Nilotinib Effects on Safety, Tolerability, and Biomarkers in Alzheimer's Disease

Raymond S Turner, Michaeline L Hebron, Abigail Lawler, Elizabeth E Mundel, Nadia Yusuf, J Nathan Starr, Muhammad Anjum, Fernando Pagan, Yasar Torres-Yaghi, Wangke Shi, Sanjana Mulki, Dalila Ferrante, Sara Matar, Xiaoguang Liu, Giuseppe Esposito, Frank Berkowitz, Xiong Jiang, Jaeil Ahn, Charbel Moussa, Raymond S Turner, Michaeline L Hebron, Abigail Lawler, Elizabeth E Mundel, Nadia Yusuf, J Nathan Starr, Muhammad Anjum, Fernando Pagan, Yasar Torres-Yaghi, Wangke Shi, Sanjana Mulki, Dalila Ferrante, Sara Matar, Xiaoguang Liu, Giuseppe Esposito, Frank Berkowitz, Xiong Jiang, Jaeil Ahn, Charbel Moussa

Abstract

Objective: Preclinical evidence with nilotinib, a US Food and Drug Administration (FDA)-approved drug for leukemia, indicates improvement in Alzheimer's disease phenotypes. We investigated whether nilotinib is safe, and detectable in cerebrospinal fluid, and alters biomarkers and clinical decline in Alzheimer's disease.

Methods: This single-center, phase 2, randomized, double-blind, placebo-controlled study investigated the safety, tolerability, and pharmacokinetics of nilotinib, and measured biomarkers in participants with mild to moderate dementia due to Alzheimer's disease. The diagnosis was supported by cerebrospinal fluid or amyloid positron emission tomography biomarkers. Nilotinib 150 mg versus matching placebo was taken orally once daily for 26 weeks followed by nilotinib 300 mg versus placebo for another 26 weeks.

Results: Of the 37 individuals enrolled, 27 were women and the mean (SD) age was 70.7 (6.48) years. Nilotinib was well-tolerated, although more adverse events, particularly mood swings, were noted with the 300 mg dose. In the nilotinib group, central nervous system (CNS) amyloid burden was significantly reduced in the frontal lobe compared to the placebo group. Cerebrospinal fluid Aβ40 was reduced at 6 months and Aβ42 was reduced at 12 months in the nilotinib group compared to the placebo. Hippocampal volume loss was attenuated (-27%) at 12 months and phospho-tau-181 was reduced at 6 months and 12 months in the nilotinib group.

Interpretation: Nilotinib is safe and achieves pharmacologically relevant cerebrospinal fluid concentrations. Biomarkers of disease were altered in response to nilotinib treatment. These data support a larger, longer, multicenter study to determine the safety and efficacy of nilotinib in Alzheimer's disease. ANN NEUROL 2020 ANN NEUROL 2020;88:183-194.

Conflict of interest statement

C.M. is an inventor on several US and international Georgetown University patents to use nilotinib and other tyrosine kinase inhibitors as a treatment for neurodegenerative diseases. C.M., his laboratory, and Georgetown University previously received some income from licensing of the technology to Axovant Science. Georgetown University spun out the technology (April 2020) to a start‐up company (KeifeRX LLC), from which C.M. receives consulting fees and Georgetown University, C.M., and F.L.P. receive equities and Y.T.‐Y. is an advisor. An individual and institutional conflict management plan (CMP) determines that C.M. can design a study as the (inventor) person who discovered the potential effects of nilotinib in neurodegenerative diseases (thus conceived of the study), can serve as a corresponding principal investigator (PI) and senior author but cannot serve as a clinical PI of any nilotinib clinical study. C.M. must not determine participants' eligibility and must not consent participants. C.M. does not perform primary clinical data analysis and all primary CSF biomarkers analyzed in C.M. laboratory and Georgetown University by blinded investigators must be validated by an external and independent organization using comparable technologies and methods (see Acknowledgments). All investigators must be blinded to treatments until all data are analyzed per group and unblinded by an externally independent DSMB (see Acknowledgments). An external independent study monitor follows study progress and adherence to the protocol. Study visits, scheduled events, and adherence to study protocol are verified by independent nurses and staff of the CRU (see Acknowledgments), which is a Translational Science Awards Program (CTSA). No other authors have any related conflicts of interest. C.M. discovered the potential use of nilotinib in preclinical models and conceived of this phase 2 clinical study and contributed with R.S.T. to the protocol and study design based on his understanding of nilotinib mechanisms of action in preclinical models, especially in relation to nilotinib effects on CNS biomarkers in AD, hence, he is a corresponding and senior author. In adherence with the CMP, C.M. did not provide any scientific or statistical analysis (J.A., R.S.T., and M.L.H.) of the data and the other authors who performed the experiments (R.S.T., G.E., F.B., and X.L.) have analyzed the data with other members (M.L.H., X.J., S.M., M.A., M.S., and S.M.) of the study team who managed the data (S.M., D.F., W.S., M.L.H., M.A., M.S.) and prepared the figures and performed statistical analysis (J.A. and M.L.H.). C.M. was not the clinical principal investigator (PI) who (R.S.T.) executed the protocol and monitored patients (N.J.S., A.L., N.Y., A.L.M., M.A., and Y.T.‐Y.). All safety, biomarkers, and clinical data were unblinded by the DMSB and analyzed by S.M., D.F., M.A., M.L.H., W.S., and S.M., and figures and tables were prepared by M.L.H. and J.A. by the study team. C.M. contributed to manuscript drafting and all authors contributed to the interpretation of the data and approved the final version of manuscript.

© 2020 The Authors. Annals of Neurology published by Wiley Periodicals, Inc. on behalf of American Neurological Association.

Figures

FIGURE 1
FIGURE 1
CONSORT Flow Diagram. Phase 2, randomized, double‐blind, placebo‐controlled study to evaluate nilotinib effects on safety, tolerability, pharmacokinetics, biomarkers, and potential clinical outcomes in Alzheimer's disease. [Color figure can be viewed at www.annalsofneurology.org]
FIGURE 2
FIGURE 2
Quantitative Regional Analysis of (A) standardized regions of interests displayed on fused positron emission tomography (PET) computed tomography (CT) images. Representative PET CT images at baseline and at 12 months of amyloid deposition in the reference cerebellum, frontal, and temporal lobes in participants who (B) received placebo or (C) nilotinib. The graph shows reduction in standardized uptake value ratio (SUVR) from baseline to 12 months in: (D) frontal lobe, (E) temporal lobe, and (F) whole brain (composite) in nilotinib versus placebo groups. *p < 0.05, **p < 0.01 within the group or as indicated between groups.
FIGURE 3
FIGURE 3
Graphs represent, (A) volumetric hippocampal volume and CSF levels of (B) Aβ40, (C) Aβ42, (D) Aβ42/Aβ40 ratio, (E) total tau, (F) phospho‐tau‐181, and (G) ratio of ptau‐181/total tau. The level of dopamine metabolites (H) homovanillic acid (HVA) and (I) 3,4‐hydroxyphenylacetic acid (DOPAC). Graphs represent the mean difference in (J) Abelson phosphorylation at tyrosine 412, (K) pan‐tyrosine phosphorylation of Abelson, and (L) the ratio of Abelson phosphorylation (activation) at tyrosine 412/pan‐tyrosine in the cerebrospinal fluid (CSF) of patients with Alzheimer's disease (AD) treated with nilotinib versus placebo. *p < 0.05, **p < 0.01, and ***p < 0.001 within the group or as indicated between groups (% change is included only with nonsignificant differences).
FIGURE 4
FIGURE 4
Graphs represent exploratory clinical outcomes and their 95% confidence interval between the placebo and nilotinib‐treated groups in (A) Mini‐Mental Status Examination (MMSE), (B) Alzheimer Disease Assessment Scale–cognitive subscale (ADAS‐Cog), (C) ADAS‐Cog subscale 13 (Maze) or time to complete task, (D) Global Clinical Dementia Rating–Sum of Boxes (CDR‐SOB), (E) Alzheimer's Disease Cooperative Study–Instrumental Activities of Daily Living Inventory (ADCS‐ADL), and (F) severity and frequency of behavioral symptoms as measured by Neuropsychiatric Inventory (NPI). *p < 0.05.

References

    1. Reitz C, Brayne C, Mayeux R. Epidemiology of Alzheimer disease. Nat Rev Neurol 2011. Mar;7:137–152.
    1. Govaerts L, Schoenen J, Bouhy D. Pathogenesis of Alzheimer's disease: molecular and cellular mechanisms. Rev Med Liege 2007. Apr;62:209–216.
    1. Roberson ED, Scearce‐Levie K, Palop JJ, et al. Reducing endogenous tau ameliorates amyloid beta‐induced deficits in an Alzheimer's disease mouse model. Science 2007. May 4;316:750–754.
    1. Deremer DL, Ustun C, Natarajan K. Nilotinib: a second‐generation tyrosine kinase inhibitor for the treatment of chronic myelogenous leukemia. Clin Ther 2008. Nov;30:1956–1975.
    1. Skorski T. BCR‐ABL1 kinase: hunting an elusive target with new weapons. Chem Biol 2011. Nov 23;18:1352–1353.
    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. Dec 1;68:9809–9816.
    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. Mar;27:149–166.
    1. Jeitany M, Leroy C, Tosti P, et al. Inhibition of DDR1‐BCR signalling by nilotinib as a new therapeutic strategy for metastatic colorectal cancer. EMBO Mol Med 2018. Apr;10:e7918
    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. Feb 8;288:4103–4115.
    1. Hebron ML, Lonskaya I, Moussa CE. Nilotinib reverses loss of dopamine neurons and improves motor behavior via autophagic degradation of alpha‐synuclein in Parkinson's disease models. Hum Mol Genet 2013. Aug 15;22:3315–3328.
    1. Hebron ML, Lonskaya I, Olopade P, et al. Tyrosine kinase inhibition regulates early systemic immune changes and modulates the neuroimmune response in alpha‐synucleinopathy. J Clin Cell Immunol 2014. Sep 30;5:259.
    1. Lonskaya I, Hebron M, Desforges NM, et al. Nilotinib‐induced autophagic changes increase endogenous parkin level and ubiquitination, leading to amyloid clearance. J Mol Med 2014;92:373–386.
    1. Hebron ML, Javidnia M, Moussa CE. Tau clearance improves astrocytic function and brain glutamate‐glutamine cycle. J Neurol Sci 2018. Aug 15;391:90–99.
    1. Lonskaya I, Hebron ML, Selby ST, et al. Nilotinib and bosutinib modulate pre‐plaque alterations of blood immune markers and neuro‐inflammation in Alzheimer's disease models. Neuroscience 2015. Sep 24;304:316–327.
    1. Lonskaya I, Hebron M, Chen W, et al. Tau deletion impairs intracellular beta‐amyloid‐42 clearance and leads to more extracellular plaque deposition in gene transfer models. Mol Neurodegener 2014. Nov 10;9:46.
    1. Lonskaya I, Hebron ML, Desforges NM, et al. Nilotinib‐induced autophagic changes increase endogenous parkin level and ubiquitination, leading to amyloid clearance. J Mol Med (Berl) 2014. Apr;92:373–386.
    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. Apr;7:e00470.
    1. Pagan F, Hebron M, Valadez EH, et al. Nilotinib effects in Parkinson's disease and dementia with Lewy bodies. J Parkinsons Dis 2016. Jul 11;6:503–517.
    1. Pagan FL, Hebron ML, Wilmarth B, et al. Nilotinib effects on safety, tolerability, and potential biomarkers in Parkinson disease: a phase 2 randomized clinical trial. JAMA Neurol 2019. Dec;16:503–517.
    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. Jun;19:149–166.
    1. Hebron M, Peyton M, Liu X, et al. Discoidin domain receptor inhibition reduces neuropathology and attenuates inflammation in neurodegeneration models. J Neuroimmunol 2017. Oct 15;311:1–9.
    1. McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging‐Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement 2011. May;7:263–269.
    1. Schulz KF, Grimes DA. Unequal group sizes in randomised trials: guarding against guessing. Lancet 2002. Mar 16;359:966–970.
    1. Barthel H, Gertz HJ, Dresel S, et al. Cerebral amyloid‐beta PET with florbetaben (18F) in patients with Alzheimer's disease and healthy controls: a multicentre phase 2 diagnostic study. Lancet Neurol 2011. May;10:424–435.
    1. Ashok AH, Marques TR, Jauhar S, et al. The dopamine hypothesis of bipolar affective disorder: the state of the art and implications for treatment. Mol Psychiatry 2017. May;22:666–679.
    1. Lonskaya I, Hebron M, Desforges NM, et al. Tyrosine kinase inhibition increases functional parkin‐Beclin‐1 interaction and enhances amyloid clearance and cognitive performance. EMBO Mol Med 2013;5:1247–1262.
    1. Karuppagounder SS, Brahmachari S, Lee Y, et al. The c‐Abl inhibitor, nilotinib, protects dopaminergic neurons in a preclinical animal model of Parkinson's disease. Sci Rep 2014;4:4874.
    1. Mahul‐Mellier AL, Fauvet B, Gysbers A, et al. 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 2014. Jun 1;23:2858–2879.
    1. Khandelwal PJ, Herman AM, Hoe HS, et al. Parkin mediates beclin‐dependent autophagic clearance of defective mitochondria and ubiquitinated Abeta in AD models. Hum Mol Genet 2011. Jun 1;20:2091–2102.
    1. Rebeck GW, Hoe HS, Moussa CE. Beta‐amyloid1‐42 gene transfer model exhibits intraneuronal amyloid, gliosis, tau phosphorylation, and neuronal loss. J Biol Chem 2010. Mar 5;285:7440–7446.
    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. Nov;139:610–623.
    1. Jack CR Jr, Bennett DA, Blennow K, et al. A/T/N: an unbiased descriptive classification scheme for Alzheimer disease biomarkers. Neurology 2016. Aug 2;87:539–547.
    1. Skoog I, Kern S, Zetterberg H, et al. Low cerebrospinal fluid Abeta42 and Abeta40 are related to white matter lesions in cognitively normal elderly. J Alzheimers Dis 2018;62:1877–1886.
    1. Iturria‐Medina Y, Sotero RC, Toussaint PJ, et al. Alzheimer's disease neuroimaging I. Early role of vascular dysregulation on late‐onset Alzheimer's disease based on multifactorial data‐driven analysis. Nat Commun 2016. Jun 21;7:11934.

Source: PubMed

3
Abonnere