade P, Selby ST, Pagan F, Moussa CE. Tyrosine kinase inhibition regulates early systemic immune changes and modulates the neuroimmune response in α-synucleinopathy. J Clin Cell Immunol. 2014;5:259. doi:10.4172/2155-9899.1000259
    1. Lonskaya I, Hebron ML, Desforges NM, Schachter JB, Moussa CE. Nilotinib-induced autophagic changes increase endogenous parkin level and ubiquitination, leading to amyloid clearance. J Mol Med (Berl). 2014;92(4):373-386. doi:10.1007/s00109-013-1112-3
    1. Pagan F, Hebron M, Valadez EH, et al. . Nilotinib effects in Parkinson’s disease and dementia with Lewy bodies. J Parkinsons Dis. 2016;6(3):503-517. doi:10.3233/JPD-160867
    1. World Medical Association World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191-2194. doi:10.1001/jama.2013.281053
    1. Schulz KF, Grimes DA. Unequal group sizes in randomised trials: guarding against guessing. Lancet. 2002;359(9310):966-970. doi:10.1016/S0140-6736(02)08029-7
    1. Pagan FL, Hebron ML, Wilmarth B, et al. . Pharmacokinetics and pharmacodynamics of a single dose nilotinib in individuals with Parkinson’s disease. Pharmacol Res Perspect. 2019;7(2):e00470. doi:10.1002/prp2.470
    1. Novartis. Talinga (nilotinib). . Accessed November 6, 2019.
    1. Kopin IJ. Catecholamine metabolism: basic aspects and clinical significance. Pharmacol Rev. 1985;37(4):333-364.
    1. Eisenhofer G, Kopin IJ, Goldstein DS. Catecholamine metabolism: a contemporary view with implications for physiology and medicine. Pharmacol Rev. 2004;56(3):331-349. doi:10.1124/pr.56.3.1
    1. Lonskaya I, Hebron ML, Desforges NM, Franjie A, Moussa CE. Tyrosine kinase inhibition increases functional parkin-beclin-1 interaction and enhances amyloid clearance and cognitive performance. EMBO Mol Med. 2013;5(8):1247-1262. doi:10.1002/emmm.201302771
    1. Karuppagounder SS, Brahmachari S, Lee Y, Dawson VL, Dawson TM, Ko HS. The c-Abl inhibitor, nilotinib, protects dopaminergic neurons in a preclinical animal model of Parkinson’s disease. Sci Rep. 2014;4:4874. doi:10.1038/srep04874
    1. Mahul-Mellier AL, Fauvet B, Gysbers A, et al. . c-Abl phosphorylates α-synuclein and regulates its degradation: implication for α-synuclein clearance and contribution to the pathogenesis of Parkinson’s disease. Hum Mol Genet. 2014;23(11):2858-2879. doi:10.1093/hmg/ddt674
    1. El-Agnaf OM, Salem SA, Paleologou KE, et al. . Detection of oligomeric forms of alpha-synuclein protein in human plasma as a potential biomarker for Parkinson’s disease. FASEB J. 2006;20(3):419-425. doi:10.1096/fj.03-1449com
    1. Mollenhauer B, El-Agnaf OM, Marcus K, Trenkwalder C, Schlossmacher MG. Quantification of α-synuclein in cerebrospinal fluid as a biomarker candidate: review of the literature and considerations for future studies. Biomark Med. 2010;4(5):683-699. doi:10.2217/bmm.10.90
    1. Park MJ, Cheon SM, Bae HR, Kim SH, Kim JW. Elevated levels of α-synuclein oligomer in the cerebrospinal fluid of drug-naïve patients with Parkinson’s disease. J Clin Neurol. 2011;7(4):215-222. doi:10.3988/jcn.2011.7.4.215
    1. Hebron ML, Javidnia M, Moussa CE. Tau clearance improves astrocytic function and brain glutamate-glutamine cycle. J Neurol Sci. 2018;391:90-99. doi:10.1016/j.jns.2018.06.005
    1. Heyburn L, Hebron ML, Smith J, et al. . Tyrosine kinase inhibition reverses TDP-43 effects on synaptic protein expression, astrocytic function and amino acid dis-homeostasis. J Neurochem. 2016;139(4):610-623. doi:10.1111/jnc.13763
    1. Lonskaya I, Hebron ML, Selby ST, Turner RS, Moussa CE. Nilotinib and bosutinib modulate pre-plaque alterations of blood immune markers and neuro-inflammation in Alzheimer’s disease models. Neuroscience. 2015;304:316-327. doi:10.1016/j.neuroscience.2015.07.070
    1. Fahn S; Parkinson Study Group . Does levodopa slow or hasten the rate of progression of Parkinson’s disease? J Neurol. 2005;252(suppl 4):IV37-IV42. doi:10.1007/s00415-005-4008-5

Source: PubMed

3
구독하다