Nilotinib Effects in Parkinson's disease and Dementia with Lewy bodies

Fernando Pagan, Michaeline Hebron, Ellen H Valadez, Yasar Torres-Yaghi, Xu Huang, Reversa R Mills, Barbara M Wilmarth, Hellen Howard, Connell Dunn, Alexis Carlson, Abigail Lawler, Sean L Rogers, Ramsey A Falconer, Jaeil Ahn, Zhaoxia Li, Charbel Moussa, Fernando Pagan, Michaeline Hebron, Ellen H Valadez, Yasar Torres-Yaghi, Xu Huang, Reversa R Mills, Barbara M Wilmarth, Hellen Howard, Connell Dunn, Alexis Carlson, Abigail Lawler, Sean L Rogers, Ramsey A Falconer, Jaeil Ahn, Zhaoxia Li, Charbel Moussa

Abstract

Background: We evaluated the effects of low doses of the tyrosine kinase Abelson (Abl) inhibitor Nilotinib, on safety and pharmacokinetics in Parkinson's disease dementia or dementia with Lewy bodies.

Objectives: The primary outcomes of this study were safety and tolerability; pharmacokinetics and target engagement were secondary, while clinical outcomes were exploratory.

Methods: Twelve subjects were randomized into 150 mg (n = 5) or 300 mg (n = 7) groups and received Nilotinib orally every day for 24 weeks.

Results: This study shows that 150 mg and 300 mg doses of Nilotinib appear to be safe and tolerated in subjects with advanced Parkinson's disease. Nilotinib is detectable in the cerebrospinal fluid (CSF) and seems to engage the target Abl. Motor and cognitive outcomes suggest a possible beneficial effect on clinical outcomes. The CSF levels of homovanillic acid are significantly increased between baseline and 24 weeks of treatment. Exploratory CSF biomarkers were measured.

Conclusions: This small proof-of-concept study lacks a placebo group and participants were not homogenous, resulting in baseline differences between and within groups. This limits the interpretations of the biomarker and clinical data, and any conclusions should be drawn cautiously. Nonetheless, the collective observations suggest that it is warranted to evaluate the safety and efficacy of Nilotinib in larger randomized, double-blind, placebo-controlled trials.

Keywords: Lewy bodies; Nilotinib; Parkinson; dopamine; homovanillic acid; synuclein; tau.

Figures

Fig.1
Fig.1
Design and milestones of an open-label, phase I clinical trial to evaluate the safety and efficacy of 150 mg and 300 mg Nilotinib for 24 weeks in patients with Parkinson’s disease (PD) with dementia (PDD) or PD with mild cognitive impairment (PD-MCI) or dementia with Lewy bodies (DLB). Cerebrospinal fluid (CSF), Mini Mental State Examination (MMSE), Abelson (Abl), homovanillic acid (HVA), β-amyloid (Aβ), Scales for Outcomes in Parkinson’s Disease-Cognition (SCOPA-Cog), Neuron Specific Enolase (SNE).
Fig.2
Fig.2
Pharmacokinetics and pharmacodynamics. Graph shows Nilotinib levels in (A) plasma, (B) CSF and (C) ratio of CSF: plasma of Nilotinib in a population analysis (N = 33 data points) collected at baseline, 1, 2, 3, 4 or 5hrs after oral administration. Plasma was collected 30 min before CSF. Graph (D) is level of Abl inhibition via de-phosphorylation in the same sample population showing the level of pan-tyrosine phosphorylated Abl (active) in the CSF. (E) is non-compartmental analysis of 150 mg and 300 mg of CSF and plasma Nilotinib. Nilotinib was quantified in reference to 13C Nilotinib standard and biological samples were centrifuged to obtain unbound Nilotinib. Cerebrospinal fluid (CSF), Abelson (Abl). Graph shows absorbance levels (ABS) of pan-tyrosine phosphorylated Abl in the CSF of individual participants at baseline and after oral dosing of Nilotinib in the 150 mg (F) and 300 mg (G) groups.

References

    1. Boland B, Kumar A, Lee S, Platt FM, Wegiel J, Yu WH, & Nixon RA (2008) Autophagy induction and autophagosome clearance in neurons: Relationship to autophagic pathology in Alzheimer’s disease. J Neurosci 28, 6926–6937.
    1. Kegel KB, Kim M, Sapp E, McIntyre C, Castano JG, Aronin N, & DiFiglia M (2000) Huntingtin expression stimulates endosomal-lysosomal activity, endosome tubulation, and autophagy. J Neurosci 20, 7268–7278.
    1. Nixon RA, Wegiel J, Kumar A, Yu WH, Peterhoff C, Cataldo A, & Cuervo AM (2005) Extensive involvement of autophagy in Alzheimer disease: An immuno-electron microscopy study. J Neuropathol Exp Neurol 64, 113–122.
    1. Ravikumar B, Duden R, & Rubinsztein DC (2002) Aggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagy. Hum Mol Genet 11, 1107–1117.
    1. Sabatini DM (2006) mTOR and cancer: Insights into a complex relationship. Nat Rev Cancer 6, 729–734.
    1. Stefanis L, Larsen KE, Rideout HJ, Sulzer D, & Greene LA (2001) Expression of A53T mutant but not wild-type alpha-synuclein in PC12 cells induces alterations of the ubiquitin-dependent degradation system, loss of dopamine release, and autophagic cell death. J Neurosci 21, 9549–9560.
    1. Webb JL, Ravikumar B, Atkins J, Skepper JN, & Rubinsztein DC (2003) Alpha-Synuclein is degraded by both autophagy and the proteasome. J Biol Chem 278, 25009–25013.
    1. Nixon RA (2013) The role of autophagy in neurodegenerative disease. Nat Med 19, 983–997.
    1. Deremer DL, Ustun C, & Natarajan K (2008) Nilotinib: A second-generation tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia. Clin Ther 30, 1956–1975.
    1. Skorski T (2011) BCR-ABL1 kinase: Hunting an elusive target with new weapons. Chem Biol 18, 1352–1353.
    1. Mahon FX, Hayette S, Lagarde V, Belloc F, Turcq B, Nicolini F, Belanger C, Manley PW, Leroy C, Etienne G, Roche S, & Pasquet JM (2008) Evidence that resistance to nilotinib may be due to BCR-ABL, Pgp, or Src kinase overexpression. Cancer Res 68, 9809–9816.
    1. Salomoni P, & Calabretta B (2009) Targeted therapies and autophagy: New insights from chronic myeloid leukemia. Autophagy 5, 1050–1051.
    1. Hebron ML, Lonskaya I, & Moussa CE (2013) Nilotinib reverses loss of dopamine neurons and improves motor behavior via autophagic degradation of alpha-synuclein in Parkinson’s disease models. Hum Mol Genet 22, 3315–3328.
    1. Lonskaya I, Hebron ML, Desforges NM, Franjie A, & Moussa CE (2013) Tyrosine kinase inhibition increases functional parkin-Beclin-1 interaction and enhances amyloid clearance and cognitive performance. EMBO Mol Med 5, 1247–1262.
    1. Hebron ML, Lonskaya I, Sharpe K, Weerasinghe PP, Algarzae NK, Shekoyan AR, & Moussa CE (2013) Parkin ubiquitinates Tar-DNA binding protein-43 (TDP-43) and promotes its cytosolic accumulation via interaction with histone deacetylase 6 (HDAC6). J Biol Chem 288, 4103–4115.
    1. Hebron ML, Lonskaya I, Olopade P, Selby ST, Pagan F, & Moussa CE (2014) Tyrosine Kinase Inhibition Regulates Early Systemic Immune Changes and Modulates the Neuroimmune Response in alpha-Synucleinopathy. J Clin Cell Immunol 5, 259.
    1. Lonskaya I, Hebron M, Desforges NM, Schachter JB, & Moussa CE (2014) Nilotinib-induced autophagic changes increase endogenous parkin level and ubiquitination, leading to amyloid clearance. J Mol Med 92, 373–386.
    1. Lonskaya I, Desforges NM, Hebron ML, & Moussa CE (2013) Ubiquitination increases parkin activity to promote autophagic alpha-synuclein clearance. e. PLoS One 8, 83914.
    1. Lonskaya I, Hebron ML, Desforges NM, Schachter JB, & Moussa CE (2014) Nilotinib-induced autophagic changes increase endogenous parkin level and ubiquitination, leading to amyloid clearance. J Mol Med (Berl) 92, 373–386.
    1. Lonskaya I, Hebron M, Chen W, Schachter J, & Moussa C (2014) Tau deletion impairs intracellular beta-amyloid-42 clearance and leads to more extracellular plaque deposition in gene transfer models. Mol Neurodegener 9, 46.
    1. Karuppagounder SS, Brahmachari S, Lee Y, Dawson VL, Dawson TM, & Ko HS (2014) The c-Abl inhibitor, nilotinib, protects dopaminergic neurons in a preclinical animal model of Parkinson’s disease. Sci Rep 4, 4874.
    1. Mahul-Mellier AL, Fauvet B, Gysbers A, Dikiy I, Oueslati A, Georgeon S, Lamontanara AJ, Bisquertt A, Eliezer D, Masliah E, Halliday G, Hantschel O, & Lashuel HA (2014) c-Abl phosphorylates alpha-synuclein and regulates its degradation: Implication for alpha-synuclein clearance and contribution to the pathogenesis of Parkinson’s disease. Hum Mol Genet 23, 2858–2879.
    1. Ko HS, Lee Y, Shin JH, Karuppagounder SS, Gadad BS, Koleske AJ, Pletnikova O, Troncoso JC, Dawson VL, & Dawson TM (2010) Phosphorylation by the c-Abl protein tyrosine kinase inhibits parkin’s ubiquitination and protective function. Proc Natl Acad Sci U S A 107, 16691–16696.
    1. Imam SZ, Zhou Q, Yamamoto A, Valente AJ, Ali SF, Bains M, Roberts JL, Kahle PJ, Clark RA, & Li S (2011) Novel regulation of parkin function through c-Abl-mediated tyrosine phosphorylation: Implications for Parkinson’s disease. J Neurosci 31, 157–163.
    1. Lev N, Ickowicz D, Melamed E, & Offen D (2008) Oxidative insults induce DJ-1 upregulation and redistribution: Implications for neuroprotection. Neurotoxicology 29, 397–405.
    1. Taira T, Saito Y, Niki T, Iguchi-Ariga SM, Takahashi K, & Ariga H (2004) DJ-1 has a role in antioxidative stress to prevent cell death. EMBO Rep 5, 213–218.
    1. Wang JY (2014) The capable ABL: What is its biological function? Mol Cell Biol 34, 1188–1197.
    1. Petzold A, Groves M, Leis AA, Scaravilli F, & Stokic DS (2010) Neuronal and glial cerebrospinal fluid protein biomarkers are elevated after West Nile virus infection. Muscle Nerve 41, 42–49.
    1. Petzold A, Michel P, Stock M, & Schluep M (2008) Glial and axonal body fluid biomarkers are related to infarct volume, severity, and outcome. J Stroke Cerebrovasc Dis 17, 196–203.
    1. Jauch EC, Lindsell C, Broderick J, Fagan SC, Tilley BC, Levine SR, & Group Nr-PSS (2006) Association of serial biochemical markers with acute ischemic stroke: The National Institute of Neurological Disorders and Stroke recombinant tissue plasminogen activator Stroke Study. Stroke 37, 2508–2513.
    1. Persson L, Hardemark HG, Gustafsson J, Rundstrom G, Mendel-Hartvig I, Esscher T, & Pahlman S (1987) S-100 protein and neuron-specific enolase in cerebrospinal fluid and serum: Markers of cell damage in human central nervous system. Stroke 18, 911–918.
    1. Mollenhauer B, Locascio JJ, Schulz-Schaeffer W, Sixel-Doring F, Trenkwalder C, & Schlossmacher MG (2011) alpha-Synuclein and tau concentrations in cerebrospinal fluid of patients presenting with parkinsonism: A cohort study. Lancet Neurol 10, 230–240.
    1. Parnetti L, Chiasserini D, Bellomo G, Giannandrea D, De Carlo C, Qureshi MM, Ardah MT, Varghese S, Bonanni L, Borroni B, Tambasco N, Eusebi P, Rossi A, Onofrj M, Padovani A, Calabresi P, & El-Agnaf O (2011) Cerebrospinal fluid Tau/alpha-synuclein ratio in Parkinson’s disease and degenerative dementias. Mov Disord 26, 1428–1435.
    1. Tateno F, Sakakibara R, Kawai T, Kishi M, & Murano T (2012) Alpha-synuclein in the cerebrospinal fluid differentiates synucleinopathies (Parkinson Disease, dementia with Lewy bodies, multiple system atrophy) from Alzheimer disease.. Alzheimer Dis Assoc Disord 26, 213–216.
    1. Bakhsheshian J, Wei BR, Chang KE, Shukla S, Ambudkar SV, Simpson RM, Gottesman MM, & Hall MD (2013) Bioluminescent imaging of drug efflux at the blood-brain barrier mediated by the transporter ABCG2. Proc Natl Acad SciU S A 110, 20801–20806.
    1. Zubenko GS, Marquis JK, Volicer L, Direnfeld LK, Langlais PJ, & Nixon RA (1986) Cerebrospinal fluid levels of angiotensin-converting enzyme, acetylcholinesterase, and dopamine metabolites in dementia associated with Alzheimer’s disease and Parkinson’s disease: A correlative study. Biol Psychiatry 21, 1365–1381.
    1. Goldstein DS, Holmes C, & Sharabi Y (2012) Cerebrospinal fluid biomarkers of central catecholamine deficiency in Parkinson’s disease and other synucleinopathies. Brain 135, 1900–1913.
    1. Ren Y, Jiang H, Ma D, Nakaso K, & Feng J (2011) Parkin degrades estrogen-related receptors to limit the expression of monoamine oxidases. Hum Mol Genet 20, 1074–1083.
    1. Mollenhauer B, Trautmann E, Otte B, Ng J, Spreer A, Lange P, Sixel-Doring F, Hakimi M, Vonsattel JP, Nussbaum R, Trenkwalder C, & Schlossmacher MG (2012) alpha-Synuclein in human cerebrospinal fluid is principally derived from neurons of the central nervous system. J Neural Transm 119, 739–746.
    1. Mollenhauer B, Cullen V, Kahn I, Krastins B, Outeiro TF, Pepivani I, Ng J, Schulz-Schaeffer W, Kretzschmar HA, McLean PJ, Trenkwalder C, Sarracino DA, Vonsattel JP, Locascio JJ, El-Agnaf OM, & Schlossmacher MG (2008) Direct quantification of CSF alpha-synuclein by ELISA and first cross-sectional study in patients with neurodegeneration. Exp Neurol 213, 315–325.
    1. Mollenhauer B, Trautmann E, Taylor P, Manninger P, Sixel-Doring F, Ebentheuer J, Trenkwalder C, & Schlossmacher MG (2013) Total CSF alpha-synuclein is lower in de novo Parkinson patients than in healthy subjects. Neurosci Lett 532, 44–48.
    1. Hong Z, Shi M, Chung KA, Quinn JF, Peskind ER, Galasko D, Jankovic J, Zabetian CP, Leverenz JB, Baird G, Montine TJ, Hancock AM, Hwang H, Pan C, Bradner J, Kang UJ, Jensen PH, & Zhang J (2010) DJ-1 and alpha-synuclein in human cerebrospinal fluid as biomarkers of Parkinson’s disease. Brain 133, 713–726.
    1. Ohrfelt A, Grognet P, Andreasen N, Wallin A, Vanmechelen E, Blennow K, & Zetterberg H (2009) Cerebrospinal fluid alpha-synuclein in neurodegenerative disorders-a marker of synapse loss? Neurosci Lett 450, 332–335.
    1. Kasuga K, Tokutake T, Ishikawa A, Uchiyama T, Tokuda T, Onodera O, Nishizawa M, & Ikeuchi T (2010) Differential levels of alpha-synuclein, beta-amyloid42 and tau in CSF between patients with dementia with Lewy bodies and Alzheimer’s disease. J Neurol Neurosurg Psychiatry 81, 608–610.
    1. Lleo A, Cavedo E, Parnetti L, Vanderstichele H, Herukka SK, Andreasen N, Ghidoni R, Lewczuk P, Jeromin A, Winblad B, Tsolaki M, Mroczko B, Visser PJ, Santana I, Svenningsson P, Blennow K, Aarsland D, Molinuevo JL, Zetterberg H, & Mollenhauer B (2015) Cerebrospinal fluid biomarkers in trials for Alzheimer and Parkinson diseases. Nat Rev Neurol 11, 41–55.
    1. Mollenhauer B, Bibl M, Wiltfang J, Steinacker P, Ciesielczyk B, Neubert K, Trenkwalder C, & Otto M (2006) Total tau protein, phosphorylated tau (181p) protein, beta-amyloid(1-42), and beta-amyloid(1-40) in cerebrospinal fluid of patients with dementia with Lewy bodies. Clin Chem Lab Med 44, 192–195.
    1. Parnetti L, Tiraboschi P, Lanari A, Peducci M, Padiglioni C, D’Amore C, Pierguidi L, Tambasco N, Rossi A, & Calabresi P (2008) Cerebrospinal fluid biomarkers in Parkinson’s disease with dementia and dementia with Lewy bodies. Biol Psychiatry 64, 850–855.
    1. Kang JH, Irwin DJ, Chen-Plotkin AS, Siderowf A, Caspell C, Coffey CS, Waligorska T, Taylor P, Pan S, Frasier M, Marek K, Kieburtz K, Jennings D, Simuni T, Tanner CM, Singleton A, Toga AW, Chowdhury S, Mollenhauer B, Trojanowski JQ, Shaw LM, & Parkinson’s Progression Markers I (2013) Association of cerebrospinal fluid beta-amyloid 1-42, T-tau, P-tau181, and alpha-synuclein levels with clinical features of drug-naive patients with early Parkinson disease. JAMA Neurol 70, 1277–1287.

Source: PubMed

3
購読する